US20050215490A1 - Polyene polyketides and methods of treating bacterial infections - Google Patents

Polyene polyketides and methods of treating bacterial infections Download PDF

Info

Publication number
US20050215490A1
US20050215490A1 US10/975,545 US97554504A US2005215490A1 US 20050215490 A1 US20050215490 A1 US 20050215490A1 US 97554504 A US97554504 A US 97554504A US 2005215490 A1 US2005215490 A1 US 2005215490A1
Authority
US
United States
Prior art keywords
compound
alkyl
pharmaceutically acceptable
compounds
bacterial infection
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/975,545
Inventor
Chris Farnet
Anne-Marie Alarco
Emmanuel Zazopoulos
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Thallion Pharmaceuticals Inc
Original Assignee
Ecopia Biosciences Inc
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 Ecopia Biosciences Inc filed Critical Ecopia Biosciences Inc
Priority to US10/975,545 priority Critical patent/US20050215490A1/en
Assigned to ECOPIA BIOSCIENCES INC. reassignment ECOPIA BIOSCIENCES INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALARCO, ANNE-MARIE, FARNET, CHRIS M., ZAZOPOULOS, EMMANUEL
Publication of US20050215490A1 publication Critical patent/US20050215490A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C279/00Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups
    • C07C279/04Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of guanidine groups bound to acyclic carbon atoms of a carbon skeleton
    • C07C279/14Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of guanidine groups bound to acyclic carbon atoms of a carbon skeleton being further substituted by carboxyl groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/10Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H5/00Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium
    • C07H5/04Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium to nitrogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H5/00Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium
    • C07H5/04Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium to nitrogen
    • C07H5/06Aminosugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • C12N1/205Bacterial isolates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P13/00Preparation of nitrogen-containing organic compounds
    • C12P13/001Amines; Imines
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P13/00Preparation of nitrogen-containing organic compounds
    • C12P13/02Amides, e.g. chloramphenicol or polyamides; Imides or polyimides; Urethanes, i.e. compounds comprising N-C=O structural element or polyurethanes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/02Oxygen as only ring hetero atoms
    • C12P17/06Oxygen as only ring hetero atoms containing a six-membered hetero ring, e.g. fluorescein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/16Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms containing two or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/44Preparation of O-glycosides, e.g. glucosides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/40Preparation of oxygen-containing organic compounds containing a carboxyl group including Peroxycarboxylic acids
    • C12P7/42Hydroxy-carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/10Systems containing only non-condensed rings with a five-membered ring the ring being unsaturated
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/821Microorganisms used in the destruction of hazardous or toxic waste

