WO2018195536A1 - Antibacterial compounds - Google Patents

Antibacterial compounds Download PDF

Info

Publication number
WO2018195536A1
WO2018195536A1 PCT/US2018/028868 US2018028868W WO2018195536A1 WO 2018195536 A1 WO2018195536 A1 WO 2018195536A1 US 2018028868 W US2018028868 W US 2018028868W WO 2018195536 A1 WO2018195536 A1 WO 2018195536A1
Authority
WO
WIPO (PCT)
Prior art keywords
bacteria
infection
compound
group
bacterial
Prior art date
Application number
PCT/US2018/028868
Other languages
French (fr)
Inventor
Shen YU
Rhonda Feinbaum
Laurence Rahme
Terence Moy
Frederick M. Ausubel
Original Assignee
Yu Shen
Rhonda Feinbaum
Laurence Rahme
Terence Moy
Ausubel Frederick M
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 Yu Shen, Rhonda Feinbaum, Laurence Rahme, Terence Moy, Ausubel Frederick M filed Critical Yu Shen
Publication of WO2018195536A1 publication Critical patent/WO2018195536A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • A61K31/055Phenols the aromatic ring being substituted by halogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/131Amines acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/191Carboxylic acids, e.g. valproic acid having two or more hydroxy groups, e.g. gluconic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/7056Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing five-membered rings with nitrogen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • This invention relates to compounds useful in treating bacterial infections.
  • the bacterial species Pseudomonas aeruginosa and Acinetobacter baumanii have emerged as significant Gram-negative bacterial pathogens, presenting multi-drug resistant strains and intrinsic antibiotic resistance. Numerous life-threating infections are attributed to P. aeruginosa and A. baumanii. In one example, these nosocomial pathogens cause outbreaks in hospitals all over the world, colonizing patients in dialysis units, neonatal units, hematology/oncology wards, and liver transplant units. The current arsenal of drugs is not sufficient to treat these infections.
  • the present disclosure shows, inter alia, that the human opportunistic pathogen Pseudomonas aeruginosa catabolizes the anthranilate analog 2-amino-6- fluorobenzoic acid (6-FABA) to 3-fluorocatechol (FCAT) and then to 2-fluoro-cis,cis- muconate (FMUC).
  • 6-FABA 2-amino-6- fluorobenzoic acid
  • FCAT 3-fluorocatechol
  • FMUC 2-fluoro-cis,cis- muconate
  • the present application provides halogenated compounds, including halogenated derivatives of catechol, that are potent anti- infectives that show in vivo antibacterial activity at concentrations ⁇ 20% of their in vitro minimal inhibitory concentrations (MIC).
  • MIC in vitro minimal inhibitory concentrations
  • gentamicin and other traditional antibiotics are only active in vivo at concentrations five to ten times higher than their in vitro MICs, suggesting that the mode of action of the compounds of the present disclosure is not mere inhibition of bacterial growth.
  • the present application provides antibacterial compounds that efficiently block bacterial virulence. This is despite the fact that some of the exemplified compounds have unfavorably high in vitro MICs.
  • these compounds include ribavirin, which was previously approved by FDA as an antiviral medication used to treat RSV infection, hepatitis C, and viral hemorrhagic fever.
  • ribavirin which was previously approved by FDA as an antiviral medication used to treat RSV infection, hepatitis C, and viral hemorrhagic fever.
  • the results presented herein shown that the effect of ribavirin against P. aeruginosa was comparable to that of the broad-spectrum, high-potency carbapenem antibiotic meropenem, and that ribavirin was very efficacious against a meropenem-resistant baumanii strain.
  • the present application provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound selected from the group consisting of:
  • the present disclosure provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound selected from the group consisting of:
  • the present disclosure provides a method of treating a bacterial infection comprising administering to a subject in need thereof a
  • a 2-amino-6-fluorobenzoic acid (6-FABA) having the following structure:
  • bacterial infection is caused by a bacterium that does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
  • QS quorum sensing
  • HAQ 4-hydroxy-2-alkylquinoline
  • the present disclosure provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a thera eutically effective amount of a compound selected from:
  • the bacterial infection is caused by Gram-positive bacteria. In some embodiments, the bacterial infection is caused by Gram-negative bacteria.
  • the bacterial infection is caused by an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or
  • the ESKAPE pathogen is selected from P. aeruginosa and baumannii. In some embodiments, the ESKAPE pathogen is P. aeruginosa. In some embodiments, the ESKAPE pathogen is baumannii. In some embodiments, the baumannii strain is meropen em -resistant. In some embodiments, the bacterial infection is caused by a bacterium selected from the group consisting of:
  • the bacterial infection is selected from the group consisting of: nosocomial infection, skin infection, respiratory infection, wound infection, endovascular infection, CNS infection, abdominal infection, blood stream infection, urinary tract infection, pelvic infection, invasive systemic infection, gastrointestinal infection, dental infection, zoonotic infection, and connective tissue infection.
  • the bacterial infection is selected from the group consisting of: atopic dermatitis, sinusitis, food poisoning, abscess, pneumonia, meningitis, osteomyelitis, endocarditis, bacteremia, sepsis, and urinary tract infection.
  • the compound is administered to the subject by a route selected from the group consisting of: oral, sublingual, gastrointestinal, rectal, topical, intradermal, subcutaneous, nasal, intravenous, and intramuscular.
  • the subject has been identified as having a lung disease.
  • the lung disease is a structural lung disease.
  • the lung disease is selected from the group consisting of: cystic fibrosis, bronchiectasis, emphysema, and chronic obstructive pulmonary disease, and bronchiectasis.
  • the lung disease is cystic fibrosis.
  • the subject is pediatric.
  • the compound is administered to the subject in combination with at least one additional therapeutic agent.
  • the additional therapeutic agent is an antibiotic.
  • the antibiotic is selected from the group consisting of: a quinolone, a ⁇ -lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof.
  • the aminoglycoside antibiotic is tobramycin.
  • the compound and the additional therapeutic agent are administered consecutively. In some embodiments, the compound and the additional therapeutic agent are administered simultaneously.
  • the therapeutically effective amount of the compound is in a range of about 4 mg/kg to about 45 mg/kg.
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
  • bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
  • QS quorum sensing
  • HAQ 4-hydroxy-2-alkylquinoline
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a 2-amino-6-fluorobenzoic acid (6-FABA) having the following structure:
  • bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
  • QS quorum sensing
  • HAQ 4-hydroxy-2-alkylquinoline
  • the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from:
  • the present disclosure provides a method of inhibiting inosine-monophosphate dehydrogenase (EVIPDH) in a bacteria, the method comprising contacting the bacteria with a compound selected from:
  • bacteria is Gram-positive. In some embodiments, the bacteria is Gram-negative.
  • the bacteria is an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or Enter obacter).
  • the bacteria e.g., ESKAPE pathogen
  • the bacteria is selected from P. aeruginosa and baumannii.
  • he ESKAPE pathogen is J 3 , aeruginosa.
  • the ESKAPE pathogen is A. baumannii.
  • the baumannii strain is meropenem-resistant.
  • the bacteria is selected from the group consisting of: S. aureus and E. faecalis.
  • the bacteria is contacted in vitro. In some embodiments, the bacteria is contacted in vitro.
  • the bacteria is contacted in vivo. In some embodiments, the bacteria is contacted ex vivo. In some embodiments, the effective amount of the compound is at least 20% less than MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
  • the effective amount of the compound is about 5-fold lower than MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
  • the effective amount of the compound is about 30 to about 300 times below the MIC of the test compound determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound selected from:
  • the pharmaceutical composition comprises at least one additional therapeutic agent.
  • the additional therapeutic agent is an antibiotic.
  • the additional therapeutic agent is an antibiotic selected from the group consisting of: a quinolone, a ⁇ -lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof.
  • the aminoglycoside is tobramycin.
  • FIG. 1 is a line plot showing that 6-F ABA blocks PA14-mediated killing of C. elegans. Wild-type N2 worms fed E. coli OP50 or PA14 + 6-F ABA at different concentrations.
  • FIG. 2 is a line plot showing that 6-F ABA decreases the ability of a PA14 pqsA mutant to kill C. elegans.
  • FIG. 3 is a line plot showing that m59, a specific MvfR inhibitor, blocks PA14-mediated killing of C. elegans similarly to PA14 mvfl or pqsA mutants.
  • FIG. 4 is a line plot showing that tryptophan does not enhance the ability of
  • FIG. 5 is a line plot showing that VAA AtrpE AtrpG AphnA (tryptophan auxotroph) is not blind to FAB A.
  • FIG. 6 is a line plot showing that 6-FABA significantly extends the lifespan of a C. elegans fer-15, fem-1 (FF) sterile mutant.
  • FIG. 7 is a line plot showing that FABA's effect is not likely due to generating reactive oxygen species: P. aeruginosa catalase or superoxide dismutase mutants respond to FABA similarly to wild-type PA14.
  • FIG. 8 is a line plot showing that. 6-FABA has a much more dramatic effect on blocking PA14-mediated killing than mutating mvfli.
  • FIG. 9 is a line plot showing that m50, a specific MvfR inhibitor, blocks PA14-mediated killing of C. elegans similarly to PA14 mvfl or pqsA mutants.
  • FIG. 10 is an image showing that 6-FABA is metabolized to a brown pigment by PA14.
  • FIG. 11 is an image showing that antA, antB and antC mutations block 6- FABA catabolism to brown pigment.
  • FIG. 12 is a scheme showing simplified anthranilate and 6-FABA catabolic pathways.
  • FIG. 13 is a line plot showing that a PA14 antA mutant is only modestly rescued by 6-FABA, similar to the extent of killing observed with mvfR in the absence of 6-FABA.
  • FIG. 14 is a line plot showing that a PA14 antC mutant is only modestly rescued by 6-FABA, similar to the extent of killing observed with mvfR in the absence of 6-FABA.
  • FIG. 15 is a line plot showing that FCAT rescues PA14 antA and PA14 mvfR- mediated killing to a significantly greater extent than a PA14 mvfR mutant without FCAT.
  • FIG. 16 is a scheme showing catabolism of 6-FABA.
  • FIG. 17 is a line plot showing dose response growth curves of PA14 in different concentrations of FCAT. Rate of growth is not affected at 1 mM, but bacteria are killed at 4 mM.
  • FIG. 18 is a line plot showing that rates of growth of both WT PA14 and
  • PA14 catA are not affected at 1 mM FCAT, but bacteria are killed at 4 mM, showing that the killing effect is not due to FMUC, but rather most likely due to a high concentration of catechols (which generate reactive oxygen species).
  • FIG. 19 is a line plot showing dose response curves of PA14-mediated killing of C. elegans in different concentrations of FCAT.
  • FIG. 20 is a line plot showing comparison of the abilities of various catechol derivatives and 6-FABA to block the ability of P. aeruginosa PA14 to kill C. elegans.
  • FIG. 21 is a line plot showing that double-substituted catechol derivatives also block the ability of PA14 to kill C. elegans.
  • FIG. 22 is a line plot showing that mutation of P. aeruginosa PA14 catA attenuates the ability of FCAT to rescue C. elegans from P. aeruginosa PA14- mediated killing, suggesting that FCAT-mediated rescue of PA14 killing of C.
  • FIG. 23 is a line plot showing that mutation of catB does not affect the ability of FCAT to rescue C. elegans from P. aeruginosa PA14-mediated killing.
  • FIG. 24 is a line plot showing that mutation of PA2682 does not affect the ability of FCAT to rescue C. elegans from P. aeruginosa PA14-mediated killing. Several mutant alleles corresponding to 5 independent PA2682 transposon mutations were tested.
  • FIG. 25 is a line plot showing that FCAT does not rescue C. elegans from Enterococcus faecalis-mediated killing; and that bromopyruvate rescues C. elegans from Enterococcus faecalis-mediated killing.
  • FIG. 26 is a line plot showing that FCAT does not rescue C. elegans from
  • Staphylococcus aureus-mediated killing and that bromopyruvate rescues C. elegans from Enterococcus faecalis-mediated killing.
  • FIG. 27 is a line plot showing that immunocompromised worms (pmk-1 and fshr-1) are rescued by FCAT from P. aeruginosa PA14-mediated killing.
  • FIG. 28 is a line plot showing that immunocompromised worms (zip-2) are rescued by FCAT from P. aeruginosa PA14-mediated killing.
  • FIG. 29 is a line plot showing that FCAT attenuates the virulence of the PA14 quorum-sensing mutants: mvfR, lasR and the double mvfR;lasR mutant.
  • FIG. 30 is a line plot showing that the effect of FCAT (at the same dose) is time-dependent, suggesting the compound is converted relatively slowly to FMUC.
  • FIG. 31 is a line plot showing that MICs are not always good indicators of in vivo efficacy: carbenicillin and gentamicin are dosed at IX, 10X and 50X MIC, respectively. FCAT was dosed at 0.2X and IX MIC.
  • FIG. 32 is a line plot showing that bromopyruvate blocks PA14 virulence, similarly to FCAT.
  • FIG. 33 is a line plot showing that the alkylating agent iodoacetamide (IAM) is also a potent inhibitor of P. aeruginosa PA14 virulence.
  • IAM alkylating agent iodoacetamide
  • FIG. 34 is a bar graph showing that ribavirin is active against P. aeruginosa in a mouse thigh model.
  • FIG. 35 is a line plot showing that ribavirin is active against an ESBL expressing baumannii strain in a mouse thigh model.
  • FIG. 36 is a line plot showing that 4-hydroxy-3-nitrophenylacetic acid (40H) and nisoldipine exhibited activity against PA14 in the C. elegans infection model.
  • FIG. 37 is a line plot showing that ribavirin and mechlorethamine exhibited dose-dependent activity against PA14 infection in the C. elegans infection model.
  • FIG. 38 is a line plot showing percent survival of C. elegans worms in response to treatment with various concentrations of FCAT, BP, ribavirin, and mechlorethamine HC1.
  • FIG. 39 contains (A) a scheme showing nucleotide metabolism; and (B) heat charts showing the effect of adenosine (upper, A) and guanosine (G) against ribavirin.
  • FIG. 40 contains a scheme showing the mechanism of ribavirin's anti -viral activity.
  • FIG. 41 contains (A) a predicted 153AA C-terminal kinase domain of
  • PA14_62230 using the Phyre 2 server; (B) a heat chart showing bacterial growth; and (C) a line plot showing survival of C. elegans on PA14 wild-type and two transposon insertion mutants in PA14 62230.
  • FIG. 42 is a line plot showing PA14 wildtype (PA14) and 3 ribavirin resistant mutants (RR1, RR2, and RR3) in the C. elegans assay.
  • a cornerstone assay in canonical antibiotic discovery is to evaluate putative antimicrobials in a growth inhibition (also called minimum inhibitory concentration, MIC) assay. These assays are typically carried out in a media that contains amino acids, nucleic acids, lipids, and carbohydrates, and that promotes rapid bacterial growth. In a typical growth inhibition assay, potent antibiotic molecules must be highly toxic to bacteria. That is, the compound must block the ability of the bacteria to utilize a wide range of nutrients.
  • MIC minimum inhibitory concentration
  • a compound that may not be sufficiently toxic to bacteria in a growth inhibition assay may still be useful for treating a bacterial infection, for example, by inhibiting bacterial virulence. That is, a compound that does not affect essential bacterial processes required in vitro for growth, and that does not have antimicrobial activity against free-living planktonic bacteria in vitro, may nevertheless target bacterial virulence or growth in vivo during an infection, for example, by modulating quorum sensing signaling pathways, pathogen-synthesized toxins, or other types of virulence-related factors.
  • the virulence blockers may be suitable to treat persisting pathogen populations that are hard to eradicate with conventional antibiotics.
  • the present application provides a method of inhibiting virulence of bacteria, the method comprising contacting the bacteria with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof.
  • the effective amount of the compound is at least about 20%, about 30%), about 40%, or about 50% less than the MIC of the same compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
  • the effective amount of the compound is about 2-fold, about 5-fold, about 10-fold, or about 15-fold lower than the MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
  • the compound is 2-amino-6-fluorobenzoic acid (6- FABA):
  • the compound is selected from:
  • the com ound is ribavirin:
  • the com ound is ribavirin-5'-monophosphate: or a pharmaceutically acceptable salt thereof.
  • the compound is nisoldipine:
  • the compound is cr v v i
  • the compound is obatoclax:
  • the com ound is chlorpromazine:
  • the compound is HMS3408N17 (l'-([l, l'-biphe yl)-[l,4'-bipiperidin]-4-amin
  • the compound is MolPort-008-370-584 ( ⁇ -(l-p' fluoro-[l, -biphenyl]-4-yl)piperidin-4-yl)propane-l,3-diamine):
  • the compound is dimetndazole:
  • the compound is mangafodipir sodium:
  • the compound is otenzepad:
  • the compound is ZM 39923 :
  • the compound is PPT:
  • the compound is 4-hydroxy-3-nitrophenylacetic acid:
  • the com ound is clonidine:
  • the compound is betonicine:
  • the com ound is clotrimazole:
  • the compound is ammonium
  • the compound is otilonium bromide:
  • the com ound is FPA 124:
  • the compound is HMS3604J07 ((E)-3-bromo-N'-((3,5- dibromo-4-hydroxy-6-oxocyclohexa-2,4-dien-l-ylidene)methyl)benzohydrazide):
  • the com ound is lomofungin:
  • a salt (e.g., pharmaceutically acceptable salt) of a compound of the present disclosure is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
  • the compound is a pharmaceutically acceptable acid addition salt.
  • acids commonly employed to form pharmaceutically acceptable salts of the compounds of the present disclosure include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para- toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids.
  • inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenyl acetate, pheny
  • pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid.
  • bases commonly employed to form pharmaceutically acceptable salts of the compounds of the present disclosure include hydroxides of alkali metals, including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydroxyl- substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine;
  • piperidine pyrrolidine
  • amino acids such as arginine, lysine, and the like.
  • a compound may inhibit bacterial virulence by inhibiting inosine- monophosphate dehydrogenase (IMPDH) in a bacteria.
  • IMPDH inosine- monophosphate dehydrogenase
  • the present disclosure provides a method of inhibiting inosine-monophosphate dehydrogenase (IMPDH) in a bacteria, the method comprising contacting the bacteria with an effective amount of a compound as described herein.
  • the compound is ribavirin or ribavirin 5 '-phosphate.
  • the bacteria is at least 2-fold, 4-fold, 8-fold, 10-fold, 24-fold, 48-fold, 100-fold, 256-fold, 512-fold or 1000-fold resistant to one or more of other antibiotic agents.
  • the bacteria is multi-drug resistant (MDR).
  • MDR multi-drug resistant
  • the bacteria is resistant to methicillin, vancomycin, rifampicin, linezolid, daptomycin, gentamicin and/or ciprofloxacin.
  • the bacteria uses quorum sensing (QS).
  • QS quorum sensing
  • the bacteria uses quorum sensing mediated by N-acyl-homoserine lactone (AHL) (e.g., when the bacteria is Gram-negative bacteria), or by an autoinducing peptide (e.g., when the bacteria is Gram-negative bacteria).
  • AHL N-acyl-homoserine lactone
  • the bacteria does not use quorum sensing activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ), such as 3,4-dihydroxy-2-heptylquinoline (PQS), 4-hydroxy-2-heptylquinoline, or 3,4-dihydroxy-2-nonylquinoline.
  • HAQ 4-hydroxy-2-alkylquinoline
  • PQS 3,4-dihydroxy-2-heptylquinoline
  • PQS 4,hydroxy-2-heptylquinoline
  • 3,4-dihydroxy-2-nonylquinoline 3,4-dihydroxy
  • the bacteria is Gram-positive bacteria.
  • the bacteria is a member of a genus selected from the group consisting of Staphylococcus (including coagulase negative and coagulase positive), Streptococcus, Peptococcus, Enterococcus, and Bacillus.
  • the bacteria is a member of the Staphylococcus genus and the species of bacteria is selected from the group consisting of S. aureus, methicillin-susceptible S. aureus ⁇ SSA), coagulase negative staphylococci, methicillin-resistant S. aureus (MRSA), vancomycin-resistant S. aureus (VRSA), S. arlettae, S. agnetis, S. auricularis, S. capitis, S. caprae, S. carnosus, S. caseolyticus, S. chromogenes, S. cohnii, S. condimenti, S. delphini, S. devriesei, S. epidermidis, S.
  • S. aureus methicillin-susceptible S. aureus ⁇ SSA
  • MRSA methicillin-resistant S. aureus
  • VRSA vancomycin-resistant S. aureus
  • S. arlettae S. agnetis, S.
  • the bacteria is a member of the Peptococcus genus and the species of bacteria is P. magnus.
  • the bacteria is a member of the Streptococcus genus and the species of bacteria is selected from the group consisting of S. agalactiae, S. anginosus, S. bovis, S. canis, S. constellatus, S. dysgalactiae, S. equinus, S. iniae, S. intermedius, S. milleri, S. mitis, S. mutans, S. oralis, S. parasanguinis, S. peroris, S. pneumoniae, S. pseudopneumoniae, S. pyogenes, S. ratti, S. salivarius, S. tigurinus, S. thermophilus, S. sanguinis, S. sobrinus, S. suis, S. uberis, S. vestibularis, S. viridans, and S. zooepidemicus .
  • the bacteria is a member of the Enterococcus genus and the species of bacteria is selected from the group consisting of E. avium, E. durans, E. faecalis, E. gallinarum, E. haemoperoxidus, E. hirae, E. malodoratus, E. moraviensis, E. mundtii, E. pseudoavium, E. raffinosus, E. solitaries, and E. faecium.
  • the bacteria is a member of the Propionibacterium genus. In such embodiments, the bacteria is P. acnes.
  • the bacteria is a Gram-negative bacteria.
  • the bacteria is a member of a family selected from the group consisting of Enterobacteriaceae, Helicobacter aceae, Campylobacter aceae, Neisseriaceae, Pseudomonadaceae, Moraxellaceae, Xanthomonadaceae,
  • the bacteria is a member of a genus selected from the group consisting of Citrobacter, Enterobacter, Escherichia, Klebsiella, Pantoea, Proteus, Salmonella, Serratia, Shigella, Yersinia, Helicobacter, Wolinella,
  • Campylobacter Arcobacter, Neisseria, Francisella, Pseudomonas, Acinetobacter, Moraxella, Stenotrophomonas, Haemophilus, Pasteurella, and Legionella.
  • the bacteria is a member of the Citrobacter genus and the species of bacteria is selected from the group consisting of C. amalonaticus, C braakii, C. diver sus, C. farmer, C. freundii, C. gillenii, C. koseri, C. murliniae, C. rodentium, C. sedlakii, C. werkmanii, and C. youngae.
  • the bacteria is a member of the Enterobacter genus and the species of bacteria is selected from the group consisting of E. aerogenes, E.
  • amnigenus E. agglomerans, E. arachidis, E. asburiae, E. cancerogenous, E. cloacae, E. cowanii, E. dissolvens, E. gergoviae, E. helveticus, E. hormaechei, E. intermedius, E. kobei, E. ludwigii, E. mori, E. nimipressuralis, E. oryzae, E. pulveris, E. pyrinus, E. radicincitans, E. taylorae, E. turicensis, E. sakazakii, and E. spp.
  • the bacteria is a member of the Escherichia genus and the species of bacteria is selected from the group consisting of E. albertii, E. blattae, E. coli, E. fergusonii, E. hermannii, and E. vulneris.
  • the bacteria is a member of the Klebsiella genus and the species of bacteria is selected from the group consisting of K. granulomatis, K. oxytoca, K. pneumoniae, K. terrigena, and K. planticola.
  • the bacteria is a member of the Pantoea genus and the species of bacteria is selected from the group consisting of P. agglomerans, P.
  • the bacteria is a member of the Proteus genus and the species of bacteria is selected from the group consisting of P. hauseri, P. mirabilis, P. myxofaciens, P. penneri, and P. vulgaris.
  • the bacteria is a member of the Salmonella genus and the species of bacteria is selected from the group consisting of S. bongori, and S. enterica.
  • the bacteria is a member of the Serratia genus and the species of bacteria is selected from the group consisting of S. entomophila, S. ficaria, S. fonticola, S. grimesii, S. liquefaciens, S. marcescens, S. odorifera, S. plymuthica, S. proteamaculans, S. quinivorans, S. rubidaea, and S. symbiotica.
  • the bacteria is a member of the Shigella genus and the species of bacteria is selected from the group consisting of S. boydii, S. dysenteriae, S. flexneri, and S. sonnei.
  • the bacteria is a member of the Yersinia genus and the species of bacteria is selected from the group consisting of Y pestis, Y
  • the bacteria is a member of the Helicobacter genus and the species of bacteria is selected from the group consisting of H. acinonychis, H. anseris, H. aurati, H. baculiformis, H. bilis, H. bizzozeronii, H. brantae, H.
  • canadensis H. canis, H. cetorum, H. cholecystus, H. cinaedi, H. cynogastricus, H. equorum, H. felis, H. fennelliae, H. ganmani, H. heilmannii, H. hepaticus, H.
  • mesocricetorum H. macacae, H. marmotae, H. mastomyrinus, H. mesocricetorum, H. muridarum, H. mustelae, H. pametensis, H. pullorum, H. pylori, H. rappini, H.
  • the bacteria is a member of the Campylobacter genus and the species of bacteria is selected from the group consisting of C. avium, C.
  • cryaerophilus C. cuniculorum, C. curvus, C. fennelliae, C. fetus, C. gracilis, C.
  • the bacteria is a member of the Arcobacter genus and the species of bacteria is selected from the group consisting of bivalviorum, A. butzleri, A. cibarius, A. cryaerophilus, A. defluvii, A. ellisii, A. halophilus, A. marinus, A. molluscorum, A. mytili, A. nitrofigilis, A. skirrowii, A. thereius, A. trophiarum, and A. venerupis.
  • the bacteria is a member of the Neisseria genus and the species of bacteria is selected from the group consisting of N. bacilliformis, N.
  • cinerea N. denitrificans, N. elongata, N. flavescens, N. gonorrhoeae, N. lactamica, N. macacae, N. meningitidis, N. mucosa, N. pharyngis, N. polysaccharea, N. sicca, N. subflava, and N. weaver.
  • the bacteria is a member of the Francisella genus and the species of bacteria is selected from the group consisting of F. tularensis, F novicida, F hispaniensis, W. persica, F noatunensis, F philomiragia, F halioticida, F endociliophora, and F guangzhouensis .
  • the bacteria is a member of the Pseudomonas genus and the species of bacteria is selected from the group consisting of P. aeruginosa, P. oryzihabitans, and ! plecoglossicida. In some embodiments, the bacteria is a member of the Acinetobacter genus and the species of bacteria is baumannii.
  • the bacteria is a member of the Moraxella genus and the species of bacteria is selected from the group consisting ofM catarrhalis, M. lacunata, and M bovis.
  • the bacteria is a member of the Stenotrophomonas genus and the species of bacteria is S. maltophilia.
  • the bacteria is a member of the Haemophilus genus and the species of bacteria is selected from the group consisting of H aegyptius, H aphrophilus, H. avium, H. ducreyi, H. felis, H. haemolyticus, H. influenzae, H.
  • the bacteria is a member of the Pasteurella genus and the species of bacteria is selected from the group consisting of P. multocida, P.
  • the bacteria is a member of the Legionella genus and the species of bacteria is selected from the group consisting of L. pneumophila, L. anisa, L. bozemanae, L. multiplinnatiensis, L. gormanii, L. jordani, L. longbeachae, L. maceachernii, L. micdadei, L. sainthelensi, L. wadsworthii, and L. waltersii.
  • the bacteria is a member of the Mycobacterium genus and the species of bacteria is selected from a group consisting ofM tuberculosis and M. smegmatic.
  • the bacteria is a member of a genus selected from: Acinetobacter, Burkholderia, Acinetobacter, Burkholderia, Klebsiella, Pseudomonas, and Escherichia.
  • the bacteria is a member of a species selected from: K pneumoniae, P. aeruginosa, Enterobacteriaceae, a d E. coli.
  • the bacteria is an ESKAPE pathogen (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacte ). Treating bacterial infections
  • the present disclosure provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof.
  • the bacterial infection is caused by any one of the bacteria described herein.
  • the bacterial infection is resistant to treatment with one or more of the antibiotic agents described herein (e.g., bacterial infection is resistant to treatment with methicillin, vancomycin, rifampicin, gentamicin and/or ciprofloxacin).
  • the bacterial infection is characterized as resistant to treatment with one or more available antibiotic agents.
  • the bacterial infection is a skin infection.
  • the skin infection is selected from the group consisting of acne, pimples, impetigo, boils, cellulitis, folliculitis, carbuncles, scalded skin syndrome, skin abscesses, atopic dermatitis, and typhoid fever.
  • the bacterial infection is a skin infection caused by P. acnes.
  • the skin infection is acne.
  • the bacterial infection is a skin and soft tissue infection (e.g., acne).
  • the bacterial infection is a respiratory infection.
  • the respiratory infection is selected from the group consisting of upper respiratory tract infection, bronchopneumonia, atypical pneumonia, tuberculosis, mycobacterium tuberculosis, pneumonia, anaerobic pleuropulmonary infection, ventilator-associated pneumonia, aspiration pneumonia, lung abscess, bronchitis, chronic obstructive pulmonary disease, obstructive pulmonary disease, Pontiac fever, and legionellosis.
  • the bacterial infection is a wound infection. In some aspects of these embodiments, the wound infection is a postsurgical wound infection. In some embodiments, the bacterial infection is a blood stream infection. In some aspects of these embodiments, the blood stream infection is bacteremia or sepsis. In some embodiments, the bacterial infection is a pelvic infection. In some aspects of the embodiments, the pelvic infection is bacterial vaginosis.
  • the bacterial infection is a gastrointestinal infection.
  • the gastrointestinal infection is selected from the group consisting of peptic ulcer, chronic gastritis, duodenitis, gastroenteritis, diarrhea, dysentery, diphtheria, food poisoning and foodborne illness.
  • the bacterial infection is a bone, joint or muscle infection.
  • the bone, joint or muscle infection is selected from the group consisting of tetanus, secondary meningitis, meningitis, neonatal meningitis, sinusitis, laryngitis, arthritis, septic arthritis, Bartholin gland abscess, chancroid, osteomyelitis, endocarditis, mediastinitis, pericarditis, peritonitis, otitis media, blepharoconjunctivitis, keratoconjunctivitis, and conjunctivitis.
