WO2012085177A1 - Nemaucin, an antibiotic produced by entomopathogenic xenorhabdus cabanillasii - Google Patents

Nemaucin, an antibiotic produced by entomopathogenic xenorhabdus cabanillasii Download PDF

Info

Publication number
WO2012085177A1
WO2012085177A1 PCT/EP2011/073738 EP2011073738W WO2012085177A1 WO 2012085177 A1 WO2012085177 A1 WO 2012085177A1 EP 2011073738 W EP2011073738 W EP 2011073738W WO 2012085177 A1 WO2012085177 A1 WO 2012085177A1
Authority
WO
WIPO (PCT)
Prior art keywords
nemaucin
compound according
xaa
antibiotic
xenorhabdus
Prior art date
Application number
PCT/EP2011/073738
Other languages
French (fr)
Inventor
Maxime Gualtieri
Philippe Villain-Guillot
Alain Givaudan
Sylvie Pages
Original Assignee
Nosopharm
Institut National De La Recherche Agronomique
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nosopharm, Institut National De La Recherche Agronomique filed Critical Nosopharm
Publication of WO2012085177A1 publication Critical patent/WO2012085177A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • C07K7/56Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring the cyclisation not occurring through 2,4-diamino-butanoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • C12N1/205Bacterial isolates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/16Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms containing two or more hetero rings
    • C12P17/165Heterorings having nitrogen atoms as the only ring heteroatoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales

Definitions

  • the present invention relates to new antibiotic compounds, to a strain of Xenorhabdus cabanillasii producing said antibiotic compounds and to the use of these compounds in the treatment of microbial disease in humans and animals.
  • Antimicrobial resistance is a major public health problem with a significant impact on morbidity, mortality and healthcare-associated costs. The problem has been worsened by the restriction of antibiotic drug discovery and development programs.
  • MRSA methicillin-resistant Staphylococcus aureus
  • VRE vancomycin-resistant enterococci
  • ESBL extended spectrum ⁇ -lactamase formers
  • Antimicrobial peptides comprise a diverse class of molecules used in host defense by bacteria, plants, insects, and animals.
  • polypeptide antibiotics are polymyxin B and gramicidin S produced respectively by Bacillus polymyxa and Bacillus brevis.
  • the present application is based on research on an underexploited bacterial genus: Xenorhabdus.
  • Gram-negative bacterial strains of the genus Xenorhabdus are known to be symbiotically associated with soil dwelling nematodes of the Steinernematidae family. After entering the insect larvae via natural openings, nematodes release bacteria from their intestine to the host's hemocoel. Bacteria are involved in killing the insect host by producing insecticidal proteins and inhibitors of the insect immune system.
  • Boemare et al. Boemare N.E. et al, Appl. Environ. Microbiol. 58, 3032-3037 (1992) classified the antibiotic activities of Xenorhabdus into two categories: antimicrobial molecules with broad spectrum of activity, and bacteriocins with very narrow spectrum of activity and active only against bacteria closely related to X. nematophila.
  • Nemaucins produced by fermentation of a Xenorhabdus cabanillasii strain, and purified by cation exchange chromatography and reversed phase chromatography.
  • the chemical structure of Nemaucins was obtained from the analysis of homo and heteronuclear 2D NMR and MS- MS experiments.
  • a first object of the present invention is a compound of formula (I):
  • Xaai is aspartic acid or glutamic acid
  • Xaa 2 is arginine, histidine or lysine
  • Xaa 3 is serine, threonine, asparagine or glutamine,
  • Xaa 4 is selected in the group consisting of
  • R is H, OH or CH2-OH
  • n 1, 2, 3, 4 or 5.
  • Xaai is aspartic acid.
  • Xaa 2 is histidine.
  • Xaa 3 is asparagine.
  • Xaa 4 is
  • R is OH
  • n 4.
  • the compound of the present invention is the compound of formula (II):
  • n 1, 2, 3, 4 or 5.
  • n 4.
  • the compounds of the present invention are for use as a medicament.
  • the compounds of the present invention are for use as an antibiotic agent.
  • the compounds of the present invention are for use in treatment of microbial disease.
  • the compounds of the present invention are for use for treatment of a microbial disease caused by a microorganism selected from Staphylococcus, Enterococcus, Streptococcus, Bacillus, Clostridium, Chlamydia, Mycobacterium, Pasteurella, Escherichia, Enterobacter, Klebsiella, Citrobacter, Proteus, Serratia, Pseudomonas, Acinetobacter, Shigella, Salmonella, Stenotrophomonas, Yersinia, Burkholderia, Morganella, Moraxella, Propionibacterium, Listeria, Corynebacterium, Bifidobacterium, Lactobacillus, and Brucella.
  • the compound of the present invention is for treatment of a microbial disease caused by a microorganism selected from Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens and Pseudomonas aeruginosa.
  • a microorganism selected from Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens
  • Another object of the present invention is a composition comprising a compound of the present invention in association with another antibiotic compound.
  • the present invention is also directed to a pharmaceutical composition
  • a pharmaceutical composition comprising:
  • Xaai is aspartic acid or glutamic acid
  • Xaa 2 is arginine, histidine or lysine
  • Xaa 3 is serine, threonine, asparagine or glutamine,
  • Xaa 4 is selected in the group consisting of
  • R is H, OH or -CH2-OH
  • n 1, 2, 3, 4 or 5
  • Xaai is aspartic acid.
  • Xaa 2 is histidine.
  • Xaa 3 is asparagine.
  • Xaa 4 is
  • R is OH
  • n 4.
  • the compound of the present invention is the compound of formula (II):
  • n 1, 2, 3, 4 or 5.
  • n is 1, 2, 3, 4 or 5.
  • a further object of the present invention is a method for producing a compound according to formula (II) comprising the following steps:
  • the present invention is also directed to the Xenorhabdus cabanillasii strain deposited at CNCM on 15-12-2010 having the accession number CNCM 1-4418.
  • Another object of the present invention is a culture supernatant from the
  • Xenorhabdus cabanillasii strain of the present invention having antibiotic activity.
  • a further object of the present invention is an extract from the Xenorhabdus cabanillasii strain of the present invention having antibiotic activity. Description of the invention
  • the present invention is related to Xenorhabdus cabanillasii strain JM26 deposited at CNCM (Collection Nationale de Cultures de Microorganismes) in the name of INRA (Institut National de la Recherche Agronomique) on 15-12-2010 having the accession number CNCM 1-4418.
  • the antibiotic compound of the present invention is secreted into the culture supernatant.
  • the present invention is related to a culture supernatant from Xenorhabdus cabanillasii strain CNCM 1-4418 exhibiting antibiotic activity.
  • Xenorhabdus cabanillasii strain CNCM 1-4418 is grown in a liquid culture medium under standard conditions, the bacterial cells are removed and the supernatant is recovered. The bacterial cells may for example be removed by centrifugation or filtration.
  • the invention is related to extracts from Xenorhabdus cabanillasii strain CNCM 1-4418 showing antibiotic activity.
  • Cell extracts from Xenorhabdus cabanillasii may be prepared according to any appropriate method known to the skilled person.
  • the present invention also encompasses culture supernatant and extracts from
  • the present invention also encompasses culture supernatant and extracts from Xenorhabdus cabanillasii strain CNCM 1-4418 for use as an antibiotic agent.
  • antibiotic refer generally to an effect in which a reduction, inhibition or a halt in the growth of a microorganism is achieved. Antibiotic activity may be tested according to any known method such as a microdilution method.
  • the substances and compounds of the present invention inhibit or kill microorganism and more particularly kill or inhibit bacteria.
  • the substances and compounds of the present invention inhibit or kill multidrug resistant bacteria which exhibit resistance to known antibiotics.
  • Xenorhabdus cabanillasii strain CNCM 1-4418 culture supernatant from this strain and cell extracts derived from this strain exhibit antibiotic activity against different microorganism including human bacterial pathogens.
  • Xenorhabdus cabanillasii strain CNCM 1-4418 culture supernatant from this strain and cell extracts derived from this strain exhibit antibiotic activity against Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens and Pseudomonas aeruginosa.
  • the present invention is related to a compound of formula (I):
  • Xaai is aspartic acid or glutamic acid
  • Xaa 2 is arginine, histidine or lysine
  • Xaa 3 is serine, threonine, asparagine or glutamine,
  • Xaa 4 is selected in the group consisting of
  • R is H, OH or CH2- n is 1, 2, 3, 4 or 5, a salt, a hydrate or a solvate thereof.
  • These compounds have been named "Nemaucins".
  • the term “Nemaucins” refers to the compounds of formula I, a salt, a hydrate or a solvate thereof.
  • Xaai is aspartic acid.
  • Xaa 2 is histidine.
  • Xaa 3 is asparagine.
  • Xaa 4 is
  • R is OH
  • n 4.
  • the compound of the present invention is the compound of formula (II):
  • n 1, 2, 3, 4 or 5
  • n 4.
  • This compound has been named "Nemaucin”.
  • the antibiotic compounds according to the present invention are for use as a medicament, for use as an antibiotic agent, for use as an antimicrobial agent, or for use in the treatment of microbial disease in particular of bacterial infection caused by pathogenic bacteria.
  • the compounds of the present invention are preferably for use in the treatment of bacterial infection and in particular in the treatment of hospital-acquired infections or nosocomial bacterial infections.
  • the invention is also related to the use of compounds of formula (I) for the manufacture of a medicament for treatment of microbial infection or microbial disease.
  • the invention is also related to the use of compounds of formula (I) for the manufacture of an antibiotic.
  • the present invention also provides methods for treating microbial/bacterial infection and/or microbial/bacterial disease including administering an effective amount of a compound of formula (I) to a human or to a patient in need thereof.
  • the invention relates to methods for treatment of bacterial disease and or bacterial infection.
  • the patient is a mammal, i.e., a living mammal.
  • the patient is a human, i.e., a living human, including a living human child and a living human adult.
  • treatment refers generally to treatment and therapy, whether of a human or an animal (e.g., in veterinary applications), in which some desired therapeutic effect is achieved, for example, the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, and cure of the condition.
  • Treatment as a prophylactic measure i.e., prophylaxis
  • use with subjects who have not yet developed the condition, but who are at risk of developing the condition is encompassed by the term "treatment.”
  • terapéuticaally-effective amount refers to the amounts of the active agents (compounds of formula (I) or a salt, hydrate, or solvate thereof) that is effective for producing some desired therapeutic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.
  • treatment includes combination treatments and therapies, in which two or more treatments or therapies are combined, for example, sequentially or simultaneously.
  • the active agents of the present invention may also be used in further combination therapies, e.g., in conjunction with other agents, for example, other antimicrobial or antibiotic agents, etc.
  • Another aspect of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising:
  • Xaai is aspartic acid or glutamic acid
  • Xaa 2 is arginine, histidine or lysine
  • Xaa 3 is serine, threonine, asparagine or glutamine,
  • Xaa 4 is selected in the group consisting of
  • R is H, OH or CH2-OH
  • n 1, 2, 3, 4 or 5
  • Xaai is aspartic acid.
  • Xaa 2 is histidine.
  • Xaa 3 is asparagine.
  • Xaa 4 is
  • R is OH
  • n 4;
  • the compound is the compound of formula (II):
  • n 1, 2, 3, 4 or 5
  • n 4.
  • pharmaceutically acceptable carriers includes any and all solvents, dispersion media, coatings, and the like that are physiologically compatible.
  • compositions of the invention may be in a variety of forms. These include for example liquid, semi-solid, and solid dosage forms, but the preferred form depends on the intended mode of administration and therapeutic application.
  • compositions are preferably for oral, topical, percutaneous or parenteral administration.
  • compositions as described herein may be orally administered.
  • compositions for oral administration tablets, pills, powders (gelatine capsules, sachets) or granules may be used.
  • the active ingredient according to the invention is mixed with one or more inert diluents, such as starch, cellulose, sucrose, lactose or silica, under an argon stream.
  • these compositions may also comprise substances other than diluents, for example one or more lubricants such as magnesium stearate or talc, a coloring, a coating (sugar-coated tablet) or a glaze.
  • Preferred oral compositions include coated and uncoated tablets, hard and soft gelatin capsules, sugar-coated pills, lozenges, pellets, and powders.
  • compositions for oral administration there may be used pharmaceutically acceptable solutions, suspensions, emulsions, syrups and elixirs containing inert diluents such as water, ethanol, glycerol, vegetable oils or paraffin oil.
  • inert diluents such as water, ethanol, glycerol, vegetable oils or paraffin oil.
  • These compositions may comprise substances other than diluents, for example wetting, sweetening, thickening, flavouring or stabilizing products.
  • the pharmaceutical compositions for oral use contain Nemaucin together with the usual excipients as diluting agents like mannitol, lactose and sorbitol; binding agents like starchs, gelatins, sugars, cellulose derivatives, natural gums and polyvinylpyrrolidone; lubricating agents like talc, stearates, hydrogenated vegetable oils, polyethylenglycol and colloidal silicon dioxide; disintegrating agents like starcks, celluloses, alginates, gums and reticulated polymers; coloring, flavoring and sweetening agents.