Definitions

  • This invention relates to methods of treating bacterial infections in a mammal using a compound of using a polyene polyketide of Formula I or its pharmaceutically acceptable salts.
  • Microbial natural products have been a reliable source of antimicrobial drugs. Streptomycin, erythromycin, tetracycline, pristinamycin, vancomycin and lincomycin are examples of antibiotics produced by microorganisms.
  • Streptomycin, erythromycin, tetracycline, pristinamycin, vancomycin and lincomycin are examples of antibiotics produced by microorganisms.
  • problems related to resistance to current antibiotics by severe pathogens such as VRE and MRSA have been on the rise.
  • MRSA infections are common causes of hospital acquired infections, and a limited number of drugs remain effective against these infections.
  • the invention encompasses a method of inhibiting bacterial infection, the method comprising contacting the bacterial cells with a therapeutically effective amount of a compound selected from Compounds 1, 2, 3, 4, 5, 6, 7 and pharmaceutically acceptable salts or prodrugs thereof: such that growth of the bacterial infection is inhibited.
  • the invention provides a method of inhibiting the growth of a bacterial infection, the method comprising contacting a bacterial cell with a therapeutically effective amount of a compound of Formula I: such that growth of the bacterial infection is inhibited; wherein,
  • a of the compound is —C(NH)NH 2 .
  • a of the compound is H.
  • a of the compound is —C(NH)NHC(O)CH 3 .
  • a of the compound is In another embodiment of the compound R 4 is CH 3 .
  • R 4 of the compound is H.
  • Z is In a sub-class of this embodiment, R 6 , R 7 and R 8 are each H, and R 5 is COOH or CO 2 CH 3 .
  • the invention further contemplates an embodiment of this aspect in which Z is OH.
  • the invention further contemplates an embodiment of this aspect in which D is In a subclass of this embodiment R 11 is H.
  • the invention further contemplates an embodiment of this aspect in which D In a subclass of this embodiment R 11 is CH 3.
  • D is OH.
  • Specific embodiments of compounds of Formula I useful to inhibit the growth of bacterial infection are as follows: or a pharmaceutically acceptable salt or prodrug of any one of Compounds 1-28.
  • the invention relates to a method of inhibiting the growth of a bacterial infection, the method comprising contacting a bacterial cell with a pharmaceutical composition comprising of a compound of Formula I as defined above, or a pharmaceutically acceptable salt or prodrug thereof, with a pharmaceutically acceptable carrier, such as growth of the bacterial infection is inhibited.
  • a pharmaceutical composition comprising of a compound of Formula I as defined above, or a pharmaceutically acceptable salt or prodrug thereof, with a pharmaceutically acceptable carrier, such as growth of the bacterial infection is inhibited.
  • a pharmaceutically acceptable carrier such as growth of the bacterial infection is inhibited.
  • the invention further encompasses a method of treating a bacterial infection in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of a compound selected from the Compounds 1, 2, 3, 4, 5, 6, 7 and pharmaceutically acceptable salts or prodrugs thereof: such that the bacterial infection is treated.
  • the invention provides a method of treating a bacteria infection in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of a therapeutically effective amount of a compound of Formula I: such that the bacterial infection is treated; wherein,
  • Embodiment of Formula I useful in treating bacterial infection in a mammal are compounds of Formula I wherein A of the compound is —C(NH)NH 2 .
  • a of the compound is H.
  • a of the compound is —C(NH)NHC(O)CH 3 .
  • a of the compound is In another embodiment of the compound R 4 is CH 3 .
  • R 4 of the compound is H.
  • Z is In a sub-class of this embodiment, R 6 , R 7 and R 8 are each H, and R 5 is COOH or CO 2 CH 3 .
  • the invention further contemplates an embodiment of this aspect in which Z is OH.
  • the invention further contemplates an embodiment of this aspect in which D is In a subclass of this embodiment R 11 is H.
  • the invention further contemplates an embodiment of this aspect in which D is In a subclass of this embodiment R 11 is CH 3 .
  • D is OH.
  • Specific compounds of Formula I useful to treat bacterial infection in a mammal are compound 1-28.
  • the invention relates to a method of treating bacterial infection in a mammal, the method comprising contacting a bacterial cell with a pharmaceutical composition comprising of a compound of Formula I as defined above, or a pharmaceutically acceptable salt or prodrug thereof, with a pharmaceutically acceptable carrier, such that the bacterial infection is treated.
  • a pharmaceutical composition comprising of a compound of Formula I as defined above, or a pharmaceutically acceptable salt or prodrug thereof, with a pharmaceutically acceptable carrier, such that the bacterial infection is treated.
  • the bacterial infection is from a bacteria selected from the group consisting of Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp., Proteus spp., Pseudomonas aeruginosa, E. coli, Serratia marcesens, Staphylococcus aureus, Coagulase negative Staphylococcus, Haemophilus infuenzae, Bacillus anthracis, Mycoplasma pneumoniae, and Staphylococcus epidermidis.
  • the bacterial infection is caused by Gram-positive bacteria.
  • the Gram-positive bacteria is a Staphylococcus or Enterococcus species.
  • the compound is administered orally. In another embodiment, the compound is administered intravenously. In still another embodiment, the compound is administered intraperitoneally.
  • FIG. 1 Electrospray mass spectrum (Positive Ion Mode) of Compound 1.
  • FIG. 2 Electrospray mass (ms/ms of m/z:837.50) spectrum (Positive Ion Mode) of Compound 1.
  • FIG. 3 Electrospray mass spectrum (Negative Ion Mode) of Compound 1.
  • FIG. 4 500 MHz proton nuclear magnetic resonance ( 1 H-NMR) spectrum of Compound 1 in d 4 -MeOH.
  • FIG. 5 500 MHz proton nuclear magnetic resonance ( 1 H-NMR) spectrum of Compound 2 in d 4 -MeOH.
  • FIG. 6 500 MHz proton nuclear magnetic resonance ( 1 H NMR) spectrum of Compound 3 in d 4 -MeOH.
  • FIG. 7 500 MHz proton nuclear magnetic resonance ( 1 H NMR) spectrum of Compound 4 in d 4 -MeOH.
  • FIG. 8 500 MHz proton nuclear magnetic resonance ( 1 H NMR) spectrum of Compound 5 in d 4 -MeOH.
  • FIG. 9 500 MHz proton nuclear magnetic resonance ( 1 H NMR) spectrum of Compound 6 in d 4 -MeOH.
  • FIG. 10 500 MHz proton nuclear magnetic resonance ( 1 H NMR) spectrum of Compound 7 in d 4 -MeOH.
  • the present invention relates to methods of inhibiting the growth of bacterial cells.
  • the method comprtises contacting bacterial cells with a polyene polyketide compound of Formula I, as exemplified herein as Compound 1, Compound 2 and Compound 7, which were isolated from strains of actinomycetes, Amycolatopsis sp. such as Amycolatopsis orientalis ATCC 43491, Amycolatopsis orientalis IDAC 220604-01, or a mutant or a variant thereof.
  • Amycolatopsis orientalis ATCC 43491 may be obtained from the American Type Culture Collection (ATCC) P.O. Box 1549, Manassas, Va. 20108 USA;
  • Amycolatopsis orientalis IDAC 220604-01 may be obtained from the International Depositary Authority of Canada (IDAC), Bureau of Microbiology, Health Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2.
  • the present invention further related to methods of treating a bacterial infection in a mammal, the method comprising administering a therapeutically effective amount of a polyene polyketide compound of the invention, such that the bacterial infection is treated.
  • Compound 1, Compound 2 and Compound 7 are useful as a pharmaceutical, in particular for use as in inhibitor of bacterial cell growth.
  • a compound of Formula I can be used. Accordingly, certain aspects of the present invention relate to use of the polyene polyketide compounds of the present invention in to inhibit bacterial growth, and use of the polyene polyketides of the invention in the preparation of a medicament to treat bacterial infection.
  • the invention further related to pharmaceutically acceptable salts and derivatives of the compounds 1-7 and to methods for obtaining such compounds.
  • One method of obtaining the compounds is by cultivating Amycolatopsis orientalis ATCCTM 43491, or a mutant or a variant thereof, under suitable Amycolatopsis sp. culture conditions preferably using the fermentation protocol described herein below.
  • the invention also relates to pharmaceutical compositions comprising a compound selected from 1-28 and its pharmaceutically acceptable salts and derivatives. Accordingly, certain aspects of the present invention relate to use to inhibit bacterial growth of a pharmaceutical compositions comprising a polyene polyketide compound of the present invention together with a pharmaceutically acceptable carrier, and use in the preparation of medicament to treat bacterial infection of a pharmaceutical compositions comprising a polyene polyketide compound of the present invention together with a pharmaceutically acceptable carrier.
  • compositions comprising the polyene polyketide compounds of the present invention together with a pharmaceutically acceptable carrier, methods of using the compositions to inhibit bacterial growth, and methods of using the pharmaceutical compositions to treat bacterial infections.
  • Me refers to methyl (CH 3 )
  • Et refers to ethyl (CH 2 CH 3 )
  • Pr refers to n-propyl (CH 2 CH 2 CH 3 )
  • Ac refers to acetyl (C(O)CH 3 ).
  • alkyl refers to a linear or branched hydrocarbon groups.
  • alkyl groups include, without limitation, methyl, ethyl, n-propyl, isopropyl, n-butyl, pentyl, hexyl, heptyl, cyclopentyl, cyclohexyl, cyclohexymethyl, and the like.
  • Alkyl may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro,.thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl, oxo, guanidino and formyl.
  • substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro,.thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sul
  • alkenyl refers to linear, branched or cyclic hydrocarbon groups containing at least one carbon-carbon double bond.
  • alkenyl groups include, without limitation, vinyl, 1-propene-2-yl, 1-butene-4-yl, 2-butene-4-yl, 1-pentene-5-yl and the like.
  • Alkenyl may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, alkoxy, aryloxy, sulfinyl, sulfonyl, formyl, oxo and guanidino.
  • the double bond portion(s) of the unsaturated hydrocarbon chain may be either in the cis or trans configuration.
  • cycloalkyl or cycloalkyl ring refers to a saturated or partially unsaturated carbocyclic ring in a single or fused carbocyclic ring system having from three to fifteen ring members.
  • cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclohexyl, and cycloheptyl.
  • Cycloalkyl may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • heterocyclyl, heterocyclic or heterocyclyl ring refers to a saturated or partially unsaturated ring containing one to four hetero atoms or hetero groups selected from O, N, NH, NR x , PO 2 , S, SO or SO 2 in a single or fused heterocyclic ring system having from three to fifteen ring members.
  • heterocyclyl, heterocyclic or heterocyclyl ring include, without limitation, morpholinyl, piperidinyl, and pyrrolidinyl.
  • Heterocyclyl, heterocyclic or heterocyclyl ring may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, oxo, thiocarbonyl, imino, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • substituents selected from acyl, amino, acylamino, acyloxy, oxo, thiocarbonyl, imino, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycl
  • amino acid refers to any natural amino acid, all natural amino acids are well known to a person skilled in the art.
  • halo is defined as a bromine, chlorine, fluorine or iodine.
  • aryl or aryl ring refers to common aromatic groups having “4n+2” electrons, wherein n is an integer from 1 to 3, in a monocyclic or conjugated polycyclic system and having from five to fifteen ring atoms.
  • Aryl ring may include from 1 to 3 heteroatoms such as nitrogen, oxygen and sulphur atoms. Examples of aryl include, without limitation, phenyl, naphthyl, biphenyl, terphenyl, furyl, pyrrollyl, thienyl, pyridyl, oxazolyl, imidazolyl, pyrazolyl and indolyl groups.
  • Aryl may optionally be substituted with one or more substituent group selected from acyl, amino, acylamino, acyloxy, azido, alkythio, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • substituent group selected from acyl, amino, acylamino, acyloxy, azido, alkythio, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, s
  • the compounds of the present invention can possess one or more asymetric carbon atoms and can exist as optical isomers forming mixtures of racemic or non-racemic compounds.
  • the compounds of the present invention are useful as a single isomer or as a mixture of stereochemical isomeric forms.
  • Diastereoisomers, i.e., nonsuperimposable stereochemical isomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes.
  • the invention embraces isolated compounds.
  • An isolated compound refers to a compound which represents at least 10%, 20%, 50% and 80% of the compound of the present invention present in a mixture, provided that the mixture comprising the compound of the invention-has demonstrable (i.e. statistically significant) biological activity including antimicrobial activity when tested in conventional biological assays known to a person skilled in the art.
  • treatment refers to the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disorder, e.g., a disease or condition, a symptom of disease, or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disease, the symptoms of disease, or the predisposition toward disease.
  • a disorder e.g., a disease or condition, a symptom of disease, or a predisposition toward a disease
  • a “pharmaceutical composition” comprises a pharmacologically effective amount of a compound of the invention and a pharmaceutically acceptable carrier.
  • pharmaceutically effective amount refers to that amount of polyene polyketide effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 25% reduction in that parameter.
  • pharmaceutically acceptable carrier refers to a carrier for administration of a therapeutic agent.
  • Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the term specifically excludes cell culture medium.
  • pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • salts include acid addition salts and base addition salts.
  • the nature of the salt is not critical, provided that it is pharmaceutically-acceptable.
  • examples of acid addition salts include hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulphuric, phosphoric, formic, acetic, citric, tartaric, succinic, oxalic, malic, glutamic, propionic, glycolic, gluconic, maleic, embonic (pamoic), methanesulfonic, ethanesulfonic, 2-hydroxyethanesulfonic, pantothenic, benzenesulfonic, toluenesulfonic, sulfanilic, mesylic, cyclohexylaminosulfonic, stearic, algenic, ⁇ -hydroxybutyric, malonic, galactantic, galacturonic acid and the like.
  • Suitable pharmaceutically-acceptable base addition salts of compounds of the invention include, but are not limited to, metallic salts made from aluminium, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, lysine, procaine and the like. Additional examples of pharmaceutically acceptable salts are listed in Berge S M et al., Journal of Pharmaceutical Sciences, (1977) Vol. 66 no 1, pp. 1-19. All of these salts may be prepared by conventional means form the corresponding compounds of Formula I by treating with the appropriate acid or base.
  • the invention relates to novel polyene polyketides, referred to herein as Compounds 1-7: or, to a pharmaceutically acceptable salt or prodrug of any of Compounds 1 to 7.
  • Compounds 1 to 7 may be characterized by any one or more of their physicochemical and spectral properties given below, such as mass, UV, and NMR spectroscopic data.
  • the invention provides compounds of Formula I, wherein A is —C(NH)NH 2 ; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein A is —C(NH)NHC(O)CH 3 ; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein A is and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein R 4 is CH 3 ; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein R 4 is H; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein Z is and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 6 , R 7 and R 8 are each H, and R 5 is COOH, all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein Z is and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 6 , R 7 and Ra are each H, and R 5 is CO 2 CH 3 , all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein Z is OH and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein D is and all other groups are as previously defined or a pharmaceutically acceptable salt or prodrug thereof.
  • R 11 is H and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein D is and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 11 is CH 3 and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides compounds of Formula I, wherein D is OH; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compounds of the invention may be formulated into pharmaceutical compositions comprised compounds of Formula I in combination with a pharmaceutical acceptable carrier as discussed in Section IV below.
  • compounds 1-7 may be obtained by cultivating a strain of actinomycetes, Amycolatopsis sp. such as Amycolatopsis orientalis ATCC 43491, Amycolatopsis orientalis IDAC 220604-01, or a mutant or a variant thereof.
  • Amycolatopsis orientalis ATCC 43491 may be obtained from the American Type Culture Collection (ATCC) P.O. Box 1549, Manassas, Va. 20108 USA;
  • Amycolatopsis orientalis IDAC 220604-01 may be obtained from the International Depositary Authority of Canada (IDAC), Bureau of Microbiology, Health Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2.
  • Amycolatopsis orientalis IDAC 220604-01 was made under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for Purposes of Patent Procedure.
  • the deposited strain will be irrevocably and without restriction or conditions released to the public upon the issuance of a patent.
  • the deposited strains are provided merely as a convenience to those skilled in the art and are not an admission that a deposit is required for enablement, such as that required under 35 U.S.C. ⁇ 112. Indeed it is to be understood that the present invention is not limited to use of a particular strain of Amycolatopsis orientalis.
  • the invention contemplates the use of other Formula I producing organisms, such as mutant or variants of Amycolatopsis orientalis ATCC 43491 or Amycolatopsis orientalis IDAC 220604-01 that can be derived by well known means such as X-ray irradiation, ultraviolet irradiation, treatment with nitrogen mustard, phage exposure, antibiotic selection and the like, or through the use of recombinant genetic engineering as described in co-pending parent application U.S. Ser. No. 10/937,379.
  • the polyene polyketides of the invention may be biosynthesized by various microorganisms.
  • Microorganisms that may synthesize the compounds of the present invention include but are not limited to bacteria of the order Actinomycetales, also referred to as actinomycetes.
  • Non-limiting examples of members belonging to the genera of Actinomycetes include Nocardia, Geodermatophilus, Actinoplanes, Micromonospora, Nocardioides, Saccharothrix, Amycolatopsis, Kutzneria, Saccharomonospora, Saccharopolyspora, Kitasatospora, Streptomyces, Microbispora, Streptosporangium, Actinomadura.
  • Microorganisms producing polyene polyketides of the invention are cultivated in culture media containing known nutritional sources for actinomycetes, having assimilable sources of carbon, nitrogen plus optional inorganic salts and other known growth factors, at a pH of about 6 to about 9. Suitable media include, without limitation, growth media provided in Table 1. Microorganisms are cultivated at incubation temperatures of about 20° C. to about 40° C. for about 3 to about 40 days. The culture media inoculated with the polyene polyketide-producing microorganisms, may be aerated by incubating the inoculated culture media with agitation, for example shaking on a rotary shaker, or a shaking water bath.
  • Aeration may also be achieved by the injection of air, oxygen or an appropriate gaseous mixture to the inoculated culture media during incubation.
  • the polyene polyketide compounds can be extracted and isolated from the cultivated culture media by techniques known to a skilled person in the art and/or disclosed herein, including for example centrifugation, chromatography, adsorption, filtration.
  • the cultivated culture media can be mixed with a suitable organic solvent such as n-butanol, n-butyl acetate and 4-methyl-2-pentanone, the organic layer can be separated for example, by centrifugation followed by the removal of the solvent, by evaporation to dryness or by evaporation to dryness under vacuum.
  • the resulting residue can optionally be reconstituted with for example water, ethyl ether, ethanol acetate, methanol or a mixture thereof, and re-extracted in a two-phase system with a suitable organic solvent such as hexane, carbon tetrachloride, methylene chloride or a mixture thereof.
  • a suitable organic solvent such as hexane, carbon tetrachloride, methylene chloride or a mixture thereof.
  • the compound can be further purified by the use of standard techniques such as chromatography.
  • polyene polyketides-biosynthesized by microorganism may optionally be subjected to random and/or directed chemical modifications to form compounds that are derivatives or structural analogs. Such derivative or structural analogs having similar functional activity are within the scope of the present invention.
  • Polyene polyketides may optionally be modified using methods known in the art and described in co-pending application U.S. Ser. No. 10/937,379.
  • Solution of trace elements contains: ZnCl 2 40 mg; FeCl 3 .6H 2 O (200 mg); CuCl 2 .2H 2 O (10 mg); (NH 4 ) 6 MO 7 O 24 .4H 2 O (10 mg) per litre.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a polyene polyketide, as described in Section II and a pharmaceutically acceptable carrier as described below.
  • the pharmaceutical composition comprising the polyene polyketide is useful for treating a variety of bacterial infections.
  • the compounds of the present invention can be formulated for oral, intravenous, intramuscular, subcutaneous, topical or parenteral administration for the therapeutic or prophylactic treatment of diseases, particularly bacterial infections.
  • compounds of the present invention can be mixed with conventional pharmaceutical carriers and excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, wafers and the like.
  • the compositions comprising a compound of this present invention will contain from about 0.1% to about 99.9%, about 1% to about 98%, about 5% to about 95%, about 10% to about 80% or about 15% to about 60% by weight of the active compound.
  • compositions of the present invention are prepared in accordance with standard procedures and are administered at dosages that are selected to reduce, prevent, or eliminate bacterial infection (See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. and Goodman and Gilman's the Pharmaceutical Basis of Therapeutics, Pergamon Press, New York, N.Y. for a general description of the methods for administering various antimicrobial agents for human therapy, the contents of which are incorporated herein by reference).
  • the compositions of the present invention can be delivered using controlled (e.g., capsules) or sustained release delivery systems (e.g., bioerodable matrices).
  • compositions of the invention preferably of Formula I
  • U.S. Pat. No. 4,452,775 issued to Kent
  • U.S. Pat. No. 5,039,660 issued to Leonard
  • U.S. Pat. No. 3,854,480 issued to Zaffaroni
  • compositions of the present invention comprise one or more compounds of the present invention in association with one or more non-toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants and/or excipients, collectively referred to herein as “carrier” materials, and if desired other active ingredients.
  • carrier materials
  • the compositions may contain common carriers and excipients, such as corn starch or gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid.
  • the compositions may contain crosarmellose sodium, microcrystalline cellulose, sodium starch glycolate and alginic acid.
  • Tablet binders that can be included are acacia, methylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone (Providone), hydroxypropyl methylcellulose, sucrose, starch and ethylcellulose.
  • Lubricants that can be used include magnesium stearate or other metallic stearates, stearic acid, silicon fluid, talc, waxes, oils and colloical silica.
  • Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring or the like can also be used. It may also be desirable to add a coloring agent to make the dosage form more esthetic in appearance or to help identify the product comprising a compound of the present invention.
  • the pharmaceutical compositions are in the form of, for example, a tablet, capsule, suspension or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a therapeutically-effective amount of the active ingredient. Examples of such dosage units are tablets and capsules.
  • the tablets and capsules which can contain, in addition to the active ingredient, conventional carriers such as binding agents, for example, acacia gum, gelatin, polyvinylpyrrolidone, sorbitol, or tragacanth; fillers, for example, calcium phosphate, glycine, lactose, maize-starch, sorbitol, or sucrose; lubricants, for example, magnesium stearate, polyethylene glycol, silica or talc; disintegrants, for example, potato starch, flavoring or coloring agents, or acceptable wetting agents.
  • binding agents for example, acacia gum, gelatin, polyvinylpyrrolidone, sorbitol, or tragacanth
  • fillers for example, calcium phosphate, glycine, lactose, maize-starch, sorbitol, or sucrose
  • lubricants for example, magnesium stearate, polyethylene glycol, silica or talc
  • disintegrants for example
  • Oral liquid preparations generally are in the form of an aqueous or oily solution, suspensions, emulsions, syrups or elixirs may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous agents, preservatives, coloring agents and flavoring agents.
  • additives for liquid preparations include acacia, almond oil, ethyl alcohol, fractionated coconut oil, gelatin, glucose syrup, glycerin, hydrogenated edible fats, lecithin, methyl cellulose, methyl or propyl para-hydroxybenzoate, propylene glycol, sorbitol, or sorbic acid.
  • IV intravenous
  • compounds of the present invention can be dissolved or suspended in any of the commonly used intravenous fluids and administered by infusion.
  • Intravenous fluids include, without limitation, physiological saline or Ringer's solution.
  • Formulations for parental administration can be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions or suspensions can be prepared from sterile powders or granules having one or more of the carriers mentioned for use in the formulations for oral administration.
  • the compounds can be dissolved in polyethylene glycol, propylene glycol, ethanol, corn oil, benzyl alcohol, sodium chloride, and/or various buffers.
  • a sterile formulation of compounds of the present invention or-suitable soluble salts forming the compound can be dissolved and administered in a pharmaceutical diluent such as Water-for-Injection (WFI), physiological saline or 5% glucose.
  • WFI Water-for-Injection
  • a suitable insoluble form of the compound may be prepared and administered as a suspension in an aqueous base or a pharmaceutically acceptable oil base, e.g. an ester of a long chain fatty acid such as ethyl oleate.
  • the compounds of present invention can also be prepared in suitable forms to be applied to the skin, or mucus membranes of the nose and throat, and can take the form of creams, ointments, liquid sprays or inhalants, lozenges, or throat paints.
  • suitable forms can include chemical compounds such as dimethylsulfoxide (DMSO) to facilitate surface penetration of the active ingredient.
  • DMSO dimethylsulfoxide
  • the compounds of the present invention can be presented in liquid or semi-liquid form formulated in hydrophobic or hydrophilic bases as ointments, creams, lotions, paints or powders.
  • the compounds of the present invention can be administered in the form of suppositories admixed with conventional carriers such as cocoa butter, wax or other glyceride.
  • the compounds of the present invention can be in powder form for reconstitution in the appropriate pharmaceutically acceptable carrier at the time of delivery.
  • the unit dosage form of the compound can be a solution of the compound or a salt thereof in a suitable diluent in sterile, hermetically sealed ampoules.
  • the amount of the compound of the present invention in a unit dosage comprises a therapeutically-effective amount of at least one active compound of the present invention which may vary depending on the recipient subject, route and frequency of administration.
  • a recipient subject refers to a plant, a cell culture or an animal such as an ovine or a mammal including a human.
  • the novel compositions disclosed herein are placed in a pharmaceutically acceptable carrier and are delivered to a recipient subject (including a human subject) in accordance with known methods of drug delivery.
  • the methods of the invention for delivering the compositions of the invention in vivo utilize art-recognized protocols for delivering the agent with the only substantial procedural modification being the substitution of the compounds of the present invention for the drugs in the art-recognized protocols.
  • the methods for using the claimed composition for treating cells in culture for example, to eliminate or reduce the level of bacterial contamination of a cell culture, utilize art-recognized protocols for treating cell cultures with antibacterial agents with the only substantial procedural modification being the substitution of the compounds of the present invention for the agents used in the art-recognized protocols.
  • the compounds of the present invention provide a method for treating microbial infections.
  • unit dosage refers to a quantity of a therapeutically-effective amount of a compound of the present invention that elicits a desired therapeutic response.
  • therapeutically-effective amount means an amount of a compound of the present invention that prevents the onset, alleviates the symptoms, or stops the progression of a bacterial infection.
  • treating is defined as administering, to a subject, a therapeutically-effective amount of at least one compound of the present invention, both to prevent the occurrence of a bacterial infection, or to control or eliminate a bacterial infection.
  • desired therapeutic response refers to treating a recipient subject with a compound of the present invention such that a bacterial infection is reversed, arrested or prevented in a recipient subject.
  • the compounds of the present invention can be administered as a single daily dose or in multiple doses per day.
  • the treatment regime may require administration over extended periods of time, e.g., for several days or for from two to four weeks.
  • the amount per administered dose or the total amount administered will depend on such factors as the nature and severity of the infection, the age and general health of the recipient subject, the tolerance of the recipient subject to the compound and the type of the bacterial infection.
  • a compound according to this invention may also be administered in the diet or feed of a patient or animal.
  • the diet for animals can be normal foodstuffs to which the compound can be added or it can be added to a premix.
  • the compounds of the present invention may be taken in combination, together or separately with any known clinically approved antibiotic to treat a recipient subject in need of such treatment.
  • the present invention relates to a method of inhibiting bacterial growth.
  • Compounds described herein can possess antbacterial activity.
  • the compounds are effective in inhibiting bacterial infections in vivo.
  • bacterial organisms that may be controlled by the compositions and methods of this invention include, but are not limited to the following organisms: Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp., Proteus spp., Pseudomonas aeruginosa, Escherichia coli, Serratia marcesens, Staphylococcus aureus, Haemophilus infuenzae, Bacillus anthracis, Mycoplasma pneumoniae, and Coagulase negative Staphylococcus including Staphylococcus epidermidis.
  • compositions and compounds of the invention will be useful in controlling or treating infections caused by Gram-positive bacteria. None limiting examples of Gram-positive bacterial growth that may be inhibited by the compounds and pharmaceutical compositions of the invention include Staphylococci and Enterococci. Compositions and methods will be useful for controlling, or reducing the advancement, severity or effects of nosocomial or non-nosocomial infections. Examples of nosocomial uses include, but are not limited to, urinary tract infections, pneumonia, surgical wound infections, bacteremia and therapy for febrile neutropenic patients. Examples of non-nosocomial uses include but are not limited to urinary tract infections, pneumonia, prostatitis, skin and soft tissue infections and intra-abdominal infections.
  • the term “inhibition” when used in conjunction the antibacterial methods refers to a suppression, killing, stasis or destruction of bacterial cells.
  • the antibacterial methods preferably result in prevention, reduction or elimination of invasive activity and related growth of bacterial cells.
  • the term “effective amount” when used in conjunction with antibacterial methods refers to the amount of the compound sufficient to result in the inhibition of growth bacterial cells.
  • the inhibition of bacterial growth according to this method can be monitored in several ways. First bacterial cells grown in vitro can be treated with the compound and monitored for growth or death relative to the same cells cultured in the absence of the compound. A cessation of growth or a slowing of the growth rate, e.g. by 10% of more, is indicative of bacterial cell inhibition.
  • bacterial cell inhibition can be monitored by administering the compound to an animal model having a bacterial infection of interest.
  • experimental aminal bacterial models are known in the art and described in the examples herein.
  • a convenient means to measure antibacterial activity is by Minimal Inhibitory Concentration (MIC), which is the minimal concentration of an agent necessary to inhibit a standard (control) dose of bacteria.
  • MIC Minimal Inhibitory Concentration
  • MMC Minimal Bactericidal Concentration
  • the invention provides a method of decreasing bacterial quantity in a biological sample.
  • This method comprises the step of contacting the biological sample with a polyene polyketide of Formula I, a compound as described herein, or a pharmaceutically acceptable derivative or prodrug thereof.
  • This method is effective if the number of bacteria decreases by at least 10%, and preferably more, e.g., 25%, 50%, 75% or even 100% after contacting the biological sample with a polyene polyketide of Formula I, a compound as described herein, or a pharmaceutically acceptable derivative or prodrug thereof.
  • the present invention relates to a method for treating bacterial infection in a mammalian subject in need thereof, comprising the step of administering to the mammal a therapeutically effective amount of a polyene poketide of Formula I, a compound described herein or a pharmaceutically acceptable derivative or prodrug thereof in combination with a physiologically acceptable carrier.
  • the compound is represented by Formula I.
  • the compound is selected from Compounds 1-27.
  • compositions effective to treat or prevent a bacterial infection comprise any one of Compounds 1 to 7, a compound of Formula I, or a pharmaceutically acceptable derivative or prodrug thereof, in an amount sufficient to measurably decrease bacterial quantity, and a pharmaceutically acceptable carrier.
  • measurably decrease bacterial quantity means a measurable change in the number of bacteria between a sample containing the inhibitor and a sample not containing the inhibitor.
  • a typical effective unit dose of any one of Compounds 1 to 7, a compound of Formula I as described herein or a pharmaceutically acceptable derivative or prodrug thereof given orally or parenterally would be from about 5 to about 100 mg/kg of body weight of the subject with a daily dose ranging from about 15 to about 300 mg/kg of body weight of the subject.
  • Another preferred embodiment of this invention relates to a method, as described above, of treating a bacterial infection in a mammal in need thereof, but further comprising the step of administering to the mammal an agent which increases the susceptibility of bacterial organisms to antibiotics.
  • the invention provides a method, as described above, of decreasing bacterial quantity in a biological sample, but further comprising the step of contacting the biological sample with an agent which increases the susceptibility of bacterial organisms to antibiotics.
  • Methods of decreasing bacterial quantity are effective if the number of bacteria decreases at least 10%, and preferably more, e.g., 25%, 50%, 75% or even 100% after contacting the biological sample with any one of Compounds 1 to 7, a compound of Formula I as described herein, or a pharmaceutically acceptable derivative or prodrug thereof.
  • pharmaceutically acceptable derivatives or prodrugs of the compounds of this invention may also be employed in compositions to treat or prevent the above-identified disorders.
  • a “pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • Pharmaceutically acceptable prodrugs of the compounds of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters.
  • Amycolatopsis orientalis ATCCTM 43491 was cultivated under aerobic conditions in an aqueous nutrient medium containing assimilable sources of carbon, assimilable sources of nitrogen, inorganic salts and vitamins.
  • Preferred carbon sources are glucose, glycerol and the like.
  • Preferred nitrogen sources are beef extract, malt extract, yeast extract, and the like. Representative media are provided in Table 1.
  • Applied and Environmental Microbiology 76, 377-386 composed of 30 g trypticase soy broth (BactoTM), 3 g yeast extract, 2 g MgSO 4 , 5 g glucose, 4 g maltose made up to one liter with distilled water. The flasks are shaken (250 rpm) for about 60 hours at 28° C.
  • Compounds 1, 2, 7, 8 and 15 could also be produced in other media including JA, GA, RM, NA, CA, and CB (Table 1). Compounds 1 and 2 were further produced as described above using a preferred strain, namely Amycolatopsis orientalis IDAC 220604-01.
  • the residue was further purified by HPLC (Waters Autopurification System with ACD), using a Waters Xterra MS C18 column (5 ⁇ , 19 ⁇ 150 mm), and a gradient of 10 mM aqueous NH 4 HCO 3 , pH 10/acetonitrile 85:15 to 25:75 over 30 min at 19 mL/min, UV detector set at 261 nm.
  • the sample was loaded as a suspension in DMSO/MeOH (3:1).
  • the pooling of eluate gave samples of Compound 7 ( ⁇ 1 mg, RT: 16-17 min) and a mixture of Compounds 1 and 2 (1.04 g, non-freezedried, RT: 11.8-12.1 min).
  • the first purification (step 2) was also accomplished using a Phenomenex Max-RP C12 column (4 ⁇ , 21.2 ⁇ 250 mm) using the gradient buffer (as above) and acetonitrile 89.5:10.5 to 20:80 over 25 minutes, with the same flow and UV detection. Fractions were collected at a retention time of 16.5-17 minutes (mixture of Compounds 1 and 2) and at RT: 21-22 minutes (Compound 7).
  • the mixture of Compounds 1 and 2 is further purified by HPLC (Waters Autopurification System with ACD), using a RCM Column (Novapak C-18, 6 ⁇ , 40 ⁇ 200 mm), and a gradient of 10 mM aqueous ammonium acetate (NH 4 OAc) to pH 5 with glacial acetic acid/acetonitrile 80:20 to 20:80 over 25 min at 35 mL/min. Fractions were collected and freeze-dried to give pure Compound 1 (RT: 18.5-18.8 minutes, 224.5 mg) and pure Compound 2 (RT: 17.3-17.6 minutes, 34.6 mg).
  • HPLC Waters Autopurification System with ACD
  • the crude residue of 1 a) was partitioned between 100 mL(per litre of fermentation) of hexanes and 100 mL(per litre of fermentation) of methanol (MeOH) in water (3:2) buffered to pH 10 with ammonium hydroxide (NH 4 OH) and at 10 mM ammonium bicarbonate (NH 4 HCO 3 ) salt concentration. The two layers were separated and the methanol:water layer evaporated to dryness. The procedure was repeated and concentrated to dryness.
  • the mixture of Compounds 1 and 2 was further purified by HPLC (Waters Autopurification System with ACD), using a Symmetry C18 column (5 ⁇ , 30 ⁇ 100 mm), and a gradient of 10 mM aqueous NH 4 OAc, made to pH 5 with glacial acetic acid/acetonitrile 74:26 to 50:50 over 20 min at 39 mL/min.
  • the collection was triggered by UV 261 nm (PDA).
  • the sample was loaded as a suspension in DMSO:MeOH (3:1).
  • the pooling of eluates gave pure Compound 1 (RT: 14.9-15.2 min) and pure Compound 2 (RT: 14.1-14.2 min), generally with a ratio of 5:1.
  • Compound 7 had a retention time of 13-14 minutes using the same conditions.
  • Compounds 1, 2 and 7 were produced by fermentation as described in Example 1 and isolated as described in Example 2.
  • Compound 1 is named 3,4,5-trihydroxy-6-[1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid.
  • Compound 1 structure determination was based on mass, UV and NMR data.
  • the NMR data detailed in Table 2 was collected at 500 MHz in d 4 -MeOH including 1 H-NMR spectrum of FIG. 4 , and the multidimensional pulse sequences gCOSY, gDQCOSY, gHSQC, and gHMBC.
  • the molecular formula of C 46 H 68 N 4 O 10 and the chemical structure were established based the 1 H-NMR data, the COSY, HSQC and HMBC measured on about 500 ⁇ g of very pure material of structure illustrated above.
  • the carbon assignments shown in Table 2 were made by virtue of the HSQC and HMBC.
  • UV spectrum for Compound 1 exhibited UV ⁇ max at 258.77 and 362.77 nm in methanol in accordance with the methylpentaene amide of the 2-aminocyclopenta-1,3-dione tautomer.
  • Compound 2 is named 6- ⁇ 16-guanidino-1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethylhexadeca-2,8,12-trienyloxy ⁇ -3,4,5-trihydroxy-tetrahydro-pyran-2-carboxylic acid.
  • Compound 7 is named 6-[1-(11-carboxy-1-methyldodeca-2,4,5,8,10-pentaenyl)-2,4,10-trimethyl-16-(N-methylguanidino)-hexadeca-2,8,12-trienyloxy]-3,4,5-trihydroxy-tetrahydropyran-2-carboxylic acid.
  • Compound 3 is named 3,4,5-trihydroxy-6-[1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid methyl ester.
  • Compound 4 is named 3,4,5-trihydroxy-6-[1-[11 -(2-methoxy-5-oxo-cyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid.
  • Compound 5 is named 3,4,5-trihydroxy-6-[1-[11 -(2-methoxy-5-oxo-cyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid methyl ester.
  • Diazomethane was generated by standard methods in an Aldrich diazomethane-generator [Z41, 173-6]. Approx. 2 mL 5N sodium hydroxide was added dropwise to 450 mg 1-methyl-3-nitro-1-nitrosoguanidine and the generated gaseous diazomethane was condensed in 4 mL of diethyl ether at 0° C. The diazomethane solution was added dropwise to a solution of Compound 1 (10 mg) in 6 mL of methanol. The reaction was carried out at room temperature with magnetic stirring maintained for 15 minutes until completion. The reaction product was evaporated at 30° C. under a gentle stream of N 2 .
  • the crude material was purified by multiple injections on HPLC using a YMC ODS-A 10 ⁇ 250 mm column and a gradient of 5 mM NH 4 OAc/acetonitrile at 5 mL/min (90:10 for 1 min, 90:10-30:70 o. 20 min).
  • the combined yield of pure Compound 3 derived from two separate 10 mg batches was 0.99 mg.
  • Compound 6 is named 6- ⁇ 16-(N′-acetyl-N-methylguanidino)-1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethylhexadeca-2,8,12-trienyloxy ⁇ -3,4,5-trihydroxytetrahydropyran-2-carboxylic acid.
  • Compound 6 was prepared by acetylation of Compound 1 using the following procedure.
  • Acetic anhydride (3.38 ⁇ L, Sigma) was added to a solution of Compound 1 (20 mg) in methanol (2 mL) and the reaction stirred at room temperature for 72 hours. Additional portions of acetic anhydride (20 ⁇ L) were added after 24 and 48 hours.
  • the reaction was monitored by TLC (Merck Silica gel 60 F 254 , eluting with 7% methanol in chloroform) visualized under UV. The reaction was concentrated in vacuo.
  • culture media (ex: MH, BHI and Agar) were supplied by Quelab Laboratories Inc, Montreal, Canada. Indicator strains were supplied by American Type Culture Collection (ATCCTM, Manassas, Va., USA).
  • Antibacterial activity of Compounds 1 and 2 was measured by determining the minimal inhibitory concentration (MIC) necessary to obtain a complete inhibition of bacteria growth in eight indicator strains, namely Staphylococcus aureus (ATCCTM 6538P), Staphylococcus aureus MRS3 (ATCCTM 700699), Staphylococcus epidermidis (ATCCTM 12228), Bacillus subtilis (ATCCTM 23857), Bacillus megaterium (ATCCTM 14581), Enterococcus faecalis VRE-1 (ATCCTM 29212), Enterococcus faecalis VRE-2 (ATCCTM 51299) and Micrococcus luteus (ATCCTM 9341).
  • MIC minimal inhibitory concentration
  • NCCLS National Committee for Clinical Laboratory Standards
  • M7-A5 Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard - Fifth Edition.
  • NCCLS document M7-A5 ISBN 1-56238-394-9; NCCLS, 940 West Valley Road, Suite 1400, Wayne, Pa. 19087-1898 USA), the content of which is incorporated herein by reference.
  • Compounds 1 and 2 were prepared as 100 ⁇ stock solution in DMSO, with concentrations ranging from 3.2 mg/ml to 0.00625 mg/ml (a two-fold dilution series over 12 points). An aliquot of each 100 ⁇ stock solution was diluted 50-fold in test medium described below to give a set of ten (10) 2 ⁇ solutions ranging from 64 to 0.125 ⁇ g/mL. 50 ⁇ l of each of the ten 2 ⁇ solutions was aliquoted into the corresponding well of a 12-well row, with the final well reserved for medium alone control. Each test was made in duplicate.
  • Vancomycin used as positive control compounds, was prepared as 2 ⁇ stock solutions in sterile, double-distilled water (ddH 2 O) ranging from 64 ⁇ g/ml to 0.00125 ⁇ g/ml (a two-fold dilution series over 10 points). An aliquot of 50 ⁇ l corresponding to each concentration (at 2 ⁇ ) was then transferred to 96-well microplates to obtain a series of eleven two-fold dilutions.
  • the indicator strains were incubated with 11 concentrations of each of Compounds 1 and 2, Vancomycin (SigmaTM) control compound and one media alone control. For MIC determination, assay plates were incubated at 35° C. for 18 hours. The MIC for each indicator was assessed as the lowest concentration of compound resulting in total absence of growth and is shown below in Table 4. TABLE 4 Antibacterial activity of Compounds 1 and 2, MIC ( ⁇ g/ml) Strain Compound 1 Compound 2 Vancomycin S. aureus ATCC TM 6538P 2 1 2 S. aureus ATCC TM 700699 4 1 4 S. epidermidis 4 1 2 ATCC TM 12228 B. subtilis ATCC TM 23857 1 2 0.25 B.
  • Antibacterial activitiy of Compounds.3, 4, 5, 6 and 7 are accomplished using the same method in a panel of bacterial strains and Vancomycin as positive control.
  • MBC minimal bactericidal concentration
  • NCCLS guideline M26-A Methods for Determining Bactericidal Activity of Antimicrobial Agents (NCCLS document M7-A5, ISBN 1-56238-394-9; NCCLS, 940 West Valley Road, Suite 1400, Wayne, Pa. 19087-1898 USA), the content of which is incorporated herein by reference.
  • Microdilutions broth assays were performed using the same procedure as for MIC determination, Example 6. 24 hours post-incubation, cells were collected in wells corresponding to the MIC, 2 ⁇ MIC and 4 ⁇ MIC. Collected cells were plated on agar media. MBC were determined as the drug concentration at which more than 99.9% growth inhibition was observed. As a reference for 100% growth, cells grown in media only were also plated.
  • Acute toxicities were determined using CD-1 female mice (supplied by Charles River Laboratories, Wilmington, Mass.) having a bodyweight of 18-25 g, and about 6 weeks of age.
  • Compounds 1 and 2 at doses ranging from 1 to 70 mg/kg (formulated in 20% propylene glycol/water for a total volume of 10 ⁇ L/g of weight) were administered intraperitoneal (i.p.) and intravenous (i.v.) to groups of 5 mice (5 mice per concentration, per mode of administration). Two groups of 3 mice were injected with vehicle only. Mice were observed immediately after injection, within 4 hours and once a day for the subsequent days.
  • mice supplied by Charles River Laboratories, Wilmington, Mass. having a bodyweight of 20 ⁇ 2 g.
  • Mice (38) were inoculated i.p. with an LD 90-100 dose (1 ⁇ 10 6 CFU/mouse) of Staphylococcus aureus (Smith strain, ATCCTM 13709) suspended in 0.5 mL of BHI containing 5% mucin.
  • Compound 1 at doses 20, 30, 40, 50, 60 and 70 mg/kg (formulated in 20% propylene glycol/water for a total volume of 10 ⁇ L/g of weigth) was administered i.p. one hour post infection (5 mice for each concentration).
  • mice Another group of 5 mice was treated i.v. with a 10 mg/kg dose of Vancomycin. A last group of 3 mice was administered vehicle only. Mortality was recorded for 7 days.
  • In vivo efficacy studies showed 100% protection at the lowest dose tested (20 mg/kg i.p.) against an acute lethal infection with S. aureus. TABLE 7 Tolerability and efficacy of Compound 1 In vitro Hemolytic activity (EC 50 )* >128 ⁇ g/ml In vivo Mouse MTD 70 mg/kg - i.p. In vivo efficacy: S.
  • LAH lithium aluminum hydride
  • THF tetrahydrofuran
  • 1M hydrochloric solution is used to acidify the solution carefully.
  • the mixture is extracted three times with ethyl acetate. Organic layers are combined and washed with saturated aqueous sodium bicarbonate, water and brine, dried over magnesium sulfate, filtered and concentrated in vacuo. Pure Compound 11 is obtained by pooling and concentrating the appropriate fractions of HPLC purification according to Example 4.
  • Compound 23 is prepared by modification of the guanidino group of Compound 1 according to the procedure described in Argoudelis et al., J. Antibiotics, Vol. XL, No. 6, June 1987, pp 750-760.
  • a mixture of 200 mg of Compound 1 in 1.2 ml water, 1.0 ml of absolute ethanol,1.0 ml of 2,4-pentadione and 120 mg of sodium bicarbonate is stirred at 90° C. for 3 hours. The mixture is allowed to cool to room temperature, concentrated to dryness, dissolved in 5 ml of 2N acetic acid, and purified by HPLC as described in Example 4. Pure Compound 23 is obtained by pooling and concentrating the appropriate eluate fractions.
  • Compound 24 is prepared as follows. See Bartzatt et al., Biotechnol. Appl. Biochem. (2002) 36, 89-93.
  • a Wheaton-type double-chamber device (Wheaton Co., Millville, N.J., U.S.A.) is utilized to generate diazopropane (CH 3 CH 2 CHN 2 ).
  • Approx. 5.0 mg of Compound 1 is placed into an organic solvent (ethyl acetate/diethyl ether, 1:1, v/v) and the diazopropane gas formed is allowed to dissolve in the mixture.
  • Diazopropane is generated by mixing 0.15 ml of 5 M NaOH with 0.15 g of 3-nitro-1-nitroso-1-propylguanidine.
  • Compound 25 is prepared by acylation of Compound 1 as follows. Acetic anhydride (4.5 equivalents) is added dropwise to a solution of 50 mg/ml of Compound 1 and in acetonitrile and pyridine (9:1). The mixture is stirred under reflux and monitored by TLC (see example 5). The solvent is removed under vacuum and the residue is dissolved in methanol. The methanol solution is purified by HPLC as described in Example 4. Pure Compound 25 is obtained by pooling and concentratin the appropriate eluate fractions.
  • Compound 27 is prepared by acylation of Compound 1 as follows. Acetic anhydride (3.2 equivalents) is added dropwise to a solution of 50 mg/ml of Compound 1 in acetonitrile. The mixture is stirred under reflux and monitored by TLC (see Example 5). The solvent is removed under vacuum and the residue is dissolved in methanol. The methanol solution is purified by HPLC as described in Example 4. Pure Compound 27 is obtained by pooling and concentrating the appropriate eluate fractions.