  • the joint infection is an infection of a shoulder, a knee, a hip, or an elbow.
  • the bacterial infection is septic arthritis (e.g., septic arthritis caused by P. acnes or septic arthritis caused by S. aureus).
  • the bacterial infection is selected from the group consisting of a dental infection, a zoonotic infection, an invasive systemic infection, a urinary tract infection, an abdominal infection, a CNS infection, an endovascular infection, a connective tissue infection, and a nosocomial infection.
  • the bacterial infection is selected from the group consisting of syphilis, leprosy, abscesses, sepsis, empyema, and tularemia.
  • the bacterial infection is associated with implanted devices (e.g., catheter, ballon catheter, stent, pacer etc).
  • the bacterial infection is osteomyelitis, endocarditis, or an infection associated with an implanted device, which is caused by a S. aureus persister, P. acnes, P. aeruginosa, or
  • a composition of the present application further comprises one or more additional therapeutic agents.
  • the additional therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound of the present disclosure.
  • the second therapeutic agent is a virulence blocker. In some embodiments, the second therapeutic agent is m50 or m59, or a
  • a pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof, also optionally contains at least one additional therapeutic agent, or a pharmaceutically acceptable salt thereof.
  • the additional therapeutic agent in the composition is any one of the antibiotics described herein (e.g., gentamicin or defensin 1).
  • the second therapeutic agent may be present in the composition in a therapeutically effective amount.
  • an effective amount of the additional therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent. For example, an effective amount is between about 70% and 100% of the normal monotherapeutic dose.
  • the effective amount of gentamicin is lower than the amount that causes
  • a method of treating a subject in need thereof as disclosed herein comprises administering to the subject one or more additional therapeutic agents.
  • the additional therapeutic agent may be administered to the subject in a separate pharmaceutical composition or dosage form (e.g., any one of the compositions, formulation, routes and dosage forms described herein).
  • a compound as provided herein, or a pharmaceutically acceptable salt thereof can be used in combination with an antibiotic.
  • a compound as provided herein, or a pharmaceutically acceptable salt thereof can be used in combination with a cationic antimicrobial peptide (CAMP).
  • the cationic antimicrobial peptide is a defensin peptide (e.g., defensin 1 such as beta-defensin 1 or alpha- defensin 1), or cecropin, andropin, moricin, ceratotoxin, melittin, magainin, dermaseptin, bombinin, brevinin (e.g., brevinin-1), esculentin, buforin II (e.g., from amphibians), CAP18 (e.g., from rabbits), LL37 (e.g., from humans), abaecin, apidaecins (e.g., from honeybees), prophenin (e.g., from pigs), indolicidin (e.g.,
  • defensin 1 such as beta
  • the antibiotic is selected from the quinolone class of antibiotic compounds.
  • the antibiotic is selected from the group consisting of levofloxacin, norfloxacin, ofloxacin, ciprofloxacin, perfloxacin, lomefloxacin, fleroxacin, sparfloxacin, grepafloxacin, trovafloxacin, clinafloxacin, gemifloxacin, enoxacin, sitafloxacin, nadifloxacin, tosulfloxacin, cinnoxacin, rosoxacin, miloxacin, moxifloxacin, gatifloxacin, cinnoxacin, enoxacin, fleroxacin, lomafloxacin, lomefloxacin, miloxacin, nalidixic acid, nadifloxacin, oxolinic acid, pefloxacin, pirimidic
  • the antibiotic is selected from a ⁇ -lactam, a
  • the antibiotic is selected from the cephalosporin class of antibiotic compounds.
  • the antibiotic is selected from the group consisting of cefazolin, cefuroxime, ceftazidime, cephalexin, cephaloridine, cefamandole, cefsulodin, cefonicid, cefoperazine, cefoprozil, and ceftriaxone.
  • the antibiotic is selected from the penicillin class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of penicillin G, penicillin V, procaine penicillin, and benzathine penicillin, ampicillin, and amoxicillin, benzylpenicillin,
  • phenoxymethylpenicillin oxacillin, methicillin, dicloxacillin, flucloxacillin, temocillin, azlocillin, carbenicillin, ricarcillin, mezlocillin, piperacillin, apalcillin, hetacillin, bacampicillin, sulbenicillin, mecicilam, pevmecillinam, ciclacillin, talapicillin, aspoxicillin, cloxacillin, nafcillin, and pivampicillin.
  • the antibiotic is selected from the carbapenem class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of thienamycin, tomopenem, lenapenem, tebipenem, razupenem, imipenem, meropenem, ertapenem, doripenem, panipenem (betamipron), and biapenem. In some embodiments, the antibiotic is selected from the lipopeptide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of polymyxin B, colistin (polymyxin E), and daptomycin.
  • the antibiotic is selected from the aminoglycoside class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of gentamicin, amikacin, tobramycin, debekacin, kanamycin, neomycin, netilmicin, paromomycin, sisomycin, spectinomycin, and streptomycin.
  • the antibiotic is selected from the glycopeptide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of vancomycin, teicoplanin, telavancin,
  • the antibiotic is selected from the macrolide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of azithromycin, clarithromycin, erythromycin, fidaxomicin, telithromycin, carbomycin A, josamycin, kitasamycin,
  • oleandomycin midecamycin/midecamycinacetate, oleandomycin, solithromycin, spiramycin, troleandomycin, tylosin/tylocine, roxithromycin, dirithromycin, troleandomycin, spectinomycin, methymycin, neomethymycin, erythronolid, megalomycin, picromycin, narbomycin, oleandomycin, triacetyl-oleandomycin, laukamycin, kujimycin A, albocyclin and cineromycin B.
  • the antibiotic is selected from the ansamycin class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of streptovaricin, geldanamycin, herbimycin, rifamycin, rifampin, rifabutin, rifapentine and rifamixin.
  • the antibiotic is selected from the sulfonamide class of antibiotic compounds.
  • the antibiotic is selected from the group consisting of sulfanilamide, sulfacetamide, sulfapyridine, sulfathiazole, sulfadiazine, sulfamerazine, sulfadimidine, sulfasomidine, sulfasalazine, mafenide, sulfamethoxazole, sulfamethoxypyridazine, sulfadimethoxine,
  • the antibiotic is selected from the group consisting of quinolones, fluoroquinolones, ⁇ -lactams, cephalosporins, penicillins, carbapenems, lipopeptide antibiotics, glycopeptides, macrolides, ansamycins, sulfonamides, and combinations of two or more thereof.
  • the present application provides separate dosage forms of a compound described herein, or a pharmaceutically acceptable salt thereof, and one or more of any of the above-described second therapeutic agents.
  • the separate dosage forms may be administered together consecutively (e.g., within less than 24 hours of one another) or simultaneously (e.g., administered to the patient within 5 minutes of one another).
  • compositions comprising, formulations, and routes of administration
  • the present application also provides pharmaceutical compositions comprising an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
  • the carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of the present application include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, di sodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances,
  • polyethylene glycol sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
  • solubility and bioavailability of the compounds of the present application in pharmaceutical compositions may be enhanced by methods well-known in the art.
  • One method includes the use of lipid excipients in the formulation. See “Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water- Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley-Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of the present application optionally formulated with a poloxamer, such as LUTROLTM and PLURONICTM (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • compositions of the present application include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compound of the present disclosure is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques).
  • Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • compositions of the present application suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc.
  • Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • compositions suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3- butanediol.
  • the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • compositions of the present application may be administered in the form of suppositories for rectal administration.
  • compositions can be prepared by mixing a compound of the present application with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax, and polyethylene glycols.
  • compositions of the present application may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, for example, U.S. Patent No. 6,803,031.
  • Topical administration of the pharmaceutical compositions of the present application is especially useful when the desired treatment involves areas or organs readily accessible by topical application (e.g., skin and soft tissues).
  • the topical compositions of the present disclosure can be prepared and used in the form of an aerosol spray, cream, emulsion, solid, liquid, dispersion, foam, oil, gel, hydrogel, lotion, mousse, ointment, powder, patch, pomade, solution, pump spray, stick, towelette, soap, or other forms commonly employed in the art of topical administration and/or cosmetic and skin care formulation.
  • the topical compositions can be in an emulsion form, as a cream or a paste.
  • the topical composition comprises a combination of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and one or more additional ingredients, carriers, excipients, or diluents including, but not limited to, absorbents, anti-irritants, anti-acne agents, preservatives, antioxidants, coloring agents/pigments, emollients (moisturizers), emulsifiers, film-forming/holding agents, fragrances, leave-on exfoliants, prescription drugs, preservatives, scrub agents, silicones, skin-identical/repairing agents, slip agents, sunscreen actives,
  • additional ingredients, carriers, excipients, or diluents including, but not limited to, absorbents, anti-irritants, anti-acne agents, preservatives, antioxidants, coloring agents/pigments, emollients (moisturizers), emulsifiers, film-forming/holding agents, fragrances, leave-on exfoliants, prescription drugs, preservatives
  • diluents, carriers, and excipients may include, but are not limited to, polyethylene glycols (such as PEG200, PEG300, PEG400, PEG540, PEG600, PEG1450 or mixtures thereof) and coconut oils (such as propylene glycol dicaprate, coco-caprylate/caprate, propylene glycol dicaprylate/dicaprate, caprylic/capric triglyceride, caprylic/capric/lauric triglyceride, caprylic/capric/linoleic triglyceride, tricaprin, tricaprylin, glyceryl trioleate, neopentyl glycol dicaprylate/dicaprate, caprylic/capric/palmitic/stearic triglceride, or mixtures thereof).
  • polyethylene glycols such as PEG200, PEG300, PEG400, PEG540, PEG600, PEG1450 or mixtures thereof
  • coconut oils such as prop
  • suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water.
  • preservatives may include, but are not limited to, 1,2-hexanediol, benzoic acid, benzothonium chloride, borax, bronopol, butylparaben, caprylyl glycol, chlorophene, chloroxylenol, chlorphenesin, dehydroacetic acid, diazolidinyl urea, DMDM hydantoin, ethylhexylglycerin, ethylparaben, formaldehyde-releasing preservative, Germaben II, hoelen, imidazolidinyl urea, iodopropynyl butylcarbamate,
  • phenoxyisopropanol phytosphingosine, polyaminopropyl biguanide, potassium sorbate, propylparaben, quaternium-15, sodium benzoate, sodium citrate, sodium dehydroacetate, sodium hexametaphosphate, sodium hydroxymethylglycinate, sodium lactobionate, sodium metabi sulfite, sodium sulfite, sorbic acid, and styrax benzoin.
  • slip agents may include, but are not limited to, amodimethicone, fos-PEG-18 methyl ether dimethyl silane, fos-phenylpropyl dimethicone, butylene glycol, cetyl dimethicone, cetyl dimethicone copolyol, cetyl PEG/PPG- 10/1- dimethicone, cyclohexasiloxane, cyclomethicone, cyclopentasiloxane,
  • cyclotetrasiloxane decylene glycol, diisostearoyl trimethylolpropane siloxy silicate, dimethicone, dimethicone copolyol, dimethicone crosspolymer, dimethiconol, dipropylene glycol, hexylene glycol, hydrolyzed silk, isododecane, methicone, methyl trimethicone, methylsilanol mannuronate, methylsilanol PEG-7 glyceryl cocoate, PEG- 10 dimethicone, PEG- 10 dimethicone/vinyl dimethicone crosspolymer, PEG- 12 dimethicone, PEG/PPG-18/18 dimethicone, PEG/PPG-20/15 dimethicone, pentylene glycol, phenyl trimethicone, polymethylsilsesquioxane, PPG-3 benzyl ether myristate
  • sunscreen actives may include, but are not limited to, avobenzone, benzephenone-3, benzophenones, bumetrizole, butyl methoxydibenzoylmethane, ecamsule, ensulizole, ethylhexyl methoxycinnamate, homosalate, menthyl anthranilate, meradmiate, Mexoryl SX, octinoxate, octisalate, octocrylene, octyl methoxycinnamate, octyl salicylate, oxybenzone, padimate O, para- aminobenzoic acid (PABA), Parsol 1789, terephthalylidine dicamphor sulfonic acid, Tinosorb M, Tinosorb S, and titanium dioxide.
  • PABA para- aminobenzoic acid
  • emulsifiers, surfactants, and detergents may include, but are not limited to, ammonium laureth sulfate, ammonium lauryl sulfate, arachidyl glucoside, behenic acid, bis-PEG-lS methyl ether dimethyl silane, C20-40 pareth-40, cocamidopropyl betaine,
  • Application of the subject therapeutics may be local, so as to be administered at the site of interest (e.g., infected area of skin, or an infected joint or other connective tissue).
  • site of interest e.g., infected area of skin, or an infected joint or other connective tissue.
  • Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
  • the compounds of the present application may be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, or catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents, or catheters.
  • Suitable coatings and the general preparation of coated implantable devices are known in the art and are exemplified in U.S. Patent Nos. 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccharides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Coatings for invasive devices are to be included within the definition of pharmaceutically acceptable carrier, adjuvant or vehicle, as those terms are used herein.
  • the present application provides a method of coating an implantable medical device comprising the step of contacting said device with the coating composition described above. It will be obvious to those skilled in the art that the coating of the device will occur prior to implantation into a mammal.
  • the present application provides a method of impregnating an implantable drug release device comprising the step of contacting said drug release device with a compound or composition of the present application.
  • Implantable drug release devices include, but are not limited to, biodegradable polymer capsules or bullets, non-degradable, diffusible polymer capsules and biodegradable polymer wafers.
  • the present application provides an implantable medical device coated with a compound or a composition comprising a compound of the present application, such that said compound is therapeutically active.
  • organ or tissue may be bathed in a medium containing a composition of the present application
  • a composition of the present application may be painted onto the organ, or a composition of the present application may be applied in any other convenient way.
  • compositions of the present application a compound of the present disclosure, or a pharmaceutically available salt thereof, is present in an effective amount (e.g., a therapeutically effective amount).
  • Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
  • an effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof can range, for example, from about lmg to about 200 mg, from about 1 to about 100 mg, from about 1 to about 50 mg, from about 1 mg to about 30 mg, from about 1 mg to about 15 mg, from about 10 mg to about 2000 mg, from about 10 mg to about 1900 mg, from about 10 mg to about 1800 mg, from about 10 mg to about 1700 mg, from about 10 mg to about 1600 mg, from about 10 mg to about 1500 mg, from about 10 mg to about 1400 mg, from about 10 mg to about 1300 mg, from about 10 mg to about 1200 mg, from about 10 mg to about 1100 mg, from about 10 mg to about 1000 mg, from 10 mg about to about 900 mg, from about 10 mg to about 800 mg, from about 10 mg to about 700 mg, from about 10 mg to about 600 mg, from about 10 mg to about 500 mg, from about 10 mg to about 400 mg, from about 10 mg to about 300 mg, from about 10 mg to about 200 mg, from
  • pharmaceutically acceptable salt thereof is 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, or 10 mg.
  • an effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof can range, for example, from about 1 mg/kg to about 1000 mg/kg.
  • an effective amount can range from about 1 mg/kg to about 50 mg/kg, about 4 mg/kg to about 45 mg/kg, or about 50 mg/kg to about 500 mg/kg.
  • the composition containing an effective amount of the compound, or a pharmaceutically acceptable salt thereof is administered once daily. In some embodiments, the composition containing an effective amount of the compound, or a pharmaceutically acceptable salt thereof, is administered twice daily. In some embodiments, the composition containing an effective amount of the compound, or a pharmaceutically acceptable salt thereof, is administered thrice daily.
  • Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of administration, and
  • any one of compounds described herein, or a salt thereof may be used inhibiting virulence of bacteria or reducing bacteria growth on a surface (e.g., for disinfecting or sanitizing a surface).
  • the surface may be metallic, plastic, ceramic, or wooden, for example, the surface is a floor, a table, a kitchen counter, a cutting board, or a medical instrument.
  • any one of the compounds of the present application may be used in a commercial setting for general disinfecting, e.g., in medical and food industries.
  • the compound may be provided in a cleaning composition comprising an acceptable carrier.
  • the carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the cleaning composition.
  • Acceptable carriers that may be used in a cleaning composition of the present application include, but are not limited to, alcohols, water, surfactants, emollients, stabilizers, thickeners, viscosifiers, and fragrances.
  • the term “about” means “approximately” (e.g., plus or minus approximately 10% of the indicated value).
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal.
  • an in vitro cell can be a cell in a cell culture.
  • an in vivo cell is a cell living in an organism such as a mammal.
  • the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • contacting" the inosine-monophosphate dehydrogenase (IMPDH) with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having IMPDH, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the IMPDH.
  • the term "subject" does not refer to any animal other than human.
  • the subject is pediatric (e.g., from birth through age 21).
  • the subject has been identified as having a lung disease.
  • the lung disease is a structural lung disease.
  • the lung disease is selected from the group consisting of: cystic fibrosis, bronchiectasis, emphysema, and chronic obstructive pulmonary disease, and bronchiectasis.
  • the lung disease is cystic fibrosis.
  • the phrase “effective amount” or “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • treating refers to 1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder ⁇ i.e., arresting further development of the pathology and/or symptomatology), or 2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
  • preventing or prevention of a disease, condition or disorder refers to decreasing the risk of occurrence of the disease, condition or disorder in a subject or group of subjects (e.g., a subject or group of subjects predisposed to or susceptible to the disease, condition or disorder). In some embodiments, preventing a disease, condition or disorder refers to decreasing the possibility of acquiring the disease, condition or disorder and/or its associated symptoms. In some embodiments, preventing a disease, condition or disorder refers to completely or almost completely stopping the disease, condition or disorder from occurring.
  • resistant and “persistent” refer to bacterial strains that exhibit a high level of tolerance to one or more antibiotics.
  • the bacterial strain is resistant when the MIC of the bacterial strain is at least 2x (2 -fold) of the MIC for the non-resistant strain.
  • the x-fold resistant bacterial strain may be determined by the following steps: (i) MIC is determined for a non-resistant bacterial strain; (ii) the non-resistant bacterial strain is treated in a multi- well plate with an antibiotic at 2x, 5x, lOx etc, of the minimal inhibitory concentration (MIC); (iii) bacterial culture treated with the highest concentration that permitted bacterial growth is taken for serial passage for 100 days; and (iv) MIC of the bacterial culture after 100 days of serial passage is determined. If MIC of the bacterial culture after 100 days of serial passage is at least 2x of the MIC of the non-resistant strain, then the bacterial culture is at least 2x resistant to the antibiotic.
  • MIC minimal inhibitory concentration
  • C. elegans anti -infective activity assay the assay was performed according to the methods and procedures similar to those previously described in Yuen, G.L. and F.M. Ausubel (2016) Both live and dead Enterococci activate Caenorhabditis elegans host defense via immune and stress pathways. Virulence PMID: 29436902. DOI: 10.1080/21505594.2018.1438025; Sifri, CD., J. Begun, F.M. Ausubel and S.B.
  • aeruginosa PA14 overnight cultures are plated on SK agar and incubated at 37 °C for 24 hours.
  • Test compound is added to the agar, after which time plates are incubated at 25 °C for 24 hours.
  • L4 stage C. elegans are transferred onto these plates (30-50 worms per plate) and survival is monitored until all of the worms are dead. Compounds resulting in significantly improved survival compared to mock-treated controls are considered active.
  • C. elegans is a natural bacterivore and can be raised easily in the laboratory by feeding on lawns non-pathogenic strains of Escherichia coli, especially E. coli strain OP50.
  • a large variety of human pathogens kill C. elegans, including ! aeruginosa, Enterococcus faecalis, and S. aureus.
  • P. aeruginosa, E. faecalis, and S. aureus kill C. elegans in liquid medium
  • mice Female 5 to 6-week-old CD-I mice (18 - 22 gm) were used.
  • mice were administered test compounds at times and route of administration as appropriate. There were 3 dose groups for each Test Compound. Mice were euthanized by C0 2 inhalation at 24 hours post infection and thigh samples were taken. Thighs were aseptically removed, placed in 1-2 mL sterile PBS, homogenized, serially diluted and plated to determine CFU counts. Plates were incubated 18-24 hours under the appropriate strain conditions prior to counting. Colony counts were performed on agar plates.
  • the number of colonies were converted to CFU/thigh by multiplying the number of colonies by the volume of the thigh homogenate spotted and the dilution at which the colonies were counted (5-50 colonies/spot). All count data were transformed into loglO CFU/thigh for calculation of means and standard deviations.
  • Example 1 6-fluoroaminobenzoic acid (6-FABA) dramatically attenuates the ability of P. aeruginosa to kill C. elegans.
  • 6-FABA is much more effective at inhibiting P. aeruginosa strain PA14-mediated killing of C. elegans than would be expected if its sole mode of action is to block PQS biosynthesis and MvfR-mediated QS signaling.
  • 6-FAB A inhibits the biosynthesis of PQS, thereby disrupting MvfR-dependent gene expression, and blocks bacterial virulence in a murine thermal injury model of P. aeruginosa infection (See Ref. 53). That is, C. elegans infected with wild-type P. aeruginosa in the presence of 6-FABA survived much longer than C. elegans infected with P. aeruginosa mvfl or pqsA mutants (genes that encode the transcription factor and the first synthetic enzyme in the PQS system, respectively) in the absence of the drug.
  • PA14 P. aeruginosa strain PA14 (hereafter PA14) to kill C. elegans animals
  • PA14 was used in "slow killing" assay as described by Tan et al (See Ref. 47). If 6-FABA completely blocked MvfR-mediated signaling, it would diminish the ability of PA14 to kill similarly to the level of killing that would be observed by infecting C. elegans with PA14 carrying null mutations in mvfl or pqsA.
  • 6-FABA is much more effective at inhibiting ! aeruginosa strain PA14-mediated killing of C. elegans than would be expected if the sole mode of action of 6-FABA is to inhibit the MvfR quorum sensing regulon.
  • 6-FABA did not activate the expression of a panel of C. elegans immune response reporter genes (including the genes daf-16, clec-60, irg-1, T24B8.5, sod-3, gst-4, F35E12.5, hspl6.2), suggesting that it does not target the C. elegans immune system.
  • Fluorinated compounds such as the fluorouracil may cause C. elegans infertility issues (See Ref. 59), and C. elegans sterile mutants or C. elegans animals treated with FUdR are more restistant to bacterial infections than fertile worms (Ref. 60).
  • C. elegans ferl5-feml mutant is slightly more resistant to PA14 infection than wild-type worms
  • 6-FABA dramatically extend the lifespan of both the wild-type and sterile mutant on P. aeruginosa PA14 lawn ( Figure 6).
  • Example 2 - 6-FABA is catabolized to fluorocatechol (FCAT), which also attenuates C. elegans killing
  • FCAT is a potent anti-infective compound in the C. elegans PA14 killing assay, similar to 6-FABA.
  • 3-fluorocatechol FCAT
  • FCAT was also able to inhibit killing of C. elegans by the antA mutant, attenuating the virulence of this mutant to a similar level as observed with wild type PA14 ( Figure 15).
  • FCAT has rescuing activity in the C.
  • FCAT is able to extend the life span of C. elegans pmk-1, fshr-1 and zip-2 mutant worms, which are immunocompromised and hyper- susceptible to P. aeruginosa PA14-mediated killing.
  • the mode of action of FCAT is also orthogonal to known QS mechanisms in P. aeruginosa, as lasR, mvfli and the lasR-mvfR mutants are attenuated in the C. elegans assay but the worm lifespan is further extended in presence of FCAT (Figure 29).
  • FCAT functions as a potent anti-infective below its in vitro MIC.
  • 0.5 mM FCAT which is -20% of its in vitro MIC of -2.5 mM
  • carbenicillin or gentamicin were added to the P.
  • Example 3 catechol derivatives attenuate P. aeruginosa killing
  • Catechol derivatives including catechol itself, exhibited anti-virulence effect in the C. elegans assay ( Figures 20-21), indicating the effect is not specific for fluorine substitution at the 3' position:
  • Example 4 - FCAT is catabolized to fluoro-cis,cis-muconate (FMUC), which also attenuates P. aeruginosa killing
  • Figures 12 and 16 show that catechol is catabolized in P. aeruginosa by catechol 1,2-di oxygenase (CatA) to cis,cis-muconate, by CatB to muconolactone, and by CatC to 3-oxoadipate enol-lactone, which is then likely converted to either succinyl- or acetyl-CoA which feed into the citric acid cycle (See Ref. 61).
  • CatA catechol 1,2-di oxygenase
  • CatB catechol 1,2-di oxygenase
  • CatC 3-oxoadipate enol-lactone
  • 3-bromopyruvate also blocks P. aeruginosa virulence in the C. elegans model ( Figure 32).
  • Iodoacetate and iodoacetamide Figure 33 have strong activity in the C. elegans assay.
  • Table 1 shows MICs and in vivo effective doses of halogenated compounds, FCAT and gentamicin for inhibiting growth of P. aeruginosa in vitro and blocking the ability of P. aeruginosa to kill C. elegans in vivo.
  • Table 1 shows MICs and in vivo effective doses of halogenated compounds, FCAT and gentamicin for inhibiting growth of P. aeruginosa in vitro and blocking the ability of P. aeruginosa to kill C. elegans in vivo.
  • Test compounds were tested in C. elegans assay at 100 ⁇ (20-30 ⁇ g/ml). At this concentration, all compounds were shown to significantly inhibit bacterial virulence and reduce bacterial growth to > 3 sigma.
  • the structures of the test compounds are shown in Table 2, and the in vitro MIC data for the test compounds is
  • Ribavirin was tested (via single bolus intraperitoneal administration) in a standard neutropenic mice thigh infection model.
  • the thigh model is broadly accepted as a model for antibiotic in vivo efficacy (See Refs. 44 and 66-69). As shown in
  • ribavirin is active against P. aeruginosa PA14 in the thigh model, at 150 mg/kg. The effect is comparable to 75 mg/kg meropenem. At 300 mg/kg which is still well-tolerated by the animal, the drug knocks down the bacterial titer in the thigh to the detection limit. Ribavirin is also active against an extended spectrum beta- lactamase (ESBL) expressing baumannii strain UNT190-1 ( Figure 35). The strain is resistant to 300 mg/kg meropenem treatment.
  • ESBL beta- lactamase
  • Ribavirin at 75 mg/kg, which is within the achievable dose of its current anti-viral indication, knocks down the bacterial titer in the thigh to the detection limit, which is superior to the levofloxacin control at 200 mg/kg.
  • the effective dose of ribavirin for P. aeruginosa and A. baumannii was about 0.15 mg/ml and about 0.075 mg/ml, respectively, 50-100 fold below the MICs.
  • the effective concentration of ribavirin against P. aeruginosa was 0.025-0.25 mg/ml, 30-300 fold below the MIC.
  • TOB commercial antibiotic tobramycin
  • MLB Mueller-Hinton Broth
  • Figure 41 contains (A) a predicted 153 AA C-terminal kinase domain of PA14 62230, using the Phyre 2 server; (B) heat chart showing bacterial growth in a media, where ribavirin is diluted from 10 mM to about 20 ⁇ in 2X serial dilution steps from column 2 to 11; and (C) a line plot showing survival of C. elegans on PA14 wild-type and two transposon insertion mutants in PA14 62230.
  • FMPDH inosine-monophosphate dehydrogenase
  • Figure 39 contains (A) a scheme showing nucleotide metabolism; and (B) heat charts showing the effect of adenosine (upper, A) and guanosine (G) against ribavirin. As the results show, adenosine has no significant antagonistic effect whereas guanosine exhibits a concentration dependent antagonistic effect.
  • Muschiol, S., et al. A small-molecule inhibitor of type III secretion inhibits different stages of the infectious cycle of Chlamydia trachomatis. Proc Natl Acad Sci U S A, 2006. 103(39): p. 14566-71.
  • GPDH dehydrogenase
  • Aerosolized 3-bromopyruvate inhibits lung tumorigenesis without causing liver toxicity. Cancer Prev Res (Phila) 5:717-25.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present application provides antibacterial compounds. Pharmaceutical compositions containing these compounds, and methods of using these compounds for inhibiting bacterial virulence and treating bacterial infections are also provided.