  • diluting agents like mannitol, lactose and sorbitol
  • binding agents like starchs, gelatins, sugars, cellulose derivatives, natural gums and polyvinylpyrrolidone
  • lubricating agents like talc, stearates, hydrogenated vegetable oils, polyethylenglycol and colloidal silicon dioxide
  • disintegrating agents like starcks, celluloses
  • compositions comprise Nemaucin according to the present invention with carriers or excipients suitable for topical administration.
  • Preferred compositions for topical administration of Nemaucin according to the invention include ointments, pomades, creams, gels, and lotions.
  • compositions for topical use contain Nemaucin together with the usual excipients like white petrolatum, white wax, lanolin and derivatives thereof, stearylic alcohol, propylenglycol, sodium lauryl sulfate, ethers of the fatty polyoxy ethylene alcohols, sorbitan monostearate, glyceryl monostearate, propylene glycol monostearate, polyetilene glycols, methylcellulose, hydroxymethylpropylcellulose, sodium carboxymethylcellulose, colloidal aluminium and magnesium silicate, sodium alginate.
  • Any topical preparation may be used in the present invention, for instance ointments, pomades, creams, gels and lotions.
  • Nemaucin The doses of Nemaucin depend on the desired effect, the duration of the treatment and the route of administration used.
  • the pharmaceutical compositions according to the present invention are for use as an antimicrobial agent, for use as antibiotic or for use in the treatment of microbial disease in particular of microbial disease caused by bacteria.
  • compositions of the present invention are preferably for use in the treatment of bacterial infection and in particular for use in the treatment of hospital- acquired infections or nosocomial bacterial infections.
  • the compound of the present invention is for use for treatment of a microbial disease caused by a microorganism selected from Staphylococcus, Enter ococcus, Streptococcus, Bacillus, Clostridium, Chlamydia, Mycobacterium, Pasteurella, Escherichia, Enter obacter, Klebsiella, Citrobacter, Proteus, Serratia, Pseudomonas, Acinetobacter, Shigella, Salmonella, Stenotrophomonas, Yersinia, Burkholderia, Morganella, Moraxella, Propionibacterium, Listeria, Corynebacterium, Bifidobacterium, Lactobacillus, and Brucella.
  • a microorganism selected from Staphylococcus, Enter ococcus, Streptococcus, Bacillus, Clostridium, Chlamydia, Mycobacterium, Pasteurella, Escherichia, Enter
  • the compound of the present invention is for treatment of a microbial disease caused by a microorganism selected from Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens and Pseudomonas aeruginosa.
  • the compounds of the present invention may be combined with other active compounds exhibiting an antimicrobial/antibiotic activity.
  • the pharmaceutical compositions encompassed by the present invention may also contain a further therapeutic agent for the treatment of bacterial disease or bacterial infection.
  • Another aspect of the present invention is a method for producing a compound according to formula (II) comprising the following steps:
  • the active compounds according to the present invention may be purified from the Xenorhabdus cabanillasii cells of the present invention.
  • the active compounds of the present invention may be purified from the culture supernatant after removal of the Xenorhabdus cabanillasii cells.
  • Xenorhabdus cabanillasii strain CNCM 1-4418 is grown in a liquid culture medium under standard conditions, the bacterial cells are removed and the supernatant is recovered.
  • the bacterial cells may for example be removed by centrifugation or filtration.
  • the active compound is purified from the culture supernatant of Xenorhabdus cabanillasii strain CNCM 1-4418 by successive cation- exchange chromatography, reversed-phase chromatography and reverse phase HPLC.
  • Figure 1 HPLC chromatogram of the Xenorhabdus cabanillasii growth medium
  • Figure 2 Bactericidal effects of Nemaucin on growing S. aureus CIP 76.24
  • Xenorhabdus cabanillasi CMCM 1-4418 was grown on Luria-Bertani medium (LB, composed of bactotryptone 10 g/L, yeast extract 5 g/L and NaCl 10 g/L) for liquid culture and on LB-agar for solid cultures.
  • LB Luria-Bertani medium
  • the phase status (I or II) of this strain was determined by culturing on NBTA (Nutrient agar (Difco) 31 g/L, bromothymol blue 25 mg/L and 2, 3, 5-triphenyl tetrazolium chloride 1% 40 mg/L) and measuring antibacterial activity against Micrococcus luteus.
  • NBTA Nutrient agar (Difco) 31 g/L, bromothymol blue 25 mg/L and 2, 3, 5-triphenyl tetrazolium chloride 1% 40 mg/L
  • Xenorhabdus exhibit two colony forms or variants when cultured in vitro
  • Phase I variants absorb dyes and are blue on NBTA plates, while phase II colonies are red.
  • Phases I and II of strains are indicated as suffixes ⁇ l ⁇ and 12, respectively) attached to strain designations. This strain was maintained at 15°C on NBTA medium.
  • Antibiotics were tested at final concentrations (prepared from serial twofold dilutions) ranging from 0.01 to 100 ⁇ g/mL.
  • the minimal inhibitory concentration (MIC) was defined as the lowest antibiotic concentration which yielded no visible growth.
  • the test medium was Mueller-Hinton broth (MHB), the inoculum was 5xl0 5 CFU/mL.
  • the inoculated microplates were incubated at 37°C for 18 h before reading.
  • Minimal bactericidal concentrations (MBCs) were established by extending the MIC procedure to the evaluation of bactericidal activity. After 24 hours, 10 ⁇ _, were drawn from the wells, serially diluted and then spotted onto suitable agar plates. The plates were incubated at 37°C overnight. The MBC was read 18 h later as the lowest concentration of antibiotic which resulted in 0.1 % survival in the subculture. All the experiments were done in triplicates.
  • HSA human serum albumin
  • Microplates were incubated at 37°C for 48h with 5% of C0 2 . After 48h of culture, the medium was removed by aspiration and 100 ⁇ _, of RPMI + MTT (0.5 mg/mL) were added to each wells. Microplates were inoculated at 37°C during 150 min. Next, all the medium was removed by inverting and tapping the plate and 100 ⁇ _, of DMSO were added in each plate. The spectrophotometric absorbance was then measured at 550 nm.
  • XI represents the concentration for which viability was superior to 50%
  • X2 represents the concentration for which viability was inferior to 50%
  • Yl represents the percentage of viability at the concentration XI
  • Y2 the percentage of viability at the concentration X2.
  • Bacterial killing curves were carried out by inoculating S. aureus CIP 76.24 with Nemaucin concentrations equal to the MIC, two-fold the MIC and four-fold the MIC. Vancomycin was used at four-fold its MIC.
  • the S. aureus inoculums was prepared from colonies grown overnight in MHB. Antibiotics concentrations in the flask were adjusted in MHB according to the desired concentration.
  • Culture tubes containing 10 mL were inoculated with S. aureus at an approximate inoculum of 10 5 cfu/mL. Samples were drawn and bacteria were counted at 0, 1, 2, 3, 4, 6 and 24h of incubation at 37°C. Thus, after vortexing the culture tubes, two 50 ⁇ .
  • the purified compound was analyzed by Mass Spectroscopy and NMR to determine its chemical structure.
  • the NMR study was carried out on Bruker Avance 500 and 700 MHz spectrometers equipped with cryoprobes.
  • the sample (5-7 mM) was solubilized in water
  • Nemaucin MS-MS fragmentation was then carried out on the Nemaucin.
  • ESI-LC-MS data were obtained in the positive mode on a Waters alliance LC-MS system (Waters ZQ mass detector, Waters photodiode array detector 2696, Waters alliance HPLC systems 2790).
  • the HPLC column used was a C18 column (Waters Symmetry C18 5 ⁇ 4.6X150 mm) maintained at 35 °C. Solvents were (A) water + 0.1% TFA (B) acetonitrile + 0.1% TFA and the flow rate was 1 mL/min. The mobile phase composition was 100% A at 0 min, ramped to 30% B at 30 min. Samples were dissolved in solvent A (100 L). Sample injection volume was 10 iL. UV- Visible detection was by absorbance at 200-400 nm.
  • MS-MS fragmentation data were obtained on a Waters Micromass Q-Tof micro mass spectrometer.
  • Xenorhabdus cabanillasii CNCM 1-4418 was cultivated for 48 h, at 28°C with shaking in a 2 L Erlenmeyer flask containing 500 mL of medium broth composed of bactopeptone 10 g/L, K 2 HP0 4 5 g/L, MgS0 4 , 7H 2 0 1 g/L, (NH 4 ) 2 S04 2g/L, CaC0 3 2 g/L, and NaCl 10 g/L.
  • the culture was inoculated with 0.1% (v/v) of a 24 h preculture in the same medium.
  • the antibiotic production was controlled by analytical HPLC.
  • Bacterial cells were removed by low-speed centrifugation (6000 ⁇ g, 10 min at 4°C) and supernatant was sterilized onto 0.22 ⁇ pore size filter. Supernatant was added (1 : 1; v/v) to a 0.1 M NaCl-0.02 M Tris buffer (pH 7), and subjected to cation-exchange chromatography on a Sep Pack CarboxyMethyl cartridge (AcellTM Plus CM, Waters). Unbound material was removed by washes with a 0.1 M NaCl-0.02 M Tris buffer (pH 7) and the antibiotic activity eluted with 0.5 M NaCl-0.02 M Tris buffer (pH 7).
  • This eluate was acidified with 0.1% (v/v) trifluoroacetic acid (TFA) and was then subjected to reverse- phase chromatography on a Sep Pack C18 cartridge (Sep-Pak Plus CI 8, Waters). Unbound material was removed by washing with H 2 0-TFA 0.1% and the antibiotic pool was eluted with acetonitrile. The eluate was freeze-dried then resuspended in water (1 :5; v/v).
  • TFA trifluoroacetic acid
  • Nemaucin compounds demonstrate strong antibacterial activities.
  • the Nemaucins were tested for antimicrobial activity against a wide range of bacteria involved in nosocomial and animal infection.
  • Nemaucin has strong activity against S. aureus CIP 76.24 and E. coli CIP 76.25 (inferior to ⁇ g/mL) but has moderate activity against P. aeruginosa CIP 76.110.
  • Nemaucin was also tested against multi-resistant bacteria including Gram-positive and Gram-negative bacteria. They also have a very strong activity against Gram-positive multi-resistant bacteria such as MRSA, VRE and Gram-negative multi-resistant bacteria such as ESBL Enterobacter cloacae, E. coli, Klebsiella pneumonia, Klebsiella oxytoca, Klebsiella singaponasis or carbapenem resistant K. pneumoniae.
  • Nemaucin has a wide spectrum antibacterial activity including multi-resistant bacteria.
  • Table 1 MIC ⁇ g/mL of Nemaucin, Polymyxin, and Vancomycin against bacterial strains, including multiresistant clinical isolates (Nimes University Hospital)
  • NEB 2 0.78 0.39 - pneumoniae Klebsiella
  • Nemaucin is bactericidal (Table 2).
  • Table 2 The Bactericidal effects of Nemaucin on growing S. aureus CIP 76.24 was determined. At twofold and four-fold the MIC, Nemaucin kills 100% of bacteria within 6 h, making this molecule a fast and powerful bactericidal agent. ( Figure 2)
  • Nemaucin One compound referred to as Nemaucin was isolated, purified to homogeneity as a white powder and characterized by mass spectrometry. ESI-MS experiments revealed the molecular weight of Nemaucin. ( Figures 5 and 6)
  • Nemaucin A White powder; UV MeOHmax nm 214; ESI-MS (m/z) 1303 [M+H] + ;
  • the 1H spectrum revealed the features of a peptidic moiety with amide signals in the 9.1-6.9 ppm area, alpha proton signals in the 4.8-3.5 ppm, beta proton signals in the 3.7-2.0 ppm and methyl signals at 1.2-1.1 ppm.
  • an homonuclear data set (COSY, TOCSY, NOESY and ROESY) was recorded, as well as 1H- 13 C data set (HSQC, HSQC-TOCSY, HMBC) and a 1H- 15 N HSQC in natural abundance.
  • the combined analysis of all these data allowed us to identify spin systems of the peptidic sequence which contains at least 6 residues, including 2 unusual, derived from Threonine (Thr) and Proline (Pro) alterations.
  • Thr derived residue located in position 5 of the sequence results from the change of the ⁇ oxygen atom for a nitrogen atom, giving rise to an unusual ⁇ amino group. Moreover, this amino group is involved in a peptidic bond. This is clearly supported first, by the TOCSY and COSY data and confirmed by the 1H- 15 N HSQC which shows the two H-N cross peaks. Such an unusual residue was referred to later as Thn.
  • the Pro derived residue in position 6 was shown to contain an additional -CH(OH)- CH2- moiety between the alpha and carbonyl carbons.
  • the molecular weight of such a residue is 141.1 Da (159.1 Da in its carboxylic and amino form). It was referred to later as P141.
  • the 1H- 13 C HSQC and HSQC-TOCSY data were particularly helpful to identify its spin system. The configuration of the asymmetric carbons of these two non-natural residues was not determined.
  • the peptidic sequence was found to contain 6 residues as following:

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Antibiotic compounds purified from Xenorhabdus cabanillasii strain CNCM I-4418 and methods for producing said compounds and pharmaceutical compositions comprising said compounds and a pharmaceutically acceptable carrier for use in the treatment of fungal disease. Xenorhabdus cabanillasii strain CNCM I-4418.

Description

NEMAUCIN, AN ANTIBIOTIC PRODUCED BY
ENTOMOPATHOGENIC XENORHABDUS CABANILLASII
The present invention relates to new antibiotic compounds, to a strain of Xenorhabdus cabanillasii producing said antibiotic compounds and to the use of these compounds in the treatment of microbial disease in humans and animals.
Antimicrobial resistance is a major public health problem with a significant impact on morbidity, mortality and healthcare-associated costs. The problem has been worsened by the restriction of antibiotic drug discovery and development programs. Nowadays, the most relevant multiresistant bacterial pathogens are methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococci (VRE), extended spectrum β-lactamase formers (ESBL), multiresistant Pseudomonas and Acinetobacter species. For these bacteria, only a few of the existing antibiotics are efficient. There is an urgent need for new antibacterial compounds to ensure that bacterial infections can be effectively treated in the future. Living organisms have proven to be the most reliable source of bioactive chemicals with antimicrobial activity. Environmental microbes continue to be outstanding resources for the identification of new molecules. In this context, the discovery of new groups of antimicrobial peptides makes natural antibiotics the basic element of a novel generation of drugs for the treatment of bacterial and fungal infections. Antimicrobial peptides comprise a diverse class of molecules used in host defense by bacteria, plants, insects, and animals. Two examples of polypeptide antibiotics are polymyxin B and gramicidin S produced respectively by Bacillus polymyxa and Bacillus brevis.
The present application is based on research on an underexploited bacterial genus: Xenorhabdus. Gram-negative bacterial strains of the genus Xenorhabdus are known to be symbiotically associated with soil dwelling nematodes of the Steinernematidae family. After entering the insect larvae via natural openings, nematodes release bacteria from their intestine to the host's hemocoel. Bacteria are involved in killing the insect host by producing insecticidal proteins and inhibitors of the insect immune system. The bacteria proliferate in the corpse of the host and favour the reproduction of the nematode by degrading the insect biomass and by producing antibiotics that inhibit the development of the other microorganisms present in the corpse of the insect (bacteria, fungi). Boemare et al. (Boemare N.E. et al, Appl. Environ. Microbiol. 58, 3032-3037 (1992)) classified the antibiotic activities of Xenorhabdus into two categories: antimicrobial molecules with broad spectrum of activity, and bacteriocins with very narrow spectrum of activity and active only against bacteria closely related to X. nematophila. Only a few families of compounds have been described from Xenorhabdus in the literature: xenocoumacins (Gregson R.P. et al, WO 86/01509; Mclnerney B.V. et al, J. Nat. Prod. 54, 774-84 (1991)), xenorhabdins (Rhodes S.H. et al,WO 84/01775; Mclnerney B.V. et al, J. Nat. Prod. 54, 785-95 (1991)), indole derivatives (Webster J.M. et al, US patent 5,569,668; Li J. Et al, J. Nat. Prod. 58, 1081-6 (1995); Li J. et al, Can. J. Microbiol. 43, 770-3 (1997)), puromycin (Webster J.M. et al, Entomopathogenic hematology. 99-114 (2002)), benzylidenacetone (Ji D. et al, FEMS Microbiol. Lett. 239, 241-8 (2004)), proteinaceous bacteriocins (Boemare N.E. et al, Appl. Environ. Microbiol. 58, 3032-7 (1992); Thaler J.O. et al, Appl. Environ. Microbiol. 61, 2049-52 (1995)), xenematide (Lang G. et al, J. Nat. Prod. 71 (6), 1074-1077 (2008) and PAX (Gualtieri M. et al, J.Antibiot. 62(6) 295-302 (2009)).
In the present application, we describe new antimicrobial compounds, Nemaucins, produced by fermentation of a Xenorhabdus cabanillasii strain, and purified by cation exchange chromatography and reversed phase chromatography. The chemical structure of Nemaucins was obtained from the analysis of homo and heteronuclear 2D NMR and MS- MS experiments.
These molecules showed strong activity against Gram-positive and Gram-negative human bacterial pathogens, including some multi-resistant strains.
Summary
A first object of the present invention is a compound of formula (I):
Figure imgf000003_0001
wherein
Xaai is aspartic acid or glutamic acid,
Xaa2 is arginine, histidine or lysine,
Xaa3 is serine, threonine, asparagine or glutamine,
Xaa4 is selected in the group consisting of
Figure imgf000004_0001
R is H, OH or CH2-OH,
n is 1, 2, 3, 4 or 5.
Preferably, Xaai is aspartic acid.
Preferably, Xaa2 is histidine.
Preferably, Xaa3 is asparagine.
Preferably, Xaa4 is
Figure imgf000004_0002
Preferably, R is OH.
Preferably, n=4.
In preferred embodiments the compound of the present invention is the compound of formula (II):
Figure imgf000004_0003
(Π)
wherein n is 1, 2, 3, 4 or 5.
Preferably, n=4.
Preferably, the compounds of the present invention are for use as a medicament. Preferably, the compounds of the present invention are for use as an antibiotic agent.
Preferably, the compounds of the present invention are for use in treatment of microbial disease. Preferably, the compounds of the present invention are for use for treatment of a microbial disease caused by a microorganism selected from Staphylococcus, Enterococcus, Streptococcus, Bacillus, Clostridium, Chlamydia, Mycobacterium, Pasteurella, Escherichia, Enterobacter, Klebsiella, Citrobacter, Proteus, Serratia, Pseudomonas, Acinetobacter, Shigella, Salmonella, Stenotrophomonas, Yersinia, Burkholderia, Morganella, Moraxella, Propionibacterium, Listeria, Corynebacterium, Bifidobacterium, Lactobacillus, and Brucella.
More preferably, the compound of the present invention is for treatment of a microbial disease caused by a microorganism selected from Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens and Pseudomonas aeruginosa.
Another object of the present invention is a composition comprising a compound of the present invention in association with another antibiotic compound.
The present invention is also directed to a pharmaceutical composition comprising:
An effective amount of a compound of formula (I)
Figure imgf000005_0001
wherein
Xaai is aspartic acid or glutamic acid,
Xaa2 is arginine, histidine or lysine,
Xaa3 is serine, threonine, asparagine or glutamine,
Xaa4 is selected in the group consisting of
Figure imgf000005_0002
R is H, OH or -CH2-OH,
n is 1, 2, 3, 4 or 5,
A pharmaceutically acceptable
Preferably, Xaai is aspartic acid.
Preferably, Xaa2 is histidine.
Preferably, Xaa3 is asparagine.
Preferably, Xaa4 is
Figure imgf000006_0001
Preferably, R is OH
Preferably, n=4.
In preferred embodiments the compound of the present invention is the compound of formula (II):
Figure imgf000006_0002
(Π)
wherein n is 1, 2, 3, 4 or 5. Preferably, n=4.
A further object of the present invention is a method for producing a compound according to formula (II) comprising the following steps:
Growing Xenorhabdus cabanillasii strain CNCM 1-4418 in a liquid culture medium, - Purifying a compound according to formula (II)
Figure imgf000006_0003
wherein n=4. In preferred embodiments, the compound of formula (II) wherein n=4, is purified from the culture supernatant after removal of the Xenorhabdus cabanillasii cells.
The present invention is also directed to the Xenorhabdus cabanillasii strain deposited at CNCM on 15-12-2010 having the accession number CNCM 1-4418.
Another object of the present invention is a culture supernatant from the
Xenorhabdus cabanillasii strain of the present invention having antibiotic activity.
A further object of the present invention is an extract from the Xenorhabdus cabanillasii strain of the present invention having antibiotic activity. Description of the invention
In a first aspect, the present invention is related to Xenorhabdus cabanillasii strain JM26 deposited at CNCM (Collection Nationale de Cultures de Microorganismes) in the name of INRA (Institut National de la Recherche Agronomique) on 15-12-2010 having the accession number CNCM 1-4418.
It has been found that Xenorhabdus cabanillasii strain CNCM 1-4418 produces a compound exhibiting antibiotic or antimicrobial activity.
When Xenorhabdus cabanillasii strain CNCM 1-4418 is grown in a liquid culture medium, the antibiotic compound of the present invention is secreted into the culture supernatant. In another aspect, the present invention is related to a culture supernatant from Xenorhabdus cabanillasii strain CNCM 1-4418 exhibiting antibiotic activity. For the preparation of a culture supernatant having antibiotic activity Xenorhabdus cabanillasii strain CNCM 1-4418 is grown in a liquid culture medium under standard conditions, the bacterial cells are removed and the supernatant is recovered. The bacterial cells may for example be removed by centrifugation or filtration.
In another aspect, the invention is related to extracts from Xenorhabdus cabanillasii strain CNCM 1-4418 showing antibiotic activity. Cell extracts from Xenorhabdus cabanillasii may be prepared according to any appropriate method known to the skilled person.
The present invention also encompasses culture supernatant and extracts from
Xenorhabdus cabanillasii strain CNCM 1-4418 for use as medicament.
The present invention also encompasses culture supernatant and extracts from Xenorhabdus cabanillasii strain CNCM 1-4418 for use as an antibiotic agent.
The terms "antibiotic", "antibiotic activity" and "antimicrobial activity" as used herein refer generally to an effect in which a reduction, inhibition or a halt in the growth of a microorganism is achieved. Antibiotic activity may be tested according to any known method such as a microdilution method. The substances and compounds of the present invention inhibit or kill microorganism and more particularly kill or inhibit bacteria. Interestingly, the substances and compounds of the present invention inhibit or kill multidrug resistant bacteria which exhibit resistance to known antibiotics.
Xenorhabdus cabanillasii strain CNCM 1-4418, culture supernatant from this strain and cell extracts derived from this strain exhibit antibiotic activity against different microorganism including human bacterial pathogens.
More particularly, Xenorhabdus cabanillasii strain CNCM 1-4418, culture supernatant from this strain and cell extracts derived from this strain exhibit antibiotic activity against Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens and Pseudomonas aeruginosa.
Compounds having antibiotic activity as described above have been purified from the culture supernatant of Xenorhabdus cabanillasii strain CNCM 1-4418.
In another aspect, the present invention is related to a compound of formula (I):
Figure imgf000008_0001
(I) wherein
Xaai is aspartic acid or glutamic acid,
Xaa2 is arginine, histidine or lysine,
Xaa3 is serine, threonine, asparagine or glutamine,
Xaa4 is selected in the group consisting of
Figure imgf000008_0002
R is H, OH or CH2- n is 1, 2, 3, 4 or 5, a salt, a hydrate or a solvate thereof. These compounds have been named "Nemaucins". The term "Nemaucins" refers to the compounds of formula I, a salt, a hydrate or a solvate thereof.
Preferably, Xaai is aspartic acid.
Preferably, Xaa2 is histidine.
Preferably, Xaa3 is asparagine.
Preferably, Xaa4 is
Figure imgf000009_0001
Preferably, R is OH
Preferably, n=4.
In preferred embodiments the compound of the present invention is the compound of formula (II):
Figure imgf000009_0002
(Π)
wherein n is 1, 2, 3, 4 or 5,
a salt, a hydrate or a solvate thereof.
Preferably, n=4. This compound has been named "Nemaucin".
The term "Nemaucin" refers to the compound of formula II wherein n=4, a salt, a hydrate or a solvate thereof.
In preferred embodiments, the antibiotic compounds according to the present invention are for use as a medicament, for use as an antibiotic agent, for use as an antimicrobial agent, or for use in the treatment of microbial disease in particular of bacterial infection caused by pathogenic bacteria.
The compounds of the present invention are preferably for use in the treatment of bacterial infection and in particular in the treatment of hospital-acquired infections or nosocomial bacterial infections.
The invention is also related to the use of compounds of formula (I) for the manufacture of a medicament for treatment of microbial infection or microbial disease. The invention is also related to the use of compounds of formula (I) for the manufacture of an antibiotic.
The present invention also provides methods for treating microbial/bacterial infection and/or microbial/bacterial disease including administering an effective amount of a compound of formula (I) to a human or to a patient in need thereof. In a preferred embodiment, the invention relates to methods for treatment of bacterial disease and or bacterial infection.