Abstract

This invention related to methods of inhibiting the growth of bacterial cells and methods of treating bacterial infections in mammals using a novel polyene polyketide.

Description

    RELATED APPLICATIONS
  • This application is a continuation-in-part application of U.S. application Ser. No. 10/937,379 filed Sep. 10, 2004, which claims priority from U.S. provisional application 60/501,821 filed Sep. 11, 2003, U.S. provisional application 60/574,922 filed May 28, 2004 and U.S. provisional application 60/581,707 filed Jun. 23, 2004. The entire teachings of the above application are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • This invention relates to methods of treating bacterial infections in a mammal using a compound of using a polyene polyketide of Formula I or its pharmaceutically acceptable salts.
  • BACKGROUND OF THE INVENTION
  • Microbial natural products have been a reliable source of antimicrobial drugs. Streptomycin, erythromycin, tetracycline, pristinamycin, vancomycin and lincomycin are examples of antibiotics produced by microorganisms. However, the discovery of new antibiotics has dropped significantly over the past few decades. At the same time, problems related to resistance to current antibiotics by severe pathogens such as VRE and MRSA have been on the rise. According to the U.S. Center for Disease Control and Prevention, MRSA infections are common causes of hospital acquired infections, and a limited number of drugs remain effective against these infections. The number of hospital-acquired infections is growing as hospitals are increasingly treating patients suffering from compromised immune systems caused by such things as more cancer patients and the associated chemotherapy treatments, aging of the population, and increasing numbers of surgical operations such as organ transplants that require immunosuppressants. From the foregoing, there is a need for new antibiotic therapeutics that have higher efficacy, specificity or reduced side effects.
  • SUMMARY OF THE INVENTION
  • The invention encompasses a method of inhibiting bacterial infection, the method comprising contacting the bacterial cells with a therapeutically effective amount of a compound selected from Compounds 1, 2, 3, 4, 5, 6, 7 and pharmaceutically acceptable salts or prodrugs thereof:
    Figure US20050215490A1-20050929-C00001
    Figure US20050215490A1-20050929-C00002

    such that growth of the bacterial infection is inhibited.
  • In another aspect the invention provides a method of inhibiting the growth of a bacterial infection, the method comprising contacting a bacterial cell with a therapeutically effective amount of a compound of Formula I:
    Figure US20050215490A1-20050929-C00003

    such that growth of the bacterial infection is inhibited;
    wherein,
      • A is selected from —C(NH)NHR1, CH3, H or
        Figure US20050215490A1-20050929-C00004
      • R1 is selected from H, C1-6alkyl, C6-10aryl, C(O)C1-6alkyl and C(O)C6-10aryl;
      • R2 and R3 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl;
      • R4is selected from H or CH3;
      • Z is OH or O when taken with adjacent carbon atom to form a carbonyl; or
      • Z may be a tetrahydropyranoxy of formula:
        Figure US20050215490A1-20050929-C00005
      • R5is selected from H, COOH, C1-6 alkyl or C(O)OC1-6 alkyl;
      • R6, R7 and R8 are each independently selected from H, C1-6 alkyl and C(O)C1-6 alkyl; or
      • R6, R7 and R8 may each independently be absent when the adjacent oxygen and carbon atoms are taken together to form a carbonyl; or
      • R6, R7 and R8 may each independently be a bond when any of two neighboring R6, R7 and R8 are taken together with attached oxygen and carbon atoms to form a 1,3-dioxolane ring of formula:
        Figure US20050215490A1-20050929-C00006
      • R9 and R10 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl; or
      • R9 and R10 are taken together with adjacent carbon atom to form a ring having from 5 to 7 carbons;
      • D is selected from OH, NH2, NH(C1-3alkyl), N(C1-3alkyl)2, OC1-3alkyl or
        Figure US20050215490A1-20050929-C00007
      • R11 is selected from H or C1-3 alkyl;
        or a pharmaceutically acceptable salt or prodrug thereof.
  • In one embodiment of this aspect, A of the compound is —C(NH)NH2. Alternatively, in another embodiment, A of the compound is H. Alternatively, in still another embodiment, A of the compound is —C(NH)NHC(O)CH3. Alternatively, in yet another embodiment, A of the compound is
    Figure US20050215490A1-20050929-C00008