Description

Antibacterial compounds
CLAIM OF PRIORITY
This application claims priority to U.S. Patent Application Serial No.
62/488,259, filed on April 21, 2017, the entire contents of which are hereby incorporated by reference.
TECHNICAL FIELD
This invention relates to compounds useful in treating bacterial infections.
BACKGROUND
The bacterial species Pseudomonas aeruginosa and Acinetobacter baumanii have emerged as significant Gram-negative bacterial pathogens, presenting multi-drug resistant strains and intrinsic antibiotic resistance. Numerous life-threating infections are attributed to P. aeruginosa and A. baumanii. In one example, these nosocomial pathogens cause outbreaks in hospitals all over the world, colonizing patients in dialysis units, neonatal units, hematology/oncology wards, and liver transplant units. The current arsenal of drugs is not sufficient to treat these infections.
SUMMARY
The present disclosure shows, inter alia, that the human opportunistic pathogen Pseudomonas aeruginosa catabolizes the anthranilate analog 2-amino-6- fluorobenzoic acid (6-FABA) to 3-fluorocatechol (FCAT) and then to 2-fluoro-cis,cis- muconate (FMUC). Each of these compounds, 6-FABA, FCAT, and FMUC, independently decreases the ability of P. aeruginosa to kill the nematode
Caenorhabditis elegans. The experiments presented herein demonstrate that 6-FABA, FCAT and FMUC function independently of quorum sensing (QS) to block P.
aeruginosa virulence.
Accordingly, in one aspect the present application provides halogenated compounds, including halogenated derivatives of catechol, that are potent anti- infectives that show in vivo antibacterial activity at concentrations < 20% of their in vitro minimal inhibitory concentrations (MIC). In contrast, gentamicin and other traditional antibiotics are only active in vivo at concentrations five to ten times higher than their in vitro MICs, suggesting that the mode of action of the compounds of the present disclosure is not mere inhibition of bacterial growth.
In another aspect, the present application provides antibacterial compounds that efficiently block bacterial virulence. This is despite the fact that some of the exemplified compounds have unfavorably high in vitro MICs. Examples of these compounds include ribavirin, which was previously approved by FDA as an antiviral medication used to treat RSV infection, hepatitis C, and viral hemorrhagic fever. The results presented herein shown that the effect of ribavirin against P. aeruginosa was comparable to that of the broad-spectrum, high-potency carbapenem antibiotic meropenem, and that ribavirin was very efficacious against a meropenem-resistant baumanii strain.
In a first general aspect, the present application provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound selected from the group consisting of:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof.
In a second general aspect, the present disclosure provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound selected from the group consisting of:
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof.
In a third general aspect, the present disclosure provides a method of treating a bacterial infection comprising administering to a subject in need thereof a
therapeutically effective amount of a 2-amino-6-fluorobenzoic acid (6-FABA) having the following structure:
Figure imgf000004_0002
or a pharmaceutically acceptable salt thereof,
wherein the bacterial infection is caused by a bacterium that does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
In a fourth general aspect, the present disclosure provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a thera eutically effective amount of a compound selected from:
Figure imgf000004_0003
Figure imgf000005_0001
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof. Certain implementations of the first, second, third and fourth general aspects are described below:
In some embodiments, the bacterial infection is caused by Gram-positive bacteria. In some embodiments, the bacterial infection is caused by Gram-negative bacteria.
In some embodiments, the bacterial infection is caused by an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or
Enter obacter). In some embodiments, the ESKAPE pathogen is selected from P. aeruginosa and baumannii. In some embodiments, the ESKAPE pathogen is P. aeruginosa. In some embodiments, the ESKAPE pathogen is baumannii. In some embodiments, the baumannii strain is meropen em -resistant. In some embodiments, the bacterial infection is caused by a bacterium selected from the group consisting of:
S. aureus and E. faecalis.
In some embodiments, the bacterial infection is selected from the group consisting of: nosocomial infection, skin infection, respiratory infection, wound infection, endovascular infection, CNS infection, abdominal infection, blood stream infection, urinary tract infection, pelvic infection, invasive systemic infection, gastrointestinal infection, dental infection, zoonotic infection, and connective tissue infection.
In some embodiments, the bacterial infection is selected from the group consisting of: atopic dermatitis, sinusitis, food poisoning, abscess, pneumonia, meningitis, osteomyelitis, endocarditis, bacteremia, sepsis, and urinary tract infection. In some embodiments, the compound is administered to the subject by a route selected from the group consisting of: oral, sublingual, gastrointestinal, rectal, topical, intradermal, subcutaneous, nasal, intravenous, and intramuscular.
In some embodiments, the subject has been identified as having a lung disease. In some embodiments, the lung disease is a structural lung disease. In some embodiments, the lung disease is selected from the group consisting of: cystic fibrosis, bronchiectasis, emphysema, and chronic obstructive pulmonary disease, and bronchiectasis. In some embodiments, the lung disease is cystic fibrosis. In some embodiments, the subject is pediatric.
In some embodiments, the compound is administered to the subject in combination with at least one additional therapeutic agent. In some embodiments, the additional therapeutic agent is an antibiotic. In some embodiments, the antibiotic is selected from the group consisting of: a quinolone, a β-lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof. In some embodiments, the aminoglycoside antibiotic is tobramycin. In some
embodiments, the compound and the additional therapeutic agent are administered consecutively. In some embodiments, the compound and the additional therapeutic agent are administered simultaneously.
In some embodiments, the therapeutically effective amount of the compound is in a range of about 4 mg/kg to about 45 mg/kg.
In a fifth general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof. In a sixth general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
Figure imgf000008_0001
or pharmaceutically acceptable salt thereof.
In a seventh general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
Figure imgf000008_0002
or a pharmaceutically acceptable salt thereof.
In an eighth general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
Figure imgf000008_0003
or pharmaceutically acceptable salt thereof,
wherein the bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ). In a ninth general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a com ound selected from the group consisting of:
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof,
wherein the bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
In a tenth general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a 2-amino-6-fluorobenzoic acid (6-FABA) having the following structure:
Figure imgf000009_0002
or a pharmaceutically acceptable salt thereof,
wherein the bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
In an eleventh general aspect, the present disclosure provides a method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from:
Figure imgf000009_0003
Figure imgf000010_0001
Figure imgf000011_0001
In a twelfth general aspect, the present disclosure provides a method of inhibiting inosine-monophosphate dehydrogenase (EVIPDH) in a bacteria, the method comprising contacting the bacteria with a compound selected from:
Figure imgf000011_0002
harmaceutically acceptable salt thereof.
Certain implementations of the fifth, sixth, seventh, eighth, ninth, tenth, eleventh, and twelfth general aspects are described below:
In some embodiments, bacteria is Gram-positive. In some embodiments, the bacteria is Gram-negative.
In some embodiments, the bacteria is an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or Enter obacter). In some embodiments, the bacteria (e.g., ESKAPE pathogen) is selected from P. aeruginosa and baumannii. In some embodiments, he ESKAPE pathogen is J3, aeruginosa. In some embodiments, the ESKAPE pathogen is A. baumannii. In some embodiments, the baumannii strain is meropenem-resistant. In some embodiments, the bacteria is selected from the group consisting of: S. aureus and E. faecalis.
In some embodiments, the bacteria is contacted in vitro. In some
embodiments, the bacteria is contacted in vivo. In some embodiments, the bacteria is contacted ex vivo. In some embodiments, the effective amount of the compound is at least 20% less than MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
In some embodiments, the effective amount of the compound is about 5-fold lower than MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
In some embodiments, the effective amount of the compound is about 30 to about 300 times below the MIC of the test compound determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
In a thirteenth general aspect, the present disclosure provides a pharmaceutical composition comprising a compound selected from:
Figure imgf000012_0001
or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
Certain implementations of the thirteenth general aspect are described below: In some embodiments, the pharmaceutical composition comprises at least one additional therapeutic agent. In some embodiments, the additional therapeutic agent is an antibiotic. In some embodiments, the additional therapeutic agent is an antibiotic selected from the group consisting of: a quinolone, a β-lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof. In some embodiments, the aminoglycoside is tobramycin. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present application belongs. Methods and materials are described herein for use in the present application; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the present application will be apparent from the following detailed description and figures, and from the claims.
DESCRIPTION OF DRAWINGS
FIG. 1 is a line plot showing that 6-F ABA blocks PA14-mediated killing of C. elegans. Wild-type N2 worms fed E. coli OP50 or PA14 + 6-F ABA at different concentrations.
FIG. 2 is a line plot showing that 6-F ABA decreases the ability of a PA14 pqsA mutant to kill C. elegans.
FIG. 3 is a line plot showing that m59, a specific MvfR inhibitor, blocks PA14-mediated killing of C. elegans similarly to PA14 mvfl or pqsA mutants.
FIG. 4 is a line plot showing that tryptophan does not enhance the ability of
PA14 to kill worms in the presence of 6-FABA.
FIG. 5 is a line plot showing that VAA AtrpE AtrpG AphnA (tryptophan auxotroph) is not blind to FAB A.
FIG. 6 is a line plot showing that 6-FABA significantly extends the lifespan of a C. elegans fer-15, fem-1 (FF) sterile mutant.
FIG. 7 is a line plot showing that FABA's effect is not likely due to generating reactive oxygen species: P. aeruginosa catalase or superoxide dismutase mutants respond to FABA similarly to wild-type PA14.
FIG. 8 is a line plot showing that. 6-FABA has a much more dramatic effect on blocking PA14-mediated killing than mutating mvfli.
FIG. 9 is a line plot showing that m50, a specific MvfR inhibitor, blocks PA14-mediated killing of C. elegans similarly to PA14 mvfl or pqsA mutants. FIG. 10 is an image showing that 6-FABA is metabolized to a brown pigment by PA14.
FIG. 11 is an image showing that antA, antB and antC mutations block 6- FABA catabolism to brown pigment.
FIG. 12 is a scheme showing simplified anthranilate and 6-FABA catabolic pathways.
FIG. 13 is a line plot showing that a PA14 antA mutant is only modestly rescued by 6-FABA, similar to the extent of killing observed with mvfR in the absence of 6-FABA.
FIG. 14 is a line plot showing that a PA14 antC mutant is only modestly rescued by 6-FABA, similar to the extent of killing observed with mvfR in the absence of 6-FABA.
FIG. 15 is a line plot showing that FCAT rescues PA14 antA and PA14 mvfR- mediated killing to a significantly greater extent than a PA14 mvfR mutant without FCAT.
FIG. 16 is a scheme showing catabolism of 6-FABA.
FIG. 17 is a line plot showing dose response growth curves of PA14 in different concentrations of FCAT. Rate of growth is not affected at 1 mM, but bacteria are killed at 4 mM.
FIG. 18 is a line plot showing that rates of growth of both WT PA14 and
PA14 catA are not affected at 1 mM FCAT, but bacteria are killed at 4 mM, showing that the killing effect is not due to FMUC, but rather most likely due to a high concentration of catechols (which generate reactive oxygen species).
FIG. 19 is a line plot showing dose response curves of PA14-mediated killing of C. elegans in different concentrations of FCAT.
FIG. 20 is a line plot showing comparison of the abilities of various catechol derivatives and 6-FABA to block the ability of P. aeruginosa PA14 to kill C. elegans.
FIG. 21 is a line plot showing that double-substituted catechol derivatives also block the ability of PA14 to kill C. elegans.
FIG. 22 is a line plot showing that mutation of P. aeruginosa PA14 catA attenuates the ability of FCAT to rescue C. elegans from P. aeruginosa PA14- mediated killing, suggesting that FCAT-mediated rescue of PA14 killing of C.
elegans is a consequence of FCAT being catabolized to fluoro-cis,cis-muconate. FIG. 23 is a line plot showing that mutation of catB does not affect the ability of FCAT to rescue C. elegans from P. aeruginosa PA14-mediated killing.
FIG. 24 is a line plot showing that mutation of PA2682 does not affect the ability of FCAT to rescue C. elegans from P. aeruginosa PA14-mediated killing. Several mutant alleles corresponding to 5 independent PA2682 transposon mutations were tested.
FIG. 25 is a line plot showing that FCAT does not rescue C. elegans from Enterococcus faecalis-mediated killing; and that bromopyruvate rescues C. elegans from Enterococcus faecalis-mediated killing.
FIG. 26 is a line plot showing that FCAT does not rescue C. elegans from
Staphylococcus aureus-mediated killing; and that bromopyruvate rescues C. elegans from Enterococcus faecalis-mediated killing.
FIG. 27 is a line plot showing that immunocompromised worms (pmk-1 and fshr-1) are rescued by FCAT from P. aeruginosa PA14-mediated killing.
FIG. 28 is a line plot showing that immunocompromised worms (zip-2) are rescued by FCAT from P. aeruginosa PA14-mediated killing.
FIG. 29 is a line plot showing that FCAT attenuates the virulence of the PA14 quorum-sensing mutants: mvfR, lasR and the double mvfR;lasR mutant.
FIG. 30 is a line plot showing that the effect of FCAT (at the same dose) is time-dependent, suggesting the compound is converted relatively slowly to FMUC.
FIG. 31 is a line plot showing that MICs are not always good indicators of in vivo efficacy: carbenicillin and gentamicin are dosed at IX, 10X and 50X MIC, respectively. FCAT was dosed at 0.2X and IX MIC.
FIG. 32 is a line plot showing that bromopyruvate blocks PA14 virulence, similarly to FCAT.
FIG. 33 is a line plot showing that the alkylating agent iodoacetamide (IAM) is also a potent inhibitor of P. aeruginosa PA14 virulence.
FIG. 34 is a bar graph showing that ribavirin is active against P. aeruginosa in a mouse thigh model.
FIG. 35 is a line plot showing that ribavirin is active against an ESBL expressing baumannii strain in a mouse thigh model.
FIG. 36 is a line plot showing that 4-hydroxy-3-nitrophenylacetic acid (40H) and nisoldipine exhibited activity against PA14 in the C. elegans infection model. FIG. 37 is a line plot showing that ribavirin and mechlorethamine exhibited dose-dependent activity against PA14 infection in the C. elegans infection model.
FIG. 38 is a line plot showing percent survival of C. elegans worms in response to treatment with various concentrations of FCAT, BP, ribavirin, and mechlorethamine HC1.
FIG. 39 contains (A) a scheme showing nucleotide metabolism; and (B) heat charts showing the effect of adenosine (upper, A) and guanosine (G) against ribavirin.
FIG. 40 contains a scheme showing the mechanism of ribavirin's anti -viral activity.
FIG. 41 contains (A) a predicted 153AA C-terminal kinase domain of
PA14_62230, using the Phyre 2 server; (B) a heat chart showing bacterial growth; and (C) a line plot showing survival of C. elegans on PA14 wild-type and two transposon insertion mutants in PA14 62230.
FIG. 42 is a line plot showing PA14 wildtype (PA14) and 3 ribavirin resistant mutants (RR1, RR2, and RR3) in the C. elegans assay.
DETAILED DESCRIPTION
A cornerstone assay in canonical antibiotic discovery is to evaluate putative antimicrobials in a growth inhibition (also called minimum inhibitory concentration, MIC) assay. These assays are typically carried out in a media that contains amino acids, nucleic acids, lipids, and carbohydrates, and that promotes rapid bacterial growth. In a typical growth inhibition assay, potent antibiotic molecules must be highly toxic to bacteria. That is, the compound must block the ability of the bacteria to utilize a wide range of nutrients.
However, a compound that may not be sufficiently toxic to bacteria in a growth inhibition assay, may still be useful for treating a bacterial infection, for example, by inhibiting bacterial virulence. That is, a compound that does not affect essential bacterial processes required in vitro for growth, and that does not have antimicrobial activity against free-living planktonic bacteria in vitro, may nevertheless target bacterial virulence or growth in vivo during an infection, for example, by modulating quorum sensing signaling pathways, pathogen-synthesized toxins, or other types of virulence-related factors. The virulence blockers may be suitable to treat persisting pathogen populations that are hard to eradicate with conventional antibiotics. In some embodiments, the present application provides a method of inhibiting virulence of bacteria, the method comprising contacting the bacteria with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the effective amount of the compound is at least about 20%, about 30%), about 40%, or about 50% less than the MIC of the same compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay. In some embodiments, the effective amount of the compound is about 2-fold, about 5-fold, about 10-fold, or about 15-fold lower than the MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
Antibacterial compounds
In some embodiments, the compound is 2-amino-6-fluorobenzoic acid (6- FABA):
Figure imgf000017_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments the compound is catechol compound selected from:
Figure imgf000017_0002
(BCAT), (3,5-CCAT), (4,5-CCAT),
Figure imgf000017_0003
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is selected from:
Figure imgf000018_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is ribavirin:
Figure imgf000018_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is ribavirin-5'-monophosphate:
Figure imgf000018_0003
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is nisoldipine:
Figure imgf000019_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is cr v v i
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is obatoclax:
Figure imgf000019_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is chlorpromazine:
Figure imgf000019_0003
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is HMS3408N17 (l'-([l, l'-biphe yl)-[l,4'-bipiperidin]-4-amin
Figure imgf000019_0004
or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is MolPort-008-370-584 (^-(l-p' fluoro-[l, -biphenyl]-4-yl)piperidin-4-yl)propane-l,3-diamine):
Figure imgf000020_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is dimetndazole:
Figure imgf000020_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is mangafodipir sodium:
Figure imgf000020_0003
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is otenzepad:
Figure imgf000020_0004
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is ZM 39923 :
Figure imgf000020_0005
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is PPT:
Figure imgf000020_0006
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is 4-hydroxy-3-nitrophenylacetic acid:
Figure imgf000021_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is clonidine:
Figure imgf000021_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is betonicine:
Figure imgf000021_0003
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is clotrimazole:
Figure imgf000021_0004
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is ammonium
pyrrolidinedithiocarbamate (APDC):
Figure imgf000021_0005
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is otilonium bromide:
Figure imgf000022_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is FPA 124:
Figure imgf000022_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is HMS3604J07 ((E)-3-bromo-N'-((3,5- dibromo-4-hydroxy-6-oxocyclohexa-2,4-dien-l-ylidene)methyl)benzohydrazide):
Figure imgf000022_0003
or a pharmaceutically acceptable salt thereof.
In some embodiments, the com ound is lomofungin:
Figure imgf000022_0004
or a pharmaceutically acceptable salt thereof.
In some embodiments, a salt (e.g., pharmaceutically acceptable salt) of a compound of the present disclosure is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group. According to another embodiment, the compound is a pharmaceutically acceptable acid addition salt.
In some embodiments, acids commonly employed to form pharmaceutically acceptable salts of the compounds of the present disclosure include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para- toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids. Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenyl acetate, phenylpropionate, phenylbutyrate, citrate, lactate, β-hydroxybutyrate, glycolate, maleate, tartrate, methanesulfonate, propanesulfonate, naphthalene- 1 -sulfonate, naphthalene-2- sulfonate, mandelate and other salts. In one embodiment,
pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid.
In some embodiments, bases commonly employed to form pharmaceutically acceptable salts of the compounds of the present disclosure include hydroxides of alkali metals, including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydroxyl- substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine;
pyridine; N-methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris- (2-OH-(Cl-C6)-alkylamine), such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri- (2-hydroxyethyl)amine; N-methyl -D-glucamine; morpholine; thiomorpholine;
piperidine; pyrrolidine; and amino acids such as arginine, lysine, and the like.
In some embodiments, the compounds of the present disclosure, or
pharmaceutically acceptable salts thereof, are substantially isolated. A compound may inhibit bacterial virulence by inhibiting inosine- monophosphate dehydrogenase (IMPDH) in a bacteria. Hence, in some embodiments, the present disclosure provides a method of inhibiting inosine-monophosphate dehydrogenase (IMPDH) in a bacteria, the method comprising contacting the bacteria with an effective amount of a compound as described herein. In some aspects of these embodiments, the compound is ribavirin or ribavirin 5 '-phosphate.
Bacterial pathogens
In some embodiments, the bacteria is at least 2-fold, 4-fold, 8-fold, 10-fold, 24-fold, 48-fold, 100-fold, 256-fold, 512-fold or 1000-fold resistant to one or more of other antibiotic agents. In some embodiments, the bacteria is multi-drug resistant (MDR). For example, the bacteria is resistant to methicillin, vancomycin, rifampicin, linezolid, daptomycin, gentamicin and/or ciprofloxacin.
In some embodiments, the bacteria uses quorum sensing (QS). In these embodiments, the bacteria uses quorum sensing mediated by N-acyl-homoserine lactone (AHL) (e.g., when the bacteria is Gram-negative bacteria), or by an autoinducing peptide (e.g., when the bacteria is Gram-negative bacteria). In some embodiments, the bacteria does not use quorum sensing activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ), such as 3,4-dihydroxy-2-heptylquinoline (PQS), 4-hydroxy-2-heptylquinoline, or 3,4-dihydroxy-2-nonylquinoline.
In some embodiments, the bacteria is Gram-positive bacteria.
In some embodiments, the bacteria is a member of a genus selected from the group consisting of Staphylococcus (including coagulase negative and coagulase positive), Streptococcus, Peptococcus, Enterococcus, and Bacillus.
In some embodiments, the bacteria is a member of the Staphylococcus genus and the species of bacteria is selected from the group consisting of S. aureus, methicillin-susceptible S. aureus ^SSA), coagulase negative staphylococci, methicillin-resistant S. aureus (MRSA), vancomycin-resistant S. aureus (VRSA), S. arlettae, S. agnetis, S. auricularis, S. capitis, S. caprae, S. carnosus, S. caseolyticus, S. chromogenes, S. cohnii, S. condimenti, S. delphini, S. devriesei, S. epidermidis, S. equorum, S. felis, S. fleurettii, S. gallinarum, S. haemolyticus, S. hominis, S. hyicus, S. intermedius, S. kloosii, S. leei, S. lentus, S. lugdunensis, S. lutrae, S. massiliensis, S. microti, S. muscae, S. nepalensis, S. pasteuri, S. pettenkoferi, S. piscifermentans, S. pseudintermedius, S. pseudolugdunensis, S. pulvereri, S. rostri, S. saccharolyticus, S. saprophytics, S. schleiferi, S. sciuri, S. simiae, S. simulans, S. stepanovicii, S.
succinus, S. vitulinus, S. warneri, and S. xylosus.
In some embodiments, the bacteria is a member of the Peptococcus genus and the species of bacteria is P. magnus.
In some embodiments, the bacteria is a member of the Streptococcus genus and the species of bacteria is selected from the group consisting of S. agalactiae, S. anginosus, S. bovis, S. canis, S. constellatus, S. dysgalactiae, S. equinus, S. iniae, S. intermedius, S. milleri, S. mitis, S. mutans, S. oralis, S. parasanguinis, S. peroris, S. pneumoniae, S. pseudopneumoniae, S. pyogenes, S. ratti, S. salivarius, S. tigurinus, S. thermophilus, S. sanguinis, S. sobrinus, S. suis, S. uberis, S. vestibularis, S. viridans, and S. zooepidemicus .
In some embodiments, the bacteria is a member of the Enterococcus genus and the species of bacteria is selected from the group consisting of E. avium, E. durans, E. faecalis, E. gallinarum, E. haemoperoxidus, E. hirae, E. malodoratus, E. moraviensis, E. mundtii, E. pseudoavium, E. raffinosus, E. solitaries, and E. faecium.
In some embodiments, the bacteria is a member of the Propionibacterium genus. In such embodiments, the bacteria is P. acnes.
In some embodiments, the bacteria is a Gram-negative bacteria.
In some embodiments, the bacteria is a member of a family selected from the group consisting of Enterobacteriaceae, Helicobacter aceae, Campylobacter aceae, Neisseriaceae, Pseudomonadaceae, Moraxellaceae, Xanthomonadaceae,
Pasteurellaceae, and Legionellaceae .
In some embodiments, the bacteria is a member of a genus selected from the group consisting of Citrobacter, Enterobacter, Escherichia, Klebsiella, Pantoea, Proteus, Salmonella, Serratia, Shigella, Yersinia, Helicobacter, Wolinella,
Campylobacter, Arcobacter, Neisseria, Francisella, Pseudomonas, Acinetobacter, Moraxella, Stenotrophomonas, Haemophilus, Pasteurella, and Legionella.
In some embodiments, the bacteria is a member of the Citrobacter genus and the species of bacteria is selected from the group consisting of C. amalonaticus, C braakii, C. diver sus, C. farmer, C. freundii, C. gillenii, C. koseri, C. murliniae, C. rodentium, C. sedlakii, C. werkmanii, and C. youngae.
In some embodiments, the bacteria is a member of the Enterobacter genus and the species of bacteria is selected from the group consisting of E. aerogenes, E.
amnigenus, E. agglomerans, E. arachidis, E. asburiae, E. cancerogenous, E. cloacae, E. cowanii, E. dissolvens, E. gergoviae, E. helveticus, E. hormaechei, E. intermedius, E. kobei, E. ludwigii, E. mori, E. nimipressuralis, E. oryzae, E. pulveris, E. pyrinus, E. radicincitans, E. taylorae, E. turicensis, E. sakazakii, and E. spp.
In some embodiments, the bacteria is a member of the Escherichia genus and the species of bacteria is selected from the group consisting of E. albertii, E. blattae, E. coli, E. fergusonii, E. hermannii, and E. vulneris.
In some embodiments, the bacteria is a member of the Klebsiella genus and the species of bacteria is selected from the group consisting of K. granulomatis, K. oxytoca, K. pneumoniae, K. terrigena, and K. planticola.
In some embodiments, the bacteria is a member of the Pantoea genus and the species of bacteria is selected from the group consisting of P. agglomerans, P.
ananatis, P. citrea, P. dispersa, P. punctata, P. stewartii, P. terrea, and P. vagans.
In some embodiments, the bacteria is a member of the Proteus genus and the species of bacteria is selected from the group consisting of P. hauseri, P. mirabilis, P. myxofaciens, P. penneri, and P. vulgaris.
In some embodiments, the bacteria is a member of the Salmonella genus and the species of bacteria is selected from the group consisting of S. bongori, and S. enterica.
In some embodiments, the bacteria is a member of the Serratia genus and the species of bacteria is selected from the group consisting of S. entomophila, S. ficaria, S. fonticola, S. grimesii, S. liquefaciens, S. marcescens, S. odorifera, S. plymuthica, S. proteamaculans, S. quinivorans, S. rubidaea, and S. symbiotica.
In some embodiments, the bacteria is a member of the Shigella genus and the species of bacteria is selected from the group consisting of S. boydii, S. dysenteriae, S. flexneri, and S. sonnei.
In some embodiments, the bacteria is a member of the Yersinia genus and the species of bacteria is selected from the group consisting of Y pestis, Y
pseudotuberculosis, and Y. enterocolitica. In some embodiments, the bacteria is a member of the Helicobacter genus and the species of bacteria is selected from the group consisting of H. acinonychis, H. anseris, H. aurati, H. baculiformis, H. bilis, H. bizzozeronii, H. brantae, H.
canadensis, H. canis, H. cetorum, H. cholecystus, H. cinaedi, H. cynogastricus, H. equorum, H. felis, H. fennelliae, H. ganmani, H. heilmannii, H. hepaticus, H.
mesocricetorum, H. macacae, H. marmotae, H. mastomyrinus, H. mesocricetorum, H. muridarum, H. mustelae, H. pametensis, H. pullorum, H. pylori, H. rappini, H.
rodentium, H. salomonis, H. suis, H. trogontum, H. typhlonius, and H. winghamensis.
In some embodiments, the bacteria is a member of the Campylobacter genus and the species of bacteria is selected from the group consisting of C. avium, C.
butzleri, C. canadensis, C. cinaedi, C. coli, C. concisus, C. corcagiensis, C.
cryaerophilus, C. cuniculorum, C. curvus, C. fennelliae, C. fetus, C. gracilis, C.
helveticus, C. hominis, C. hyoilei, C. hyointestinalis, C. insulaenigrae, C. jejuni, C. lanienae, C. lari, C. mucosalis, C. mustelae, C. nitrofigilis, C. peloridis, C. pylori, C. rectus, C. showae, C. sputorum, C. subantarcticus, C. upsaliensis, C. ureolyticus, and
C. volucris.
In some embodiments, the bacteria is a member of the Arcobacter genus and the species of bacteria is selected from the group consisting of bivalviorum, A. butzleri, A. cibarius, A. cryaerophilus, A. defluvii, A. ellisii, A. halophilus, A. marinus, A. molluscorum, A. mytili, A. nitrofigilis, A. skirrowii, A. thereius, A. trophiarum, and A. venerupis.
In some embodiments, the bacteria is a member of the Neisseria genus and the species of bacteria is selected from the group consisting of N. bacilliformis, N.
cinerea, N. denitrificans, N. elongata, N. flavescens, N. gonorrhoeae, N. lactamica, N. macacae, N. meningitidis, N. mucosa, N. pharyngis, N. polysaccharea, N. sicca, N. subflava, and N. weaver.
In some embodiments, the bacteria is a member of the Francisella genus and the species of bacteria is selected from the group consisting of F. tularensis, F novicida, F hispaniensis, W. persica, F noatunensis, F philomiragia, F halioticida, F endociliophora, and F guangzhouensis .
In some embodiments, the bacteria is a member of the Pseudomonas genus and the species of bacteria is selected from the group consisting of P. aeruginosa, P. oryzihabitans, and ! plecoglossicida. In some embodiments, the bacteria is a member of the Acinetobacter genus and the species of bacteria is baumannii.
In some embodiments, the bacteria is a member of the Moraxella genus and the species of bacteria is selected from the group consisting ofM catarrhalis, M. lacunata, and M bovis.
In some embodiments, the bacteria is a member of the Stenotrophomonas genus and the species of bacteria is S. maltophilia.
In some embodiments, the bacteria is a member of the Haemophilus genus and the species of bacteria is selected from the group consisting of H aegyptius, H aphrophilus, H. avium, H. ducreyi, H. felis, H. haemolyticus, H. influenzae, H.
parainfluenzae, H. paracuniculus, H. parahaemolyticus, H. pittmaniae, Haemophilus segnis, and H. somnus.
In some embodiments, the bacteria is a member of the Pasteurella genus and the species of bacteria is selected from the group consisting of P. multocida, P.
stomatis, P. dagmatis, P. canis, P. bettyae, and P. anatis.
In some embodiments, the bacteria is a member of the Legionella genus and the species of bacteria is selected from the group consisting of L. pneumophila, L. anisa, L. bozemanae, L. cincinnatiensis, L. gormanii, L. jordani, L. longbeachae, L. maceachernii, L. micdadei, L. sainthelensi, L. wadsworthii, and L. waltersii.
In some embodiments, the bacteria is a member of the Mycobacterium genus and the species of bacteria is selected from a group consisting ofM tuberculosis and M. smegmatic.
In some embodiments, the bacteria is a member of a genus selected from: Acinetobacter, Burkholderia, Acinetobacter, Burkholderia, Klebsiella, Pseudomonas, and Escherichia. In such embodiments, the bacteria is a member of a species selected from: K pneumoniae, P. aeruginosa, Enterobacteriaceae, a d E. coli.
In some embodiments, the bacteria is an ESKAPE pathogen (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacte ). Treating bacterial infections
In some embodiments, the present disclosure provides a method of treating a bacterial infection in a subject, the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the bacterial infection is caused by any one of the bacteria described herein. In some embodiments, the bacterial infection is resistant to treatment with one or more of the antibiotic agents described herein (e.g., bacterial infection is resistant to treatment with methicillin, vancomycin, rifampicin, gentamicin and/or ciprofloxacin). In these embodiments, the bacterial infection is characterized as resistant to treatment with one or more available antibiotic agents.
In some embodiments, the bacterial infection is a skin infection. In some aspects of these embodiments, the skin infection is selected from the group consisting of acne, pimples, impetigo, boils, cellulitis, folliculitis, carbuncles, scalded skin syndrome, skin abscesses, atopic dermatitis, and typhoid fever. In some embodiments, the bacterial infection is a skin infection caused by P. acnes. In such embodiments, the skin infection is acne. In some embodiments, the bacterial infection is a skin and soft tissue infection (e.g., acne).
In some embodiments, the bacterial infection is a respiratory infection. In some aspects of these embodiments, the respiratory infection is selected from the group consisting of upper respiratory tract infection, bronchopneumonia, atypical pneumonia, tuberculosis, mycobacterium tuberculosis, pneumonia, anaerobic pleuropulmonary infection, ventilator-associated pneumonia, aspiration pneumonia, lung abscess, bronchitis, chronic obstructive pulmonary disease, obstructive pulmonary disease, Pontiac fever, and legionellosis.
In some embodiments, the bacterial infection is a wound infection. In some aspects of these embodiments, the wound infection is a postsurgical wound infection. In some embodiments, the bacterial infection is a blood stream infection. In some aspects of these embodiments, the blood stream infection is bacteremia or sepsis. In some embodiments, the bacterial infection is a pelvic infection. In some aspects of the embodiments, the pelvic infection is bacterial vaginosis.
In some embodiments, the bacterial infection is a gastrointestinal infection. In some aspects of these embodiments, the gastrointestinal infection is selected from the group consisting of peptic ulcer, chronic gastritis, duodenitis, gastroenteritis, diarrhea, dysentery, diphtheria, food poisoning and foodborne illness.
In some embodiments, the bacterial infection is a bone, joint or muscle infection. In some aspects of these embodiments, the bone, joint or muscle infection is selected from the group consisting of tetanus, secondary meningitis, meningitis, neonatal meningitis, sinusitis, laryngitis, arthritis, septic arthritis, Bartholin gland abscess, chancroid, osteomyelitis, endocarditis, mediastinitis, pericarditis, peritonitis, otitis media, blepharoconjunctivitis, keratoconjunctivitis, and conjunctivitis. In some embodimetns, the joint infection is an infection of a shoulder, a knee, a hip, or an elbow. In some embodiments, the bacterial infection is septic arthritis (e.g., septic arthritis caused by P. acnes or septic arthritis caused by S. aureus).
In some embodiments, the bacterial infection is selected from the group consisting of a dental infection, a zoonotic infection, an invasive systemic infection, a urinary tract infection, an abdominal infection, a CNS infection, an endovascular infection, a connective tissue infection, and a nosocomial infection. In some embodiments, the bacterial infection is selected from the group consisting of syphilis, leprosy, abscesses, sepsis, empyema, and tularemia.
In some embodiments, the bacterial infection is associated with implanted devices (e.g., catheter, ballon catheter, stent, pacer etc). In some embodiments, the bacterial infection is osteomyelitis, endocarditis, or an infection associated with an implanted device, which is caused by a S. aureus persister, P. acnes, P. aeruginosa, or
A. baumannii.
Additional therapeutic agents
In some embodiments, a composition of the present application further comprises one or more additional therapeutic agents. The additional therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound of the present disclosure.
In some embodiments, the second therapeutic agent is a virulence blocker. In some embodiments, the second therapeutic agent is m50 or m59, or a
pharmaceutically acceptable salt thereof. In some embodiments, a pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof, also optionally contains at least one additional therapeutic agent, or a pharmaceutically acceptable salt thereof. In these embodiments, the additional therapeutic agent in the composition is any one of the antibiotics described herein (e.g., gentamicin or defensin 1). The second therapeutic agent may be present in the composition in a therapeutically effective amount. For pharmaceutical compositions that comprise an additional therapeutic agent, or for methods that comprise using an additional therapeutic agent, an effective amount of the additional therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent. For example, an effective amount is between about 70% and 100% of the normal monotherapeutic dose. The normal monotherapeutic dosages of these additional therapeutic agents are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda,
Calif. (2000), each of which references are incorporated herein by reference in their entirety. In some embodiments, when the additional therapeutic agent is gentamicin, the effective amount of gentamicin is lower than the amount that causes
nephrotoxicity in a subject.
In some embodiments, a method of treating a subject in need thereof as disclosed herein comprises administering to the subject one or more additional therapeutic agents. The additional therapeutic agent may be administered to the subject in a separate pharmaceutical composition or dosage form (e.g., any one of the compositions, formulation, routes and dosage forms described herein). In these embodiments, a compound as provided herein, or a pharmaceutically acceptable salt thereof, can be used in combination with an antibiotic.
In some embodiments, a compound as provided herein, or a pharmaceutically acceptable salt thereof, can be used in combination with a cationic antimicrobial peptide (CAMP). In some aspects of these embodiments, the cationic antimicrobial peptide is a defensin peptide (e.g., defensin 1 such as beta-defensin 1 or alpha- defensin 1), or cecropin, andropin, moricin, ceratotoxin, melittin, magainin, dermaseptin, bombinin, brevinin (e.g., brevinin-1), esculentin, buforin II (e.g., from amphibians), CAP18 (e.g., from rabbits), LL37 (e.g., from humans), abaecin, apidaecins (e.g., from honeybees), prophenin (e.g., from pigs), indolicidin (e.g., from cattle), brevinins, protegrin (e.g., from pig), tachyplesins (e.g., from horseshoe crabs), or drosomycin (e.g., from fruit flies).
In some embodiments, the antibiotic is selected from the quinolone class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of levofloxacin, norfloxacin, ofloxacin, ciprofloxacin, perfloxacin, lomefloxacin, fleroxacin, sparfloxacin, grepafloxacin, trovafloxacin, clinafloxacin, gemifloxacin, enoxacin, sitafloxacin, nadifloxacin, tosulfloxacin, cinnoxacin, rosoxacin, miloxacin, moxifloxacin, gatifloxacin, cinnoxacin, enoxacin, fleroxacin, lomafloxacin, lomefloxacin, miloxacin, nalidixic acid, nadifloxacin, oxolinic acid, pefloxacin, pirimidic acid, pipemidic acid, rosoxacin, rufloxacin, temafloxacin, tosufloxacin, trovafloxacin, and besifloxacin.
In some embodiments, the antibiotic is selected from a β-lactam, a
monobactam, oxazolidinone, and lipopeptide.
In some embodiments, the antibiotic is selected from the cephalosporin class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of cefazolin, cefuroxime, ceftazidime, cephalexin, cephaloridine, cefamandole, cefsulodin, cefonicid, cefoperazine, cefoprozil, and ceftriaxone.
In some embodiments, the antibiotic is selected from the penicillin class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of penicillin G, penicillin V, procaine penicillin, and benzathine penicillin, ampicillin, and amoxicillin, benzylpenicillin,
phenoxymethylpenicillin, oxacillin, methicillin, dicloxacillin, flucloxacillin, temocillin, azlocillin, carbenicillin, ricarcillin, mezlocillin, piperacillin, apalcillin, hetacillin, bacampicillin, sulbenicillin, mecicilam, pevmecillinam, ciclacillin, talapicillin, aspoxicillin, cloxacillin, nafcillin, and pivampicillin.
In some embodiments, the antibiotic is selected from the carbapenem class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of thienamycin, tomopenem, lenapenem, tebipenem, razupenem, imipenem, meropenem, ertapenem, doripenem, panipenem (betamipron), and biapenem. In some embodiments, the antibiotic is selected from the lipopeptide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of polymyxin B, colistin (polymyxin E), and daptomycin.
In some embodiments, the antibiotic is selected from the aminoglycoside class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of gentamicin, amikacin, tobramycin, debekacin, kanamycin, neomycin, netilmicin, paromomycin, sisomycin, spectinomycin, and streptomycin.
In some embodiments, the antibiotic is selected from the glycopeptide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of vancomycin, teicoplanin, telavancin,
ramoplanin, daptomycin, decaplanin, and bleomycin.
In some embodiments, the antibiotic is selected from the macrolide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of azithromycin, clarithromycin, erythromycin, fidaxomicin, telithromycin, carbomycin A, josamycin, kitasamycin,
midecamycin/midecamycinacetate, oleandomycin, solithromycin, spiramycin, troleandomycin, tylosin/tylocine, roxithromycin, dirithromycin, troleandomycin, spectinomycin, methymycin, neomethymycin, erythronolid, megalomycin, picromycin, narbomycin, oleandomycin, triacetyl-oleandomycin, laukamycin, kujimycin A, albocyclin and cineromycin B.
In some embodiments, the antibiotic is selected from the ansamycin class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of streptovaricin, geldanamycin, herbimycin, rifamycin, rifampin, rifabutin, rifapentine and rifamixin.
In some embodiments, the antibiotic is selected from the sulfonamide class of antibiotic compounds. In some aspects of these embodiments, the antibiotic is selected from the group consisting of sulfanilamide, sulfacetamide, sulfapyridine, sulfathiazole, sulfadiazine, sulfamerazine, sulfadimidine, sulfasomidine, sulfasalazine, mafenide, sulfamethoxazole, sulfamethoxypyridazine, sulfadimethoxine,
sulfasymazine, sulfadoxine, sulfametopyrazine, sulfaguanidine, succinylsulfathiazole and phthalylsulfathiazole. In some embodiments, the antibiotic is selected from the group consisting of quinolones, fluoroquinolones, β-lactams, cephalosporins, penicillins, carbapenems, lipopeptide antibiotics, glycopeptides, macrolides, ansamycins, sulfonamides, and combinations of two or more thereof.
In some embodiments, the present application provides separate dosage forms of a compound described herein, or a pharmaceutically acceptable salt thereof, and one or more of any of the above-described second therapeutic agents. The separate dosage forms may be administered together consecutively (e.g., within less than 24 hours of one another) or simultaneously (e.g., administered to the patient within 5 minutes of one another).
Compositions, formulations, and routes of administration
In some embodiments, the present application also provides pharmaceutical compositions comprising an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier. The carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of the present application include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, di sodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances,
polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
If required, the solubility and bioavailability of the compounds of the present application in pharmaceutical compositions may be enhanced by methods well-known in the art. One method includes the use of lipid excipients in the formulation. See "Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water- Soluble Drugs (Drugs and the Pharmaceutical Sciences)," David J. Hauss, ed. Informa Healthcare, 2007; and "Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley-Interscience, 2006.
Another known method of enhancing bioavailability is the use of an amorphous form of a compound of the present application optionally formulated with a poloxamer, such as LUTROL™ and PLURONIC™ (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent 7,014,866; and United States patent publications 20060094744 and 20060079502.
The pharmaceutical compositions of the present application include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. In certain embodiments, the compound of the present disclosure is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques). Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
In some embodiments, the compound of the present disclosure, or a pharmaceutically acceptable salt thereof, is administered orally. Compositions of the present application suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc. Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption. In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
Compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
Compositions suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
The pharmaceutical compositions of the present application may be administered in the form of suppositories for rectal administration. These
compositions can be prepared by mixing a compound of the present application with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax, and polyethylene glycols.
The pharmaceutical compositions of the present application may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, for example, U.S. Patent No. 6,803,031.
Topical administration of the pharmaceutical compositions of the present application is especially useful when the desired treatment involves areas or organs readily accessible by topical application (e.g., skin and soft tissues).
The topical compositions of the present disclosure can be prepared and used in the form of an aerosol spray, cream, emulsion, solid, liquid, dispersion, foam, oil, gel, hydrogel, lotion, mousse, ointment, powder, patch, pomade, solution, pump spray, stick, towelette, soap, or other forms commonly employed in the art of topical administration and/or cosmetic and skin care formulation. The topical compositions can be in an emulsion form, as a cream or a paste.
In some embodiments, the topical composition comprises a combination of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and one or more additional ingredients, carriers, excipients, or diluents including, but not limited to, absorbents, anti-irritants, anti-acne agents, preservatives, antioxidants, coloring agents/pigments, emollients (moisturizers), emulsifiers, film-forming/holding agents, fragrances, leave-on exfoliants, prescription drugs, preservatives, scrub agents, silicones, skin-identical/repairing agents, slip agents, sunscreen actives,
surfactants/detergent cleansing agents, penetration enhancers, and thickeners. Lists of ingredients, which are well known in the art, are disclosed, for example, in "Cosmetics: Science and Technology," edited by M. S. Balsam and E. Sagarin, 2nd Edition, 1972, Wiley Pub. Co.; "The Chemistry and Manufacture of Cosmetics" by M. G. DeNavasse; and "Harry's Cosmeticology," J.B. Wilkinson et al., 7th Edition, 1982, Chem. Pub. Co.; the disclosures of each of the above being incorporated herein by reference in their entirety. In some embodiments, diluents, carriers, and excipients may include, but are not limited to, polyethylene glycols (such as PEG200, PEG300, PEG400, PEG540, PEG600, PEG1450 or mixtures thereof) and coconut oils (such as propylene glycol dicaprate, coco-caprylate/caprate, propylene glycol dicaprylate/dicaprate, caprylic/capric triglyceride, caprylic/capric/lauric triglyceride, caprylic/capric/linoleic triglyceride, tricaprin, tricaprylin, glyceryl trioleate, neopentyl glycol dicaprylate/dicaprate, caprylic/capric/palmitic/stearic triglceride, or mixtures thereof). In some embodiments, suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water. In some embodiments, preservatives may include, but are not limited to, 1,2-hexanediol, benzoic acid, benzothonium chloride, borax, bronopol, butylparaben, caprylyl glycol, chlorophene, chloroxylenol, chlorphenesin, dehydroacetic acid, diazolidinyl urea, DMDM hydantoin, ethylhexylglycerin, ethylparaben, formaldehyde-releasing preservative, Germaben II, hoelen, imidazolidinyl urea, iodopropynyl butylcarbamate,
isobutylparaben, methylchloroisothiazolinone, methyldibromo glutaronitrile,
Methylisothiazolinone, methylparaben, o-cymen-5-ol, phenoxyethanol,
phenoxyisopropanol, phytosphingosine, polyaminopropyl biguanide, potassium sorbate, propylparaben, quaternium-15, sodium benzoate, sodium citrate, sodium dehydroacetate, sodium hexametaphosphate, sodium hydroxymethylglycinate, sodium lactobionate, sodium metabi sulfite, sodium sulfite, sorbic acid, and styrax benzoin. In some embodiments, slip agents may include, but are not limited to, amodimethicone, fos-PEG-18 methyl ether dimethyl silane, fos-phenylpropyl dimethicone, butylene glycol, cetyl dimethicone, cetyl dimethicone copolyol, cetyl PEG/PPG- 10/1- dimethicone, cyclohexasiloxane, cyclomethicone, cyclopentasiloxane,
cyclotetrasiloxane, decylene glycol, diisostearoyl trimethylolpropane siloxy silicate, dimethicone, dimethicone copolyol, dimethicone crosspolymer, dimethiconol, dipropylene glycol, hexylene glycol, hydrolyzed silk, isododecane, methicone, methyl trimethicone, methylsilanol mannuronate, methylsilanol PEG-7 glyceryl cocoate, PEG- 10 dimethicone, PEG- 10 dimethicone/vinyl dimethicone crosspolymer, PEG- 12 dimethicone, PEG/PPG-18/18 dimethicone, PEG/PPG-20/15 dimethicone, pentylene glycol, phenyl trimethicone, polymethylsilsesquioxane, PPG-3 benzyl ether myristate, silica dimethyl silylate, silk powder, siloxane, simethicone, sorbitol, stearyl dimethicone, stearyl methicone, triethoxycaprylylsilane, trimethylsiloxysilicate, xylitol, and zinc stearate. In some embodiments, sunscreen actives may include, but are not limited to, avobenzone, benzephenone-3, benzophenones, bumetrizole, butyl methoxydibenzoylmethane, ecamsule, ensulizole, ethylhexyl methoxycinnamate, homosalate, menthyl anthranilate, meradmiate, Mexoryl SX, octinoxate, octisalate, octocrylene, octyl methoxycinnamate, octyl salicylate, oxybenzone, padimate O, para- aminobenzoic acid (PABA), Parsol 1789, terephthalylidine dicamphor sulfonic acid, Tinosorb M, Tinosorb S, and titanium dioxide. In some embodiments, emulsifiers, surfactants, and detergents may include, but are not limited to, ammonium laureth sulfate, ammonium lauryl sulfate, arachidyl glucoside, behenic acid, bis-PEG-lS methyl ether dimethyl silane, C20-40 pareth-40, cocamidopropyl betaine,
cocamidopropyl dimethylamine, cocamidopropyl hydroxysultaine, coco-glucoside, coconut oil, decyl glucoside, dicetyl phosphate, dihydrocholeth-30, disodium cocoamphodi acetate, disodium cocoyl glutamate, disodium lauraminopropionate, glyceryl behanate, hydrogenated vegetable glycerides citrate, isohexadecane, isostearamide DEA, lauramphocarboxyglycinate, laureth-23, laureth-4, laureth-7, lauryl PEG-9 polydimethylsiloxy ethyl dimethicone, lauryl alcohol, lauryl glucoside, magnesium laureth sulfate, magnesium oleth sulfate, myristic acid, nonoxynols, oleic acid, oleth 10, palm kernel acid, palmitic acid, PEG-60 almond glycerides, PEG-75 shea butter glycerides, PEG 90M, PEG- 10 dimethicone, PEG- 10 dimethicone/vinyl dimethicone crosspolymer, PEG- 10 rapeseed sterol, PEG- 100 stearate, PEG- 12 dimethicone, PEG- 120 methyl glucose dioleate, PEG-20 methyl glucose
sesqui stearate, PEG-40 stearate, PEG-60 hydrogenated castor oil, PEG-7 glyceryl cocoate, PEG-8, PEG-80 sorbitan laurate, PEG/PPG- 17/6 copolymer (polyethylene glycol/polypropylene glycol-17/6 copolymer), PEG/PPG-18/18 dimethicone,
PEG/PPG-20/15 dimethicone, poloxamer 184, Poloxamer 407, poloxamers, polyglyceryl-3 beeswax, polyglyceryl-4 isostearate, polyglyceryl-6 isostearate, polysorbate 20, polysorbate 60, polysorbate 80, potassium cetyl phosphate, potassium hydroxide, potassium myristate, PPG- 12 buteth-16, PPG-26-Buteth-26, Salvia officinalis, Saponaria officinalis extract, soapwort, sodium C14.16 olefin sulfonate, sodium cetearyl sulfate, sodium cocoamphoacetate, sodium cocoate, sodium cocoyl glutamate, sodium cocoyl isethionate, sodium dilauramidoglutamide lysine, sodium hexametaphosphate, sodium hydroxide, sodium laureth sulfate, sodium laureth-13 carboxylate, sodium lauroamphoacetate, sodium lauroyl lactylate, sodium lauroyl sarcosinate, sodium lauryl glucose carboxylate, sodium lauryl sulfate, sodium methyl cocoyl taurate, sodium methyl taurate, sodium myreth sulfate, sodium palm kernelate, sodium palmate, sodium PEG-7 olive oil carboxylate, sodium trideceth sulfate, steareth-20, TEA-lauryl sulfate (triethanolamine- lauryl sulfate), and tribehenin PEG- 20 esters.
Application of the subject therapeutics may be local, so as to be administered at the site of interest (e.g., infected area of skin, or an infected joint or other connective tissue). Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
Thus, according to yet another embodiment, the compounds of the present application may be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, or catheters.
Suitable coatings and the general preparation of coated implantable devices are known in the art and are exemplified in U.S. Patent Nos. 6,099,562; 5,886,026; and 5,304,121. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccharides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition. Coatings for invasive devices are to be included within the definition of pharmaceutically acceptable carrier, adjuvant or vehicle, as those terms are used herein.
According to another embodiment, the present application provides a method of coating an implantable medical device comprising the step of contacting said device with the coating composition described above. It will be obvious to those skilled in the art that the coating of the device will occur prior to implantation into a mammal.
According to another embodiment, the present application provides a method of impregnating an implantable drug release device comprising the step of contacting said drug release device with a compound or composition of the present application. Implantable drug release devices include, but are not limited to, biodegradable polymer capsules or bullets, non-degradable, diffusible polymer capsules and biodegradable polymer wafers.
According to another embodiment, the present application provides an implantable medical device coated with a compound or a composition comprising a compound of the present application, such that said compound is therapeutically active.
Where an organ or tissue is accessible because of removal from the subject, such organ or tissue may be bathed in a medium containing a composition of the present application, a composition of the present application may be painted onto the organ, or a composition of the present application may be applied in any other convenient way.
In the pharmaceutical compositions of the present application, a compound of the present disclosure, or a pharmaceutically available salt thereof, is present in an effective amount (e.g., a therapeutically effective amount).
The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., Cancer Chemother. Rep, 1966, 50: 219. Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
In some embodiments, an effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, can range, for example, from about lmg to about 200 mg, from about 1 to about 100 mg, from about 1 to about 50 mg, from about 1 mg to about 30 mg, from about 1 mg to about 15 mg, from about 10 mg to about 2000 mg, from about 10 mg to about 1900 mg, from about 10 mg to about 1800 mg, from about 10 mg to about 1700 mg, from about 10 mg to about 1600 mg, from about 10 mg to about 1500 mg, from about 10 mg to about 1400 mg, from about 10 mg to about 1300 mg, from about 10 mg to about 1200 mg, from about 10 mg to about 1100 mg, from about 10 mg to about 1000 mg, from 10 mg about to about 900 mg, from about 10 mg to about 800 mg, from about 10 mg to about 700 mg, from about 10 mg to about 600 mg, from about 10 mg to about 500 mg, from about 10 mg to about 400 mg, from about 10 mg to about 300 mg, from about 10 mg to about 200 mg, from about 10 mg to about 100 mg, and from about 10 mg to about 50 mg. In some embodiments, an effective amount of the compound, or a
pharmaceutically acceptable salt thereof, is 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, or 10 mg.
In some embodiments, an effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, can range, for example, from about 1 mg/kg to about 1000 mg/kg. For example, an effective amount can range from about 1 mg/kg to about 50 mg/kg, about 4 mg/kg to about 45 mg/kg, or about 50 mg/kg to about 500 mg/kg.
In some embodiments, the composition containing an effective amount of the compound, or a pharmaceutically acceptable salt thereof, is administered once daily. In some embodiments, the composition containing an effective amount of the compound, or a pharmaceutically acceptable salt thereof, is administered twice daily. In some embodiments, the composition containing an effective amount of the compound, or a pharmaceutically acceptable salt thereof, is administered thrice daily.
Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of
administration, the sex, age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician.
Cleaning compositions
In some embodiments, any one of compounds described herein, or a salt thereof, may be used inhibiting virulence of bacteria or reducing bacteria growth on a surface (e.g., for disinfecting or sanitizing a surface). The surface may be metallic, plastic, ceramic, or wooden, for example, the surface is a floor, a table, a kitchen counter, a cutting board, or a medical instrument. Hence, any one of the compounds of the present application may be used in a commercial setting for general disinfecting, e.g., in medical and food industries. For these purposes, the compound may be provided in a cleaning composition comprising an acceptable carrier. The carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the cleaning composition. Acceptable carriers that may be used in a cleaning composition of the present application include, but are not limited to, alcohols, water, surfactants, emollients, stabilizers, thickeners, viscosifiers, and fragrances.
Definitions
As used herein, the term "about" means "approximately" (e.g., plus or minus approximately 10% of the indicated value).
The term "compound" as used herein is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified.
The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, N=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. In some embodiments, the compound has the
Figure imgf000043_0001
In some embodiments, the compound has the (^-configuration.
Compounds provided herein also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
As used herein, the term "cell" is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal.
As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example,
"contacting" the inosine-monophosphate dehydrogenase (IMPDH) with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having IMPDH, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the IMPDH.
As used herein, the term "individual", "patient", or "subject" used
interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans. In some embodiment, the term "subject" does not refer to any animal other than human. In some embodiments, the subject is pediatric (e.g., from birth through age 21). In some embodiments, the subject has been identified as having a lung disease. In some embodiments, the lung disease is a structural lung disease. In some embodiments, the lung disease is selected from the group consisting of: cystic fibrosis, bronchiectasis, emphysema, and chronic obstructive pulmonary disease, and bronchiectasis. In some embodiments, the lung disease is cystic fibrosis.
As used herein, the phrase "effective amount" or "therapeutically effective amount" refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
As used herein the term "treating" or "treatment" refers to 1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder {i.e., arresting further development of the pathology and/or symptomatology), or 2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
As used herein, the term "preventing" or "prevention" of a disease, condition or disorder refers to decreasing the risk of occurrence of the disease, condition or disorder in a subject or group of subjects (e.g., a subject or group of subjects predisposed to or susceptible to the disease, condition or disorder). In some embodiments, preventing a disease, condition or disorder refers to decreasing the possibility of acquiring the disease, condition or disorder and/or its associated symptoms. In some embodiments, preventing a disease, condition or disorder refers to completely or almost completely stopping the disease, condition or disorder from occurring.
As used herein, the terms "resistant" and "persistent" (or "persister") refer to bacterial strains that exhibit a high level of tolerance to one or more antibiotics. In some embodiments, the bacterial strain is resistant when the MIC of the bacterial strain is at least 2x (2 -fold) of the MIC for the non-resistant strain. The x-fold resistant bacterial strain may be determined by the following steps: (i) MIC is determined for a non-resistant bacterial strain; (ii) the non-resistant bacterial strain is treated in a multi- well plate with an antibiotic at 2x, 5x, lOx etc, of the minimal inhibitory concentration (MIC); (iii) bacterial culture treated with the highest concentration that permitted bacterial growth is taken for serial passage for 100 days; and (iv) MIC of the bacterial culture after 100 days of serial passage is determined. If MIC of the bacterial culture after 100 days of serial passage is at least 2x of the MIC of the non-resistant strain, then the bacterial culture is at least 2x resistant to the antibiotic.
EXAMPLES
Materials and methods
C. elegans anti -infective activity assay: the assay was performed according to the methods and procedures similar to those previously described in Yuen, G.L. and F.M. Ausubel (2018) Both live and dead Enterococci activate Caenorhabditis elegans host defense via immune and stress pathways. Virulence PMID: 29436902. DOI: 10.1080/21505594.2018.1438025; Sifri, CD., J. Begun, F.M. Ausubel and S.B.
Calderwood (2003) Caenorhabditis elegans as a model host for Staphylococcus aureus pathogenesis. Infection and Immunity 71 :2208-2217. PMID: 12654843; PMCID: PMC 152095; Garsin, D.A., CD. Sifri, E. Mylonakis, X. Qin, K.V. Singh, B E.
Murray, S B. Calderwood and EM. Ausubel (2001) A simple model host for identifying gram-positive virulence factors. Proc. Natl. Acad. Sci. USA 98: 10892- 10897. PMID: 11535834; PMCID: PMC58570; Tan MW., and in Mahajan-Miklos S, Ausubel FM. Killing of Caenorhabditis elegans by Pseudomonas aeruginosa used to model mammalian bacterial pathogenesis. PNAS. 1999, 96(2), 715-20. Exemplary protocol is as follows: P. aeruginosa PA14 overnight cultures are plated on SK agar and incubated at 37 °C for 24 hours. Test compound is added to the agar, after which time plates are incubated at 25 °C for 24 hours. L4 stage C. elegans are transferred onto these plates (30-50 worms per plate) and survival is monitored until all of the worms are dead. Compounds resulting in significantly improved survival compared to mock-treated controls are considered active.
C. elegans is a natural bacterivore and can be raised easily in the laboratory by feeding on lawns non-pathogenic strains of Escherichia coli, especially E. coli strain OP50. A large variety of human pathogens kill C. elegans, including ! aeruginosa, Enterococcus faecalis, and S. aureus. In addition, P. aeruginosa, E. faecalis, and S. aureus kill C. elegans in liquid medium,
Animals: Female 5 to 6-week-old CD-I mice (18 - 22 gm) were used.
Animals were obtained from Harlan Laboratories and cared for in accordance with Guide for Care and Use of Laboratory Animals" (National Academy Press,
Washington DC, 2011). The health status of the animals was evaluated in accordance with accepted veterinary practice. Only animals considered acceptable for use in these studies were released from acclimation. Analgesia and anesthesia procedures were used whenever necessary to minimize discomfort, distress, pain, and injury of the mice. All mice were be euthanized by C02 inhalation prior to tissue collection.
Thigh infection assay: The mice were housed in groups of 3 with free access to food and water during the studies. The animals were made neutropenic by administration of cyclophosphamide (Cytoxan) on Days -4 and -1. Days listed are referenced from the date of infection (study day = Day 0). On Day 0, animals were inoculated intramuscularly (0.1 ml/thigh) with ~1 x 105 CFU/mouse of Pseudomonas aerginosa strain PA14, and Acinetobacter baumannii strain UNT 190-1 (expressing OXA-66 and OXA-23, two extended-spectrum beta-lactamases), into the right thigh. One group did not receive drug treatment and their thighs were harvested at 1 hour post-infection. The remaining mice were be administered test compounds at times and route of administration as appropriate. There were 3 dose groups for each Test Compound. Mice were euthanized by C02 inhalation at 24 hours post infection and thigh samples were taken. Thighs were aseptically removed, placed in 1-2 mL sterile PBS, homogenized, serially diluted and plated to determine CFU counts. Plates were incubated 18-24 hours under the appropriate strain conditions prior to counting. Colony counts were performed on agar plates. The number of colonies were converted to CFU/thigh by multiplying the number of colonies by the volume of the thigh homogenate spotted and the dilution at which the colonies were counted (5-50 colonies/spot). All count data were transformed into loglO CFU/thigh for calculation of means and standard deviations.
Example 1 - 6-fluoroaminobenzoic acid (6-FABA) dramatically attenuates the ability of P. aeruginosa to kill C. elegans.
Unexpectedly, it was found that 6-FABA is much more effective at inhibiting P. aeruginosa strain PA14-mediated killing of C. elegans than would be expected if its sole mode of action is to block PQS biosynthesis and MvfR-mediated QS signaling. This is unexpected, because Lesic et al. showed that 6-FAB A inhibits the biosynthesis of PQS, thereby disrupting MvfR-dependent gene expression, and blocks bacterial virulence in a murine thermal injury model of P. aeruginosa infection (See Ref. 53). That is, C. elegans infected with wild-type P. aeruginosa in the presence of 6-FABA survived much longer than C. elegans infected with P. aeruginosa mvfl or pqsA mutants (genes that encode the transcription factor and the first synthetic enzyme in the PQS system, respectively) in the absence of the drug.
To test whether 6-FABA blocks the ability of P. aeruginosa strain PA14 (hereafter PA14) to kill C. elegans animals, PA14 was used in "slow killing" assay as described by Tan et al (See Ref. 47). If 6-FABA completely blocked MvfR-mediated signaling, it would diminish the ability of PA14 to kill similarly to the level of killing that would be observed by infecting C. elegans with PA14 carrying null mutations in mvfl or pqsA. Unexpectedly, however, it was found that 4 mM 6-FABA, the maximum concentration achievable in agar (Figure 1), had a much more dramatic effect on blocking PA14-mediated killing than mutating mvfli or pqsA (Figures 8 and 2). The mvfR and pqsA mutations increased the LT50 (time until 50% killing) of the nematodes when feeding on PA 14 to approximately 90 hours compared to an LT50 of 50 hours when feeding on wild-type PA14, whereas 4 mM 6-FABA dramatically increased the LT50 to -160 hours. Further, addition of 4 mM 6-FABA to C. elegans killing assays conducted with PA 14 mvfii or pqsA mutants resulted in an additional attenuation of virulence, virtually identical to that of wild-type PA14 with the same amount of the drug (Figures 8 and 2). In addition, two compounds that were identified in a direct screen for inhibitors of pqsA expression, m50 and m59 (See Ref.
54)
Figure imgf000048_0001
inhibited C. elegans killing similarly to the pqsA and mvfR mutants and did not further cause an extension of the C. elegans lifespan (Figures 9 and 3). Thus, 6-FABA is much more effective at inhibiting ! aeruginosa strain PA14-mediated killing of C. elegans than would be expected if the sole mode of action of 6-FABA is to inhibit the MvfR quorum sensing regulon.
Supplementation of SK agar with tryptophan did not enhance the ability of PA14 to kill worms in the presence of 6-FABA (Figure 4). Moreover, a PA14 anthranilate auxotroph (PA14 Δ trpE Δ trpG AphnA), and consequentially a tryptophan auxotroph (See Refs. 55-56), was indistinguishable from wild-type PA14 in the C. elegans PA14 killing assay (Figure 5), suggesting that 6-FABA does not attenuate PA14 virulence by disrupting tryptophan synthesis. The fact that the anthanilate mutant phenocopied wild-type PA14 rather than an mvfR or pqsA mutant suggested that there was sufficient tryptophan in SK agar to support the growth of the mutant, consistent with the fact that it made a blue lawn similar to wild-type PA14 on SK agar, indicating the normal production of pyocyanin whose synthesis is also dependent on anthranilate (See Refs. 57-58). In combination, these data suggest that the effect of 6-FABA on blocking PA14-mediated killing of C. elegans is not likely a consequence of tryptophan limitation, but these experiments do not exclude the possibility that there is some level of tryptophan limitation that affects PA14 virulence but not growth and that cannot be rescued by exogenous tryptophan.
6-FABA did not activate the expression of a panel of C. elegans immune response reporter genes (including the genes daf-16, clec-60, irg-1, T24B8.5, sod-3, gst-4, F35E12.5, hspl6.2), suggesting that it does not target the C. elegans immune system.
Fluorinated compounds such as the fluorouracil may cause C. elegans infertility issues (See Ref. 59), and C. elegans sterile mutants or C. elegans animals treated with FUdR are more restistant to bacterial infections than fertile worms (Ref. 60). However, although a C. elegans ferl5-feml mutant is slightly more resistant to PA14 infection than wild-type worms, 6-FABA dramatically extend the lifespan of both the wild-type and sterile mutant on P. aeruginosa PA14 lawn (Figure 6).
Also, P. aeruginosa catalase mutants as well as a P. aeruginosa superoxide dismutase mutant were attenuated to the same extent of the wild-type PA14 in presence of 6-FABA (Figure 7), suggesting that the 6-FABA mode of action does not likely involve the generation of reactive oxygen species.
Example 2 - 6-FABA is catabolized to fluorocatechol (FCAT), which also attenuates C. elegans killing
When 6-FABA, which is colorless, is added to a lawn of PA14 on NGM agar, the agar accumulates a brown pigment over the course of 3 days (Figure 10). As shown in Figure 12 and in more detail in Figure 16, it was shown that anthranilate is catabolized to catechol via AntA, AntB, and AntC (See Refs. 57-58), suggesting that 6-FABA might be catabolized to 3-flourocatechol (FCAT) by the same enzymes. If catabolism of 6-FABA to FCAT contributes its higher than expected antivirulence activity, antA, antB, or antC mutants should be much less susceptible than wild-type PA14 to the ability of 6-FABA to rescue C. elegans from PA14-mediated killing. That is, the extent of killing observed by an antA, antB, or antC mutant in the presence of 6-FABA should be comparable to that observed with an mvjR mutant in the absence of 6-FABA, because 6-FABA should still be able to block MvfR-mediated signaling in an antABC mutant. It was observed that a PA 14 antA mutant was only modestly rescued by 6-FABA (LT50 = ~90 hours), similar to the extent of killing observed with mvjR in the absence of 6-FABA (LT50 = ~90 hours) (Figure 13). A similar result was obtained with a PA14 antC mutant (Figure 14). In addition, in contrast to wild-type PA14, lawns of PA14 antA, antB or antC mutants did not accumulate the brown- colored products in the presence of 6-FABA, in contrast to trpA and trpB mutants which still accumulated the brown pigment(s) (Figure 11).
The results in Figures 13 and 14 suggest that 6-FABA is catabolized by the antABC operon to 3-fluorocatechol (FCAT)
Figure imgf000050_0001
and that FCAT is a potent anti-infective compound in the C. elegans PA14 killing assay, similar to 6-FABA. Indeed, 3-fluorocatechol (FCAT) also rescued both wild- type PA14- and PA14 wv R-mediated killing to a significantly greater extent than a PA14 mvfl mutant without FCAT (Figure 15). FCAT was also able to inhibit killing of C. elegans by the antA mutant, attenuating the virulence of this mutant to a similar level as observed with wild type PA14 (Figure 15). Importantly, FCAT has rescuing activity in the C. elegans PA14 killing assay at a concentration that is 2-4 fold lower than the concentration required to affect the growth of PA14 in NGM media (Figures 13; 14; 17). These results suggest that FCAT itself is the active product downstream of 6-FABA that reduces PA14 virulence.
As shown in Figures 27-28, FCAT is able to extend the life span of C. elegans pmk-1, fshr-1 and zip-2 mutant worms, which are immunocompromised and hyper- susceptible to P. aeruginosa PA14-mediated killing. Importantly, the mode of action of FCAT is also orthogonal to known QS mechanisms in P. aeruginosa, as lasR, mvfli and the lasR-mvfR mutants are attenuated in the C. elegans assay but the worm lifespan is further extended in presence of FCAT (Figure 29).
An important feature of FCAT is that it functions as a potent anti-infective below its in vitro MIC. As shown in Figure 31, in the C. elegans assay, 0.5 mM FCAT (which is -20% of its in vitro MIC of -2.5 mM) exhibited potent activity. In contrast, when the traditional antibiotics carbenicillin or gentamicin were added to the P.
aeruginosa PA14 lawn (similarly to FCAT), carbenicillin failed to exhibit any anti- infective effect at 50X (7.5 mg/ml) its MIC (150 μg/ml) and gentamicin exhibited only moderate effects at IX, 10X and 50X of its MIC (0.15 μg/ml). The lack of efficacy of carbenicillin could be explained by lack of growth of the fully-grown bacterial lawn after the addition of the compound, as it is known that penicillin antibiotics only kill growing bacterial cells. In any case, the observation that FCAT has significant activity in the C. elegans killing assay at concentrations below its in vitro MIC, whereas carbenicillin has no effect and gentamicin only a modest effect, suggests that FCAT functions via a mode of action distinct from penicillins and aminoglycosides.
Example 3 - catechol derivatives attenuate P. aeruginosa killing
Catechol derivatives, including catechol itself, exhibited anti-virulence effect in the C. elegans assay (Figures 20-21), indicating the effect is not specific for fluorine substitution at the 3' position:
Figure imgf000051_0001
(BCAT), (3,5-CCAT), (4,5-CCAT),
Figure imgf000051_0002
(4-CCAT), and (4-FCAT).
Example 4 - FCAT is catabolized to fluoro-cis,cis-muconate (FMUC), which also attenuates P. aeruginosa killing
Figures 12 and 16 show that catechol is catabolized in P. aeruginosa by catechol 1,2-di oxygenase (CatA) to cis,cis-muconate, by CatB to muconolactone, and by CatC to 3-oxoadipate enol-lactone, which is then likely converted to either succinyl- or acetyl-CoA which feed into the citric acid cycle (See Ref. 61). FCAT is catabolized by CatAto fluoro-cis,cis-muconate FMUC)
Figure imgf000052_0001
and by CatB to 5-fluoro-muconolactone, which is then further converted to 2- maleylacetate by the P. aeruginosa enzyme encoded by PA2682 (See Ref. 62).
Mutation of PA 14 catA (Figure 22), but not catB, catC, a catBC double mutant, or PA2682 (Figures 23-24), attenuated the ability of FCAT to rescue C. elegans from P. aeruginosa killing. The LT50 of the catA mutant with FABA/FCAT exhibited dose dependency. The LT50 of catA with 4 mM FCAT was higher than the LT50 of wildtype PA14 with 4 mM FABA, but the LT50 of catA with 4 mM FABA was lower than the
LT50 of wildtype PA14 with 4 mM FABA (Figure 22). In addition, FCAT is not active against bacterial species that lack the catA gene, such as E. faecalis and S. aureus (Figures 25-26), These data suggest that fluoro-cis,cis-muconate derived from 6- FAB A or FCAT is active compound that contributes to the anti-infective activities independently shown for 6-FAB A and FCAT.
Kinetics of the conversion of FCAT to FMUC and its effect on FCAT efficacy are shown in Figure 30. When FCAT is added to the bacterial lawn simultaneously with the worms or the worms were added 6 hours after the addition of FCAT, the LT50 increase was moderate (-50%). However, with increasing times of incubation of FCAT with the bacterial lawn, the large the observed increase in LT50, reaching
-150% if bacteria were pre-treated with FCAT for 24 hours before the worms were added. This data indicate that the uptake of FCAT and its conversion to FMUC is a relatively slow process. Example 5 - halogenated compounds inhibit bacterial virulence
3-bromopyruvate (BP) also blocks P. aeruginosa virulence in the C. elegans model (Figure 32). Iodoacetate and iodoacetamide (Figure 33) have strong activity in the C. elegans assay. Table 1 shows MICs and in vivo effective doses of halogenated compounds, FCAT and gentamicin for inhibiting growth of P. aeruginosa in vitro and blocking the ability of P. aeruginosa to kill C. elegans in vivo. Table 1
Figure imgf000053_0001
Example 6 - antibacterial compounds
Test compounds were tested in C. elegans assay at 100 μΜ (20-30 μg/ml). At this concentration, all compounds were shown to significantly inhibit bacterial virulence and reduce bacterial growth to > 3 sigma. The structures of the test compounds are shown in Table 2, and the in vitro MIC data for the test compounds is
Figure imgf000053_0002
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Table 3
Figure imgf000056_0002
clonidine
>50 >50 >50 >50 >50 >50 >50 hydrochloride
betonicine 75 >300 300 75 75 >300 >300
Several compounds, including 4-hydroxy-3-nitrophenyl acetic acid, nisodipine, mechlorethamine, dimetridazole, and HMS3408N17 exhibited in vivo efficacy in the C. elegans model, similarly to ribavirin (Figures 36-37). 4-hydroxy-3- nitrophenyl acetic acid and mangafodipir exhibited excellent MICs against E.coli and K. pneumoniae. Figure 38 shows percent survival of C. elegans worms in response to treatment with various concentrations of FCAT, BP, ribavirin, and mechlorethamine HC1.
Example 7 - Antibacterial activity of ribavirin
Ribavirin was tested (via single bolus intraperitoneal administration) in a standard neutropenic mice thigh infection model. The thigh model is broadly accepted as a model for antibiotic in vivo efficacy (See Refs. 44 and 66-69). As shown in
Figure 34, ribavirin is active against P. aeruginosa PA14 in the thigh model, at 150 mg/kg. The effect is comparable to 75 mg/kg meropenem. At 300 mg/kg which is still well-tolerated by the animal, the drug knocks down the bacterial titer in the thigh to the detection limit. Ribavirin is also active against an extended spectrum beta- lactamase (ESBL) expressing baumannii strain UNT190-1 (Figure 35). The strain is resistant to 300 mg/kg meropenem treatment. Ribavirin, at 75 mg/kg, which is within the achievable dose of its current anti-viral indication, knocks down the bacterial titer in the thigh to the detection limit, which is superior to the levofloxacin control at 200 mg/kg.
It is noteworthy that the published MIC concentrations for ribavirin are very high, such that based on the published data ribavirin would not be selected for a development as antibiotic drug. Typically, it is necessary to achieve a concentration higher or equal to the MIC for an antibiotic compound to be efficacious in a patient. The published IP LD50 of ribavirin in mice is 0.9-1.3 mg/ml. Thus, based on published MICs for ribavirin of 3-10 mg/ml (See Refs. 78-79), ribavirin would likely be eliminated from any further studies because the MIC is not only very high but also in the toxic range.
Surprisingly and unexpectedly, in the neutropenic mouse deep thigh infection study, the effective dose of ribavirin for P. aeruginosa and A. baumannii was about 0.15 mg/ml and about 0.075 mg/ml, respectively, 50-100 fold below the MICs.
Similarly, in the C. elegans assay, the effective concentration of ribavirin against P. aeruginosa was 0.025-0.25 mg/ml, 30-300 fold below the MIC.
To verify the MIC of ribavirin published by Kruszewska (See Refs. 78-79), MIC measurements were carried out according to the standard CLSI protocol (Ml 00) in Mueller-Hinton broth (MHB). In these MIC studies, same P. aeruginosa strain was used as was used by Kruszewska (ATCC 15442), two benchmark strains (P.
aeruginosa ATCC 27853 a d E. coli ATCC 25922), two additional P. aeruginosa strains (PA 14 and PA3924) and two ,4. baumannii strains (AB190 and AB 197).
Commercial antibiotic tobramycin (TOB) was used as a control. The MIC of TOB against the benchmark strains in Mueller-Hinton Broth (MHB) was in accordance with the published range of 0.25 - 1 μg/ml. The results are summarized in the Table 4.
Table 4
Figure imgf000058_0001
Kruszewska determined MICs in MHB2 (Cation-adjusted MHB) medium using a paper disc growth inhibition assay on agar plates. The present MIC
measurements in MHB in a microdilution format generated results similar to those obtained by Kruszewska. That is, in the present experiment, the MIC of ribavirin against the P. aeruginosa strains ranged from 2-8 mg/ml. A. baumanii was more resistant to ribavirin than P. aeruginosa with an MIC > 16 mg/ml. Based in this data, ribavirin may not be expected to act as an antibacterial agent. Yet, in sharp contrast with this conclusion, ribavirin efficiently inhibited bacterial virulence and reduced bacterial growth in the in vivo C. Elegans essay and in in vivo mouse deep thing infection model. Example 8 - Antibacterial activity of ribavirin phosphate
Spontaneous mutants of PA14 were isolated with the spontaneous mutation frequency to ribavirin resistance of approximately 1 x 10"7. 30 independent ribavirin- resistant mutants from 30 individual cultures starting from different single colonies were isolated. The ribavirin-resistant phenotype of the mutants was verified by re- streaking onto fresh pyruvate plates containing ribavirin. Three mutants were tested in the C. elegans-P. aeruginosa "slow" killing assay to confirm that the strains are "blind" to ribavirin (See Figure 42, the mutants kill C. elegans at similar rate in presence or absence of the drug). However, these mutants were not resistant to 3- fluorocatechol (FCAT). Assuming that FCAT and ribavirin have the same target as suggested in previous examples, these results indicate that the mutants are most likely not in the target of ribavirin and FCAT, but rather in a gene that encodes an enzyme that converts ribavirin to its active metabolite that further acts as an anti- !
Aeruginosa agent.
To identify the mutations in the ribavirin-resistant mutants that confer resistance to ribavirin, whole genomes of 23 mutants, plus the parent wild-type strain (that was used to generate these mutants), were sequenced following guidelines of the MGH NGS core and instructions from Illumina. Analysis of the NGS data showed that out of the 23 mutants, 21 of them have mutations in the gene PA14 62230. This gene encodes a hypothetical protein that has a predicted C-terminal kinase domain (See Figure 41, section A). Figure 41 contains (A) a predicted 153 AA C-terminal kinase domain of PA14 62230, using the Phyre 2 server; (B) heat chart showing bacterial growth in a media, where ribavirin is diluted from 10 mM to about 20 μΜ in 2X serial dilution steps from column 2 to 11; and (C) a line plot showing survival of C. elegans on PA14 wild-type and two transposon insertion mutants in PA14 62230.
Careful examination of the mutations showed that most of the mutations occur at conserved amino acid residues that are known to be important for kinase function, suggesting that the gene product of PA14 62230 is most likely phosphorylating ribavirin in vivo (Figure 41, section A). Previously-generated transposon insertion mutants of PA14 62230 were then tested. The transposon mutants were "blind" to in the C. elegans survival assay (Figure 41, section C), similarly to the spontaneous ribavirin-resistant mutants tested in Figure 42. These results suggest that for ribavirin, the molecule that has antibacterial activity in the C. elegans-P. aeruginosa killing assay is the phosphorylated form of ribavirin, ribavirin-5-monophosphate (RMP):
Figure imgf000060_0001
In investigating mechanism of anti -viral activity of RMP, it became known that RMP targets inosine-monophosphate dehydrogenase (FMPDH) (See Figure 40). As shown in Figure 39, section A, the role of FMPDH is to convert inosine- monophosphate (FMP) to xathosine-monophosphate (XMP), a precursor of GMP When FMPDH is blocked, the bacteria cannot make GMP but can still make AMP. Therefore, if guanosine is added to the assays, it is expected to antagonize the effect of FMPDH inhibitors, whereas adenosine would not affect FMPDH inhibitors. The assay confirms this result, see Figure 39, section B. That is, guanosine but not adenosine blocks the ability of ribavirin to prevent bacterial growth, suggesting that FMPDH is the likely target of ribavirin.
Figure 39 contains (A) a scheme showing nucleotide metabolism; and (B) heat charts showing the effect of adenosine (upper, A) and guanosine (G) against ribavirin. As the results show, adenosine has no significant antagonistic effect whereas guanosine exhibits a concentration dependent antagonistic effect.
REFERENCES
1. Boucher, H.W., et al., Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis, 2009. 48(1): p. 1-12.
2. Spellberg, B., et al., The epidemic of antibiotic-resistant infections: a call to action for the medical community from the Infectious Diseases Society of America. Clin Infect Dis, 2008. 46(2): p. 155-64.
3. Rice, L.B., Unmet medical needs in antibacterial therapy. Biochem
Pharmacol, 2006. 71(7): p. 991-5.
5. De Francesco, M.A., et al., Prevalence of multidrug-resistant Acinetobacter baumannii and Pseudomonas aeruginosa in an Italian hospital. J Infect Public Health, 2013. 6(3): p. 179-85. 7. Gandra, S., et al., Trends in antibiotic resistance among major bacterial pathogens isolated from blood cultures tested at a large private laboratory network in India, 2008-2014. Int J Infect Dis, 2016. 50: p. 75-82.
8. Xu, T., et al., A 4-year surveillance of antimicrobial resistance patterns of Acinetobacter baumanni in a university-affiliated hospital in China. J Thorac Dis,
2013. 5(4): p. 506-12.
9. Ventola, C.L., The antibiotic resistance crisis: part 1: causes and threats. P T, 2015. 40(4): p. 277-83.
10. Payne, D.J., et al., Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat Rev Drug Discov, 2007. 6(1): p. 29-40.
11. Cook, D., et al., Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework. Nat Rev Drug Discov, 2014. 13(6): p. 419-31.
12. Clatworthy, A.E., E. Pierson, and D.T. Hung, Targeting virulence: a new paradigm for antimicrobial therapy. Nat Chem Biol, 2007. 3(9): p. 541-8.
13. Geske, G.D., et al., Small molecule inhibitors of bacterial quorum sensing and biofilm formation. J Am Chem Soc, 2005. 127(37): p. 12762-3.
14. Hentzer, M., et al., Inhibition of quorum sensing in Pseudomonas aeruginosa biofilm bacteria by a halogenated furanone compound. Microbiology, 2002. 148(Pt 1): p. 87-102.
15. Hentzer, M., et al., Attenuation of Pseudomonas aeruginosa virulence by quorum sensing inhibitors. EM BO J, 2003. 22(15): p. 3803-15.
16. Hung, D.T., et al., Small-molecule inhibitor of Vibrio cholerae virulence and intestinal colonization. Science, 2005. 310(5748): p. 670-4.
17. Lyon, G.J., et al., Reversible and specific extracellular antagonism of receptor-histidine kinase signaling. J Biol Chem, 2002. 277(8): p. 6247-53.
18. Manefield, M., et al., Halogenated furanones inhibit quorum sensing through accelerated LuxR turnover. Microbiology, 2002. 148(Pt 4): p. 1119-27.
19. Manefield, M., et al., Halogenated furanones from the red alga, Delisea pulchra, inhibit carbapenem antibiotic synthesis and exoenzyme virulence factor production in the phytopathogen Erwinia carotovora. FEMS Microbiol Lett, 2001. 205(1): p. 131-8. 20. Mayville, P., et al., Structure-activity analysis of synthetic autoinducing thiolactone peptides from Staphylococcus aureus responsible for virulence. Proc Natl Acad Sci U S A, 1999. 96(4): p. 1218-23.
21. Muh, U., et al., Novel Pseudomonas aeruginosa quorum-sensing inhibitors identified in an ultra-high-throughput screen. Antimicrob Agents Chemother, 2006.
50(11): p. 3674-9.
22. Smith, K.M., Y. Bu, and H. Suga, Induction and inhibition of Pseudomonas aeruginosa quorum sensing by synthetic autoinducer analogs. Chem Biol, 2003. 10(1): p. 81-9.
23. Wright, J.S., 3rd, R. Jin, and R.P Novick, Transient interference with staphylococcal quorum sensing blocks abscess formation. Proc Natl Acad Sci U S A, 2005. 102(5): p. 1691-6.
24. Wu, H., et al., Synthetic furanones inhibit quorum-sensing and enhance bacterial clearance in Pseudomonas aeruginosa lung infection in mice. J Antimicrob Chemother, 2004. 53(6): p. 1054-61.
25. Moayeri, M., et al., Cisplatin inhibition of anthrax lethal toxin. Antimicrob Agents Chemother, 2006. 50(8): p. 2658-65.
26. Sarac, M.S., et al., Protection against anthrax toxemia by hexa-D-arginine in vitro and in vivo. Infect Immun, 2004. 72(1): p. 602-5.
27. Shoop, W.L., et al., Anthrax lethal factor inhibition. Proc Natl Acad Sci U S A, 2005. 102(22): p. 7958-63.
28. Turk, B.E., et al., The structural basis for substrate and inhibitor selectivity of the anthrax lethal factor. Nat Struct Mol Biol, 2004. 11(1): p. 60-6.
29. Bailey, L., et al., Small molecule inhibitors of type III secretion in Yersinia block the Chlamydia pneumoniae infection cycle. FEBS Lett, 2007. 581(4): p. 587-95.
30. Kauppi, A.M., et al., Targeting bacterial virulence: inhibitors of type III secretion in Yersinia. Chem Biol, 2003. 10(3): p. 241-9.
31. Muschiol, S., et al., A small-molecule inhibitor of type III secretion inhibits different stages of the infectious cycle of Chlamydia trachomatis. Proc Natl Acad Sci U S A, 2006. 103(39): p. 14566-71.
32. Nordfelth, R., et al., Small-molecule inhibitors specifically targeting type III secretion. Infect Immun, 2005. 73(5): p. 3104-14. 33. Hudson, D.L., et al., Inhibition of type III secretion in Salmonella enterica serovar Typhimurium by small-molecule inhibitors. Antimicrob Agents Chemother, 2007. 51(7): p. 2631-5.
34. Pinkner, J.S., et al., Rationally designed small compounds inhibit pilus biogenesis in uropathogenic bacteria. Proc Natl Acad Sci U S A, 2006. 103(47): p. 17897-902.
35. Svensson, A. , et al . , Design and evaluation of pilicides: potential novel antibacterial agents directed against uropathogenic Escherichia coli. Chembiochem, 2001. 2(12): p. 915-8.
36. Kirienko, N. V, et al., Pseudomonas aeruginosa disrupts Caenorhabditis elegans iron homeostasis, causing a hypoxic response and death. Cell Host Microbe, 2013. 13(4): p. 406-16.
37. Moy, T.I., et al., Identification of novel antimicrobials using a live-animal infection model. Proc Natl Acad Sci U S A, 2006. 103(27): p. 10414-9.
38. Moy, T.I., et al., High-throughput screen for novel antimicrobials using a whole animal infection model. ACS Chem Biol, 2009. 4(7): p. 527-33.
39. Bolz, D.D., J.L. Tenor, and A. Aballay, A conserved PMK-l/p38 MAPK is required in caenorhabditis elegans tissue-specific immune response to Yersinia pestis infection. J Biol Chem, 2010. 285(14): p. 10832-40.
40. Feinbaum, R.L., et al., Genome-wide identification of Pseudomonas aeruginosa virulence-related genes using a Caenorhabditis elegans infection model.
PLoS Pathog, 2012. 8(7): p. el002813.
41. Irazoqui, J.E., et al., Distinct pathogenesis and host responses during infection of C. elegans by P. aeruginosa and S. aureus. PLoS Pathog, 2010. 6: p. el000982. 42. Kim, D.H., et al., A conserved p38 MAP kinase pathway in Caenorhabditis elegans innate immunity. Science, 2002. 297(5581): p. 623-6.
43. Lee, D.G., et al., Genomic analysis reveals that Pseudomonas aeruginosa virulence is combinatorial. Genome Biol, 2006. 7(10): p. R90.
44. Mahajan-Miklos, S., et al., Molecular mechanisms of bacterial virulence elucidated using a Pseudomonas aeruginosa-Caenorhabditis elegans pathogenesis model. Cell, 1999. 96(1): p. 47-56.
45. Sifri, CD., et al., Caenorhabditis elegans as a model host for Staphylococcus aureus pathogenesis. Infect Immun, 2003. 71(4): p. 2208-17. 46. Sifri, CD., et al., Virulence effect of Enterococcus faecalis protease genes and the quorum-sensing locus fsr in Caenorhabditis elegans and mice. Infect Immun, 2002. 70(10): p. 5647-50.
47. Tan, M.W., S. Mahajan-Miklos, and F.M. Ausubel, Killing of Caenorhabditis elegans by Pseudomonas aeruginosa used to model mammalian bacterial
pathogenesis. Proc Natl Acad Sci U S A, 1999. 96(2): p. 715-20.
48. Conery, A.L., et al., High-throughput screening for novel anti-infectives using a C. elegans pathogenesis model. Curr Protoc Chem Biol, 2014. 6(1): p. 25-37.
49. Kim, W. , et al . , Identification of an Antimicrobial Agent Effective against Methicillin-Resistant Staphylococcus aureus Per sisters Using a Fluorescence-Based Screening Strategy. PLoS One, 2015. 10(6): p. e0127640.
50. Rajamuthiah, R., et al., Whole animal automated platform for drug discovery against multi-drug resistant Staphylococcus aureus. PLoS One, 2014. 9(2): p. e89189.
51. Walsh, C, Where will new antibiotics come from? Nat Rev Microbiol, 2003. 1(1): p. 65-70.
52. Pesci, E.C., et al., Quinolone signaling in the cell-to-cell communication system ofPseudomonas aeruginosa. Proc Natl Acad Sci U S A, 1999. 96(20): p.
11229-34.
53. Lesic, B., et al., Inhibitors of pathogen intercellular signals as selective anti- infective compounds. PLoS Pathog, 2007. 3(9): p. 1229-39.
54. Starkey, M., et al., Identification of anti-virulence compounds that disrupt quorum-sensing regulated acute and persistent pathogenicity. PLoS Pathog, 2014. 10(8): p. el004321.
55. Essar, D.W., et al., Identification and characterization of genes for a second anthranilate synthase in Pseudomonas aeruginosa: interchangeability of the two anthranilate synthases and evolutionary implications. J Bacterid, 1990. 172(2): p. 884-900.
56. Gilbert, K.B., et al., Global position analysis of the Pseudomonas aeruginosa quorum-sensing transcription factor LasR. Mol Microbiol, 2009. 73(6): p. 1072-85. 57. Mavrodi, D.V., W. Blankenfeldt, and L.S. Thomashow, Phenazine compounds in fluorescent Pseudomonas spp. biosynthesis and regulation. Annu Rev Phytopathol, 2006. 44: p. 417-45. 58. Mavrodi, D. V, et al., [A genetic locus controlling biosynthesis of phenazine antibiotic in Pseudomonas fluorescens strain 2-79]. Dokl Akad Nauk, 1997. 352(1): p. 117-20.
59. Zhang, S., et al., The Analysis of Gene Expression on Fertility Decline in Caenorhabditis elegans after the Treatment with 5-Fluorouracil. Iran J Public Health, 2015. 44(8): p. 1061-71.
60. Mitchell, D.H., et al., Synchronous growth and aging of Caenorhabditis elegans in the presence of fluorodeoxyuridine. J Gerontol, 1979. 34(1): p. 28-36.
61. Ornston, L.N., The conversion of catechol and protocatechuate to beta- ketoadipate by Pseudomonas putida. 3. Enzymes of the catechol pathway. J Biol Chem, 1966. 241(16): p. 3795-9.
62. Rosenberg, S.L. and G.D. Hegeman, Genetics of the mandelate pathway in Pseudomonas aeruginosa. J Bacterid, 1971. 108(3): p. 1270-6.
63. Ganapathy-Kanniappan, S., et al., Glyceraldehyde-3-phosphate
dehydrogenase (GAPDH) is pyruvylated during 3-bromopyruvate mediated cancer cell death. Anticancer Res, 2009. 29(12): p. 4909-18.
64. Liberati, N.T., et al., An ordered, nonredundant library of Pseudomonas aeruginosa strain PA 14 transposon insertion mutants. Proc Natl Acad Sci U S A, 2006. 103(8): p. 2833-8.
65. Tan, M.W. and F.M. Ausubel, Caenorhabditis elegans: a model genetic host to study Pseudomonas aeruginosa pathogenesis. Curr Opin Microbiol, 2000. 3(1): p. 29- 34.
66. Andes, D. and W. A. Craig, In vivo activities of amoxicillin and amoxicillin- clavulanate against Streptococcus pneumoniae: application to breakpoint
determinations. Antimicrob Agents Chemother, 1998. 42(9): p. 2375-9.
67. Drusano, G.L., Infection site concentrations: their therapeutic importance and the macrolide and macrolide -like class of antibiotics. Pharmacotherapy, 2005. 25(12 Pt 2): p. 150S-158S.
68. Nuermberger, E., Murine models of pneumococcal pneumonia and their applicability to the study of tissue-directed antimicrobials. Pharmacotherapy, 2005. 25(12 Pt 2): p. 134S-139S.
69. Retsema, J. A., et al., Preferential concentration of azithromycin in an infected mouse thigh model. J Antimicrob Chemother, 1993. 31 Suppl E: p. 5-16. 70. Graci, J.D. and C.E. Cameron, Mechanisms of action of ribavirin against distinct viruses. Rev Med Virol, 2006. 16(1): p. 37-48.
71. Paeshuyse, J., K. Dallmeier, and J. Neyts, Ribavirin for the treatment of chronic hepatitis C virus infection: a review of the proposed mechanisms of action. Curr Opin Virol, 2011. 1(6): p. 590-8.
72. Xiao, G., E. Deziel, J. He, F. Lepine, B. Lesic, M. H. Castonguay, S. Milot, A. P. Tampakaki, S. E. Stachel, and L. G. Rahme. 2006. MvfR, a key Pseudomonas aeruginosa pathogenicity LTTR-class regulatory protein, has dual ligands. Mol Microbiol 62: 1689-99.
73. Zhang, Q., J. Pan, P. E. North, S. Yang, R. A. Lubet, Y. Wang, and M. You.
2012. Aerosolized 3-bromopyruvate inhibits lung tumorigenesis without causing liver toxicity. Cancer Prev Res (Phila) 5:717-25.
74. Hedstrom, L., IMP dehydrogenase: structure, mechanism, and inhibition. Chem Rev, 2009. 109(7): p. 2903-28.
75. Hedstrom, L., et al., The antibiotic potential of prokaryotic IMP
dehydrogenase inhibitors. Curr Med Chem, 2011. 18(13): p. 1909-18.
76. Munoz-Elias, E. J. & McKinney J. D. Mycobacterium tuberculosis isocitrate lyases 1 and 2 are jointly required for in vivo growth and virulence. Nat Med 11(6): 638-644.
jointly required for in vivo growth and virulence. Nat Med 11(6): 638-644.
77. Sacchettini J. C, Rubin E.J. and Freundlich J. S. Drugs versus bugs: in pursuit of the persistent predator Mycobacterium tuberculosis. Nat Rev Microbiol 6: 41-52. 59(6): 436-9.
78. Kruszewska, H. 2002. Search of antimicrobial activity of selected non- antibiotic drugs. Acta Pol Pharm 59(6): 436-9.
79. Kruszewska. H. 2010. Examination of antimicrobial activity of selected non- antibiotics products. Acta Pol Pharm 67(6): 733-6.
80. Tan MW., Mahajan-Miklos S, Ausubel FM. Killing of Caenorhabditis elegans by Pseudomonas aeruginosa used to model mammalian bacterial pathogenesis. PNAS. 1999, 96(2), 715-20. OTHER EMBODIMENTS
It is to be understood that while the present application has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the present application, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

What is claimed is
1. A method of treating a bacterial infection in a subject, the method comprising
administering to the subject in need thereof a therapeutically effective amount of a compound selected from the group consisting of:
Figure imgf000068_0001
or a pharmaceutically acceptable salt thereof.
2. The method of claim 1, wherein the bacterial infection is caused by Gram-positive bacteria.
3. The method of claim 1, wherein the bacterial infection is caused by Gram-negative bacteria.
4. The method of any one of claims 1-3, wherein the bacterial infection is caused by an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P.
aeruginosa, or Enterobacter).
5. The method of claim 4, wherein the ESKAPE pathogen is selected from P.
aeruginosa and A. baumannii.
6. The method of claim 1 or claim 2, wherein the bacterial infection is caused by a bacterium selected from the group consisting of: S. aureus and E. faecalis.
7. A method of treating a bacterial infection in a subject, the method comprising
administering to the subject in need thereof a therapeutically effective amount of a compound selected from the group consisting of:
Figure imgf000069_0001
or a pharmaceutically acceptable salt thereof.
8. The method of claim 7, wherein the bacterial infection is caused by Gram-negative bacteria.
9. The method of claim 8, wherein the bacterial infection is caused by an ESKAPE
pathogen.
10. The method of claim 9, wherein the ESKAPE pathogen is selected from P.
aeruginosa and A. baumannii.
11. A method of treating a bacterial infection comprising administering to a subject in need thereof a therapeutically effective amount of a 2-amino-6-fluorobenzoic acid (6- FABA) having the following structure:
Figure imgf000069_0002
or a pharmaceutically acceptable salt thereof, wherein the bacterial infection is caused by a bacterium that does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
12. The method of any one of claims 1-11, wherein the bacterial infection is selected from the group consisting of: nosocomial infection, skin infection, respiratory infection, wound infection, endovascular infection, CNS infection, abdominal infection, blood stream infection, urinary tract infection, pelvic infection, invasive systemic infection, gastrointestinal infection, dental infection, zoonotic infection, and connective tissue infection.
13. The method of any one of claim 1-11, wherein the bacterial infection is selected from the group consisting of: atopic dermatitis, sinusitis, food poisoning, abscess, pneumonia, meningitis, osteomyelitis, endocarditis, bacteremia, sepsis, and urinary tract infection.
14. The method of any one of claims 1-13, wherein the compound is administered to the subject by a route selected from the group consisting of: oral, sublingual,
gastrointestinal, rectal, topical, intradermal, subcutaneous, nasal, intravenous, and intramuscular.
15. The method of any one of claims 1-14, wherein the subject has been identified as having a lung disease.
16. The method of claim 15, wherein the lung disease is a structural lung disease.
17. The method of claim 14 or claim 15, wherein the lung disease is selected from the group consisting of: cystic fibrosis, bronchiectasis, emphysema, and chronic obstructive pulmonary disease, and bronchiectasis.
18. The method of claim 17, wherein the lung disease is cystic fibrosis.
19. The method of any one of claims 1-18, wherein the subject is a pediatric subject.
20. The method of any one of claims 1-19, wherein the compound is administered to the subject in combination with at least one additional therapeutic agent.
21. The method of claim 20, wherein the additional therapeutic agent is an antibiotic.
22. The method of claim 21, wherein the antibiotic is selected from the group consisting of: a quinolone, a β-lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof.
23. The method of any one of claims 20-22, wherein the compound and the additional therapeutic agent are administered consecutively.
24. The method of any one of claims 20-22, wherein the compound and the additional therapeutic agent are administered simultaneously.
25. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
Figure imgf000071_0001
or a pharmaceutically acceptable salt thereof.
26. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
Figure imgf000071_0002
or pharmaceutically acceptable salt thereof.
27. The method of claim 25 or claim 26, wherein the bacteria is Gram-positive.
28. The method of claim 25 or claim 26, wherein the bacteria is Gram-negative
29. The method of claim 25 or claim 26, the bacteria is an ESKAPE pathogen (E.
faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or Enterobacter).
30. The method of claim 29, wherein the bacteria is selected from P. aeruginosa and baumannii.
31. The method of claim 25 or claim 26, wherein the bacteria is selected from the group consisting of: S. aureus and E. faecalis.
32. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
Figure imgf000072_0001
or a pharmaceutically acceptable salt thereof.
33. The method of claim 32, wherein the bacteria is a Gram-negative bacteria.
34. The method of claim 33, wherein the bacteria is an ESKAPE pathogen.
35. The method of claim 34, wherein the ESKAPE pathogen is selected from P.
aeruginosa and A. baumannii.
36. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
Figure imgf000073_0001
or pharmaceutically acceptable salt thereof, wherein the bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
37. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from the group consisting of:
Figure imgf000073_0002
or a pharmaceutically acceptable salt thereof, wherein the bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
38. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a 2-amino-6-fluorobenzoic acid (6-FABA) having the following structure:
Figure imgf000073_0003
or a pharmaceutically acceptable salt thereof, wherein the bacteria does not use quorum sensing (QS) activated by an extracellular 4-hydroxy-2-alkylquinoline (HAQ).
39. The method of any one of claims 25-38, wherein the bacteria is contacted in vitro.
40. The method of any one of claims 25-38, wherein the bacteria is contacted in vivo.
41. The method of any one of claims 25-38, wherein the bacteria is contacted ex vivo.
42. The method of any one of claims 25-41, wherein the effective amount of the
compound is at least 20% less than MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
43. The method of claim 42, wherein the effective amount of the compound is about 5- fold lower than MIC of the compound as determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
44. A method of treating a bacterial infection in a subject, the method comprising
administering to the subject in need thereof a therapeutically effective amount of a compound selected from:
Figure imgf000074_0001
Figure imgf000075_0001
74
Figure imgf000076_0001
Figure imgf000077_0001
or a pharmaceutically acceptable salt thereof.
45. The method of claim 44, wherein the bacterial infection is caused by Gram-positive bacteria.
46. The method of claim 44, wherein the bacterial infection is caused by Gram-negative bacteria.
47. The method of claim 44, wherein the bacterial infection is caused by an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or Enter obacter).
48. The method of claim 47, wherein the ESKAPE pathogen is P. aeruginosa
49. The method of claim 47, wherein the ESKAPE pathogen is A. baumannii.
50. The method of claim 49, wherein the baumannii strain is meropenem -resistant.
51. The method of any one of claims 44-50, wherein the therapeutically effective amount of the compound is in a range of about 4 mg/kg to about 45 mg/kg.
52. The method of any one of claims 44-51, wherein the bacterial infection is selected from the group consisting of: nosocomial infection, skin infection, respiratory infection, wound infection, endovascular infection, CNS infection, abdominal infection, blood stream infection, urinary tract infection, pelvic infection, invasive systemic infection, gastrointestinal infection, dental infection, zoonotic infection, and connective tissue infection.
53. The method of any one of claims 44-51, wherein the bacterial infection is selected from the group consisting of: atopic dermatitis, sinusitis, food poisoning, abscess, pneumonia, meningitis, osteomyelitis, endocarditis, bacteremia, sepsis, and urinary tract infection.
54. The method of any one of claims 44-53, wherein the compound is administered to the subject by a route selected from the group consisting of oral, sublingual,
gastrointestinal, rectal, topical, intradermal, subcutaneous, nasal, intravenous, and intramuscular.
55. The method of any one of claims 44-54, wherein the subject has been identified as having a lung disease.
56. The method of claim 55, wherein the lung disease is a structural lung disease.
57. The method of claim 55 or claim 56, wherein the lung disease is selected from the group consisting of cystic fibrosis, bronchiectasis, emphysema, and chronic obstructive pulmonary disease, and bronchiectasis.
58. The method of claim 56 or claim 57, wherein the lung disease is cystic fibrosis.
59. The method of any one of claims 44-58, wherein the subject is a pediatric subject.
60. The method of any one of claims 44-59, wherein the compound is administered to the subject in combination with at least one additional therapeutic agent.
61. The method of claim 60, wherein the additional therapeutic agent is an antibiotic.
62. The method of claim 61, wherein the additional therapeutic agent is an antibiotic selected from the group consisting of: a quinolone, a β-lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof.
63. The method of claim 62, wherein the aminoglycoside is tobramycin.
64. The method of any one of claims 61-63, wherein the compound and the additional therapeutic agent are administered consecutively.
65. The method of any one of claims 61-63, wherein the compound and the additional therapeutic agent are administered simultaneously.
66. A method of inhibiting inosine-monophosphate dehydrogenase (EVIPDH) in a
bacteria, the method comprising contacting the bacteria with a compound selected from:
Figure imgf000079_0001
Figure imgf000080_0001
or a pharmaceutically acceptable salt thereof.
67. The method of claim 66, wherein the bacteria is contacted in vitro.
68. The method of claim 66, wherein the bacteria is contacted in vivo.
69. The method of claim 66, wherein the bacteria is contacted ex vivo.
70. The method of any one of claims 66-69, wherein the bacteria is Gram-positive.
71. The method of any one of claims 66-69, wherein the bacteria is Gram-negative.
72. The method of any one of claims 66-69, wherein the bacteria is an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or
Enter obacter).
73. The method of claim 72, wherein the ESKAPE pathogen is P. aeruginosa.
74. The method of claim 72, wherein the ESKAPE pathogen is A. baumannii.
75. The method of claim 74, wherein the baumannii strain is meropenem -resistant.
76. A method of inhibiting virulence of a bacteria, the method comprising contacting the bacteria with an effective amount of a compound selected from:
Figure imgf000080_0002
Figure imgf000081_0001
Figure imgf000082_0001
81
Figure imgf000083_0001
77. The method of claim 76, wherein an effective amount of ribavirin is at least 20% less than MIC of the test compound determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
78. The method of claim 76, wherein the effective amount of ribavirin is about 30 to about 300 times below the MIC of the test compound determined in a conventional in vitro bacterial growth inhibition or bacterial killing assay.
79. The method of any one of claims 76-78, wherein the bacteria is contacted in vitro.
80. The method of any one of claims 76-78, wherein the bacteria is contacted in vivo.
81. The method of any one of claims 76-78, wherein the bacteria is contacted ex vivo.
82. The method of any one of claims 76-81, wherein the bacteria is Gram-positive.
83. The method of any one of claims 76-81, wherein the bacteria is Gram-negative.
84. The method of any one of claims 76-83, wherein the bacteria is an ESKAPE pathogen (E. faecium, S. aureus, K pneumoniae, A. baumannii, P. aeruginosa, or
Enter obacter).
85. The method of claim 84, wherein the ESKAPE pathogen is P. aeruginosa.
86. The method of claim 84, wherein the ESKAPE pathogen is A. baumannii.
87. The method of claim 86, wherein the baumannii strain is meropenem -resistant.
88. A pharmaceutical composition comprising a compound selected from:
Figure imgf000084_0001
Figure imgf000085_0001
or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
89. The pharmaceutical composition of claim 88, comprising at least one additional
therapeutic agent.
90. The pharmaceutical composition of claim 89, wherein the additional therapeutic agent is an antibiotic.
91. The pharmaceutical composition of claim 90, wherein the additional therapeutic agent is an antibiotic selected from the group consisting of: a quinolone, a β-lactam, a cephalosporin, a penicillin, a carbapenem, a lipopetide, an aminoglycoside, a glycopeptide, a macrolide, an ansamycin, a sulfonamide, and combinations of two or more thereof.
92. The pharmaceutical composition of claim 91, wherein the aminoglycoside is tobramycin.
PCT/US2018/028868 2017-04-21 2018-04-23 Antibacterial compounds WO2018195536A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762488259P 2017-04-21 2017-04-21
US62/488,259 2017-04-21

Publications (1)

Publication Number Publication Date
WO2018195536A1 true WO2018195536A1 (en) 2018-10-25

Family

ID=63856423

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/028868 WO2018195536A1 (en) 2017-04-21 2018-04-23 Antibacterial compounds

Country Status (1)

Country Link
WO (1) WO2018195536A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112716944A (en) * 2021-02-25 2021-04-30 成都大学 Application of nitroimidazole compounds in preparation of medicines for inhibiting bacterial quorum sensing
CN114159455A (en) * 2021-12-31 2022-03-11 山东省妇幼保健院 Application of ribavirin in preparation of anti-acinetobacter baumannii medicine
CN115400105A (en) * 2022-10-12 2022-11-29 湖北省农业科学院畜牧兽医研究所 Application of pyrogallic acid in preparation of medicines for resisting streptococcus equi subsp zooepidemicus
CN115501214A (en) * 2022-10-14 2022-12-23 湖北省农业科学院畜牧兽医研究所 Pyrogallol and sulfisoxazole composition and application thereof in inhibiting streptococcus suis type 2
CN116568297A (en) * 2020-12-09 2023-08-08 润佳(苏州)医药科技有限公司 Pharmaceutical use of fused ring phenolic compounds

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130046019A1 (en) * 2010-04-06 2013-02-21 Prescience Labs, Llc Methods of Treatment Using 3-Bromopyruvate and Other Selective Inhibitors of ATP Production
US20130177630A1 (en) * 2012-01-05 2013-07-11 Frederick Tmothy Guilford Anti-cancer composition and method utilizing 3-bp and liposomal reduced glutathione
US20130309219A1 (en) * 2011-01-31 2013-11-21 Adam J. Ratner Treatment and Prevention of Bacterial Vaginosis and Gardnerella Vaginalis Infections

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130046019A1 (en) * 2010-04-06 2013-02-21 Prescience Labs, Llc Methods of Treatment Using 3-Bromopyruvate and Other Selective Inhibitors of ATP Production
US20130309219A1 (en) * 2011-01-31 2013-11-21 Adam J. Ratner Treatment and Prevention of Bacterial Vaginosis and Gardnerella Vaginalis Infections
US20130177630A1 (en) * 2012-01-05 2013-07-11 Frederick Tmothy Guilford Anti-cancer composition and method utilizing 3-bp and liposomal reduced glutathione

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116568297A (en) * 2020-12-09 2023-08-08 润佳(苏州)医药科技有限公司 Pharmaceutical use of fused ring phenolic compounds
CN112716944A (en) * 2021-02-25 2021-04-30 成都大学 Application of nitroimidazole compounds in preparation of medicines for inhibiting bacterial quorum sensing
CN114159455A (en) * 2021-12-31 2022-03-11 山东省妇幼保健院 Application of ribavirin in preparation of anti-acinetobacter baumannii medicine
CN115400105A (en) * 2022-10-12 2022-11-29 湖北省农业科学院畜牧兽医研究所 Application of pyrogallic acid in preparation of medicines for resisting streptococcus equi subsp zooepidemicus
CN115400105B (en) * 2022-10-12 2023-08-22 湖北省农业科学院畜牧兽医研究所 Application of Jiaosuan in preparation of streptococcus equi subspecies zooepidemicus resisting medicines
CN115501214A (en) * 2022-10-14 2022-12-23 湖北省农业科学院畜牧兽医研究所 Pyrogallol and sulfisoxazole composition and application thereof in inhibiting streptococcus suis type 2
CN115501214B (en) * 2022-10-14 2023-09-22 湖北省农业科学院畜牧兽医研究所 Composition of pyrogallol and sulfamethoxazole and application thereof in inhibiting streptococcus suis type 2

Similar Documents

Publication Publication Date Title
WO2018195536A1 (en) Antibacterial compounds
US11707442B2 (en) Antibacterial and antifungal compounds
KR101148049B1 (en) Use of pyrroloquinoline compounds to kill clinically latent microorganisms
Wang et al. Staphylococcus epidermidis surfactant peptides promote biofilm maturation and dissemination of biofilm-associated infection in mice
US11278025B2 (en) Antibiotic compounds
ES2497116T3 (en) 2,4,5-trisubstituted imidazoles and their use as antimicrobial agents
TWI421097B (en) Chalcones as enhancer of antimicrobial agents
US11446280B2 (en) Compounds and methods for the treatment of microbial infections
KR101118942B1 (en) Antituberculous composition comprising oxazole compounds
JP2010509196A (en) Topical formulation
US20220324796A1 (en) Antibiotic compounds
JP2011526887A (en) Treatment of antibiotic-resistant bacterial infections
US11690824B2 (en) Antibacterial compounds
US11052078B2 (en) Compounds affecting pigment production and methods for treatment of bacterial diseases
ES2902456T3 (en) Pharmaceutical compositions comprising antibacterial agents
WO2021243015A1 (en) Antibacterial compounds
US20230041593A1 (en) Compounds and methods for the treatment of microbial infections
KR20160081605A (en) Pharmaceutical composition for preventing or treating for Helicobacter pylori infectious diseases
Endermann et al. Pharmacokinetics (PK) and in vivo efficacy of oral finafloxacin (FIN) and comparators in rodent models of systemic infection
WO2005046674A2 (en) Antibacterial compositions

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: 18786985

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: 18786985

Country of ref document: EP

Kind code of ref document: A1