In one embodiment, the patient is a mammal, i.e., a living mammal. In one embodiment, the patient is a human, i.e., a living human, including a living human child and a living human adult.
The term "treatment," as used herein in the context of treating a condition, refers generally to treatment and therapy, whether of a human or an animal (e.g., in veterinary applications), in which some desired therapeutic effect is achieved, for example, the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, and cure of the condition. Treatment as a prophylactic measure (i.e., prophylaxis) is also included. For example, use with subjects who have not yet developed the condition, but who are at risk of developing the condition, is encompassed by the term "treatment."
The term "therapeutically-effective amount," as used herein, refers to the amounts of the active agents (compounds of formula (I) or a salt, hydrate, or solvate thereof) that is effective for producing some desired therapeutic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.
The term "treatment" includes combination treatments and therapies, in which two or more treatments or therapies are combined, for example, sequentially or simultaneously. For example, the active agents of the present invention may also be used in further combination therapies, e.g., in conjunction with other agents, for example, other antimicrobial or antibiotic agents, etc.
Another aspect of the present invention is a pharmaceutical composition comprising:
- an effective amount of a compound of formula (I)
Figure imgf000011_0001
(I) wherein
Xaai is aspartic acid or glutamic acid,
Xaa2 is arginine, histidine or lysine,
Xaa3 is serine, threonine, asparagine or glutamine,
Xaa4 is selected in the group consisting of
Figure imgf000011_0002
R is H, OH or CH2-OH,
n is 1, 2, 3, 4 or 5,
and
a pharmaceutically acceptable carrier.
Preferably, Xaai is aspartic acid.
Preferably, Xaa2 is histidine.
Preferably, Xaa3 is asparagine.
Preferably, Xaa4 is
Figure imgf000011_0003
Preferably, R is OH
Preferably, n=4. In preferred embodiments the compound is the compound of formula (II):
Figure imgf000012_0001
wherein n is 1, 2, 3, 4 or 5,
a salt, a hydrate or a solvate thereof. Preferably, n=4.
As used herein, "pharmaceutically acceptable carriers" includes any and all solvents, dispersion media, coatings, and the like that are physiologically compatible.
The compositions of the invention may be in a variety of forms. These include for example liquid, semi-solid, and solid dosage forms, but the preferred form depends on the intended mode of administration and therapeutic application.
These pharmaceutical compositions are preferably for oral, topical, percutaneous or parenteral administration.
The compositions as described herein may be orally administered.
As solid compositions for oral administration, tablets, pills, powders (gelatine capsules, sachets) or granules may be used. In these compositions, the active ingredient according to the invention is mixed with one or more inert diluents, such as starch, cellulose, sucrose, lactose or silica, under an argon stream. These compositions may also comprise substances other than diluents, for example one or more lubricants such as magnesium stearate or talc, a coloring, a coating (sugar-coated tablet) or a glaze. Preferred oral compositions include coated and uncoated tablets, hard and soft gelatin capsules, sugar-coated pills, lozenges, pellets, and powders.
As liquid compositions for oral administration, there may be used pharmaceutically acceptable solutions, suspensions, emulsions, syrups and elixirs containing inert diluents such as water, ethanol, glycerol, vegetable oils or paraffin oil. These compositions may comprise substances other than diluents, for example wetting, sweetening, thickening, flavouring or stabilizing products.
In preferred embodiments, the pharmaceutical compositions for oral use contain Nemaucin together with the usual excipients as diluting agents like mannitol, lactose and sorbitol; binding agents like starchs, gelatins, sugars, cellulose derivatives, natural gums and polyvinylpyrrolidone; lubricating agents like talc, stearates, hydrogenated vegetable oils, polyethylenglycol and colloidal silicon dioxide; disintegrating agents like starcks, celluloses, alginates, gums and reticulated polymers; coloring, flavoring and sweetening agents.
In other embodiments, the compositions comprise Nemaucin according to the present invention with carriers or excipients suitable for topical administration. Preferred compositions for topical administration of Nemaucin according to the invention include ointments, pomades, creams, gels, and lotions.
The pharmaceutical compositions for topical use contain Nemaucin together with the usual excipients like white petrolatum, white wax, lanolin and derivatives thereof, stearylic alcohol, propylenglycol, sodium lauryl sulfate, ethers of the fatty polyoxy ethylene alcohols, sorbitan monostearate, glyceryl monostearate, propylene glycol monostearate, polyetilene glycols, methylcellulose, hydroxymethylpropylcellulose, sodium carboxymethylcellulose, colloidal aluminium and magnesium silicate, sodium alginate. Any topical preparation may be used in the present invention, for instance ointments, pomades, creams, gels and lotions.
The doses of Nemaucin depend on the desired effect, the duration of the treatment and the route of administration used.
In preferred embodiments, the pharmaceutical compositions according to the present invention are for use as an antimicrobial agent, for use as antibiotic or for use in the treatment of microbial disease in particular of microbial disease caused by bacteria.
The pharmaceutical compositions of the present invention are preferably for use in the treatment of bacterial infection and in particular for use in the treatment of hospital- acquired infections or nosocomial bacterial infections.
Preferably, the compound of the present invention is for use for treatment of a microbial disease caused by a microorganism selected from Staphylococcus, Enter ococcus, Streptococcus, Bacillus, Clostridium, Chlamydia, Mycobacterium, Pasteurella, Escherichia, Enter obacter, Klebsiella, Citrobacter, Proteus, Serratia, Pseudomonas, Acinetobacter, Shigella, Salmonella, Stenotrophomonas, Yersinia, Burkholderia, Morganella, Moraxella, Propionibacterium, Listeria, Corynebacterium, Bifidobacterium, Lactobacillus, and Brucella.
More preferably, the compound of the present invention is for treatment of a microbial disease caused by a microorganism selected from Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Enterobacter clocae, Klebsiella pneumoniae, Klebsellia oxytica, Klebsiella singaponasis, Citrobacter koseri, Citrobacter freundii, Proteus penneri, Serratia marescens and Pseudomonas aeruginosa. The compounds of the present invention may be combined with other active compounds exhibiting an antimicrobial/antibiotic activity. The pharmaceutical compositions encompassed by the present invention may also contain a further therapeutic agent for the treatment of bacterial disease or bacterial infection.
Another aspect of the present invention is a method for producing a compound according to formula (II) comprising the following steps:
Growing Xenorhabdus cabanillasii strain CNCM 1-4418 in a liquid culture medium, - Purifying a compound according to formula (II):
Figure imgf000014_0001
wherein n=4.
The active compounds according to the present invention may be purified from the Xenorhabdus cabanillasii cells of the present invention. Advantageously, the active compounds of the present invention may be purified from the culture supernatant after removal of the Xenorhabdus cabanillasii cells. For the preparation of a culture supernatant having antifungal activity Xenorhabdus cabanillasii strain CNCM 1-4418 is grown in a liquid culture medium under standard conditions, the bacterial cells are removed and the supernatant is recovered. The bacterial cells may for example be removed by centrifugation or filtration.
Further purification of the active compounds according to the present invention may be carried out by any known method including cation-exchange chromatography, reversed-phase chromatography and/or reverse phase HPLC.
In preferred embodiments, the active compound is purified from the culture supernatant of Xenorhabdus cabanillasii strain CNCM 1-4418 by successive cation- exchange chromatography, reversed-phase chromatography and reverse phase HPLC.
Figures
Figure 1 : HPLC chromatogram of the Xenorhabdus cabanillasii growth medium
Figure 2: Bactericidal effects of Nemaucin on growing S. aureus CIP 76.24
Figure 3 : Viability of hTERT HME-1 at 8 concentrations of Nemaucin
Figure 4: Viability of PC-3 at 8 concentrations of Nemaucin Figure 5 : ESI-MS experiments
Figure 6: UV absorption spectra of Nemaucin
Figure 7: Peptidic part of Nemaucin
Figure 8: MSMS fragmentation of Nemaucin
Figure 9: Chemical structure of Nemaucin
EXAMPLES
A. Materials and methods
Producing organism
Xenorhabdus cabanillasi CMCM 1-4418 was grown on Luria-Bertani medium (LB, composed of bactotryptone 10 g/L, yeast extract 5 g/L and NaCl 10 g/L) for liquid culture and on LB-agar for solid cultures. The phase status (I or II) of this strain was determined by culturing on NBTA (Nutrient agar (Difco) 31 g/L, bromothymol blue 25 mg/L and 2, 3, 5-triphenyl tetrazolium chloride 1% 40 mg/L) and measuring antibacterial activity against Micrococcus luteus. Xenorhabdus exhibit two colony forms or variants when cultured in vitro. Modifications of the outer membrane induce differential adsorption of dyes by variants. Phase I variants absorb dyes and are blue on NBTA plates, while phase II colonies are red. Phases I and II of strains are indicated as suffixes {l\ and 12, respectively) attached to strain designations. This strain was maintained at 15°C on NBTA medium.
Antibacterial susceptibility testing methods
The following reference strains were used for evaluation of antimicrobial activity: Pseudomonas aeruginosa CIP 76.110, Escherichia coli CIP 76.24, Staphylococcus aureus CIP 76.25, and clinical isolates obtained from patients with infection at the University Hospital of ines. Vancomycin and polymyxin (Sigma-Aldrich) were provided as standard powders by the manufacturers. MIC and MBC determination procedures were determined as recommended by the CLSI (National Committee for Clinical Laboratory Standards. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically— Sixth Edition: Approved Standard M07-A6. NCCLS, Villanova, PA, USA. (2003)).
Antibiotics were tested at final concentrations (prepared from serial twofold dilutions) ranging from 0.01 to 100 μg/mL. The minimal inhibitory concentration (MIC) was defined as the lowest antibiotic concentration which yielded no visible growth. The test medium was Mueller-Hinton broth (MHB), the inoculum was 5xl05 CFU/mL. The inoculated microplates were incubated at 37°C for 18 h before reading. Minimal bactericidal concentrations (MBCs) were established by extending the MIC procedure to the evaluation of bactericidal activity. After 24 hours, 10 μΙ_, were drawn from the wells, serially diluted and then spotted onto suitable agar plates. The plates were incubated at 37°C overnight. The MBC was read 18 h later as the lowest concentration of antibiotic which resulted in 0.1 % survival in the subculture. All the experiments were done in triplicates.
Protein binding.
The impact of serum protein binding was assessed by examining the impact of human serum albumin (HSA) on the activity of Nemaucin in vitro. The MICs for S. aureus CIP 76.25 and E. coli CIP 76.24 were determined in MHB + 4% HSA. Concentrations of HSA used for in vitro studies were selected on the basis of normal physiological concentration. A reduced potency (higher MIC) in MHB + HSA was presumed due to drug binding to serum protein.
Cytotoxicity test
100 μΙ_, of cell suspension of PC-3 (Human prostatic carcinoma) or hTERT HME-1 (Human normal mammary epithelium) prepared in RPMI + 10% SVF + 1% glutamine were inoculated into 96-well plates. The inoculating cell number was 3500 cells per well for PC-3 and 7500 cells per well for hTERT HME-1. The microplates were incubated at 37°C for 24h with 5% of C02. After 24h of culture, the medium was removed by aspiration and 100 μΐ of RPMI + 10% SVF + 1% glutamine with 8 concentrations of Nemaucin (from 0.78 μg/ml to 100 μg/mL) were added to each well. Microplates were incubated at 37°C for 48h with 5% of C02. After 48h of culture, the medium was removed by aspiration and 100 μΙ_, of RPMI + MTT (0.5 mg/mL) were added to each wells. Microplates were inoculated at 37°C during 150 min. Next, all the medium was removed by inverting and tapping the plate and 100 μΙ_, of DMSO were added in each plate. The spectrophotometric absorbance was then measured at 550 nm.
The Lethal Dose 50 corresponds to DL50 = ((50-(Y2-((Yl-Y2)/(Xl- X2))*X2))/((Y1-Y2)/(X1-X2))). XI represents the concentration for which viability was superior to 50%, X2 represents the concentration for which viability was inferior to 50%, Yl represents the percentage of viability at the concentration XI and Y2 the percentage of viability at the concentration X2.
Experiments were done in triplicate.
Bactericidal effects of Nemaucin on growing S. aureus
Bacterial killing curves were carried out by inoculating S. aureus CIP 76.24 with Nemaucin concentrations equal to the MIC, two-fold the MIC and four-fold the MIC. Vancomycin was used at four-fold its MIC. The S. aureus inoculums was prepared from colonies grown overnight in MHB. Antibiotics concentrations in the flask were adjusted in MHB according to the desired concentration. Culture tubes containing 10 mL were inoculated with S. aureus at an approximate inoculum of 105cfu/mL. Samples were drawn and bacteria were counted at 0, 1, 2, 3, 4, 6 and 24h of incubation at 37°C. Thus, after vortexing the culture tubes, two 50 μΐ. samples were removed and serially diluted with MHB. After each dilution step, 20 μΐ. was plated onto LB agar plates, which were incubated for 24 h at 37°C. Afterwards the colonies were counted and back-extrapolated to the original volume to determine the initial concentration (cfu/mL).
NMR and MS analysis
The purified compound was analyzed by Mass Spectroscopy and NMR to determine its chemical structure.
The NMR study was carried out on Bruker Avance 500 and 700 MHz spectrometers equipped with cryoprobes. The sample (5-7 mM) was solubilized in water
(95/5 H20/D20 v/v) at pH 4.3. Protons chemical shifts are expressed with respect to sodium 4,4 dimethyl-silapentane-1 -sulfonate, according to IUPAC recommendations.
LC-MS was first performed in order to obtain the m/z value of the protonated molecules of
Nemaucin. MS-MS fragmentation was then carried out on the Nemaucin. ESI-LC-MS data were obtained in the positive mode on a Waters alliance LC-MS system (Waters ZQ mass detector, Waters photodiode array detector 2696, Waters alliance HPLC systems 2790).
The HPLC column used was a C18 column (Waters Symmetry C18 5μιη 4.6X150 mm) maintained at 35 °C. Solvents were (A) water + 0.1% TFA (B) acetonitrile + 0.1% TFA and the flow rate was 1 mL/min. The mobile phase composition was 100% A at 0 min, ramped to 30% B at 30 min. Samples were dissolved in solvent A (100 L). Sample injection volume was 10 iL. UV- Visible detection was by absorbance at 200-400 nm.
Solvent flow to the MS was diverted to waste for the first 5 min to minimise salt build-up.
MS-MS fragmentation data were obtained on a Waters Micromass Q-Tof micro mass spectrometer.
B. Results
Fermentation
Xenorhabdus cabanillasii CNCM 1-4418 was cultivated for 48 h, at 28°C with shaking in a 2 L Erlenmeyer flask containing 500 mL of medium broth composed of bactopeptone 10 g/L, K2HP04 5 g/L, MgS04, 7H20 1 g/L, (NH4)2S04 2g/L, CaC03 2 g/L, and NaCl 10 g/L. The culture was inoculated with 0.1% (v/v) of a 24 h preculture in the same medium. The antibiotic production was controlled by analytical HPLC. (Figure 1) Isolation
Bacterial cells were removed by low-speed centrifugation (6000 χ g, 10 min at 4°C) and supernatant was sterilized onto 0.22 μιη pore size filter. Supernatant was added (1 : 1; v/v) to a 0.1 M NaCl-0.02 M Tris buffer (pH 7), and subjected to cation-exchange chromatography on a Sep Pack CarboxyMethyl cartridge (Acell™ Plus CM, Waters). Unbound material was removed by washes with a 0.1 M NaCl-0.02 M Tris buffer (pH 7) and the antibiotic activity eluted with 0.5 M NaCl-0.02 M Tris buffer (pH 7). This eluate was acidified with 0.1% (v/v) trifluoroacetic acid (TFA) and was then subjected to reverse- phase chromatography on a Sep Pack C18 cartridge (Sep-Pak Plus CI 8, Waters). Unbound material was removed by washing with H20-TFA 0.1% and the antibiotic pool was eluted with acetonitrile. The eluate was freeze-dried then resuspended in water (1 :5; v/v). Pure compounds were isolated from the crude extract by reverse phase HPLC using a C18 column (Waters ; Symmetry Symmetry CI 8; 5μπι; 4.6X150 mm), a linear gradient of H20, 0.1% TFA-acetonitrile starting from 0% to 30% in 30 min, a flow rate of 1 mL/min and an UV detection from 200 to 400 nm, yielding pure Nemaucin with the following FIPLC- retention time: 27.16 min.
Biological properties
Nemaucin compounds demonstrate strong antibacterial activities. The Nemaucins were tested for antimicrobial activity against a wide range of bacteria involved in nosocomial and animal infection. Regarding reference strains (Table 1), Nemaucin has strong activity against S. aureus CIP 76.24 and E. coli CIP 76.25 (inferior to ^g/mL) but has moderate activity against P. aeruginosa CIP 76.110. Nemaucin was also tested against multi-resistant bacteria including Gram-positive and Gram-negative bacteria. They also have a very strong activity against Gram-positive multi-resistant bacteria such as MRSA, VRE and Gram-negative multi-resistant bacteria such as ESBL Enterobacter cloacae, E. coli, Klebsiella pneumonia, Klebsiella oxytoca, Klebsiella singaponasis or carbapenem resistant K. pneumoniae.
Nemaucin has a wide spectrum antibacterial activity including multi-resistant bacteria. Table 1: MIC ^g/mL) of Nemaucin, Polymyxin, and Vancomycin against bacterial strains, including multiresistant clinical isolates (Nimes University Hospital)
Bacteria Strain Resistance Nemaucin Polymyxin Vancomycin
Staphylococcus
CIP 76.25 - 0.08 - 1.56 aureus
Staphylococcus
21840 MRSA 0.05 - 1.56 aureus
Staphylococcus
20681 MRSA 0.05 - 1.56 aureus
Staphylococcus
20364 MRSA 0.05 - 1.56 aureus
Staphylococcus
23305 MRSA 0.05 - 1.56 aureus
Staphylococcus
16666 MRSA 0.05 - 1.56 aureus
Enterococcus
N5 VRE 0.05 - >50 faecalis
Escherichia
CIP 76.24 - 0.78 0.31 - coli
Escherichia
PEC21 ESBL/SHV 0.78 0.31 - coli
Escherichia
NEC20 ESBL/TEM 0.39 0.02 - coli
Escherichia
MEC12 ESBL/TEM 0.78 0.07 - coli
Escherichia ESBL/CTX
MEC63 0.78 0.31 - coli M15
Escherichia ESBL/CTX
EC Abwa 0.78 0.15 - coli M15
Escherichia ESBL/CTX
AEC7 0.78 0.31 - coli M14
Enterobacter
11370 0.78 0.62 - clocae
Enterobacter
BEB 1 1.56 1.25 - clocae
Enterobacter
BEB 4 0.78 0.08 - clocae
Enterobacter
41206 1.56 0.08 - clocae
Enterobacter
36265 0.78 0.08 - clocae
Klebsiella
PEB1 0.39 0.62 - pneumoniae
Klebsiella
MEB 11 0.78 0.62 - pneumoniae
Klebsiella
NEB 2 0.78 0.39 - pneumoniae Klebsiella
NEB 4 0.78 5.00 - pneumoniae
Klebsiella
KPC2 475 KPC 1.56 1.25 - pneumoniae
Klebsiella
PEB 4 0.78 0.63 - oxytica
Klebsiella
NEB 9 0.78 0.63 - singaponasis
Citrobacter
BEB 6 0.39 0.07 - koseri
Citrobacter
37553 1.56 0.15 - freundii
Proteus
12042 0.78 >12.5 - penneri
Serratia
31674 >12.5 0.03 - marescens
Serratia
31626 >12.5 0.01 - marescens
Pseudomonas
CIP 76.110 - >12.5 0.62 - aeruginosa
Pseudomonas
5618 MDR >12.5 0.62 - aeruginosa
Pseudomonas
401681 MDR >12.5 0.31 - aeruginosa
Pseudomonas
35170 MDR >12.5 0.62 - aeruginosa
Pseudomonas
42162 MDR >12.5 0.62 - aeruginosa
Pseudomonas
512232 MDR >12.5 0.62 - aeruginosa
As evidenced by its value of MIC and MBC, Nemaucin is bactericidal (Table 2). The Bactericidal effects of Nemaucin on growing S. aureus CIP 76.24 was determined. At twofold and four-fold the MIC, Nemaucin kills 100% of bacteria within 6 h, making this molecule a fast and powerful bactericidal agent. (Figure 2)
Their activities are reduced by a factor of 2 in presence of HSA, suggesting weak interaction between HSA and Nemaucin. (Table 2)
Table 2: MIC (in μg/mL), MBC (in μg/mL) and MIC in presence of 4% of HSA (in μg/mL) against reference bacteria (*MIC/MBC)
S.aureus CIP 76.25 E.coli CIP 76.24
Nemaucin 0.04 μ /ι 1/0.08 1πά 0.78/0.78*
Nemaucin
0.08 1.56
+4% (w/v) HSA The LD50 of Nemaucin A was 36.8 ± 3.54 μ§/ιηΙ. against Human prostatic carcinoma cells (PC-3) and 28.42 ± 3.61 μg/mL against Human normal mammary epithelium cells (hTERT HME-1) (Table 3). 100 % of cell viability was observed up to 12^g/mL. (Figures 3 and 4)
Table 3: LD 50 of Nemaucin A against PC-3 and hTERT HME-1
Figure imgf000021_0001
Physico-chemical properties of nemaucins
One compound referred to as Nemaucin was isolated, purified to homogeneity as a white powder and characterized by mass spectrometry. ESI-MS experiments revealed the molecular weight of Nemaucin. (Figures 5 and 6)
Nemaucin A : White powder; UV MeOHmax nm 214; ESI-MS (m/z) 1303 [M+H]+;
Chemical structure elucidation
The 1H spectrum revealed the features of a peptidic moiety with amide signals in the 9.1-6.9 ppm area, alpha proton signals in the 4.8-3.5 ppm, beta proton signals in the 3.7-2.0 ppm and methyl signals at 1.2-1.1 ppm. To identify the spin systems of the different residues an homonuclear data set (COSY, TOCSY, NOESY and ROESY) was recorded, as well as 1H-13C data set (HSQC, HSQC-TOCSY, HMBC) and a 1H-15N HSQC in natural abundance. The combined analysis of all these data allowed us to identify spin systems of the peptidic sequence which contains at least 6 residues, including 2 unusual, derived from Threonine (Thr) and Proline (Pro) alterations.
The Thr derived residue located in position 5 of the sequence results from the change of the γ oxygen atom for a nitrogen atom, giving rise to an unusual β amino group. Moreover, this amino group is involved in a peptidic bond. This is clearly supported first, by the TOCSY and COSY data and confirmed by the 1H-15N HSQC which shows the two H-N cross peaks. Such an unusual residue was referred to later as Thn.
The Pro derived residue in position 6 was shown to contain an additional -CH(OH)- CH2- moiety between the alpha and carbonyl carbons. The molecular weight of such a residue is 141.1 Da (159.1 Da in its carboxylic and amino form). It was referred to later as P141. The 1H-13C HSQC and HSQC-TOCSY data were particularly helpful to identify its spin system. The configuration of the asymmetric carbons of these two non-natural residues was not determined. By using sequential NOEs, the peptidic sequence was found to contain 6 residues as following:
Asp^His^Asp^Asn4 Thn5-P1416-CH2-CH2-R'
Proton and 1H-13C HSQC spectra were recorded at different pH ranging from 4.34 to 2.15. Of the two Asp residues only the Asp1 was sensitive to the ionisation meaning that its γ carboxyl group is free. The signals of the Asp3 spin system were insensitive to pH suggesting that its γ carboxyl group was involved in an amide bond. The NOE between the β amide of Thn5 and the β protons of Asp3 indicates that the side chains of these two residues were bound by a peptidic bond to form a cyclic peptide.
We observed that the P1416 residue is linked to a seventh one, the amide signal of which was a triplet. Its spin system contains a -HN-CH2-CH2- moiety (COSY, TOCSY, HSQC- TOCSY).
Chemical structure of the peptidic part of Nemaucin was confirmed by MSMS fragmentation. (Table 5 and Figure 8)
Table 4: Chemical shifts of the identified peptidic part (water, 280 K)
Figure imgf000022_0001
Table 5: Key fragmentations of the 1303 [M+H]+ ions of Nemaucin A.
Figure imgf000023_0002
The 1H NMR spectra also showed 18 methylenes including 9 CH2 bonded to nitrogen. After a detailed examination of the 2D-NMR spectra (COSY, HSQC, HMBC, TOCSY), the following structure is suggested for the R'-part:
Figure imgf000023_0001
The assignment of 1H in D20 is:
1H NMR (500 MHz, D20) δ 3.38-3.21 (18H, m, H-2, 7, 10, 15, 18, 23, 26, 31, and 34), 1.75-1.58 (16H, m, H-8, 9, 16, 17, 24, 25, 32, and 33)
The complete chemical structure of Nemaucin in shown in Figure 9. This report describes the production, the purification and the characterization of a novel peptide antibiotic produced by Xenor abdus cabamllasii. Its significant antibacterial activity, including against multi-resistant bacteria, increases the potential interest of this molecule for human and animal health applications.
REFERENCES
Appl. Environ. Microbiol. 58, 3032-3037 (1992)
J. Nat. Prod. 54, 774-84 (1991)
J. Nat. Prod. 54, 785-95 (1991)
J. Nat. Prod. 58, 1081-6 (1995)
Can. J. Microbiol. 43, 770-3 (1997)
J.Antibiot. 62(6) 295-302 (2009)
Entomopathogenic Nematology. 99-114 (2002)
FEMS Microbiol. Lett. 239, 241-8 (2004)
Appl. Environ. Microbiol. 58, 3032-7 (1992)
Appl. Environ. Microbiol. 61, 2049-52 (1995)
J. Nat. Prod. 71 (6), 1074-1077 (2008) PATENT REFERENCES
WO 86/01509
WO 84/01775
US 5,569,668
Print Out (Original in Electronic Form)
0-1 Form PCT/RO/134 (SAFE)
Indications Relating to Deposited
Microorganism(s) or Other Biological
Material (PCT Rule 13bis)
0-1 -1 Prepared Using PCT Online Filing
Version 3 . 5 . 000 . 225 MT/FOP
20020701/0 . 20 . 5 . 20
0-2 International Application No.
PCT/EP201 1/073738
0-3 Applicant's or agent's file reference 359890D28953
1 The indications made below relate to
the deposited microorganism(s) or
other biological material referred to in
the description on:
1-1 page 6
1-2 line 4
1-3 Identification of deposit
1 -3-1 Name of depositary institution CNCM Collection nationale de cultures de micro-organismes
1 -3-2 Address of depositary institution Institut Pasteur, 28 , rue du Dr Roux,
75724 Paris Cedex 15 , France
1 -3-3 Date of deposit 15 December 2010 ( 15 . 12 . 2010 )
1 -3-4 Accession Number CNCM 1-4418
1-4 Additional Indications Xenorhabdus cabanillasii JM 26
1-5 Designated States for Which
Indications are Made All designations
FOR RECEIVING OFFICE USE ONLY
0-4 This form was received with the
international application: Yes
(yes or no)
0-4-1 Authorized officer
Krista Delimon
FOR INTERNATIONAL BUREAU USE ONLY
0-5 This form was received by the
international Bureau on:
0-5-1 Authorized officer