    In another embodiment of the compound R4 is CH3. Alternatively, in yet another embodiment, R4 of the compound is H. The present invention further contemplates an embodiment of this aspect in which Z is
    Figure US20050215490A1-20050929-C00009

    In a sub-class of this embodiment, R6, R7 and R8 are each H, and R5 is COOH or CO2CH3. Alternatively, the invention further contemplates an embodiment of this aspect in which Z is OH. The invention further contemplates an embodiment of this aspect in which D is
    Figure US20050215490A1-20050929-C00010

    In a subclass of this embodiment R11 is H. The invention further contemplates an embodiment of this aspect in which D
    Figure US20050215490A1-20050929-C00011

    In a subclass of this embodiment R11 is CH3. Alternative, in another embodiment of this aspect D is OH.
  • Specific embodiments of compounds of Formula I useful to inhibit the growth of bacterial infection are as follows:
    Figure US20050215490A1-20050929-C00012
    Figure US20050215490A1-20050929-C00013
    Figure US20050215490A1-20050929-C00014
    Figure US20050215490A1-20050929-C00015
    Figure US20050215490A1-20050929-C00016
    Figure US20050215490A1-20050929-C00017
    Figure US20050215490A1-20050929-C00018

    or a pharmaceutically acceptable salt or prodrug of any one of Compounds 1-28.
  • In a further aspect, the invention relates to a method of inhibiting the growth of a bacterial infection, the method comprising contacting a bacterial cell with a pharmaceutical composition comprising of a compound of Formula I as defined above, or a pharmaceutically acceptable salt or prodrug thereof, with a pharmaceutically acceptable carrier, such as growth of the bacterial infection is inhibited. Specific embodiments of this aspect include a method of inhibiting the growth of bacterial infection, the method comprising contacting a bacterial cell with a phamaceutical composition comprising a compound of any one of Compounds 1-28 or a pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable carrier such that growth of the bacterial infection is inhibited.
  • The invention further encompasses a method of treating a bacterial infection in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of a compound selected from the Compounds 1, 2, 3, 4, 5, 6, 7 and pharmaceutically acceptable salts or prodrugs thereof:
    Figure US20050215490A1-20050929-C00019
    Figure US20050215490A1-20050929-C00020

    such that the bacterial infection is treated.
  • In another aspect, the invention provides a method of treating a bacteria infection in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of a therapeutically effective amount of a compound of Formula I:
    Figure US20050215490A1-20050929-C00021

    such that the bacterial infection is treated;
    wherein,
      • A is selected from —C(NH)NHR1, CH3, H or
        Figure US20050215490A1-20050929-C00022
      • R1 is selected from H, C1-6alkyl, C6-10aryl, C(O)C1-6alkyl and C(O)C6-10aryl;
      • R2 and R3 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl;
      • R4 is selected from H or CH3;
      • Z is OH or O when taken with adjacent carbon atom to form a carbonyl; or
      • Z may be a tetrahydropyranoxy of formula:
        Figure US20050215490A1-20050929-C00023
      • R5 is selected from H, COOH, C1-6 alkyl or C(O)OC1-6 alkyl;
      • R6, R7 and R8 are each independently selected from H, C1-6 alkyl and C(O)C1-6 alkyl; or
      • R6, R7 and R8 may each independently be absent when the adjacent oxygen and carbon atoms are taken together to form a carbonyl; or
      • R6, R7 and R8 may each independently be a bond when any of two neighboring R6, R7 and R8 are taken together with attached oxygen and carbon atoms to form a 1,3-dioxolane ring of formula:
        Figure US20050215490A1-20050929-C00024
      • R9 and R10 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl; or
      • R9 and R10 are taken together with adjacent carbon atom to form a ring having from 5 to 7 carbons;
      • D is selected from OH, NH2, NH(C1-3alkyl), N(C1-3alkyl)2, OC1-3alkyl or
        Figure US20050215490A1-20050929-C00025
      • R11 is selected from H or C1-3 alkyl;
        or a pharmaceutically acceptable salt or prodrug thereof.
  • Embodiment of Formula I useful in treating bacterial infection in a mammal are compounds of Formula I wherein A of the compound is —C(NH)NH2. Alternatively, in another embodiment, A of the compound is H. Alternatively, in still another embodiment, A of the compound is —C(NH)NHC(O)CH3. Alternatively, in yet another embodiment, A of the compound is
    Figure US20050215490A1-20050929-C00026

    In another embodiment of the compound R4 is CH3. Alternatively, in yet another embodiment, R4 of the compound is H. The present invention further contemplates an embodiment of this aspect in which Z is
    Figure US20050215490A1-20050929-C00027

    In a sub-class of this embodiment, R6, R7 and R8 are each H, and R5 is COOH or CO2CH3. Alternatively, the invention further contemplates an embodiment of this aspect in which Z is OH. The invention further contemplates an embodiment of this aspect in which D is
    Figure US20050215490A1-20050929-C00028

    In a subclass of this embodiment R11 is H. The invention further contemplates an embodiment of this aspect in which D is
    Figure US20050215490A1-20050929-C00029

    In a subclass of this embodiment R11 is CH3. Alternative, in another embodiment of this aspect D is OH. Specific compounds of Formula I useful to treat bacterial infection in a mammal are compound 1-28.
  • In a further aspect, the invention relates to a method of treating bacterial infection in a mammal, the method comprising contacting a bacterial cell with a pharmaceutical composition comprising of a compound of Formula I as defined above, or a pharmaceutically acceptable salt or prodrug thereof, with a pharmaceutically acceptable carrier, such that the bacterial infection is treated. Specific embodiments of this aspect include a method of treating bacterial infection in a mammal, the method comprising contacting a bacterial cell with a phamaceutical composition comprising a compound of any one of Compounds 1-28 or a pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable carrier such that the bacterial infection is treated.
  • In one embodiment, the bacterial infection is from a bacteria selected from the group consisting of Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp., Proteus spp., Pseudomonas aeruginosa, E. coli, Serratia marcesens, Staphylococcus aureus, Coagulase negative Staphylococcus, Haemophilus infuenzae, Bacillus anthracis, Mycoplasma pneumoniae, and Staphylococcus epidermidis. In another embodiment, the bacterial infection is caused by Gram-positive bacteria. In a further embodiment, the Gram-positive bacteria is a Staphylococcus or Enterococcus species.
  • According to one embodiment of this aspect, the compound is administered orally. In another embodiment, the compound is administered intravenously. In still another embodiment, the compound is administered intraperitoneally.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1: Electrospray mass spectrum (Positive Ion Mode) of Compound 1.
  • FIG. 2: Electrospray mass (ms/ms of m/z:837.50) spectrum (Positive Ion Mode) of Compound 1.
  • FIG. 3: Electrospray mass spectrum (Negative Ion Mode) of Compound 1.
  • FIG. 4: 500 MHz proton nuclear magnetic resonance (1H-NMR) spectrum of Compound 1 in d4-MeOH.
  • FIG. 5: 500 MHz proton nuclear magnetic resonance (1H-NMR) spectrum of Compound 2 in d4-MeOH.
  • FIG. 6: 500 MHz proton nuclear magnetic resonance (1H NMR) spectrum of Compound 3 in d4-MeOH.
  • FIG. 7: 500 MHz proton nuclear magnetic resonance (1H NMR) spectrum of Compound 4 in d4-MeOH.
  • FIG. 8: 500 MHz proton nuclear magnetic resonance (1H NMR) spectrum of Compound 5 in d4-MeOH.
  • FIG. 9: 500 MHz proton nuclear magnetic resonance (1H NMR) spectrum of Compound 6 in d4-MeOH.
  • FIG. 10: 500 MHz proton nuclear magnetic resonance (1H NMR) spectrum of Compound 7 in d4-MeOH.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to methods of inhibiting the growth of bacterial cells. The method comprtises contacting bacterial cells with a polyene polyketide compound of Formula I, as exemplified herein as Compound 1, Compound 2 and Compound 7, which were isolated from strains of actinomycetes, Amycolatopsis sp. such as Amycolatopsis orientalis ATCC 43491, Amycolatopsis orientalis IDAC 220604-01, or a mutant or a variant thereof. Amycolatopsis orientalis ATCC 43491 may be obtained from the American Type Culture Collection (ATCC) P.O. Box 1549, Manassas, Va. 20108 USA; Amycolatopsis orientalis IDAC 220604-01 may be obtained from the International Depositary Authority of Canada (IDAC), Bureau of Microbiology, Health Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2.
  • The present invention further related to methods of treating a bacterial infection in a mammal, the method comprising administering a therapeutically effective amount of a polyene polyketide compound of the invention, such that the bacterial infection is treated. Compound 1, Compound 2 and Compound 7 are useful as a pharmaceutical, in particular for use as in inhibitor of bacterial cell growth. Alternatively a compound of Formula I can be used. Accordingly, certain aspects of the present invention relate to use of the polyene polyketide compounds of the present invention in to inhibit bacterial growth, and use of the polyene polyketides of the invention in the preparation of a medicament to treat bacterial infection.
  • The invention further related to pharmaceutically acceptable salts and derivatives of the compounds 1-7 and to methods for obtaining such compounds. One method of obtaining the compounds is by cultivating Amycolatopsis orientalis ATCC™ 43491, or a mutant or a variant thereof, under suitable Amycolatopsis sp. culture conditions preferably using the fermentation protocol described herein below.
  • The invention also relates to pharmaceutical compositions comprising a compound selected from 1-28 and its pharmaceutically acceptable salts and derivatives. Accordingly, certain aspects of the present invention relate to use to inhibit bacterial growth of a pharmaceutical compositions comprising a polyene polyketide compound of the present invention together with a pharmaceutically acceptable carrier, and use in the preparation of medicament to treat bacterial infection of a pharmaceutical compositions comprising a polyene polyketide compound of the present invention together with a pharmaceutically acceptable carrier.
  • Accordingly, certain aspects of the present invention relate to pharmaceutical compositions comprising the polyene polyketide compounds of the present invention together with a pharmaceutically acceptable carrier, methods of using the compositions to inhibit bacterial growth, and methods of using the pharmaceutical compositions to treat bacterial infections.
  • I. DEFINITIONS
  • Molecular terms, when used in this application, have their common meaning unless otherwise specified. The meaning of certain terms and phrases used in the specification are provided below. Abreviations, as used herein, have the following meaning: Me refers to methyl (CH3), Et refers to ethyl (CH2CH3), Pr refers to n-propyl (CH2CH2CH3) and Ac refers to acetyl (C(O)CH3).
  • The term alkyl refers to a linear or branched hydrocarbon groups. Examples of alkyl groups include, without limitation, methyl, ethyl, n-propyl, isopropyl, n-butyl, pentyl, hexyl, heptyl, cyclopentyl, cyclohexyl, cyclohexymethyl, and the like. Alkyl may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro,.thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl, oxo, guanidino and formyl.
  • The term alkenyl refers to linear, branched or cyclic hydrocarbon groups containing at least one carbon-carbon double bond. Examples of alkenyl groups include, without limitation, vinyl, 1-propene-2-yl, 1-butene-4-yl, 2-butene-4-yl, 1-pentene-5-yl and the like. Alkenyl may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, alkoxy, aryloxy, sulfinyl, sulfonyl, formyl, oxo and guanidino. The double bond portion(s) of the unsaturated hydrocarbon chain may be either in the cis or trans configuration.
  • The term cycloalkyl or cycloalkyl ring refers to a saturated or partially unsaturated carbocyclic ring in a single or fused carbocyclic ring system having from three to fifteen ring members. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclohexyl, and cycloheptyl. Cycloalkyl may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • The term heterocyclyl, heterocyclic or heterocyclyl ring refers to a saturated or partially unsaturated ring containing one to four hetero atoms or hetero groups selected from O, N, NH, NRx, PO2, S, SO or SO2 in a single or fused heterocyclic ring system having from three to fifteen ring members. Examples of a heterocyclyl, heterocyclic or heterocyclyl ring include, without limitation, morpholinyl, piperidinyl, and pyrrolidinyl. Heterocyclyl, heterocyclic or heterocyclyl ring may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, oxo, thiocarbonyl, imino, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • The term amino acid refers to any natural amino acid, all natural amino acids are well known to a person skilled in the art.
  • The term halo is defined as a bromine, chlorine, fluorine or iodine.
  • The term aryl or aryl ring refers to common aromatic groups having “4n+2” electrons, wherein n is an integer from 1 to 3, in a monocyclic or conjugated polycyclic system and having from five to fifteen ring atoms. Aryl ring may include from 1 to 3 heteroatoms such as nitrogen, oxygen and sulphur atoms. Examples of aryl include, without limitation, phenyl, naphthyl, biphenyl, terphenyl, furyl, pyrrollyl, thienyl, pyridyl, oxazolyl, imidazolyl, pyrazolyl and indolyl groups. Aryl may optionally be substituted with one or more substituent group selected from acyl, amino, acylamino, acyloxy, azido, alkythio, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • The compounds of the present invention can possess one or more asymetric carbon atoms and can exist as optical isomers forming mixtures of racemic or non-racemic compounds. The compounds of the present invention are useful as a single isomer or as a mixture of stereochemical isomeric forms. Diastereoisomers, i.e., nonsuperimposable stereochemical isomers, can be separated by conventional means such as chromatography, distillation, crystallization or sublimation. The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes.
  • The invention embraces isolated compounds. An isolated compound refers to a compound which represents at least 10%, 20%, 50% and 80% of the compound of the present invention present in a mixture, provided that the mixture comprising the compound of the invention-has demonstrable (i.e. statistically significant) biological activity including antimicrobial activity when tested in conventional biological assays known to a person skilled in the art.
  • As used herein, the term “treatment” refers to the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disorder, e.g., a disease or condition, a symptom of disease, or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disease, the symptoms of disease, or the predisposition toward disease.
  • As used herein, a “pharmaceutical composition” comprises a pharmacologically effective amount of a compound of the invention and a pharmaceutically acceptable carrier. As used herein, “pharmacologically effective amount,” “therapeutically effective amount” or simply “effective amount” refers to that amount of polyene polyketide effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 25% reduction in that parameter.
  • The term “pharmaceutically acceptable carrier” refers to a carrier for administration of a therapeutic agent. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The term specifically excludes cell culture medium. For drugs administered orally, pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • The term “pharmaceutically acceptable salts” include acid addition salts and base addition salts. The nature of the salt is not critical, provided that it is pharmaceutically-acceptable. Without being limited, examples of acid addition salts include hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulphuric, phosphoric, formic, acetic, citric, tartaric, succinic, oxalic, malic, glutamic, propionic, glycolic, gluconic, maleic, embonic (pamoic), methanesulfonic, ethanesulfonic, 2-hydroxyethanesulfonic, pantothenic, benzenesulfonic, toluenesulfonic, sulfanilic, mesylic, cyclohexylaminosulfonic, stearic, algenic, β-hydroxybutyric, malonic, galactantic, galacturonic acid and the like. Suitable pharmaceutically-acceptable base addition salts of compounds of the invention include, but are not limited to, metallic salts made from aluminium, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, lysine, procaine and the like. Additional examples of pharmaceutically acceptable salts are listed in Berge S M et al., Journal of Pharmaceutical Sciences, (1977) Vol. 66 no 1, pp. 1-19. All of these salts may be prepared by conventional means form the corresponding compounds of Formula I by treating with the appropriate acid or base.
  • Unless otherwise indicated, all numbers expressing quantities of ingredients and properties such as molecular weight, reaction conditions, MIC and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the present specification and attached claims are approximations. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of significant figures and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the examples, tables and figures are reported as precisely as possible. Any numerical values may inherently contain certain errors resulting from variations in experiments, testing measurements, statistical analysis and such.
  • II. POLYENE POLYKETIDES OF THE INVENTION
  • In one aspect of this embodiment the invention relates to novel polyene polyketides, referred to herein as Compounds 1-7:
    Figure US20050215490A1-20050929-C00030
    Figure US20050215490A1-20050929-C00031

    or, to a pharmaceutically acceptable salt or prodrug of any of Compounds 1 to 7. Compounds 1 to 7 may be characterized by any one or more of their physicochemical and spectral properties given below, such as mass, UV, and NMR spectroscopic data.
  • In another aspect the invention relates to a novel class of polyene polyketides represented by Formula I:
    Figure US20050215490A1-20050929-C00032

    wherein,
      • A is selected from —C(NH)NHR1, CH3, H or
        Figure US20050215490A1-20050929-C00033
      • R1 is selected from H, C1-6alkyl, C6-10aryl, C(O)C1-6alkyl and C(O)C6-10aryl;
      • R2 and R3 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl;
      • R4is selected from H or CH3;
      • Z is OH or O when taken with adjacent carbon atom to form a carbonyl; or
      • Z may be a tetrahydropyranoxy of formula:
        Figure US20050215490A1-20050929-C00034
      • R5 is selected from H, COOH, C1-6 alkyl or C(O)OC1-6 alkyl;
      • R6, R7 and R8 are each independently selected from H, C1-6 alkyl and C(O)C1-6 alkyl; or
      • R6, R7 and R8 may each independently be absent when the adjacent oxygen and carbon atoms are taken together to form a carbonyl; or
      • R6, R7 and R8 may each independently be a bond when any of two neighboring R6, R7 and R8 are taken together with attached oxygen and carbon atoms to form a 1,3-dioxolane ring of formula:
        Figure US20050215490A1-20050929-C00035
      • R9 and R10 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl; or
      • R9 and R10 are taken together with adjacent carbon atom to form a ring having from 5 to 7 carbons;
      • D is selected from OH, NH2, NH(C1-3alkyl), N(C1-3alkyl)2, OC1-3alkyl or
        Figure US20050215490A1-20050929-C00036
      • R11 is selected from H or C1-3 alkyl;
        or a pharmaceutically acceptable salt or prodrug thereof.
  • In one embodiment the invention provides compounds of Formula I, wherein A is —C(NH)NH2; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment of the invention provides compounds of Formula I, wherein. A is H; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein A is —C(NH)NHC(O)CH3; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein A is
    Figure US20050215490A1-20050929-C00037

    and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein R4 is CH3; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein R4 is H; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein Z is
    Figure US20050215490A1-20050929-C00038

    and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof. In a subclass of this embodiment R6, R7 and R8 are each H, and R5 is COOH, all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein Z is
    Figure US20050215490A1-20050929-C00039

    and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof. In a subclass of this embodiment R6, R7 and Ra are each H, and R5 is CO2CH3, all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein Z is OH and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein D is
    Figure US20050215490A1-20050929-C00040

    and all other groups are as previously defined or a pharmaceutically acceptable salt or prodrug thereof. In a subclass of this embodiment R11 is H and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In a further embodiment the invention provides compounds of Formula I, wherein D is
    Figure US20050215490A1-20050929-C00041

    and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof. In a subclass of this embodiment R11 is CH3 and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • In another embodiment the invention provides compounds of Formula I, wherein D is OH; and all other groups are as previously defined; or a pharmaceutically acceptable salt or prodrug thereof.
  • The following are exemplary compounds of the invention:
    Figure US20050215490A1-20050929-C00042
    Figure US20050215490A1-20050929-C00043
    Figure US20050215490A1-20050929-C00044
    Figure US20050215490A1-20050929-C00045
    Figure US20050215490A1-20050929-C00046
    Figure US20050215490A1-20050929-C00047
    Figure US20050215490A1-20050929-C00048

    or a pharmaceutically acceptable salt or prodrug of any one of Compound 1-28. Certain embodiments may exclude one or more of the compounds of Formula I. The compounds of the invention may be formulated into pharmaceutical compositions comprised compounds of Formula I in combination with a pharmaceutical acceptable carrier as discussed in Section IV below.
  • III. METHODS FOR PRODUCING THE COMPOUNDS OF THE INVENTION BY FERMENTATION
  • In one embodiment, compounds 1-7 may be obtained by cultivating a strain of actinomycetes, Amycolatopsis sp. such as Amycolatopsis orientalis ATCC 43491, Amycolatopsis orientalis IDAC 220604-01, or a mutant or a variant thereof. Amycolatopsis orientalis ATCC 43491 may be obtained from the American Type Culture Collection (ATCC) P.O. Box 1549, Manassas, Va. 20108 USA; Amycolatopsis orientalis IDAC 220604-01 may be obtained from the International Depositary Authority of Canada (IDAC), Bureau of Microbiology, Health Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2. The deposit of Amycolatopsis orientalis IDAC 220604-01 was made under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for Purposes of Patent Procedure. The deposited strain will be irrevocably and without restriction or conditions released to the public upon the issuance of a patent. The deposited strains are provided merely as a convenience to those skilled in the art and are not an admission that a deposit is required for enablement, such as that required under 35 U.S.C.§112. Indeed it is to be understood that the present invention is not limited to use of a particular strain of Amycolatopsis orientalis. Rather, the invention contemplates the use of other Formula I producing organisms, such as mutant or variants of Amycolatopsis orientalis ATCC 43491 or Amycolatopsis orientalis IDAC 220604-01 that can be derived by well known means such as X-ray irradiation, ultraviolet irradiation, treatment with nitrogen mustard, phage exposure, antibiotic selection and the like, or through the use of recombinant genetic engineering as described in co-pending parent application U.S. Ser. No. 10/937,379. In addition, the polyene polyketides of the invention may be biosynthesized by various microorganisms. Microorganisms that may synthesize the compounds of the present invention include but are not limited to bacteria of the order Actinomycetales, also referred to as actinomycetes. Non-limiting examples of members belonging to the genera of Actinomycetes include Nocardia, Geodermatophilus, Actinoplanes, Micromonospora, Nocardioides, Saccharothrix, Amycolatopsis, Kutzneria, Saccharomonospora, Saccharopolyspora, Kitasatospora, Streptomyces, Microbispora, Streptosporangium, Actinomadura. The taxonomy of actinomycetes is complex and reference is made to Goodfellow (1989) Suprageneric classification of actinomycetes, Bergey's Manual of Systematic Bacteriology, Vol. 4, Williams and Wilkins, Baltimore, pp 2322-2339, and to Embley and Stackebrandt, (1994), and The molecular phylogeny and systematics of the actinomycetes, Annu. Rev. Microbiol. 48, 257-289 (1994), for genera that may synthesize the compounds of the invention, incorporated herein in its entirety by reference.
  • Microorganisms producing polyene polyketides of the invention are cultivated in culture media containing known nutritional sources for actinomycetes, having assimilable sources of carbon, nitrogen plus optional inorganic salts and other known growth factors, at a pH of about 6 to about 9. Suitable media include, without limitation, growth media provided in Table 1. Microorganisms are cultivated at incubation temperatures of about 20° C. to about 40° C. for about 3 to about 40 days. The culture media inoculated with the polyene polyketide-producing microorganisms, may be aerated by incubating the inoculated culture media with agitation, for example shaking on a rotary shaker, or a shaking water bath. Aeration may also be achieved by the injection of air, oxygen or an appropriate gaseous mixture to the inoculated culture media during incubation. Following cultivation the polyene polyketide compounds can be extracted and isolated from the cultivated culture media by techniques known to a skilled person in the art and/or disclosed herein, including for example centrifugation, chromatography, adsorption, filtration. For example, the cultivated culture media can be mixed with a suitable organic solvent such as n-butanol, n-butyl acetate and 4-methyl-2-pentanone, the organic layer can be separated for example, by centrifugation followed by the removal of the solvent, by evaporation to dryness or by evaporation to dryness under vacuum. The resulting residue can optionally be reconstituted with for example water, ethyl ether, ethanol acetate, methanol or a mixture thereof, and re-extracted in a two-phase system with a suitable organic solvent such as hexane, carbon tetrachloride, methylene chloride or a mixture thereof. After removal of the solvent, the compound can be further purified by the use of standard techniques such as chromatography.
  • The polyene polyketides-biosynthesized by microorganism may optionally be subjected to random and/or directed chemical modifications to form compounds that are derivatives or structural analogs. Such derivative or structural analogs having similar functional activity are within the scope of the present invention. Polyene polyketides may optionally be modified using methods known in the art and described in co-pending application U.S. Ser. No. 10/937,379.
    TABLE 1
    Fermentation media
    Component CA CB GAa JA NA OA RM
    pH
    b 7 7 7.3 7 7 6.9
    Glucose 10 10 10 10
    Sucrose 20 103 100
    Cane molasses 15 5 10
    Corn starch 30
    Potato dextrin 40
    Corn steep liquor 15 3
    Yeast extract 5 3 5
    Malt extract 35 3
    Pharmamedia ™ 15
    Glycerol 20 5
    N-Z Amine A 10
    Beef extract 3
    Bacto-peptone 2 1
    Casamino acid 0.1 5 0.1
    Thiamine 0.1
    MgSO4.7H2O 1 0.2
    MgCl2.6H2O 10.12 10.13
    CaCO 3 2 5 2 4 2
    K2SO4 0.25 0.25
    FeSO4.7H2O 0.1
    KI 0.5
    MOPS 21
    Trace Elements 2 2
    Solutionc ml/L

    Unless otherwise indicated, all the components are in gm/L

    To a liter of media GA add: 10 ml KH2PO4 (0.5% solution); 80 ml CaCl2.2H2O (3.68% solution); 15 ml L-proline (20% solution); 100 ml TES buffer (5.73% solution, pH 7.2); 5 ml NaOH (1N solution).

    The pH is adjusted as marked prior to the addition of CaCO3.