Claims

1. Compound of formula (I):
Figure imgf000026_0001
(I) wherein
Xaai is aspartic acid or glutamic acid,
Xaa2 is arginine, histidine or lysine,
Xaa3 is serine, threonine, asparagine or glutamine,
Xaa4 is selected in the group consisting of
Figure imgf000026_0002
R is H, OH or -CH2- n is 1, 2, 3, 4 or 5.
2. Compound according to claim 1 wherein Xaai is aspartic acid.
3. Compound according to anyone of claims 1-2 wherein Xaa2 is histidine.
4. Compound according to anyone of claims 1-3 wherein Xaa3 is asparagine.
5. Compound according to anyone of claims 1-4 wherein Xaa4 is
Figure imgf000026_0003
6. Compound according to anyone of claims 1-5 wherein R is OH.
7. Compound according to anyone of claims 1-6 wherein n=4.
8. Compound according to anyone of claims 1-7 of formula (II):
Figure imgf000027_0001
(Π)
wherein n is 1, 2, 3, 4 or 5.
9. Compound according to claim 8 wherein n=4.
10. Compound according to anyone of claims 1-9 for use as a medicament.
11. Compound according to anyone of claims 1-10 for use as an antibiotic agent.
12. Composition comprising a compound according to anyone of claims 1-11 and an antibiotic compound.
13. Method for producing a compound according to formula (II)
comprising the following steps:
a. Growing Xenorhabdus cabanillasii strain CNCM 1-4418 in a liquid culture medium,
b. Purifying a compound according to formula (II)
Figure imgf000027_0002
wherein n=4.
14. Xenorhabdus cabanillasii strain deposited at CNCM having the accession number CNCM 1-4418.
15. Culture supernatant from the Xenorhabdus cabanillasii strain of claim 14 having antibiotic activity.
PCT/EP2011/073738 2010-12-22 2011-12-22 Nemaucin, an antibiotic produced by entomopathogenic xenorhabdus cabanillasii WO2012085177A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201061425917P 2010-12-22 2010-12-22
EP10306482.0 2010-12-22
EP10306482 2010-12-22
US61/425,917 2010-12-22

Publications (1)

Publication Number Publication Date
WO2012085177A1 true WO2012085177A1 (en) 2012-06-28

Family

ID=45497956

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/073738 WO2012085177A1 (en) 2010-12-22 2011-12-22 Nemaucin, an antibiotic produced by entomopathogenic xenorhabdus cabanillasii

Country Status (1)

Country Link
WO (1) WO2012085177A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015173778A1 (en) 2014-05-16 2015-11-19 International Centre Of Insect Physiology And Ecology Antimicrobial agents produced by xenorhabdus griffiniae strain xn45
WO2021148594A1 (en) 2020-01-23 2021-07-29 Nosopharm Chemical synthesis of the peptidic part of bioactive natural products
US11603386B2 (en) 2017-10-31 2023-03-14 Wisconsin Alumni Research Foundation Insect, tick, and mite repellent derived from Xenorhabdus budapestensis

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1984001775A1 (en) 1982-10-26 1984-05-10 Commw Scient Ind Res Org Xenorhabdin antibiotics
WO1986001509A1 (en) 1984-09-05 1986-03-13 Biotechnology Australia Pty. Ltd. Xenocoumacins
US5569668A (en) 1995-03-29 1996-10-29 Webster; John M. Indole derivatives with antibacterial and antimycotic properties

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1984001775A1 (en) 1982-10-26 1984-05-10 Commw Scient Ind Res Org Xenorhabdin antibiotics
WO1986001509A1 (en) 1984-09-05 1986-03-13 Biotechnology Australia Pty. Ltd. Xenocoumacins
US5569668A (en) 1995-03-29 1996-10-29 Webster; John M. Indole derivatives with antibacterial and antimycotic properties

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
"Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically", 2003, NATIONAL COMMITTEE FOR CLINICAL LABORATORY STANDARDS
APPL. ENVIRON. MICROBIOL., vol. 58, 1992, pages 3032 - 3037
APPL. ENVIRON. MICROBIOL., vol. 58, 1992, pages 3032 - 7
APPL. ENVIRON. MICROBIOL., vol. 61, 1995, pages 2049 - 52
BOEMARE N.E. ET AL., APPL. ENVIRON. MICROBIOL., vol. 58, 1992, pages 3032 - 3037
BOEMARE N.E. ET AL., APPL. ENVIRON. MICROBIOL., vol. 58, 1992, pages 3032 - 7
CAN. J. MICROBIOL., vol. 43, 1997, pages 770 - 3
ENTOMOPATHOGENIC NEMATOLOGY, 2002, pages 99 - 114
FEMS MICROBIOL. LETT., vol. 239, 2004, pages 241 - 8
GUALTIERI M. ET AL., J.ANTIBIOT., vol. 62, no. 6, 2009, pages 295 - 302
J. NAT. PROD., vol. 54, 1991, pages 774 - 84
J. NAT. PROD., vol. 54, 1991, pages 785 - 95
J. NAT. PROD., vol. 58, 1995, pages 1081 - 6
J. NAT. PROD., vol. 71, no. 6, 2008, pages 1074 - 1077
J.ANTIBIOT., vol. 62, no. 6, 2009, pages 295 - 302
JI D. ET AL., FEMS MICROBIOL. LETT., vol. 239, 2004, pages 241 - 8
LANG G. ET AL., J. NAT. PROD., vol. 71, no. 6, 2008, pages 1074 - 1077
LI J. ET AL., CAN. J. MICROBIOL., vol. 43, 1997, pages 770 - 3
LI J. ET AL., J. NAT. PROD., vol. 58, 1995, pages 1081 - 6
M. K. MASSAOUD ET AL: "Proteolytic Enzyme Production by Strains of the Insect Pathogen Xenorhabdus and Characterization of an Early-Log-Phase-Secreted Protease as a Potential Virulence Factor", APPLIED AND ENVIRONMENTAL MICROBIOLOGY, vol. 76, no. 20, 15 October 2010 (2010-10-15), pages 6901 - 6909, XP055001087, ISSN: 0099-2240, DOI: 10.1128/AEM.01567-10 *
MAXIME GUALTIERI ET AL: "Identification of a new antimicrobial lysine-rich cyclolipopeptide family from Xenorhabdus nematophila", THE JOURNAL OF ANTIBIOTICS, vol. 62, no. 6, 1 June 2009 (2009-06-01), pages 295 - 302, XP055001101, ISSN: 0021-8820, DOI: 10.1038/ja.2009.31 *
MCINERNEY B.V. ET AL., J. NAT. PROD., vol. 54, 1991, pages 774 - 84
MCINERNEY B.V. ET AL., J. NAT. PROD., vol. 54, 1991, pages 785 - 95
THALER J.O. ET AL., APPL. ENVIRON. MICROBIOL., vol. 61, 1995, pages 2049 - 52
WEBSTER J.M. ET AL., ENTOMOPATHOGENIC NEMATOLOGY, 2002, pages 99 - 114

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015173778A1 (en) 2014-05-16 2015-11-19 International Centre Of Insect Physiology And Ecology Antimicrobial agents produced by xenorhabdus griffiniae strain xn45
US11603386B2 (en) 2017-10-31 2023-03-14 Wisconsin Alumni Research Foundation Insect, tick, and mite repellent derived from Xenorhabdus budapestensis
WO2021148594A1 (en) 2020-01-23 2021-07-29 Nosopharm Chemical synthesis of the peptidic part of bioactive natural products

Similar Documents

Publication Publication Date Title
US11548916B2 (en) Anti-infective compound
AU2005300815B2 (en) Acylated nonadepsipeptides used as lysobactin derivatives
CN113754784A (en) Cell-penetrating antibacterial peptide and application thereof
WO2011058923A1 (en) Antibacterial agent for drug-resistant bacteria, method for screening for same, and use of same
EP2760881B1 (en) Novel peptide derivatives as antibiotics
Kurnianto et al. Antibacterial and antioxidant potential of ethyl acetate extract from Streptomyces AIA12 and AIA17 isolated from gut of Chanos chanos
CN107344958B (en) Antibacterial pentapeptide derivative and application thereof
CN117586352A (en) Antibacterial polypeptide APH220 based on salivary glands of Hirudinaria manillensis and application thereof
WO2012085177A1 (en) Nemaucin, an antibiotic produced by entomopathogenic xenorhabdus cabanillasii
RU2409661C2 (en) Enterococcus faecium lvp1073 strain, producer of bacteriocin against bacterial pathogens, bacteriocin e1073 against bacterial pathogens, lactobacillus plantarum 1 lvp7 strain - bacteriocin e1073 synthesis inducer, signal peptide sp1073 - bacteriocin e1073 synthesis regulator, method for producing bacteriocin e1073
KR101723089B1 (en) Antibacterial Composition for Antibiotics Resistant Staphylococcus aureus inhibition Comprising Shikonin
AU2007237861B2 (en) Novel antibacterial compounds
CN115850409B (en) Leader-free bacteriocin A3 resistant to multiple pathogenic bacteria, and preparation method and application thereof
CN116947999A (en) Small molecular antibacterial polypeptide, preparation method and application thereof
Murad et al. Assessment of antimicrobial, antialgal and cytotoxic activities of crude extracts from rhizospheric and freshwater cyanobacterial strains
Tehrani et al. Synthesis of a peptide derivative of microcinJ25 and evaluation of antibacterial and biological activities
Seratnahaei et al. Antimicrobial activities of the secondary metabolite extracted from a Nocardia strain
EP2468718A1 (en) Cabanillasin, a new antifungal compound, produced by entomopathogenic xenorhabdus cabanillasii
KR102342719B1 (en) Antibacterial composition comprising of Micromonospora sp. as effective component
KR102629608B1 (en) Antibacterial composition comprising a fractional extract isolated from Coturnix japonica eggs as an active ingredient
Fliss et al. Synthesis and structure-activity study of the antimicrobial lipopeptide brevibacillin
JP2022030930A (en) Peptide based antimicrobial compound
TW200812607A (en) Novel antibacterial compounds
PL223435B1 (en) TRIS peptide complex obtained from the Raoultella ornithinolytica bacterial strain and its application in treatment of human cervical carcinoma

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11810591

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11810591

Country of ref document: EP

Kind code of ref document: A1