    Solution of trace elements contains: ZnCl2 40 mg; FeCl3.6H2O (200 mg); CuCl2.2H2O (10 mg); (NH4)6MO7O24.4H2O (10 mg) per litre.
  • IV. PHARMACEUTICAL COMPOSITION COMPRISING THE COMPOUNDS OF THE INVENTION
  • The invention relates to a pharmaceutical composition comprising a polyene polyketide, as described in Section II and a pharmaceutically acceptable carrier as described below. The pharmaceutical composition comprising the polyene polyketide is useful for treating a variety of bacterial infections.
  • The compounds of the present invention, or pharmaceutically acceptable salts or prodrugs thereof, can be formulated for oral, intravenous, intramuscular, subcutaneous, topical or parenteral administration for the therapeutic or prophylactic treatment of diseases, particularly bacterial infections. For oral or parental administration, compounds of the present invention can be mixed with conventional pharmaceutical carriers and excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, wafers and the like. The compositions comprising a compound of this present invention will contain from about 0.1% to about 99.9%, about 1% to about 98%, about 5% to about 95%, about 10% to about 80% or about 15% to about 60% by weight of the active compound.
  • The pharmaceutical preparations disclosed herein are prepared in accordance with standard procedures and are administered at dosages that are selected to reduce, prevent, or eliminate bacterial infection (See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. and Goodman and Gilman's the Pharmaceutical Basis of Therapeutics, Pergamon Press, New York, N.Y. for a general description of the methods for administering various antimicrobial agents for human therapy, the contents of which are incorporated herein by reference). The compositions of the present invention can be delivered using controlled (e.g., capsules) or sustained release delivery systems (e.g., bioerodable matrices). Exemplary delayed release delivery systems for drug delivery that are suitable for administration of the compositions of the invention (preferably of Formula I) are described in U.S. Pat. No. 4,452,775 (issued to Kent), U.S. Pat. No. 5,039,660 (issued to Leonard), U.S. Pat. No. 3,854,480 (issued to Zaffaroni).
  • The pharmaceutically-acceptable compositions of the present invention comprise one or more compounds of the present invention in association with one or more non-toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants and/or excipients, collectively referred to herein as “carrier” materials, and if desired other active ingredients. The compositions may contain common carriers and excipients, such as corn starch or gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid. The compositions may contain crosarmellose sodium, microcrystalline cellulose, sodium starch glycolate and alginic acid.
  • Tablet binders that can be included are acacia, methylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone (Providone), hydroxypropyl methylcellulose, sucrose, starch and ethylcellulose.
  • Lubricants that can be used include magnesium stearate or other metallic stearates, stearic acid, silicon fluid, talc, waxes, oils and colloical silica. Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring or the like can also be used. It may also be desirable to add a coloring agent to make the dosage form more esthetic in appearance or to help identify the product comprising a compound of the present invention.
  • For oral use, solid formulations such as tablets and capsules are particularly useful. Sustained released or enterally coated preparations may also be devised. For pediatric and geriatric applications, suspension, syrups and chewable tablets are especially suitable. For oral administration, the pharmaceutical compositions are in the form of, for example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a therapeutically-effective amount of the active ingredient. Examples of such dosage units are tablets and capsules. For therapeutic purposes, the tablets and capsules which can contain, in addition to the active ingredient, conventional carriers such as binding agents, for example, acacia gum, gelatin, polyvinylpyrrolidone, sorbitol, or tragacanth; fillers, for example, calcium phosphate, glycine, lactose, maize-starch, sorbitol, or sucrose; lubricants, for example, magnesium stearate, polyethylene glycol, silica or talc; disintegrants, for example, potato starch, flavoring or coloring agents, or acceptable wetting agents. Oral liquid preparations generally are in the form of an aqueous or oily solution, suspensions, emulsions, syrups or elixirs may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous agents, preservatives, coloring agents and flavoring agents. Examples of additives for liquid preparations include acacia, almond oil, ethyl alcohol, fractionated coconut oil, gelatin, glucose syrup, glycerin, hydrogenated edible fats, lecithin, methyl cellulose, methyl or propyl para-hydroxybenzoate, propylene glycol, sorbitol, or sorbic acid.
  • For intravenous (IV) use, compounds of the present invention can be dissolved or suspended in any of the commonly used intravenous fluids and administered by infusion. Intravenous fluids include, without limitation, physiological saline or Ringer's solution.
  • Formulations for parental administration can be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions or suspensions can be prepared from sterile powders or granules having one or more of the carriers mentioned for use in the formulations for oral administration. The compounds can be dissolved in polyethylene glycol, propylene glycol, ethanol, corn oil, benzyl alcohol, sodium chloride, and/or various buffers.
  • For intramuscular preparations, a sterile formulation of compounds of the present invention or-suitable soluble salts forming the compound, can be dissolved and administered in a pharmaceutical diluent such as Water-for-Injection (WFI), physiological saline or 5% glucose. A suitable insoluble form of the compound may be prepared and administered as a suspension in an aqueous base or a pharmaceutically acceptable oil base, e.g. an ester of a long chain fatty acid such as ethyl oleate.
  • For topical use the compounds of present invention can also be prepared in suitable forms to be applied to the skin, or mucus membranes of the nose and throat, and can take the form of creams, ointments, liquid sprays or inhalants, lozenges, or throat paints. Such topical formulations further can include chemical compounds such as dimethylsulfoxide (DMSO) to facilitate surface penetration of the active ingredient.
  • For application to the eyes or ears, the compounds of the present invention can be presented in liquid or semi-liquid form formulated in hydrophobic or hydrophilic bases as ointments, creams, lotions, paints or powders.
  • For rectal administration the compounds of the present invention can be administered in the form of suppositories admixed with conventional carriers such as cocoa butter, wax or other glyceride.
  • Alternatively, the compounds of the present invention can be in powder form for reconstitution in the appropriate pharmaceutically acceptable carrier at the time of delivery. In another embodiment, the unit dosage form of the compound can be a solution of the compound or a salt thereof in a suitable diluent in sterile, hermetically sealed ampoules.
  • The amount of the compound of the present invention in a unit dosage comprises a therapeutically-effective amount of at least one active compound of the present invention which may vary depending on the recipient subject, route and frequency of administration. A recipient subject refers to a plant, a cell culture or an animal such as an ovine or a mammal including a human.
  • According to this aspect of the present invention, the novel compositions disclosed herein are placed in a pharmaceutically acceptable carrier and are delivered to a recipient subject (including a human subject) in accordance with known methods of drug delivery. In general, the methods of the invention for delivering the compositions of the invention in vivo utilize art-recognized protocols for delivering the agent with the only substantial procedural modification being the substitution of the compounds of the present invention for the drugs in the art-recognized protocols.
  • Likewise, the methods for using the claimed composition for treating cells in culture, for example, to eliminate or reduce the level of bacterial contamination of a cell culture, utilize art-recognized protocols for treating cell cultures with antibacterial agents with the only substantial procedural modification being the substitution of the compounds of the present invention for the agents used in the art-recognized protocols.
  • The compounds of the present invention provide a method for treating microbial infections. As used herein the term “unit dosage” refers to a quantity of a therapeutically-effective amount of a compound of the present invention that elicits a desired therapeutic response. As used herein the phrase “therapeutically-effective amount” means an amount of a compound of the present invention that prevents the onset, alleviates the symptoms, or stops the progression of a bacterial infection. The term “treating” is defined as administering, to a subject, a therapeutically-effective amount of at least one compound of the present invention, both to prevent the occurrence of a bacterial infection, or to control or eliminate a bacterial infection. The term “desired therapeutic response” refers to treating a recipient subject with a compound of the present invention such that a bacterial infection is reversed, arrested or prevented in a recipient subject.
  • The compounds of the present invention can be administered as a single daily dose or in multiple doses per day. The treatment regime may require administration over extended periods of time, e.g., for several days or for from two to four weeks. The amount per administered dose or the total amount administered will depend on such factors as the nature and severity of the infection, the age and general health of the recipient subject, the tolerance of the recipient subject to the compound and the type of the bacterial infection.
  • A compound according to this invention may also be administered in the diet or feed of a patient or animal. The diet for animals can be normal foodstuffs to which the compound can be added or it can be added to a premix.
  • The compounds of the present invention may be taken in combination, together or separately with any known clinically approved antibiotic to treat a recipient subject in need of such treatment.
  • V. METHODS OF INHIBITING BACTERIAL GROWTH AND METHODS OF TREATING BACTERIAL INFECTIONS
  • In one embodiment, the present invention relates to a method of inhibiting bacterial growth. Compounds described herein can possess antbacterial activity. The compounds are effective in inhibiting bacterial infections in vivo. Examples of bacterial organisms that may be controlled by the compositions and methods of this invention include, but are not limited to the following organisms: Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp., Proteus spp., Pseudomonas aeruginosa, Escherichia coli, Serratia marcesens, Staphylococcus aureus, Haemophilus infuenzae, Bacillus anthracis, Mycoplasma pneumoniae, and Coagulase negative Staphylococcus including Staphylococcus epidermidis. In a preferred embodiment, pharmaceutical compositions and compounds of the invention will be useful in controlling or treating infections caused by Gram-positive bacteria. None limiting examples of Gram-positive bacterial growth that may be inhibited by the compounds and pharmaceutical compositions of the invention include Staphylococci and Enterococci. Compositions and methods will be useful for controlling, or reducing the advancement, severity or effects of nosocomial or non-nosocomial infections. Examples of nosocomial uses include, but are not limited to, urinary tract infections, pneumonia, surgical wound infections, bacteremia and therapy for febrile neutropenic patients. Examples of non-nosocomial uses include but are not limited to urinary tract infections, pneumonia, prostatitis, skin and soft tissue infections and intra-abdominal infections.
  • The term “inhibition” when used in conjunction the antibacterial methods refers to a suppression, killing, stasis or destruction of bacterial cells. The antibacterial methods preferably result in prevention, reduction or elimination of invasive activity and related growth of bacterial cells. The term “effective amount” when used in conjunction with antibacterial methods refers to the amount of the compound sufficient to result in the inhibition of growth bacterial cells. The inhibition of bacterial growth according to this method can be monitored in several ways. First bacterial cells grown in vitro can be treated with the compound and monitored for growth or death relative to the same cells cultured in the absence of the compound. A cessation of growth or a slowing of the growth rate, e.g. by 10% of more, is indicative of bacterial cell inhibition. Alternatively, bacterial cell inhibition can be monitored by administering the compound to an animal model having a bacterial infection of interest. Examples of experimental aminal bacterial models are known in the art and described in the examples herein. A convenient means to measure antibacterial activity is by Minimal Inhibitory Concentration (MIC), which is the minimal concentration of an agent necessary to inhibit a standard (control) dose of bacteria. Another convenient means to measure antibacterial activity is Minimal Bactericidal Concentration (MBC), which is the minimal concentration of an agent necessary to kill a standard (control) dose of bacteria.
  • In a related embodiment, the invention provides a method of decreasing bacterial quantity in a biological sample. This method comprises the step of contacting the biological sample with a polyene polyketide of Formula I, a compound as described herein, or a pharmaceutically acceptable derivative or prodrug thereof. This method is effective if the number of bacteria decreases by at least 10%, and preferably more, e.g., 25%, 50%, 75% or even 100% after contacting the biological sample with a polyene polyketide of Formula I, a compound as described herein, or a pharmaceutically acceptable derivative or prodrug thereof.
  • In another embodiment, the present invention relates to a method for treating bacterial infection in a mammalian subject in need thereof, comprising the step of administering to the mammal a therapeutically effective amount of a polyene poketide of Formula I, a compound described herein or a pharmaceutically acceptable derivative or prodrug thereof in combination with a physiologically acceptable carrier. In one embodiment, the compound is represented by Formula I. In another embodiment, the compound is selected from Compounds 1-27.
  • Pharmaceutical compositions effective to treat or prevent a bacterial infection comprise any one of Compounds 1 to 7, a compound of Formula I, or a pharmaceutically acceptable derivative or prodrug thereof, in an amount sufficient to measurably decrease bacterial quantity, and a pharmaceutically acceptable carrier. The term “measurably decrease bacterial quantity”, as used herein means a measurable change in the number of bacteria between a sample containing the inhibitor and a sample not containing the inhibitor.
  • Agents which increase the susceptibility of bacterial organisms to antibiotics are known. For example, U.S. Pat. No. 5,523,288, U.S. Pat. No. 5,783,561 and U.S. Pat. No. 6,140,306 describe methods of using bactericidal/permeability-increasing protein (BPI) for increasing antibiotic susceptibility of gram-positive and gram-negative bacteria. Agents that increase the permeability of the outer membrane of bacterial organisms have been described by Vaara, M. in Microbiological Reviews (1992) pp. 395-411, and the sensitization of gram-negative bacteria has been described by Tsubery, H., et al, in J. Med. Chem. (2000) pp. 3085-3092.
  • For the method of the invention related to treatment of subjects with a bacterial infection, a typical effective unit dose of any one of Compounds 1 to 7, a compound of Formula I as described herein or a pharmaceutically acceptable derivative or prodrug thereof given orally or parenterally would be from about 5 to about 100 mg/kg of body weight of the subject with a daily dose ranging from about 15 to about 300 mg/kg of body weight of the subject.
  • Another preferred embodiment of this invention relates to a method, as described above, of treating a bacterial infection in a mammal in need thereof, but further comprising the step of administering to the mammal an agent which increases the susceptibility of bacterial organisms to antibiotics.
  • According to another preferred embodiment, the invention provides a method, as described above, of decreasing bacterial quantity in a biological sample, but further comprising the step of contacting the biological sample with an agent which increases the susceptibility of bacterial organisms to antibiotics.
  • Methods of decreasing bacterial quantity are effective if the number of bacteria decreases at least 10%, and preferably more, e.g., 25%, 50%, 75% or even 100% after contacting the biological sample with any one of Compounds 1 to 7, a compound of Formula I as described herein, or a pharmaceutically acceptable derivative or prodrug thereof.
  • In addition to the compounds of this invention, pharmaceutically acceptable derivatives or prodrugs of the compounds of this invention may also be employed in compositions to treat or prevent the above-identified disorders. A “pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof. Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species. Pharmaceutically acceptable prodrugs of the compounds of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters.
  • EXAMPLE 1 Production of Compounds 1, 2, 7, 8 and 15 by Fermentation
  • Amycolatopsis orientalis ATCC™ 43491 was cultivated under aerobic conditions in an aqueous nutrient medium containing assimilable sources of carbon, assimilable sources of nitrogen, inorganic salts and vitamins. Preferred carbon sources are glucose, glycerol and the like. Preferred nitrogen sources are beef extract, malt extract, yeast extract, and the like. Representative media are provided in Table 1.
  • Compounds 1, 2, 7, 8 and 15 were produced by the following procedure: Amycolatopsis orientalis ATCC™ 43491 was maintained and sporulated on agar plates of ISP2 medium (Difco™). The innoculum for the production phase was prepared by adding two loopfull of the spores obtained from the surface of the ISP2 agar plate to a 125-ml flask containing 25 ml of ITSB medium (Zahn et al. (2001). Applied and Environmental Microbiology 76, 377-386) composed of 30 g trypticase soy broth (Bacto™), 3 g yeast extract, 2 g MgSO4, 5 g glucose, 4 g maltose made up to one liter with distilled water. The flasks are shaken (250 rpm) for about 60 hours at 28° C. and then 10 ml of the culture is used to, inoculate each 2-L flasks containing ten glass beads and 500 ml of sterile production medium OA consisting of glucose 10 g, glycerol 5 g, corn steep liquor 3 g, beef extract 3 g, malt extract 3 g, yeast extract 3 g, calcium carbonate 2 g, thiamine 0.1 g made up to one liter with distilled water (Kanzaki et al. (1998). Biosci Biotechnol Biochem 62; 438-442). The medium was adjusted at pH 7.0, and then 1 ml of silicon defoamer-oil (Chem Service) was added to each flask before sterilization. The fermentation batches are incubated aerobically under stirring (200 rpm) at 28° C. for a period of 4 days. A fermentation period of 7 days without defoamer-oil was also used to produce Compound 1.
  • Compounds 1, 2, 7, 8 and 15 could also be produced in other media including JA, GA, RM, NA, CA, and CB (Table 1). Compounds 1 and 2 were further produced as described above using a preferred strain, namely Amycolatopsis orientalis IDAC 220604-01.
  • EXAMPLE 2 Purification of Compound 1, 2 and 7
  • Procedure 1: (for 12×500 mL of fermentation)
  • a) The whole fermentation broth at harvest was centrifuged at 3500 rpm for 20 minutes and the supernatant liquid was decanted and discarded. The residual mycelial pellet was treated with methanol (200 mL/L of original fermentation broth volume), stirred and centrifuged. The methanolic supernatant liquid was removed and the mycelial solid was extracted with acetone, the same manner as the methanol extraction. The combined methanol and acetone extracts are evaporated to dryness to a crude residue.
  • b) The crude residue of a) was partitioned between 100 mL(per litre of fermentation) of chloroform (CHCl3) and 100 mL(per litre of fermentation) of methanol (MeOH) in water (3:2) buffered to pH 10 with ammonium hydroxide (NH4OH) and at 10 mM ammonium bicarbonate (NH4HCO3) salt concentration. The two layers were separated and the methanol:water layer evaporated to dryness. The residue from the upper phase was partitioned between n-butanol (100 ml/L of fermentation) and water (100 ml/L of fermentation), buffered as above. The butanol layer was concentrated to a orange-brown residue.
  • The residue was further purified by HPLC (Waters Autopurification System with ACD), using a Waters Xterra MS C18 column (5μ, 19×150 mm), and a gradient of 10 mM aqueous NH4HCO3, pH 10/acetonitrile 85:15 to 25:75 over 30 min at 19 mL/min, UV detector set at 261 nm. The sample was loaded as a suspension in DMSO/MeOH (3:1). The pooling of eluate gave samples of Compound 7 (<1 mg, RT: 16-17 min) and a mixture of Compounds 1 and 2 (1.04 g, non-freezedried, RT: 11.8-12.1 min).
  • Alternatively, the first purification (step 2) was also accomplished using a Phenomenex Max-RP C12 column (4μ, 21.2×250 mm) using the gradient buffer (as above) and acetonitrile 89.5:10.5 to 20:80 over 25 minutes, with the same flow and UV detection. Fractions were collected at a retention time of 16.5-17 minutes (mixture of Compounds 1 and 2) and at RT: 21-22 minutes (Compound 7).
  • The mixture of Compounds 1 and 2 is further purified by HPLC (Waters Autopurification System with ACD), using a RCM Column (Novapak C-18, 6μ, 40×200 mm), and a gradient of 10 mM aqueous ammonium acetate (NH4OAc) to pH 5 with glacial acetic acid/acetonitrile 80:20 to 20:80 over 25 min at 35 mL/min. Fractions were collected and freeze-dried to give pure Compound 1 (RT: 18.5-18.8 minutes, 224.5 mg) and pure Compound 2 (RT: 17.3-17.6 minutes, 34.6 mg).
  • Procedure 2:
  • The crude residue of 1 a) (see Procedure 1) was partitioned between 100 mL(per litre of fermentation) of hexanes and 100 mL(per litre of fermentation) of methanol (MeOH) in water (3:2) buffered to pH 10 with ammonium hydroxide (NH4OH) and at 10 mM ammonium bicarbonate (NH4HCO3) salt concentration. The two layers were separated and the methanol:water layer evaporated to dryness. The procedure was repeated and concentrated to dryness.
  • The crude residue was purified by solid phase extraction. Methanol washed Diaion® HP-20 resin (30 mL) was added to the crude residue. The mixture was added to a column made of 75 mL of methanol washed HP-20 resin and eluted with a mixture of ethanol and pH 10 buffered aqueous ammonium carbonate following the following gradient:
    Fraction Ethanol Aqueous Volume
    1 10 90 500
    2 20 80 200
    3 30 70 200
    4 40 60 200
    5 50 50 200
    6 60 40 200
    7 80 20 200
    8 100 0 200
  • Fractions 4, 5 and 6 were combined and concentrated to give a mixture of Compounds 1 and 2 and Compound 7 was found in the concentrated fraction 7.
  • The mixture of Compounds 1 and 2 was further purified by HPLC (Waters Autopurification System with ACD), using a Symmetry C18 column (5μ, 30×100 mm), and a gradient of 10 mM aqueous NH4OAc, made to pH 5 with glacial acetic acid/acetonitrile 74:26 to 50:50 over 20 min at 39 mL/min. The collection was triggered by UV 261 nm (PDA). The sample was loaded as a suspension in DMSO:MeOH (3:1). The pooling of eluates gave pure Compound 1 (RT: 14.9-15.2 min) and pure Compound 2 (RT: 14.1-14.2 min), generally with a ratio of 5:1. Compound 7 had a retention time of 13-14 minutes using the same conditions.
  • EXAMPLE 3 Structure Identification of Compounds-1, 2 and 7
  • Compounds 1, 2 and 7 were produced by fermentation as described in Example 1 and isolated as described in Example 2.
    Figure US20050215490A1-20050929-C00049
  • Compound 1 is named 3,4,5-trihydroxy-6-[1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid.
  • Compound 1 structure determination was based on mass, UV and NMR data. The NMR data detailed in Table 2 was collected at 500 MHz in d4-MeOH including 1H-NMR spectrum of FIG. 4, and the multidimensional pulse sequences gCOSY, gDQCOSY, gHSQC, and gHMBC. The molecular formula of C46H68N4O10 and the chemical structure were established based the 1H-NMR data, the COSY, HSQC and HMBC measured on about 500 μg of very pure material of structure illustrated above. The carbon assignments shown in Table 2 were made by virtue of the HSQC and HMBC. The straight chain nature of Compound 1 was supported by the fact that the two protons on each of the seven methylene groups were of almost indistinguishable chemical shift.
    TABLE 2
    1H and 13C NMR (δ, ppm) Data for Compound 1 in MeOH-D4
    Assignment δH (ppm) δC (ppm) Group
     1 170.1 C
     2 138.8 C
     3 7.09 135.2 CH
     4 6.60 127.9 CH
     5 6.38 132.5 CH
     6 6.38 135.8 CH
     7 6.56 127.9 CH
     8 6.23 132.5 CH
     9 6.23 131.3 CH
    10 6.08 130.0 CH
    11 5.51 137.9 CH
    12 2.52 40.3 CH
    13 3.58 93.8 CH
    14 134.3 C
    15 5.03 136.4 CH
    16 2.32 32.2 CH
    17 1.13 37.4 CH2
    18 1.29 37.3 CH2
    19 1.87 32.9 CH2
    20 5.24 135.9 CH
    21 5.27 129.3 CH
    22 2.11 37.3 CH
    23 1.97 40.6 CH2
    24 5.41 130.0 CH
    25 5.41 130.0 CH
    26 2.01 29.6 CH2
    27 1.64 27.2 CH2
    28 3.34 48.2 CH2
    29 2.06 12.4 CH3
    30 1.21 17.4 CH3
    31 1.60 11.4 CH3
    32 0.92 20.7 CH3
    33 0.97 20.3 CH 3
     1′ 111.4 C
     2′ 199.3 C
     3′ 2.35 31.0 CH2
     4′ 2.35 31.0 CH 2
     5′ 199.3 C
     1″ 4.20 102.9 CH
     2″ 3.27 74.6 CH
     3″ 3.36 77.6 CH
     4″ 3.41 72.9 CH
     5″ 3.40 76.2 CH
     6″ 176.1 C
     1′′′ 157.7 C
     1′′′′ 3.00 35.8 CH3
  • Mass spectra (FIGS. 1, 2, and 3) analysis shown in Table 3 gave a mass (m/z) of 836.4, which confirmed a molecular formula of C46H68N4O10, and fragments that also confirmed structure assignment of Compound 1 including a sugar moiety.
    TABLE 3
    Mass Spectrometry data for Compound 1
    FIG. Ionization Mass (m/z) Fragment
    1 +mode 837.5 (M + H)+
    (+Q1) 823.7 (M − CH3)+
    2 +mode 837.3 (M + H)+
    (+EPI) 662.1 (M-sugar)+
    644.1 (M-O-sugar)+
    3 mode 835.3 (M − H)
    (−Q1) 821.5 (M − CH3)
  • The UV spectrum for Compound 1 exhibited UV λmax at 258.77 and 362.77 nm in methanol in accordance with the methylpentaene amide of the 2-aminocyclopenta-1,3-dione tautomer.
    Figure US20050215490A1-20050929-C00050
  • Compound 2 is named 6-{16-guanidino-1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethylhexadeca-2,8,12-trienyloxy}-3,4,5-trihydroxy-tetrahydro-pyran-2-carboxylic acid.
  • Structure of Compound 2 was confirmed by 1H (FIG. 5) and 13C NMR. The signals of 1″″ carbon (35.8 ppm) and protons (singlet at 3.00 ppm) of Compound 1 (see Table 5) were absent from the spectra, which confirmed the absence of this CH3 residue. Only small shifts in the guanidine area were observed in the 13C NMR of Compound 2. No further changes from the structure of Compound 1 previously described were present.
    Figure US20050215490A1-20050929-C00051
  • Compound 7 is named 6-[1-(11-carboxy-1-methyldodeca-2,4,5,8,10-pentaenyl)-2,4,10-trimethyl-16-(N-methylguanidino)-hexadeca-2,8,12-trienyloxy]-3,4,5-trihydroxy-tetrahydropyran-2-carboxylic acid.
  • Structure analysis of Compound 7 was accomplished by spectral data analysis including 1H NMR (FIG. 10) and Mass spectra. The later gave a mass of 739.37 at negative ionization (ES−) and 741.53 at positive ionization (ES+), which is consistent with a molecular formula of C41H63N3O9 and a calculated mass of 741.95.
  • Analysis of the 1H NMR (FIG. 10) showed the absence of the signal at δ 2.35 ppm (Table 5) from the NMR spectrum of Compound 1. This signal was previously assigned to the two methylene groups of the cyclopentenone of Compound 1. An effect was also observed at position 3, which doublet (found at about 7.1 ppm in the case of Compound 1) has moved 0.1 ppm (to 7.0 ppm), the rest of the spectra remaining mostly the same.
  • EXAMPLE 4 Preparation and Identification of Compounds 3, 4 and 5
  • Compounds 3, 4 and 5 were prepared according to the following procedure. A 0.1M solution of sodium hydroxide in methanol (334 μL, Fisher Chemicals) was added to Compound 1 (20 mg) in methanol (2 mL). Dimethyl sulfate (5.68 μL, Sigma) was added and the reaction stirred at room temperature for 24 hours. Additional sodium hydroxide in methanol (334 μL) and dimethyl sulfate (10 μL) were added and the reaction stirred for an additional 24 hours. A third portion of sodium hydroxide in methanol (400 μL) and dimethyl sulfate (15 μL) were added and the reaction mixture stirred for an additional 24 hours. The reaction was monitored by TLC (Merck Silica gel 60 F254, eluting with 7% methanol in chloroform) visualized under UV. The reaction was concentrated in vacuo.
  • The crude residue was purified by multiple injection on an HPLC Waters Auto-Purification System using a Symmetry (C-18, 5μ, 30×100 mm) column and the following eluent: A (10 mM ammonium acetate in water (10 mM NH4OAc)) and B acetonitrile (MeCN), 74:26 to 50:50 A:B gradient in 20 minutes, 40 mL/min. The fractions having retention times 9.4, 11.5 and 15.5 minutes were collected to give respectively Compound 4 (0.53 mg), Compound 3 (5.36 mg) and Compound 5 (4.04 mg).
  • Structures of Compounds 3, 4 and 5:
    Figure US20050215490A1-20050929-C00052
  • Compound 3 is named 3,4,5-trihydroxy-6-[1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid methyl ester.
  • Structure determination of Compound 3 was accomplished by spectral data analysis including 1H NMR (FIG. 6) and 13C NMR and mass spectrometry. The molecular ion was found at mass 852.03 (M+H)+ and 849.97 (M−H), respectively for positive and negative ionization, which confirms a calculated molecular weight of 851.08, for C47H70N4O10 as molecular formula. The methyl group (in the squares) was easily assigned to the carboxylic ester from NMR chemical shifts, of the new methyl group (singlet at δ 3.73 ppm integrating for 3 protons) and of the surrounding proton and carbon atoms. The protons most affected were 4″ and 5″ (δ 3.40-3.48 ppm (Table 5) moved to δ 3.50-3.56 ppm), 1″ (doublet δ 4.20 ppm (Table 5) moved to δ 4.26 ppm), 13 (doublet δ 4.58 ppm (Table 5) moved to δ 4.68 ppm) and 31 (δ 1.60 ppm (Table 5) moved to δ 1.52 ppm). Smaller effects were also observed at proton positions 4 to 10 and 32. These last effects might be due to a minor change in conformation and polarity difference between the carboxylic acid of Compound 1 and the ester of Compound 3.
    Figure US20050215490A1-20050929-C00053
  • Compound 4 is named 3,4,5-trihydroxy-6-[1-[11 -(2-methoxy-5-oxo-cyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid.
  • Structure determination of Compound 4 was accomplished by spectral data analysis including 1H NMR (FIG. 7) and 13C NMR and mass spectrometry. The molecular ion was found at mass 852.03 (M+H)+ and 849.98 (M−H), respectively for positive and negative ionization, which confirms a calculated molecular weight of 851.08, for C47H70N4O10 as molecular formula. The 1H NMR spectral analysis confirmed the presence of the methyl group (in the square) as a singlet integrating for three protons at 4.07 ppm. This methyl was also confirmed to be on the cyclopentenone as the two methylene groups were non-equivalent. In fact, in Compound 1, the two methylene groups from the cyclopentenone (positions 3′ and 4′) are equivalent (a 4 proton singlet at 2.35 ppm) due to the symmetry of the tautomeric forms. In Compound 4, the protons of these two positions appear as two separate triplets at 2.84 ppm and 2.52 ppm, integrating for two protons each.
    Figure US20050215490A1-20050929-C00054
  • Compound 5 is named 3,4,5-trihydroxy-6-[1-[11 -(2-methoxy-5-oxo-cyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethyl-16-(N-methyl-guanidino)-hexadeca-2,8,12-trienyloxy]-tetrahydropyran-2-carboxylic acid methyl ester.
  • Structure determination of Compound 5 was accomplished by spectral data analysis including 1H NMR (FIG. 8) and 13C NMR and mass spectrometry. The molecular ion was found at mass 866.06 (M+H)+ and 863.89 (M−H), respectively for positive and negative ionization, which confirms a calculated molecular weight of 865.11, for C48H72N4O10 as molecular formula. The 1H NMR spectral analysis confirmed the presence of two methyl groups (in the square) as singlets integrating for three protons at 3.73 ppm and 4.07 ppm. One of the methyl (4.07 ppm) was confirmed to be on the cyclopentenone as the two methylene groups, as in Compound 4, were non-equivalent and appearing as two separate triplets at 2.84 ppm and 2.52 ppm, integrating for two protons each. The other methyl group (3.73 ppm) was confirmed as the methyl ester of the glucoronic acid (see Compound 3 determination above).
  • EXAMPLE 5 Preparation of Compound 3 by Esterification of Compound 1
  • Figure US20050215490A1-20050929-C00055
  • Diazomethane was generated by standard methods in an Aldrich diazomethane-generator [Z41, 173-6]. Approx. 2 mL 5N sodium hydroxide was added dropwise to 450 mg 1-methyl-3-nitro-1-nitrosoguanidine and the generated gaseous diazomethane was condensed in 4 mL of diethyl ether at 0° C. The diazomethane solution was added dropwise to a solution of Compound 1 (10 mg) in 6 mL of methanol. The reaction was carried out at room temperature with magnetic stirring maintained for 15 minutes until completion. The reaction product was evaporated at 30° C. under a gentle stream of N2. The crude material was purified by multiple injections on HPLC using a YMC ODS-A 10×250 mm column and a gradient of 5 mM NH4OAc/acetonitrile at 5 mL/min (90:10 for 1 min, 90:10-30:70 o. 20 min). The combined yield of pure Compound 3 derived from two separate 10 mg batches was 0.99 mg.
  • EXAMPLE 6 Preparation and Identification of Compound 6
  • Figure US20050215490A1-20050929-C00056
  • Compound 6 is named 6-{16-(N′-acetyl-N-methylguanidino)-1-[11-(2-hydroxy-5-oxocyclopent-1-enylcarbamoyl)-1-methyldodeca-2,4,6,8,10-pentaenyl]-2,4,10-trimethylhexadeca-2,8,12-trienyloxy}-3,4,5-trihydroxytetrahydropyran-2-carboxylic acid.
  • Compound 6 was prepared by acetylation of Compound 1 using the following procedure. Acetic anhydride (3.38 μL, Sigma) was added to a solution of Compound 1 (20 mg) in methanol (2 mL) and the reaction stirred at room temperature for 72 hours. Additional portions of acetic anhydride (20 μL) were added after 24 and 48 hours. The reaction was monitored by TLC (Merck Silica gel 60 F254, eluting with 7% methanol in chloroform) visualized under UV. The reaction was concentrated in vacuo.
  • The crude residue was purified by multiple injection on an HPLC Waters Auto-Purification System using a Symmetry (C-18, 5μ, 30×100 mm) column and the following eluent: A (10 mM ammonium acetate in water (10 mM NH4OAc)) adjusted to pH 5 with glacial acetic acid and B acetonitrile (MeCN), 74:26 to 50:50 A:B gradient in 20 minutes, 40 mL/min. The fractions having retention times 9.4, 11.5 and 15.5 minutes were collected to give Compound 6 (6.43 mg).
  • Structure determination of Compound 6 was accomplished by spectral data analysis including 1H NMR (FIG. 9) and 13C NMR and mass spectrometry. The molecular ion was found at mass 880.03 (M+H)+ and 877.98 (M−H), respectively for positive and negative ionization, which confirms a calculated molecular weight of 879.09, for C48H70N4O11 as molecular formula. Analysis of the 1H NMR spectrum also confirmed the presence of an acetyl group as a singlet at 1.95 ppm integrating for 3 protons. No important changes were observed at other positions, which indicate this acetyl group is on the guanidine.
  • In biological Examples 7, 8 and 9, culture media (ex: MH, BHI and Agar) were supplied by Quelab Laboratories Inc, Montreal, Canada. Indicator strains were supplied by American Type Culture Collection (ATCC™, Manassas, Va., USA).
  • EXAMPLE 7 Anti-microbial Inhibitory Activity of Compounds 1 to 7
  • Antibacterial activity of Compounds 1 and 2 (Table 4) was measured by determining the minimal inhibitory concentration (MIC) necessary to obtain a complete inhibition of bacteria growth in eight indicator strains, namely Staphylococcus aureus (ATCC™ 6538P), Staphylococcus aureus MRS3 (ATCC™ 700699), Staphylococcus epidermidis (ATCC™ 12228), Bacillus subtilis (ATCC™ 23857), Bacillus megaterium (ATCC™ 14581), Enterococcus faecalis VRE-1 (ATCC™ 29212), Enterococcus faecalis VRE-2 (ATCC™ 51299) and Micrococcus luteus (ATCC™ 9341). Indicator strains preparation and MIC determination were performed according to the National Committee for Clinical Laboratory Standards (NCCLS) guideline M7-A5 Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Fifth Edition. (NCCLS document M7-A5, ISBN 1-56238-394-9; NCCLS, 940 West Valley Road, Suite 1400, Wayne, Pa. 19087-1898 USA), the content of which is incorporated herein by reference.
  • Compounds 1 and 2 were prepared as 100× stock solution in DMSO, with concentrations ranging from 3.2 mg/ml to 0.00625 mg/ml (a two-fold dilution series over 12 points). An aliquot of each 100× stock solution was diluted 50-fold in test medium described below to give a set of ten (10) 2× solutions ranging from 64 to 0.125 μg/mL. 50 μl of each of the ten 2× solutions was aliquoted into the corresponding well of a 12-well row, with the final well reserved for medium alone control. Each test was made in duplicate.
  • Vancomycin (Sigma™) used as positive control compounds, was prepared as 2× stock solutions in sterile, double-distilled water (ddH2O) ranging from 64 μg/ml to 0.00125 μg/ml (a two-fold dilution series over 10 points). An aliquot of 50 μl corresponding to each concentration (at 2×) was then transferred to 96-well microplates to obtain a series of eleven two-fold dilutions.
  • An isolated colony of each of the eight indicator strains was used to inoculate tubes containing 3 ml of test medium. Mueller-Hinton test medium was used for Staphylococcus aureus (ATCC™ 6538P), Staphylococcus aureus MRS3 (ATCC™ 700699), Staphylococcus epidermidis (ATCC™ 12228), Bacillus subtilis (ATCC™ 23857), Bacillus megaterium (ATCC™ 14581) and Micrococcus luteus (ATCC™ 9341) indicator strains, and BHI (Brain-Heart Infusion broth) test medium was used for Enterococcus faecalis VRE-1 (ATCC™ 29212) and Enterococcus faecalis VRE-2 (ATCC™ 51299) indicator strains. Cells were grown overnight at 28° C. with shaking. Inoculum density for each indicator strain was adjusted to OD600=0.1 in 5 ml 0.85% saline, then further diluted 1/100 in appropriate medium. 50 μl of the final dilution (in test medium) of each indicator strain was added to each well of a 12-well row. This brings the final dilution of the test article or control compound in solution to 1×. The final inoculum is approximately 5×105 CFU/ml.
  • The indicator strains were incubated with 11 concentrations of each of Compounds 1 and 2, Vancomycin (Sigma™) control compound and one media alone control. For MIC determination, assay plates were incubated at 35° C. for 18 hours. The MIC for each indicator was assessed as the lowest concentration of compound resulting in total absence of growth and is shown below in Table 4.
    TABLE 4
    Antibacterial activity of Compounds 1 and 2, MIC (μg/ml)
    Strain Compound 1 Compound 2 Vancomycin
    S. aureus ATCC ™ 6538P 2 1 2
    S. aureus ATCC ™ 700699 4 1 4
    S. epidermidis 4 1 2
    ATCC ™ 12228
    B. subtilis ATCC ™ 23857 1 2 0.25
    B. megaterium 1 1 0.125
    ATCC ™ 14581
    E. faecalis ATCC ™ 29212 8-16 16  4
    E. faecalis ATCC ™ 51299 16  16  8-16
    M. luteus ATCC ™ 9341 4 1-2 1
  • Antibacterial activitiy of Compounds.3, 4, 5, 6 and 7 are accomplished using the same method in a panel of bacterial strains and Vancomycin as positive control.
  • Antibacterial efficacy of Compounds 1, 2, 3, 4, 5 and 6 on bacterial strain Staphylococcus aureus NRRL B-313 (ATCC™ 6538P) was determined at different pH concentrations. These results are shown in Table 5 together with the antibacterial activity of Compound 7 on the same strain.
    TABLE 5
    Compounds 1 to 7 Antibacterial activity on S. Aureus
    (ATCC ™ 6538P), and effect of pH (MIC (μg/ml))
    1 2 3 4 5 6 7 Vancomycin
    pH 5.0 0.125 0.0625  2 2 2 0.25 ND* 1
    pH 6.0 0.25 0.125 4-8 2 4 0.5 ND* 1
    pH 7.0 1 1 16 2 4 2 32-64 1

    *ND: not determined
  • EXAMPLE 8 Bactericidal Activity of Compounds 1 and 2
  • Minimal bactericidal concentration (MBC) was also determined for Compounds 1 and 2. MBC value was measured as an indicator of cidal activity of the compound, i.e. to show that the compound actually kills bacteria instead of only inhibiting their growth. MBC were measured against three Staphylococcus strains, namely sp. aureus MSSA (ATCC™ 6538P), sp. aureus MRSA-hVISA (ATCC™ 700699) and sp. epidermidis (ATCC™ 12228). The results, shown in Table 6 below, were consistent with bactericidal activity (MBC/MIC ratio were of 1 to 2).
    TABLE 6
    Minimal Bactericidal (MBC) and Inhibitory (MIC)
    Concentrations of Compounds 1 and 2 (μg/ml)
    Compound 1 Compound 2
    Indicator Strains MIC MBC MBC/MIC MIC MBC MBC/MIC
    S. aureus MSSA 2 4 2 1 2 2
    S. aureus MRSA- 4 4 1 1 2 2
    hVISA
    S. epidermidis 4 4 1 1 1 1
  • MBC determination was performed according to the NCCLS guideline M26-A: Methods for Determining Bactericidal Activity of Antimicrobial Agents (NCCLS document M7-A5, ISBN 1-56238-394-9; NCCLS, 940 West Valley Road, Suite 1400, Wayne, Pa. 19087-1898 USA), the content of which is incorporated herein by reference.
  • Microdilutions broth assays were performed using the same procedure as for MIC determination, Example 6. 24 hours post-incubation, cells were collected in wells corresponding to the MIC, 2×MIC and 4×MIC. Collected cells were plated on agar media. MBC were determined as the drug concentration at which more than 99.9% growth inhibition was observed. As a reference for 100% growth, cells grown in media only were also plated.
  • EXAMPLE 9 In vivo Antibacterial Efficacy and Toxicity Profile
  • A. Toxicity: Acute toxicities were determined using CD-1 female mice (supplied by Charles River Laboratories, Wilmington, Mass.) having a bodyweight of 18-25 g, and about 6 weeks of age. Compounds 1 and 2 at doses ranging from 1 to 70 mg/kg (formulated in 20% propylene glycol/water for a total volume of 10 μL/g of weight) were administered intraperitoneal (i.p.) and intravenous (i.v.) to groups of 5 mice (5 mice per concentration, per mode of administration). Two groups of 3 mice were injected with vehicle only. Mice were observed immediately after injection, within 4 hours and once a day for the subsequent days. Acute toxicity studies of Compound 1 in mice indicated no observable effect and high dose tolerability: 70 mg/kg intraperitoneal and 50 mg/kg intravenous (an MTD of 250 mg/kg was also determined by oral administration). Compound 2 showed no observable effect at 25 mg/kg when administered i.v. (in 20% propylene glycol). In vitro Haemolytic activity results are also shown in Table 7, along with the in vivo toxicity and efficacy of Compound 1. No Haemolytic activity was observed for Compounds 1 and 2 at a concentration of 128 μg/ml compared to Amphotericin (EC50: 3 μg/mL).
  • B. Efficacy: In vivo efficacy studies were performed on ICR mice (supplied by Charles River Laboratories, Wilmington, Mass.) having a bodyweight of 20±2 g. Mice (38) were inoculated i.p. with an LD90-100 dose (1×106 CFU/mouse) of Staphylococcus aureus (Smith strain, ATCC™ 13709) suspended in 0.5 mL of BHI containing 5% mucin. Compound 1 at doses 20, 30, 40, 50, 60 and 70 mg/kg (formulated in 20% propylene glycol/water for a total volume of 10 μL/g of weigth) was administered i.p. one hour post infection (5 mice for each concentration). Another group of 5 mice was treated i.v. with a 10 mg/kg dose of Vancomycin. A last group of 3 mice was administered vehicle only. Mortality was recorded for 7 days. In vivo efficacy studies showed 100% protection at the lowest dose tested (20 mg/kg i.p.) against an acute lethal infection with S. aureus.
    TABLE 7
    Tolerability and efficacy of Compound 1
    In vitro Hemolytic activity (EC50)* >128 μg/ml
    In vivo Mouse MTD 70 mg/kg - i.p.
    In vivo efficacy: S. aureus (Smith) ≦20 mg/kg (100%)**

    *Concentration to achieve 50% hemolysis of SRBC, control: Amphotericin EC50: 3 μg/mL

    **Lowest dose tested i.p. gave 100% protection against S. aureus (Smith) (ATCC ™ 13709)
  • EXAMPLE 10 Preparation of Compound 10 by Oxidation of Compound 8
  • Figure US20050215490A1-20050929-C00057
  • To a methylene choride solution of Dess-Martin periodinane (1,1,1-triacetoxy-1,1-dihydro-1,2-benziodoxol-3(1H)one, Sigma-Aldrich Co.) is added a solution of Compound 8 in methylene chloride and the reaction stirred at room temperature for 1 hour. The mixture is diluted with diethyl ether and a saturated aqueous sodium bicarbonate solution containing sodium thiosulfate. Organic layer is separated and washed with saturated aqueous sodium bicarbonate, water and brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue is purified by HPLC according to the procedure described in Example 4. Pure Compound 10 is obtained by pooling and concentrating the appropriate eluate fractions.
  • EXAMPLE 11 Preparation of Compound 11 by Reduction of Compound 1
  • Figure US20050215490A1-20050929-C00058
  • LAH (lithium aluminum hydride) is added to a 0° C. solution of Compound 1 in THF (tetrahydrofuran). After hydrogen gas has stopped evolving, the reaction is allowed to warm to room temperature and stirred overnight. Water is slowly added and 1M hydrochloric solution is used to acidify the solution carefully. The mixture is extracted three times with ethyl acetate. Organic layers are combined and washed with saturated aqueous sodium bicarbonate, water and brine, dried over magnesium sulfate, filtered and concentrated in vacuo. Pure Compound 11 is obtained by pooling and concentrating the appropriate fractions of HPLC purification according to Example 4.
  • EXAMPLE 12 Preparation of Compound 23 from Compound 1
  • Figure US20050215490A1-20050929-C00059
  • Compound 23 is prepared by modification of the guanidino group of Compound 1 according to the procedure described in Argoudelis et al., J. Antibiotics, Vol. XL, No. 6, June 1987, pp 750-760. A mixture of 200 mg of Compound 1 in 1.2 ml water, 1.0 ml of absolute ethanol,1.0 ml of 2,4-pentadione and 120 mg of sodium bicarbonate is stirred at 90° C. for 3 hours. The mixture is allowed to cool to room temperature, concentrated to dryness, dissolved in 5 ml of 2N acetic acid, and purified by HPLC as described in Example 4. Pure Compound 23 is obtained by pooling and concentrating the appropriate eluate fractions.
  • EXAMPLE 13 Preparation of Compound 24 by Alkylation of Compound 1
  • Figure US20050215490A1-20050929-C00060
  • Compound 24 is prepared as follows. See Bartzatt et al., Biotechnol. Appl. Biochem. (2002) 36, 89-93. A Wheaton-type double-chamber device (Wheaton Co., Millville, N.J., U.S.A.) is utilized to generate diazopropane (CH3CH2CHN2). Approx. 5.0 mg of Compound 1 is placed into an organic solvent (ethyl acetate/diethyl ether, 1:1, v/v) and the diazopropane gas formed is allowed to dissolve in the mixture. Diazopropane is generated by mixing 0.15 ml of 5 M NaOH with 0.15 g of 3-nitro-1-nitroso-1-propylguanidine. Excess diazoalkane and the solvent are removed by nitrogen gas flow or under vacuum. The remaining residue is dissolved in methanol, and the methanol solution is purified by HPLC as described in Example 4. Pure Compound 24 is obtained by pooling and concentrating the appropriate eluate fractions.
  • EXAMPLE 14 Preparation of Compound 25 by Acylation of Compound 1
  • Figure US20050215490A1-20050929-C00061
  • Compound 25 is prepared by acylation of Compound 1 as follows. Acetic anhydride (4.5 equivalents) is added dropwise to a solution of 50 mg/ml of Compound 1 and in acetonitrile and pyridine (9:1). The mixture is stirred under reflux and monitored by TLC (see example 5). The solvent is removed under vacuum and the residue is dissolved in methanol. The methanol solution is purified by HPLC as described in Example 4. Pure Compound 25 is obtained by pooling and concentratin the appropriate eluate fractions.
  • EXAMPLE 15 Preparation of Compound 26 by Esterification of Compound 25
  • Figure US20050215490A1-20050929-C00062
  • To a solution of Compound 25 in diethyl ether/ethyl acetate (1/1) is added 1 equivalent of diazomethane in diethyl ether. The reaction mixture is allowed to stand at room temperature overnight. Excess diazomethane and the solvent are removed by nitrogen gas flow or under vacuum. The remaining residue is dissolved in methanol, and the methanol solution is purified by HPLC as described in Example 5. Pure Compound 26 is obtained by pooling and concentrating the appropriate eluate fractions.
  • EXAMPLE 16 Preparation of Compound 27 by Acylation of Compound 1
  • Figure US20050215490A1-20050929-C00063
  • Compound 27 is prepared by acylation of Compound 1 as follows. Acetic anhydride (3.2 equivalents) is added dropwise to a solution of 50 mg/ml of Compound 1 in acetonitrile. The mixture is stirred under reflux and monitored by TLC (see Example 5). The solvent is removed under vacuum and the residue is dissolved in methanol. The methanol solution is purified by HPLC as described in Example 4. Pure Compound 27 is obtained by pooling and concentrating the appropriate eluate fractions.
  • EXAMPLE 17 Preparation of Compound 28 by Esterification of Compound 27
  • Figure US20050215490A1-20050929-C00064
  • To a solution of Compound 27 in diethyl ether is added 1 equivalent of diazomethane in diethyl ether. The reaction mixture is allowed to stand at room temperature overnight. Excess diazomethane and the solvent is removed by nitrogen gas flow or under vacuum. The remaining residue is dissolved in methanol, and the methanol solution is purified by HPLC as described in Example 4. Pure Compound 28 is obtained by pooling and concentrating the appropriate eluate fractions.
  • All patents, patent applications, and published references cited herein are hereby incorporated by reference in their entirety. While this invention has been particularly shown and described with reference to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (18)

1. A method of inhibiting the growth of a bacterial infection, the method comprising contacting a bacterial cell with a therapeutically effective amount of a compound of Formula l:
Figure US20050215490A1-20050929-C00065
such that growth of the bacterial infection is inhibited;
wherein,
A is selected from —C(NH)NHR1, CH3, H or
Figure US20050215490A1-20050929-C00066
R1 is selected from H, C1-6alkyl, C6-10aryl, C(O)C1-6alkyl and C(O)C6-10aryl;
R2 and R3 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl;
R4 is selected from H or CH3;
Z is OH or O when taken with adjacent carbon atom to form a carbonyl; or
Z may be a tetrahydropyranoxy of formula:
Figure US20050215490A1-20050929-C00067
R5 is selected from H, COOH, C1-6 alkyl or C(O)OC1-6 alkyl;
R6, R7 and R8 are each independently selected from H, C1-6 alkyl and C(O)C1-6 alkyl; or
R6, R7 and R8 may each independently be absent when the adjacent oxygen and carbon atoms are taken together to form a carbonyl; or
R6, R7 and R6 may each independently be a bond when any of two neighboring R6, R7 and R8 are taken together with attached oxygen and carbon atoms to form a 1,3-dioxolane ring of formula:
Figure US20050215490A1-20050929-C00068
R9 and R10 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl; or
R9 and R10 are taken together with adjacent carbon atom to form a ring having from 5 to 7 carbons;
D is selected from OH, NH2, NH(C1-3alkyl), N(C1-3alkyl)2, OC1-3alkyl or
Figure US20050215490A1-20050929-C00069
R11 is selected from H or C13 alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
2. The method of claim 1, wherein said compound is selected from Compounds 1, 2, 3, 4, 5, 6, 7 and pharmaceutically acceptable salts or prodrugs thereof:
Figure US20050215490A1-20050929-C00070
Figure US20050215490A1-20050929-C00071
Figure US20050215490A1-20050929-C00072
3. The method of claim 1, wherein A of said compound is selected from —C(NH)NH2, —C(NH)NHC(O)CH3, and
Figure US20050215490A1-20050929-C00073
4. The method of claim 1, wherein R4 of said compound is CH3 or H.
5. The method of claim 1, wherein Z of said compound is selected from —OH,
Figure US20050215490A1-20050929-C00074
wherein R6, R7 and R8 are H;
and R5 is selected from COOH or CO2CH3.
6. The method of claim 1, wherein D of said compound is selected from the group consisting of —OH,
Figure US20050215490A1-20050929-C00075
wherein R11 is H or CH3.
7. A method of inhibiting the growth of a bacterial infection, the method comprising contacting a bacterial cell with a pharmaceutical composition comprising of a therapeutically effective amount of a compound of Formula I:
Figure US20050215490A1-20050929-C00076
wherein,
A is selected from —C(NH)NHR1, CH3, H or
Figure US20050215490A1-20050929-C00077
R1 is selected from H, C1-6alkyl, C6-10aryl, C(O)C1-6alkyl and C(O)C6-10aryl;
R2 and R3 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl;
R4is selected from H or CH3;
Z is OH or O when taken with adjacent carbon atom to form a carbonyl; or
Z may be a tetrahydropyranoxy of formula:
Figure US20050215490A1-20050929-C00078
R5is selected from H, COOH, C1-6 alkyl or C(O)OC1-6 alkyl;
R6, R7 and R8 are each independently selected from H, C1-6 alkyl and C(O)C1-6 alkyl; or
R6, R7 and R8 may each independently be absent when the adjacent oxygen and carbon atoms are taken together to form a carbonyl; or
R6, R7 and R8 may each independently be a bond when any of two neighboring R6, R7 and R8 are taken together with attached oxygen and carbon atoms to form a 1,3-dioxolane ring of formula:
Figure US20050215490A1-20050929-C00079
R9 and R10 are each independently selected from H, C1-6 alkyl, C2-7 alkene and C6-10 aryl; or
R9 and R10 are taken together with adjacent carbon atom to form a ring having from 5 to 7 carbons;
D is selected from OH, NH2, NH(C1-3alkyl), N(C1-3alkyl)2, OC1-3alkyl or
Figure US20050215490A1-20050929-C00080
R11 is selected from H or C1-3 alkyl;
or a pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable carrier, such that growth of the bacterial infection is inhibited.
8. The method of claim 7, wherein said pharmaceutical composition comprises a therapeutically effective amount of a compound selected from Compounds 1-7
Figure US20050215490A1-20050929-C00081
Figure US20050215490A1-20050929-C00082
Figure US20050215490A1-20050929-C00083
or a pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable carrier, such that growth of the bacterial infection is inhibited.
9. A method of treating a bacterial infection in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of a compound of claim 1 or a pharmaceutically acceptable salt or prodrug thereof.
10. The method of claim 9, wherein said compound is selected from the group of Compounds 1, 2, 3, 4, 5, 6, 7 and pharmaceutically acceptable salts or prodrugs thereof.
11. A method of treating a bacterial infection in a mammal, comprising the step of administering to the mammal with a pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 such that growth of the bacterial infection is inhibited;
12. The method of claim 11, wherein said bacterial infection is from a bacteria selected from the group consisting of Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp., Proteus spp., Pseudomonas aeruginosa, E. coli, Serratia marcesens, Staphylococcus aureus, Coagulase negative Staphylococcus, Haemophilus infuenzae, Bacillus anthracis, Mycoplasma pneumoniae, and Staphylococcus epidermidis.
13. The method of claim 11, wherein said the bacterial infection is caused by Gram-positive bacteria.
14. The method of claim 11, wherein said Gram-positive bacteria is selected from Staphylococci and Enterococci species.
15. The method of claim 11, wherein said bacterial infection is from a bacteria selected from the group consisting of Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp., Proteus spp., Pseudomonas aeruginosa, E. coli, Serratia marcesens, Staphylococcus aureus, Coagulase negative Staphylococcus, Haemophilus infuenzae, Bacillus anthracis, Mycoplasma pneumoniae, and Staphylococcus epidermidis.
16. The method of claim 11, wherein said compound is administered orally.
17. The method of claim 11, wherein said compound is administered intravenously.
18. The method of claim 11, wherein said compound is administered intraperitoneally.
US10/975,545 2003-09-11 2004-10-29 Polyene polyketides and methods of treating bacterial infections Abandoned US20050215490A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/975,545 US20050215490A1 (en) 2003-09-11 2004-10-29 Polyene polyketides and methods of treating bacterial infections

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US50182103P 2003-09-11 2003-09-11
US57492204P 2004-05-28 2004-05-28
US58170704P 2004-06-23 2004-06-23
US10/937,379 US7300921B2 (en) 2003-09-11 2004-09-10 Polyene polyketides and methods of production
US10/975,545 US20050215490A1 (en) 2003-09-11 2004-10-29 Polyene polyketides and methods of treating bacterial infections

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/937,379 Continuation US7300921B2 (en) 2003-09-11 2004-09-10 Polyene polyketides and methods of production

Publications (1)

Publication Number Publication Date
US20050215490A1 true US20050215490A1 (en) 2005-09-29

Family

ID=34198998

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/937,379 Expired - Fee Related US7300921B2 (en) 2003-09-11 2004-09-10 Polyene polyketides and methods of production
US10/975,545 Abandoned US20050215490A1 (en) 2003-09-11 2004-10-29 Polyene polyketides and methods of treating bacterial infections

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/937,379 Expired - Fee Related US7300921B2 (en) 2003-09-11 2004-09-10 Polyene polyketides and methods of production

Country Status (4)

Country Link
US (2) US7300921B2 (en)
EP (1) EP1539962A4 (en)
CA (1) CA2484638C (en)
WO (1) WO2005024025A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102004030371A1 (en) * 2004-06-23 2006-01-19 Dynamit Nobel Gmbh Process for the preparation of gaseous diazoalkanes
GB0805363D0 (en) 2008-03-20 2008-04-30 Sinvent As Novel NRPS-PKS gene cluster and its manipulation and utility
US8865761B1 (en) 2012-08-07 2014-10-21 The University Of Notre Dame Du Lac Regulation of cholesterol homeostasis
US20230106760A1 (en) * 2020-03-17 2023-04-06 The United States Of America,As Represented By The Secretary,Department Of Health And Human Services Antibacterial and antifungal polyketides from environmental amycolatopsis spp
JP2023524238A (en) * 2020-04-30 2023-06-09 アーチ オンコロジー,インコーポレイテッド Therapeutic SIRPα antibodies
CN114317580A (en) * 2022-01-13 2022-04-12 上海交通大学 Specific gene knockout CRISPR/Cas9 editing plasmid containing double sgRNAs and application thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3854480A (en) 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US4452775A (en) 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
GB8700513D0 (en) * 1987-01-09 1987-02-11 Pfizer Ltd Antibiotics produced by fermentation
US5039660A (en) 1988-03-02 1991-08-13 Endocon, Inc. Partially fused peptide pellet
FI903460A0 (en) * 1989-07-10 1990-07-09 Bristol Myers Squibb Co ANTIVIRAL ANTIBIOTICS BU-3889V.
CA2172245C (en) 1993-09-22 2003-04-08 Jonathan Cohen Method of treating gram-negative bacterial infection by administration of bactericidal/permeability-increasing (bpi) protein
EP0754050B1 (en) 1994-01-14 2002-06-26 XOMA Technology Ltd. Anti gram positive bacterial methods and materials
US5917084A (en) * 1996-07-03 1999-06-29 Millennium Pharmaceuticals, Inc. Antifungal agents
US6670168B1 (en) 1999-10-29 2003-12-30 Kosan Bioscience, Inc. Recombinant Streptomyces hygroscopicus host cells that produce 17-desmethylrapamycin
US6953679B2 (en) * 2000-09-19 2005-10-11 Bristol-Myers Squibb Company Method for the preparation of fused heterocyclic succinimide compounds and analogs thereof
CA2352451C (en) 2001-07-24 2003-04-08 Ecopia Biosciences Inc. High throughput method for discovery of gene clusters
EP1470241A2 (en) 2002-01-24 2004-10-27 Ecopia Biosciences Inc. Method, system and knowledge repository for identifying a secondary metabolite from a microorganism
EP1585752B1 (en) * 2003-01-21 2007-11-21 Thallion Pharmaceuticals Inc. Polyene polyketides, processes for their production and their use as a pharmaceutical

Also Published As

Publication number Publication date
US20050176653A1 (en) 2005-08-11
EP1539962A4 (en) 2006-12-27
US7300921B2 (en) 2007-11-27
CA2484638A1 (en) 2005-02-14
WO2005024025A1 (en) 2005-03-17
WO2005024025A8 (en) 2005-05-19
EP1539962A1 (en) 2005-06-15
CA2484638C (en) 2008-03-25

Similar Documents

Publication Publication Date Title
US4946941A (en) Novel glycopeptide antibiotics
SK68594A3 (en) Lipopeptides made by actinoplanes bacteries, method of their preparing and using
US20140348919A1 (en) Antibiotic compounds
US20050215490A1 (en) Polyene polyketides and methods of treating bacterial infections
EP2097423B1 (en) Aminothiazole macrocycles, their use as antibacterial compounds and processes for their production
US7271147B2 (en) Antibiotics, tripropeptins and process for producing the same
US20090069396A1 (en) Polyene Oxazoles and Processes for Their Preparation
US6387943B1 (en) Vancoresmycin, a process for its production and its use as a pharmaceutical
US20080096856A1 (en) Antibiotic Compound
US20040266008A1 (en) Polyene polyketides, processes for their production and their use as pharmaceuticals
JP3830964B2 (en) A novel thiodepsipeptide isolated from marine actinomycetes
US20060106028A1 (en) Polycyclic aromatics and derivatives thereof and processes for their preparation
DE102004010219B4 (en) HKI10311129, new antibiotic, process for its preparation and pharmaceutical compositions containing HKI10311129
JPH0558440B2 (en)
US20100144658A1 (en) Novel antibiotics, bispolides a1, a2, and a3 as well as bispolides b1, b2a, b2b and b3 and processes for producing said antibiotics
WO2011136174A1 (en) Depsipeptide compound

Legal Events

Date Code Title Description
AS Assignment

Owner name: ECOPIA BIOSCIENCES INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FARNET, CHRIS M.;ZAZOPOULOS, EMMANUEL;ALARCO, ANNE-MARIE;REEL/FRAME:016315/0950

Effective date: 20050112

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION