WO2011152720A1 - Pantothenic acid derivatives and their use in the treatment of microbial infections - Google Patents

Pantothenic acid derivatives and their use in the treatment of microbial infections Download PDF

Info

Publication number
WO2011152720A1
WO2011152720A1 PCT/NL2011/050384 NL2011050384W WO2011152720A1 WO 2011152720 A1 WO2011152720 A1 WO 2011152720A1 NL 2011050384 W NL2011050384 W NL 2011050384W WO 2011152720 A1 WO2011152720 A1 WO 2011152720A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
alkynyl
alkenyl
cycloalkenyl
cycloalkyl
Prior art date
Application number
PCT/NL2011/050384
Other languages
French (fr)
Inventor
Patrick Laurentius Johannes Maria Zeeuwen
Patrick Antonius Martinus Jansen
Josephus Schalkwijk
Floris Petrus Johannes Theodorus Rutjes
Bas Ritzen
Pedro Harold Han Hermkens
Original Assignee
Umc St. Radboud
Stichting Katholieke Universiteit, Radboud University Nijmegen
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 Umc St. Radboud, Stichting Katholieke Universiteit, Radboud University Nijmegen filed Critical Umc St. Radboud
Publication of WO2011152720A1 publication Critical patent/WO2011152720A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/164Amides, e.g. hydroxamic acids of a carboxylic acid with an aminoalcohol, e.g. ceramides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/417Imidazole-alkylamines, e.g. histamine, phentolamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/4174Arylalkylimidazoles, e.g. oxymetazolin, naphazoline, miconazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • 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
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention concerns substances and compositions having antibiotic activity as well as their use in the therapeutic and/or prophylactic treatment of humans and animals. More in particular the present invention provides novel amides derived from pantothenic acid having antibiotic activity, and novel analogues of pantetheine inhibiting the activity of pantetheinase are provided. In addition, combinations of such compounds, pharmaceutical or veterinary compositions containing them, and uses thereof as medicinal products are provided.
  • CoA Coenzyme A
  • vitamin B5 pantothenic acid
  • CoaA pantothenate kinase
  • pantothenic acid As of the early 70 ' s, amides derived from pantothenic acid (vitamin B5), have been reported to possess antibiotic activity in vitro (Clifton et al. 1 ) During the last decades many of these pantothenamides have been synthesized (Choudry et al. 2 , Mercer et al. 3 , Virga et al. 4 ) and the putative modes of action have been studied in detail (Vigra et al., Zhang et al. 5 ). Pantothenamides, of which N5Pan and N7Pan are the typical prototypes, are active against gram negative (e.g. E.coli) and gram positive (e.g. S. aureus) bacteria in vitro, and are believed to interfere with bacterial lipid synthesis through their action as CoA antimetabolites. Hitherto, no experimental results of antimicrobial action of these compounds in animals or humans (in vivo) have ever been published or presented.
  • pantothenamides are not at all antibiotically active in the presence of plasma or serum.
  • N5Pan is active as an antibiotic in 1% tryptone medium but addition of 10% plasma or serum completely abolishes antibiotic activity.
  • the present inventors surprisingly found that inhibition of pantetheinase activity prevented breakdown of pantothenamides, preserving their antibiotic activity under physiological conditions in mammals.
  • an analogue of pantetheine was provided in an attempt to obtain an inhibitor of pantetheinase activity, which fully confirmed the concept.
  • This compound was found to inhibit the action of plasma panthetheinase. It was found that, in the presence of plasma or serum, the growth inhibition of E.coli by N5Pan was restored in the presence of said inhibitor of pantetheinase activity.
  • an antimicrobial pantothenamide with an inhibitor of host pantetheinase activity provides a novel antibiotic strategy.
  • pantothenate derivatives e.g. pantothenamides
  • pantothenamides pantetheine analogues to improve antibiotic activity
  • Clifton et al 1 antibiotic pantothenamides were combined with pantothenate showing that pantothenate decreased or abolished the antibiotic activity of pantothenamides. This was taken to indicate that pantothenamides acted as pantothenic acid antimetabolites.
  • pantothenamides described in Clifton et al. nor the pantothenate has pantetheinase inhibitory activity.
  • Inhibitors of vanin-1 have been described for application in intestinal inflammation (see US 2004/247524).
  • a recent publication has identified pantetheinase inhibitors derived from various chemical scaffolds in the LOP AC library, via a high-throughput approach (Ruan et al. 10 ).
  • pantothenamide antibiotics are degraded in vivo by pantetheinases and that pantetheinase inhibitors can be used to overcome this problem have never been suggested before.
  • the present invention thus, for the first time, makes available compounds and combinations of compounds for use in therapeutic and or prophylactic treatment of bacterial infections, fungal and yeast infections in a human or animal subject in need thereof, relying on interference with microbial CoA dependent pathways and, simultaneously, inhibiting or reducing host-derived pantetheinase activity.
  • a pharmaceutical or veterinary composition comprising:
  • pantothenamide derivatives having antibiotic activity are provided as such.
  • compositions and substances for the therapeutic and/or prophylactic treatment of a human or animal subject in need thereof is provided.
  • antibiotic pantothenamide derivative refers to amide derivatives of pantothenic acid and substances structurally related to pantothenic acid, which have in vitro and/or in vivo antibiotic activity.
  • antibiotic means effective in reducing the viability or decreasing or inhibiting the growth or reproduction of a bacterium, yeast, fungi, mold, or other microrganism, in particular of a bacterium.
  • Inhibiting or reducing the growth or reproduction means increasing the generation cycle time by at least 2-fold, preferably at least 10-fold, more preferably at least 100-fold, and most preferably indefinitely, as in total cell death.
  • Terms such as antimicrobial, bacteriostatic and bactericidal are deemed synonymous to antibiotic and may be used interchangeably in the context of the present invention.
  • the antibiotic pantothenamide derivative of the invention is selected from the group of derivatives represented by formula (I) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
  • R 1 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide;
  • R 2 and R 3 independently represent a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
  • X 1 and X 2 independently represent hydrogen or a group selected from hydroxyl, thiol, cyanide, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, sulfonamide, amide, pyrazole or imidazole; and
  • X 3 represents a hydrogen atom or a group selected from hydroxyl, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide, preferably a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide.
  • the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (la) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof: wherein R 1 and X 3 have the same meaning as defined above in relation to formula (I).
  • the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (lb) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
  • R 1 represents a group selected from alkyl, alkenyl and alkynyl.
  • antibiotic pantothenamide derivative is selected from 'N5Pan', 'N7Pan' and 'N9Pan' and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof, most preferably from N5Pan and N7Pan and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof, wherein N5Pan, N7Pan and N9Pan are represented by the following formulas:
  • the term ' pantetheinase inhibitor ' typically refers to any substance somehow capable of i) reducing or inhibiting the activity of pantetheinase in viro and/or in vivo; ii) reducing or inhibiting the production of pantetheinase in vitro and/or in vivo; or iii) stimulating the inactivation or break-down of pantetheinase in vitro and/or in vivo.
  • Proteins in mammals that possess pantetheinase activity belong to the vanin (VNN) gene family (Martin et al).
  • Vanins are broadly expressed pantetheinases involved in the CoA pathway, allowing the turnover of pantetheine, thereby recycling pantothenic acid and producing cysteamine.
  • VNN1, VNN2 and VNN3 three vanin genes that are broadly expressed, as was shown before (Jansen et al). In mice only VNN1 and VNN3 are present. Both human and mouse plasma contain high levels of pantetheinase activity.
  • the pantetheinase inhibitor is selected from the group consisting of competitive enzyme inhibitors, reactive enzyme substrates capable of covalently modifying a residue of the enzyme catalytic site, antibodies capable of specifically interacting with VNN, preferably VNN1, and inhibiting its activity, and nucleic acids blocking transcription of VNN gene, preferably VNN1 gene, and/or translation of VNN mRNA, preferably VNN 1 mRNA.
  • the pantetheinase inhibitor is selected from the group consisting of VNN antagonistic agents, including VNN-1 antagonistic agents, VNN-2 antagonistic agents and VNN-3 antagonistic agents.
  • VNN antagonistic agents including VNN-1 antagonistic agents, VNN-2 antagonistic agents and VNN-3 antagonistic agents.
  • the term 'antagonistic agent' means a compound that by any means, partly or completely blocks the dissimilative pathway of CoA referred to above (allowing the turnover of pantetheine, thereby recycling pantothenic acid and producing cysteamine).
  • the pantetheinase inhibitor is selected from the group of non-hydrolysable pantothenic acid analogues and reactive enzyme substrates capable of covalently or non-covalently binding a residue of the enzyme catalytic site.
  • the pantetheinase inhibitor is selected from the group of substances represented by formula II, III or IV, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
  • R 4 , R 5 and R 6 independently represent a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
  • R 7 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
  • X 4 and X 5 independently represent hydrogen hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
  • X 6 represents sulfur, oxygen, carbon or nitrogen, said nitrogen optionally being substituted with a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, preferably sulfur, oxygen or nitrogen, said nitrogen optionally being substituted with a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
  • X 7 represents oxygen or cyanide, where the dotted line indicates the optional presence of an additional covalent bond; and m and n are integers within the range of 0-6.
  • m is not 0.
  • X 7 represents oxygen
  • R 7 does not represent hydrogen
  • the pantetheinase inhibitor is selected from substances represented by formula Ila or lib, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
  • R 4 , R 5 and R 6 independently represent hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
  • R 7 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
  • X 4 and X 5 independently represent hydrogen, hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
  • n are integers within the range of 0-6.
  • m is not 0.
  • R 7 does not represent hydrogen
  • the pantetheinase inhibitor is selected from substances represented by formula lie, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof: wherein
  • R 7 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
  • heteroarylalkyl each optionally substituted by hydroxyl, thiol, halogen and/or cyanide;
  • X 4 and X 5 independently represent hydrogen, hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
  • n are integers within the range of 0-6.
  • m is not 0.
  • R 7 does not represent hydrogen
  • the pantetheinase inhibitor is selected from substances represented by formula lie, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof, as defined above, wherein X 4 and X 5 represent hydroxyl and n and m are 1.
  • the pantetheinase inhibitor is selected from the compounds designated 'RR2', 'RR6', 'RR7' and 'RR8', more preferably the compounds designated RR2 and RR6, most preferably the compound RR6, and pharmaceutically acceptable salts, esters, amides, or prodrugs thereof:
  • the pantetheinase inhibitor is selected from the substances represented by formula Ilia or Illb, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
  • R 4 , R 5 and R 6 independently represent a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
  • R 7 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
  • X 4 and X 5 independently represent hydrogen, hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
  • X 6 represents sulfur, oxygen, carbon or nitrogen, said nitrogen optionally being substituted by a radical selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; preferably sulfur, oxygen or nitrogen, said nitrogen optionally being substituted by a radical selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; and
  • n are integers within the range of 0-6.
  • m is not 0.
  • R 7 does not represent hydrogen
  • the pantetheinase inhibitor is selected from the substances represented by formula Ila, Ilia and IV as defined herein before, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof.
  • a particularly preferred embodiment of the invention concerns a pharmaceutical or veterinary composition as defined herein before, comprising a combination of a pantothenamide derivative selected from N5Pan, N7Pan and N9Pan and a pantetheinase inhibitor selected from RR2, RR6, RR7 and RR8, such as a combination of N5Pan and RR2; a combination of N7Pan and RR2; a combination of N5Pan, N7Pan and RR2; a combination of N5Pan and RR6; a combination of N7Pan and RR6; or a combination of N5Pan, N7Pan and RR6; or pharmaceutically acceptable salts, esters, amides, or prodrugs of said compounds.
  • a pantothenamide derivative selected from N5Pan, N7Pan and N9Pan
  • a pantetheinase inhibitor selected from RR2, RR6, RR7 and RR8, such as a combination of N5
  • alkyl either alone or within other terms, means an acyclic alkyl radical, preferably containing from 1 to 10, more preferably from 1 to about 8 carbon atoms and most preferably 1 to about 6 carbon atoms. Said alkyl radicals may be optionally substituted as defined elsewhere in this document.
  • radicals include methyl, ethyl, chloroethyl, hydroxyethyl, n-propyl, oxopropyl, isopropyl, n-butyl, cyanobutyl, isobutyl, sec-butyl, tert-butyl, pentyl, aminopentyl, iso-amyl, hexyl, octyl and the like.
  • alkenyl refers to an unsaturated, acyclic hydrocarbon radical in so much as it contains at least one double bond.
  • alkenyl radicals typically contain from 2 to 10 carbon atoms, preferably from 2 to 8 carbon atoms and most preferably 2 to about 6 carbon atoms.
  • Said alkenyl radicals may be optionally substituted as defined elsewhere in this document.
  • suitable alkenyl radicals include ethenyl, 1-propenyl, 2-propenyl, 2- methyl-l-propenyl, 1-butenyl, 2-butenyl and the like.
  • alkynyl refers to an unsaturated, acyclic hydrocarbon radical in so much as it contains one or more triple bonds, such radicals typically containing from 2 to 10 carbon atoms, preferably having from 2 to 8 carbon atoms and most preferably from 2 to 6 carbon atoms. Said alkynyl radicals may be optionally substituted with groups as elsewhere in this document.
  • alkynyl radicals examples include ethynyl, propynyl, hydroxypropynyl, butyne-l-yl, butyn-2-yl, pentyne-l-yl, pentyne-2-yl, 4 methoxypentyn-2-yl, 3-methylbutyn-l- yl, hexyne-l-yl, hexyne-2-yl, hexyne-3-yl, 3,3-dimethylbutyn-l-yl radicals and the like.
  • cycloalkyl refers to carbocyclic radicals typically having 3 to 10 carbon atoms, preferably 3 to 8 carbon atoms, most preferably 5 to 8 carbon atoms. Said cycloalkyl radicals may be optionally substituted as defined elsewhere in this document. Examples of suitable cycloalkyl radicals include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • cycloalkenyl embraces carbocyclic radicals having 3 to 10 carbon atoms and one or more carbon-carbon double bonds.
  • Preferred cycloalkenyl radicals are "lower cycloalkenyl” radicals having 3-8 carbon atoms, more preferably 5-8. Examples include radicals such as cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptenyl.
  • aryl alone or in combination, means a 5-10 membered carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendant manner or may be fused.
  • fused means that a second ring is present having two adjacent atoms in common with the first ring.
  • fused is equivalent to the term “condensed”.
  • aryl embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane and biphenyl.
  • heteroaryl (on its own or in any combination, such as “heteroaryloxy”, or “heteroaryl alkyl”) is used herein to mean a 5-10 membered aromatic ring system containing one, two or three rings, which may be attached in a pendant manner or may be fused, wherein at least one of said rings contains one or more heteroatoms selected from the group consisting of N, O or S.
  • Examples include, but are not limited to, pyrrole, pyrazole, furan, thiophene, quinoline, isoquinoline, quinazolinyl, pyridine, pyrimidine, oxazole, thiazole, thiadiazole, tetrazole, triazole, imidazole, or benzimidazole.
  • cycloalkylalkyl embrace, respectively, the afore-defined cycloalkyl, cycloalkenyl, aryl and heteroaryl radicals attached to the main molecular moiety, i.e. the basic moiety depicted in the formulae, through an alkyl radical, typically an alkyl radical having 1-10, preferably 1-8, most preferably 1-6 carbon atoms, as will be understood by those skilled in the art.
  • Representative examples of arylalkyl include, but not limited to, phenylmethyl, phenylethyl and naphthylmethyl.
  • Representative examples of heteroarylalkyl groups include, but are not limited to, thiazolylmethyl, thienylmethyl, furylmethyl, imidazolylmethyl and pyridylmethyl.
  • sulfonamide includes moieties which contain a group of the formula - SO 2 NRR, where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • amide includes moieties which contain a nitrogen atom which is bound to the carbon of a carbonyl or a thiocarbonyl group.
  • alkaminocarboxy groups which include alkyl, alkenyl, or alkynyl groups bound to an amino group bound to a carboxy group. It includes arylaminocarboxy groups which include aryl or heteroaryl moieties bound to an amino group which is bound to the carbon of a carbonyl or thiocarbonyl group.
  • alkylamino carboxy include moieties wherein alkyl, alkenylaminocarboxy,” alkynylaminocarboxy, and arylaminocarboxy, respectively, are bound to a nitrogen atom which is in turn bound to the carbon of a carbonyl group.
  • the compounds of the invention can contain one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers, or diastereomers.
  • stereoisomers such as double-bond isomers (i.e., geometric isomers), enantiomers, or diastereomers.
  • the chemical structures depicted herein, and therefore the compounds of the invention encompass all of the corresponding compound's enantiomers and stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
  • a compound of the invention is considered optically active or enantiomerically pure (i.e., substantially the R-form or substantially the S-form) with respect to a chiral center when the compound is about 90% enantiomeric excess (ee) or greater, preferably, equal to or greater than 95% enantiomeric excess with respect to a particular chiral center.
  • a compound of the invention is considered to be in enantiomerically-enriched form when the compound has an enantiomeric excess of greater than about 1% ee, preferably greater than about 5% ee, more preferably, greater than about 10% ee with respect to a particular chiral center.
  • a compound of the invention is considered diastereomerically pure with respect to multiple chiral centers when the compound is about 90%> de (diastereomeric excess) or greater, preferably, equal to or greater than 95% de with respect to a particular chiral center.
  • a compound of the invention is considered to be in diastereomerically-enriched form when the compound has an diastereomeric excess of greater than about 1% de, preferably greater than about 5% de, more preferably, greater than about 10% de with respect to a particular chiral center.
  • a racemic mixture means about 50% of one enantiomer and about 50% of is corresponding enantiomer relative to all chiral centers in the molecule.
  • the invention encompasses all enantiomerically-pure, enantiomerically-enriched, diastereomerically pure, diastereomerically enriched, and racemic mixtures of compounds of Formulas I through III.
  • Enantiomeric and diastereomeric mixtures can be resolved into their component enantiomers or stereoisomers by well known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Enantiomers and diastereomers can also be obtained from diastereomerically- or enantiomerically-pure intermediates, reagents, and catalysts by well known asymmetric synthetic methods.
  • the pantothenic acid and/or pantothenamide derivatives or analogues are 'enantiomerically pure' (i.e. according to the above definitions) derivatives or analogues of D(+) pantothenic acid and/or D(+) pantothenamide, i.e. 'enantiomerically pure' substances possessing the same stereochemical arrangement as the corresponding stereocenter in D-(+) pantothenic acid and/or D(+) pantothenamide.
  • pharmaceutically acceptable salts, esters, amides, and prodrugs refers to salts, amino acid addition salts, esters, amides, and prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • salts refers to inorganic and organic acid or base addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. Depending on the particular substituents found on the compounds described herein. When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al, “Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • esters examples include compounds of the invention which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • suitable esters include Ci-C 6 alkyl esters and C5-C7 cycloalkyl esters.
  • Esters of the compounds of the invention can be prepared according to conventional methods. Pharmaceutically acceptable esters can be obtained through reaction of hydroxy groups of the compound with an organic acid, such as acetic acid or benzoic acid. In the case of compounds containing carboxylic acid groups, the pharmaceutically acceptable esters are prepared by reaction of said carboxylic acid group, as will be understood by those skilled in the art.
  • Examples of pharmaceutically acceptable, non-toxic amides of the compounds of this invention include amides derived from ammonia, primary Ci-C 6 alkyl amines and secondary Ci-C 6 dialkyl amines wherein the alkyl groups are straight or branched chain. In the case of secondary amines, the amine may also be in the form of a 5- or 6-membered heterocycle containing one nitrogen atom. Amides derived from ammonia. Amides of the compounds of the invention may be prepared according to conventional methods.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compounds of the above formula, for example, by hydrolysis in blood.
  • a thorough discussion is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • the compounds of the invention can be provided as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • Another aspect of the present invention concerns the use of the compounds of the invention, the combinations thereof or the pharmaceutical compositions containing them, as defined herein before, in a therapeutic or prophylactic method of treatment, in particular in treating or preventing a disease or condition selected from the group of bacterial infections, fungal and yeast infections and intracellular protozoan infections in a human or animal subject in need thereof.
  • a method of treating and/or preventing a disease or condition selected from the group of bacterial infections, fungal and yeast infections and intracellular protozoan infections in a human or animal subject in need thereof comprising administering to said human or animal subject an effective amount of a compound of the invention, a combination thereof or a pharmaceutical composition containing it, as defined herein before.
  • these uses and methods concern treatment of human subjects in need thereof.
  • the total daily dose of antibiotic pantothenamide derivative of the invention is typically within the range 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg depending, of course, on the mode of administration and/or the severity of the disease or condition.
  • an intravenous daily dose may only require from 0.001 mg to 40 mg.
  • the total daily dose of pantetheinase inhibitor of the invention is typically within the range 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg depending, also, on the mode of administration and/or the severity of the condition to be treated.
  • the total daily dosage(s) may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein. These dosages, furthermore, are based on an average human subject having a weight of about 65kg to 70kg. The physician will readily be able to determine appropriate doses for subjects whose bodily weight differs significantly from said average, for example for children, as will be understood by those skilled in the art.
  • the above uses and methods concern the treatment of animals in need thereof.
  • the methods and uses concern the treatment of domestic animals and/or farm animals, such as dogs, cats, horses, pigs, cattle, sheep, goats, poultry, etc.
  • a veterinarian will be able to determine appropriate doses for these different kinds of animals depending, of course, on their average body weight, the route of administration and/or the severity of the disease or condition.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
  • Parenteral administration may involve administration directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • the compounds of the invention may furthermore be administered topically or locally to the skin or mucosa. It is preferred that the compounds of the invention, the combinations thereof or the pharmaceutical or veterinary compositions containing them are administered orally or parenterally, preferably orally or intravenously.
  • the compounds of the invention, the combinations thereof or the pharmaceutical or veterinary compositions containing them are administered repeatedly, typically at least once weekly, more preferably at least once every three days, e.g. once daily. Typically, treatment lasts at least a week, more preferably at least two weeks, more preferably at least three weeks.
  • repeated and continued administration of antibiotics typically reduces the risks of development of resistance towards the antibiotic and, without wishing to be bound by any particular theory, it is hypothesized that this issue might apply to compounds and combinations of the present invention.
  • Compounds of the invention or combinations thereof may be administered to a human or animal subject either alone or as part of a pharmaceutical or veterinary composition
  • excipient is used herein to describe any ingredient other than the compound(s) of the invention.
  • excipient will, to a large extent, depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • a pharmaceutical composition for use in humans comprising one or more antibiotic pantothenamide derivatives of the invention, typically in an amount within the range of 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg and/or one or more pantetheinase inhibitors of the invention, typically in a total amount within the range of 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg; as well as one or more pharmaceutically acceptable excipients.
  • a veterinary composition for use in animals comprising one or more antibiotic pantothenamide derivatives of the invention, typically in an amount within the range of 0.0001 mg to 50000 mg, preferably 0.001 mg to 5000 mg, most preferably 0.005 mg to 2500 mg and/or one or more pantetheinase inhibitors of the invention, typically in a total amount within the range of 0.0001 mg to 50000 mg, preferably 0.001 mg to 5000 mg, most preferably 0.005 mg to 2500 mg; as well as one or more veterinary acceptable excipients.
  • compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in 'Remington's Pharmaceutical Sciences', 19th Edition (Mack Publishing Company, 1995).
  • Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including liquid- filled), chews, multi- and nano- particulates, gels, solid solution, liposome, films, ovules, sprays and liquid formulations.
  • Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet. The compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, 1 1 (6), 981- 986, by Liang and Chen (2001 ).
  • the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form.
  • tablets generally contain a disintegrant.
  • disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl -substituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate.
  • the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form.
  • Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
  • lactose monohydrate, spray-dried monohydrate, anhydrous and the like
  • mannitol xylitol
  • dextrose sucrose
  • sorbitol microcrystalline cellulose
  • starch dibasic calcium phosphate dihydrate
  • Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc.
  • surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
  • Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate.
  • Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet.
  • Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant. Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting.
  • the final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated.
  • tablets may be coated with shellac, sugar or both.
  • a syrup or elixir may contain the active compounds sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor.
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compounds may be incorporated into sustained-release preparation and formulations. The formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets, Vol. 1 , by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
  • Suitable modified release formulations for the purposes of the invention are, such as high energy dispersions and osmotic and coated particles, are to be found in Pharmaceutical Technology On-line, 25(2), 1- 14, by Verma et al (2001 ). The use of chewing gum to achieve controlled release is described in WO 00/35298.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • a coating such as lecithin
  • surfactants for example, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
  • a suitable vehicle such as sterile, pyrogen-free water.
  • the preparation of parenteral formulations under sterile conditions for example, by lyophilisation, may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.
  • the solubility of compounds used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility- enhancing agents.
  • Formulations for parenteral administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound.
  • examples of such formulations include drug-coated stents and poly(c//-lactic-coglycolic)acid (PGLA) microspheres.
  • Typical formulations for topical or local administration include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958 by Finnin and Morgan (October 1999). Formulations for topical or local administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1 , 1 , 1 ,2-tetrafluoroethane or 1,1 ,1 ,2,3,3,3-heptafluoropropane.
  • a suitable propellant such as 1 , 1 , 1 ,2-tetrafluoroethane or 1,1 ,1 ,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • the compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • Formulations for rectal/vaginal administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • the compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene glycol- containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration.
  • Drug-cyclodextrin complexes for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion complexes may be used.
  • the cyclodextrin may be used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser.
  • the present invention provides compositions comprising more than one antibiotic pantothenamide derivatives.
  • the compounds and combinations defined herein before can also be used in combination with one or more additional antibiotic agents from a different class. Such combinations may result in an antibiotic product with an increased spectrum of action and restored efficacy against resistant bacteria.
  • Suitable examples of such other (classes of) therapeutic agents which may be used in combination with the compounds or combinations of the invention include, but are by no means limited to: • aminoglycosides such as netilmicin, kanamycin, gentamycin, streptomycin, amikacin, and tobramycin;
  • macro lides such as erythromycin and lincomycin
  • tetracyclines such as tetracycline, doxycycline, chlortetracycline, and minocycline
  • ⁇ oxalidinones such as linezoloid; and fusidic acid; and chloramphenicol.
  • beta-lactam penicillins such as penicillin, dicloxacillin, and ampicillin
  • beta lactam cephalsporins such as cefepime, ceftazidime, cefotaxime, cefuroxime, cefaclor, and cetriaxone;
  • beta lactam carbapenems such as imipenem and meropenem
  • quinolones such as ciprofloxacin, moxifloxacin, and levofloxacin
  • sulfonamides such as sulfanilamide and sulfamethoxazole; metronidazole; rifampin; vancomycin; and nitrofurantoin
  • bacteriocins such as agrocin alveicin carnocin colicin curvaticin divercin enterocin enterolysin epidermin erwiniocin glycinecin halocin lactococin lacticin leucoccin mesentericin nisin pediocin plantaricin sakacin subtilin sulfolobicin vibriocin warnerin
  • Resistance modifying agents may target and inhibit multiple drug resistance (MDR) mechanisms, rendering the bacteria susceptible to antibiotics to which they were previously resistant.
  • MDR multiple drug resistance
  • These compounds include among others efflux inhibitors and Beta Lactamase inhibitors.
  • co-administration As used herein, the terms “co-administration”, “co-administered” and “in combination with”, referring to the compounds of the invention and, optionally, the one or more other therapeutic agents, is intended to mean, and does refer to and include the following:
  • kits of the invention typically comprises two or more separate pharmaceutical or veterinary composition, at least one of which contains an antibiotic pantothenamide derivative in accordance with the invention as well as means for separately retaining said composition, such as a container, divided bottle, or divided foil packet.
  • An example of such a kit is the familiar blister pack used for the packaging of tablets, capsules and the like.
  • the kit comprises two or more separate pharmaceutical compositions, at least one of which contains an antibiotic pantothenamide derivative in accordance with the invention while another one contains a pantetheinase activity reducing/inhibiting agant in accordance with the invention, as well as means for separately retaining said compositions.
  • Example 1 Synthesis of pantetheine analogues of the invention All reactions were performed under an argon atmosphere, unless stated otherwise.
  • Solvents were distilled from appropriate drying agents prior to use. Et 3 N was distilled and stored over KOH. All other chemicals were purchased from commercial suppliers and were used without further purification, unless stated otherwise. Reactions were followed, and Revalues are obtained using thin layer chromatography (TLC) on silica gel-coated plates (Merck 60 F254) with the indicated eluent and compounds were detected with UV-light and/or by charring at ca. 150 °C after dipping into a solution of potassium permanganate, or ninhydrin. Column or flash chromatography was carried out using ACROS silica gel (0.035-0.070 mm, pore diameter ca. 6 mm).
  • IR spectra were recorded on an ATI Mattson Genesis Series FTIR spectrometer. High-resolution mass spectra were recorded on a JEOL AccuTOF (ESI) or a MAT900 (EI, CI, and ESI). Melting points were analyzed with a Biichi melting point B-545 and are not corrected. NMR spectra were recorded at 298 K on a Bruker DMX 300 (300 MHz) and a Varian 400 (400 MHz) spectrometer in the solvent indicated.
  • pantothenic acid (4.47 g, 20.4 mmol) in a mixture of dry CH 2 Cl 2 /acetone (200 mL, 1 : 1 v/v) at 0 °C, were added 2- methoxyprop-l-ene (3.77 mL, 2.0 equiv) and /?TsOH (119 mg, 3 mol
  • Example 2 antibiotic and pantetheinase inhibitory activity of the substances and composition of the invention Pantheteinase activity
  • Pantetheinase activity was measured by the amount of free aminomethylcoumarin (AMC) released by the hydrolysis of the pantetheine-analogue pantothenate- AMC.
  • Pantothenate- AMC was incubated in phosphate buffer (100 mM potassium phosphate buffer pH 8.0) in the presence of serum or plasma as pantetheinase source with or without a potential pantetheinase inhibitor. In time, samples were taken and the reaction was terminated by addition of 100 mM CaC0 3 pH 10.5. Fluorescence was measured using a luminescence spectrometer (LS55, Perkin Elmer, EX 350 ⁇ 2.5 nm, EM 450 ⁇ 2.5 nm) against samples without serum or plasma as negative control.
  • LS55 luminescence spectrometer
  • pantothenamide N7Pan was incubated in phosphate buffer (500 ⁇ potassium phosphate buffer pH 8.0) for 24 hours at 37 °C with or without potential pantetheinase inhibitors in the presence of 1% human or fetal bovine serum (FBS, HyClone, Logan, UT) as pantetheinase source. Prior to analysis samples were diluted 100-fold in methanol (HPLC grade, Fisher Scientific). Mass spectrometry was performed on a JEOL JMS-T100CS (AccuTOF CS) connected to a Agilent 1100 series HPLC system.
  • Bacterial strains of Escherichia coli BL21(DE3) and Staphylococcus aureus were grown overnight in 1% Bacto Tryptone media (BD, Sparks, MD). Cultures were then diluted 1 : 1000 in fresh assay media and added to 96 well sterile ELISA plate (100 ul per well). The compounds to be tested were diluted in the same media and added to the bacteria (100 ul per well). Plates were incubated at 37 °C and bacterial growth was followed in time by reading the optical density at 490 nm using a microplate-reader (Model 450, Bio-Rad Laboratories Inc., Hercules, CA).
  • a bacterial strain of a bio luminescent Staphylococcus aureus (Xen8.1) was cultured in Mueller Hinton medium as described above in a 96-well plate with varying concentrations of the pantothenamide N7Pan and the pantetheinase inhibitor R 6, as indicated. Bio luminescence was recorded by IVIS optical imaging technology (Caliper Life Sciences). The emitted light is proportional to the number of bacteria in the wells.
  • Pantothenamide antibiotics are inactive in the presence of serum
  • Pantothenamides of which N5Pan and N7Pan are the prototypes, are active against gram-negative and gram-positive bacteria. Pantothenamides were found to be substrates of the key rate-controlling enzyme pantothenate kinase, and the rate of conversion of N5Pan was found to be more rapid than that of pantothenate itself. This led to the conclusion that the mechanism for toxicity towards E.coli is due to the formation of Co A analogs that lead to the transfer of an inactive 4'-phosphopantothenamide moiety to acyl carrier protein (ACP) which is the first step in the bacterial type II route of fatty acid synthesis (Zhang et al).
  • ACP acyl carrier protein
  • pantothenamides as such cannot be used as a viable antibiotic strategy in mammals because these compounds are hydrolyzed in serum.
  • Incubation of a prototypic pantothenamide N7Pan in serum readily generated the pantothenate and heptylamine degradation products as determined by mass spectrometry (see figures la and 2b).
  • Both pantetheinase inhibitors RR2 (figure la-b) and R 6 (figure 2a-c) inhibited the serum-mediated breakdown of N7Pan.
  • N7Pan was chosen rather than N5Pan for reasons of easy detection of the free amide.
  • FIG. 3 shows that N5Pan is active as an antibiotic against E.coli in 1% tryptone medium but addition of 10% serum completely abolishes antibiotic activity.
  • Figure 4 shows the potent inhibition of pantheteinase activity by the pantothenones RR2 and RR6.
  • the triazole analogue of pantetheine (denominated RR1), (R)-2,4-dihydroxy-N-((l-(2- hydroxyethyl)-lH-l,2,3-triazol-4-yl)methyl)-3,3-dimethylbutanamide) was found to be a poor pantetheinase inhibitor (IC 50 > 1 mM).
  • RR2 was then used to study the effect on the antimicrobial activity of N5Pan or N7Pan in vitro, using these compounds in the presence of human serum, in a 96-well plate format.
  • FIG. 5a-b shows an example of E.coli and S.aureus growth inhibition by combining RR2 with N5Pan or N7Pan, in a medium consisting of 1% tryptone and 10% complement-inactivated human serum.
  • FIG. 6 A second example (figure 6) serves to illustrate the effect of another pantetheinase inhibitor (RR6) to increase the potency of a pantothenamide antibiotic (N7Pan).
  • Figure 6 shows the effect of concentration ranges of RR6 and N7Pan on growth of a bio luminescent S.aureus strain (Xen 8.1). From this experiment it can be concluded that inhibition of pantetheinase activity will increase the potency of N7Pan, in the presence of serum, 30-60 fold, thereby bringing the MIC value in the range of currently used antibiotics.
  • pantothenamides As the putative mode of action of pantothenamides is at the level of CoA-dependent biochemical pathways, this raised the question of selectivity for microbial versus host targets. Using high concentrations of RR2 and N5Pan, which were inhibitory for E.coli, no toxicity or significant inhibition of cell growth in the human kidney cell line 293T nor in primary epidermal keratinocytes was observed (data not shown). Although the proposed mechanism of pantothenamides is at the level of bacterial fatty acid synthesis, which would also offer a good explanation for their selectivity, other mechanisms should be considered 12 .
  • Figure 1 (a+b) mass spectra of RR2 and N7Pan following incubation in 1% bovine serum.
  • Figure 1(a) shows that N7Pan is completely degraded as witnessed by the absence of the peaks at 339 and 355, and the appearance of the heptylamine peak at 116.
  • Figure 1(b) shows that addition of RR2 prevents breakdown of N7Pan as witnessed by the presence of peaks at 339 and 355, and the smaller heptylamine peak.
  • Figure 2a-c mass spectra of R 6 and N7Pan following incubation in 1% human serum.
  • Figure 2a spectrum of N7Pan (m/z of the parent compound: 339).
  • Figure 2b spectrum of N7Pan following incubation with human serum resulting in the appearance of the heptylamine hydrolysis product (m/z: 116).
  • Figure 3 Addition of serum abolishes the antibiotic effect of N5Pan on E.coli growth.
  • Figure 4 Inhibition of plasma pantetheinase activity by compounds R 1, R 2 and R 6.
  • Figure 6 Antibacterial activity of N7Pan and R 6 against a bio luminescent S. aureus strain. Wells with bacterial growth are dark to light grey here (e.g. right hand column); wells without growth are transparent (e.g. left hand column). As shown in the upper row, at the highest concentration of the pantetheinase inhibitor R 6 (128 ⁇ g/ml), a concentration of 0.5 ⁇ g/ml N7Pan is sufficient to cause complete inhibition of growth (well marked with asterisk)
  • Figure 7a+b Graphs showing decrease of plasma pantetheinase activity in vivo following administration of RR2 to mice (7a) or RR6 to rats (7b). On the Y-axis: the plasma pantetheinase activity, on the X-axis: time in minutes. A dose of 50 mg/kg in rats causes complete inhibition of plasma pantetehinase activity, which lasts up to 12 hours.
  • Vanin genes are clustered (human 6q22-24 and mouse 10A2B1) and encode isoforms of pantetheinase ectoenzymes. Immunogenetics 53: 296-306.
  • Ruan BH Cole DC, Wu P, Quazi A, Page K, Wright JF, Huang N, Stock JR, Nocka K, Aulabaugh A, Krykbaev R, Fitz LJ, Wolfman NM, Fleming ML (2009).

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention concerns novel amides derived from pantothenic acid having antibiotic activity and novel analogues of pantetheine inhibiting the activity of pantetheinase. Combinations of such substances can suitably beused in therapeutic and or prophylactic treatment of bacterial infections, fungal and yeast infections in a human or animal subject. The present invention resides in the findingthat pantothenamidesaloneare not at antibiotically active in the presence of plasma due to the breakdown of pantothenamides by enzymes with pantetheinase activityandthat inhibition of these enzymes prevented breakdown of pantothenamides,thuspreserving their antibiotic activity under physiological conditions in mammals. The present invention thus, for the first time, makes available treatment relying on interference with microbial CoA dependent pathways and, simultaneously, inhibiting or reducing pantetheinase activity.

Description

PANTOTHENIC ACID DERIVATIVES AND THEIR USE IN THE TREATMENT OF
MICROBIAL INFECTIONS
Field of the invention
The present invention concerns substances and compositions having antibiotic activity as well as their use in the therapeutic and/or prophylactic treatment of humans and animals. More in particular the present invention provides novel amides derived from pantothenic acid having antibiotic activity, and novel analogues of pantetheine inhibiting the activity of pantetheinase are provided. In addition, combinations of such compounds, pharmaceutical or veterinary compositions containing them, and uses thereof as medicinal products are provided.
Background
Increasing reports of antibiotic resistance involving opportunistic gram-positive pathogens, including methicillin-resistant and vancomycin-resistant Staphylococcus aureus strains (MRSA and VRSA), and multidrug-resistant Streptococcus pneumoniae, have emphasized the critical need for the development of antimicrobial compounds with novel modes of action. Nevertheless, since the early 1960s only a few classes of novel antibiotics have been introduced, with limited success sofar (Fischbach et al.6)
Coenzyme A (CoA), an essential co factor for maintaining life, is used in a multitude of biochemical reactions. In most bacteria, CoA is synthesized from pantothenic acid (vitamin B5) in 5 steps, with the first step being the phosphorylation of pantothenate by pantothenate kinase (CoaA). Although this pathway also exists in eukaryotes, in most cases there is no sequence homology between the prokaryotic and eukaryotic CoA biosynthetic enzymes. Thus, a potential for developing highly specific inhibitors of bacterial CoA enyzmes has long been recognized.
As of the early 70 's, amides derived from pantothenic acid (vitamin B5), have been reported to possess antibiotic activity in vitro (Clifton et al.1) During the last decades many of these pantothenamides have been synthesized (Choudry et al.2, Mercer et al.3, Virga et al.4) and the putative modes of action have been studied in detail (Vigra et al., Zhang et al.5). Pantothenamides, of which N5Pan and N7Pan are the typical prototypes, are active against gram negative (e.g. E.coli) and gram positive (e.g. S. aureus) bacteria in vitro, and are believed to interfere with bacterial lipid synthesis through their action as CoA antimetabolites. Hitherto, no experimental results of antimicrobial action of these compounds in animals or humans (in vivo) have ever been published or presented.
Given the emergence of multi-drug resistance among pathogens, the discovery and application of new treatment options is still an absolute priority. The present inventors have set out to develop new antimicrobial compositions that could aid in mitigating the development and/or consequences of increasing resistance among pathogens.
Summary of the Invention
The present inventors, to their very surprise, discovered that pantothenamides are not at all antibiotically active in the presence of plasma or serum. As will be illustrated in the experimental part, N5Pan is active as an antibiotic in 1% tryptone medium but addition of 10% plasma or serum completely abolishes antibiotic activity.
Without wishing to be bound by an particular theory, the present inventors hypothesize that this lack of activity is due to the breakdown of pantothenamides by enzymes with pantetheinase activity that are present in tissues and extracellular fluids such as plasma (Maras et al.7). The only known proteins in mammals that possess pantetheinase activity belong to the vanin (VNN) gene family (Martin et al.8). In humans, three vanin genes are known (VNN1, VNN2 and VNN3) that are ubiquitously expressed, as was shown before (Jansen et al.9). In mice only VNN1 and VNN3 are present. Both human and mouse plasma contain high levels of pantetheinase.
The present inventors surprisingly found that inhibition of pantetheinase activity prevented breakdown of pantothenamides, preserving their antibiotic activity under physiological conditions in mammals.
In accordance with the present invention an analogue of pantetheine was provided in an attempt to obtain an inhibitor of pantetheinase activity, which fully confirmed the concept. This compound was found to inhibit the action of plasma panthetheinase. It was found that, in the presence of plasma or serum, the growth inhibition of E.coli by N5Pan was restored in the presence of said inhibitor of pantetheinase activity. Thus a combination of an antimicrobial pantothenamide with an inhibitor of host pantetheinase activity provides a novel antibiotic strategy.
Combinations of pantothenate derivatives (e.g. pantothenamides) and pantetheine analogues to improve antibiotic activity have not been disclosed before. In Clifton et al1 antibiotic pantothenamides were combined with pantothenate showing that pantothenate decreased or abolished the antibiotic activity of pantothenamides. This was taken to indicate that pantothenamides acted as pantothenic acid antimetabolites. Neither the pantothenamides described in Clifton et al. nor the pantothenate has pantetheinase inhibitory activity.
Inhibitors of vanin-1 have been described for application in intestinal inflammation (see US 2004/247524). A recent publication has identified pantetheinase inhibitors derived from various chemical scaffolds in the LOP AC library, via a high-throughput approach (Ruan et al.10). The concept that pantothenamide antibiotics are degraded in vivo by pantetheinases and that pantetheinase inhibitors can be used to overcome this problem have never been suggested before.
It is furthermore assumed that certain modifications in the structure of the antibiotic pantothenamide derivative, lead to a decrease in its susceptibility to degradation by pantetheinase, thereby increasing its antibiotic potential under physiological conditions, with or without co-administration of separate pantetheinase inhibitors.
The present invention thus, for the first time, makes available compounds and combinations of compounds for use in therapeutic and or prophylactic treatment of bacterial infections, fungal and yeast infections in a human or animal subject in need thereof, relying on interference with microbial CoA dependent pathways and, simultaneously, inhibiting or reducing host-derived pantetheinase activity.
Detailed description of the invention.
Accordingly, in a first aspect of the present invention a pharmaceutical or veterinary composition is provided comprising:
one or more antibiotic pantothenamide derivatives; and
one or more pantetheinase inhibitors.
In another aspect of the invention certain novel pantothenamide derivatives having antibiotic activity are provided as such.
In another aspect of the invention certain novel pantetheinase inhibitors are provided as such.
In still other aspects of the invention the use of the aforementioned compositions and substances for the therapeutic and/or prophylactic treatment of a human or animal subject in need thereof is provided.
In the context of all aspects of the invention, the term "antibiotic pantothenamide derivative" refers to amide derivatives of pantothenic acid and substances structurally related to pantothenic acid, which have in vitro and/or in vivo antibiotic activity. As is understood by those skilled in the art, antibiotic means effective in reducing the viability or decreasing or inhibiting the growth or reproduction of a bacterium, yeast, fungi, mold, or other microrganism, in particular of a bacterium. Inhibiting or reducing the growth or reproduction means increasing the generation cycle time by at least 2-fold, preferably at least 10-fold, more preferably at least 100-fold, and most preferably indefinitely, as in total cell death. Terms such as antimicrobial, bacteriostatic and bactericidal are deemed synonymous to antibiotic and may be used interchangeably in the context of the present invention.
In a preferred embodiment the antibiotic pantothenamide derivative of the invention is selected from the group of derivatives represented by formula (I) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000005_0001
wherein:
R1 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide;
R2 and R3 independently represent a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
X1 and X2 independently represent hydrogen or a group selected from hydroxyl, thiol, cyanide, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, sulfonamide, amide, pyrazole or imidazole; and
X3 represents a hydrogen atom or a group selected from hydroxyl, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide, preferably a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide.
In a particularly preferred embodiment of the invention the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (la) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000006_0001
wherein R1 and X3 have the same meaning as defined above in relation to formula (I).
In an even more preferred embodiment of the invention the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (lb) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000006_0002
wherein R1 represents a group selected from alkyl, alkenyl and alkynyl.
Most preferably the antibiotic pantothenamide derivative is selected from 'N5Pan', 'N7Pan' and 'N9Pan' and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof, most preferably from N5Pan and N7Pan and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof, wherein N5Pan, N7Pan and N9Pan are represented by the following formulas:
Figure imgf000006_0003
In accordance with this invention the term 'pantetheinase inhibitor' typically refers to any substance somehow capable of i) reducing or inhibiting the activity of pantetheinase in viro and/or in vivo; ii) reducing or inhibiting the production of pantetheinase in vitro and/or in vivo; or iii) stimulating the inactivation or break-down of pantetheinase in vitro and/or in vivo. Proteins in mammals that possess pantetheinase activity belong to the vanin (VNN) gene family (Martin et al). Vanins are broadly expressed pantetheinases involved in the CoA pathway, allowing the turnover of pantetheine, thereby recycling pantothenic acid and producing cysteamine. In humans, three vanin genes are known (VNN1, VNN2 and VNN3) that are broadly expressed, as was shown before (Jansen et al). In mice only VNN1 and VNN3 are present. Both human and mouse plasma contain high levels of pantetheinase activity.
In a preferred embodiment of the invention, the pantetheinase inhibitor is selected from the group consisting of competitive enzyme inhibitors, reactive enzyme substrates capable of covalently modifying a residue of the enzyme catalytic site, antibodies capable of specifically interacting with VNN, preferably VNN1, and inhibiting its activity, and nucleic acids blocking transcription of VNN gene, preferably VNN1 gene, and/or translation of VNN mRNA, preferably VNN 1 mRNA.
In another preferred embodiment of the invention, the pantetheinase inhibitor is selected from the group consisting of VNN antagonistic agents, including VNN-1 antagonistic agents, VNN-2 antagonistic agents and VNN-3 antagonistic agents. As will be understood by those skilled in the art, the term 'antagonistic agent' means a compound that by any means, partly or completely blocks the dissimilative pathway of CoA referred to above (allowing the turnover of pantetheine, thereby recycling pantothenic acid and producing cysteamine).
In another preferred embodiment of the invention, the pantetheinase inhibitor is selected from the group of non-hydrolysable pantothenic acid analogues and reactive enzyme substrates capable of covalently or non-covalently binding a residue of the enzyme catalytic site.
In a particularly preferred embodiment of the invention, the pantetheinase inhibitor is selected from the group of substances represented by formula II, III or IV, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000008_0001
(TV)
R4 R5 wherein:
R4, R5 and R6 independently represent a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
R7 representsa hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide; X4 and X5 independently represent hydrogen hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
cycloalkenylalkyl, sulfonamide, amide, pyrazole and imidazole;
X6 represents sulfur, oxygen, carbon or nitrogen, said nitrogen optionally being substituted with a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, preferably sulfur, oxygen or nitrogen, said nitrogen optionally being substituted with a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
X7 represents oxygen or cyanide, where the dotted line indicates the optional presence of an additional covalent bond; and m and n are integers within the range of 0-6.
In a particularly preferred embodiment of the invention m is not 0.
In a particularly preferred embodiment of the invention X7 represents oxygen.
In a particularly preferred embodiment R7 does not represent hydrogen.
In a particularly preferred embodiment of the invention, the pantetheinase inhibitor is selected from substances represented by formula Ila or lib, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000009_0001
wherein:
R4, R5 and R6 independently represent hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
R7 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide; X4 and X5 independently represent hydrogen, hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
cycloalkenylalkyl, sulfonamide, amide, pyrazole and imidazole; and
m and n are integers within the range of 0-6.
In a particularly preferred embodiment of the invention m is not 0.
In a particularly preferred embodiment R7 does not represent hydrogen.
More preferably, the pantetheinase inhibitor is selected from substances represented by formula lie, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000010_0001
wherein
R7 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide;
X4 and X5 independently represent hydrogen, hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
cycloalkenylalkyl, sulfonamide, amide, pyrazole and imidazole; and
m and n are integers within the range of 0-6.
In a particularly preferred embodiment of the invention m is not 0.
In a particularly preferred embodiment R7 does not represent hydrogen.
Still more preferably, the pantetheinase inhibitor is selected from substances represented by formula lie, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof, as defined above, wherein X4 and X5 represent hydroxyl and n and m are 1.
In a particularly preferred embodiment of the invention the pantetheinase inhibitor is selected from the compounds designated 'RR2', 'RR6', 'RR7' and 'RR8', more preferably the compounds designated RR2 and RR6, most preferably the compound RR6, and pharmaceutically acceptable salts, esters, amides, or prodrugs thereof:
Figure imgf000010_0002
Figure imgf000010_0003
Figure imgf000011_0001
In another preferred embodiment of the invention the pantetheinase inhibitor is selected from the substances represented by formula Ilia or Illb, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000011_0002
wherein:
R4, R5 and R6 independently representa hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
R7 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and
heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide; X4 and X5 independently represent hydrogen, hydroxyl, thiol, cyanide or halogen, or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl,
cycloalkenylalkyl, sulfonamide, amide, pyrazole and imidazole; X6 represents sulfur, oxygen, carbon or nitrogen, said nitrogen optionally being substituted by a radical selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; preferably sulfur, oxygen or nitrogen, said nitrogen optionally being substituted by a radical selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; and
m and n are integers within the range of 0-6.
In a particularly preferred embodiment of the invention m is not 0.
In a particularly preferred embodiment R7 does not represent hydrogen.
In a particularly preferred embodiment of the invention the pantetheinase inhibitor is selected from the substances represented by formula Ila, Ilia and IV as defined herein before, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof.
As can be inferred from the former, a particularly preferred embodiment of the invention concerns a pharmaceutical or veterinary composition as defined herein before, comprising a combination of a pantothenamide derivative selected from N5Pan, N7Pan and N9Pan and a pantetheinase inhibitor selected from RR2, RR6, RR7 and RR8, such as a combination of N5Pan and RR2; a combination of N7Pan and RR2; a combination of N5Pan, N7Pan and RR2; a combination of N5Pan and RR6; a combination of N7Pan and RR6; or a combination of N5Pan, N7Pan and RR6; or pharmaceutically acceptable salts, esters, amides, or prodrugs of said compounds.
As utilized herein, the term "alkyl", either alone or within other terms, means an acyclic alkyl radical, preferably containing from 1 to 10, more preferably from 1 to about 8 carbon atoms and most preferably 1 to about 6 carbon atoms. Said alkyl radicals may be optionally substituted as defined elsewhere in this document. Examples of such radicals include methyl, ethyl, chloroethyl, hydroxyethyl, n-propyl, oxopropyl, isopropyl, n-butyl, cyanobutyl, isobutyl, sec-butyl, tert-butyl, pentyl, aminopentyl, iso-amyl, hexyl, octyl and the like.
The term "alkenyl" refers to an unsaturated, acyclic hydrocarbon radical in so much as it contains at least one double bond. Such alkenyl radicals typically contain from 2 to 10 carbon atoms, preferably from 2 to 8 carbon atoms and most preferably 2 to about 6 carbon atoms. Said alkenyl radicals may be optionally substituted as defined elsewhere in this document. Examples of suitable alkenyl radicals include ethenyl, 1-propenyl, 2-propenyl, 2- methyl-l-propenyl, 1-butenyl, 2-butenyl and the like. The term "alkynyl" refers to an unsaturated, acyclic hydrocarbon radical in so much as it contains one or more triple bonds, such radicals typically containing from 2 to 10 carbon atoms, preferably having from 2 to 8 carbon atoms and most preferably from 2 to 6 carbon atoms. Said alkynyl radicals may be optionally substituted with groups as elsewhere in this document. Examples of suitable alkynyl radicals include ethynyl, propynyl, hydroxypropynyl, butyne-l-yl, butyn-2-yl, pentyne-l-yl, pentyne-2-yl, 4 methoxypentyn-2-yl, 3-methylbutyn-l- yl, hexyne-l-yl, hexyne-2-yl, hexyne-3-yl, 3,3-dimethylbutyn-l-yl radicals and the like.
The term "cycloalkyl" refers to carbocyclic radicals typically having 3 to 10 carbon atoms, preferably 3 to 8 carbon atoms, most preferably 5 to 8 carbon atoms. Said cycloalkyl radicals may be optionally substituted as defined elsewhere in this document. Examples of suitable cycloalkyl radicals include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
The term "cycloalkenyl" embraces carbocyclic radicals having 3 to 10 carbon atoms and one or more carbon-carbon double bonds. Preferred cycloalkenyl radicals are "lower cycloalkenyl" radicals having 3-8 carbon atoms, more preferably 5-8. Examples include radicals such as cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptenyl.
The term "aryl", alone or in combination, means a 5-10 membered carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendant manner or may be fused. The term "fused" means that a second ring is present having two adjacent atoms in common with the first ring. The term "fused" is equivalent to the term "condensed". The term "aryl" embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane and biphenyl.
The term "heteroaryl" (on its own or in any combination, such as "heteroaryloxy", or "heteroaryl alkyl") is used herein to mean a 5-10 membered aromatic ring system containing one, two or three rings, which may be attached in a pendant manner or may be fused, wherein at least one of said rings contains one or more heteroatoms selected from the group consisting of N, O or S. Examples include, but are not limited to, pyrrole, pyrazole, furan, thiophene, quinoline, isoquinoline, quinazolinyl, pyridine, pyrimidine, oxazole, thiazole, thiadiazole, tetrazole, triazole, imidazole, or benzimidazole.
The terms "cycloalkylalkyl", "cycloalkenylalkyl", "arylalkyl" and "heteroarylalkyl" embrace, respectively, the afore-defined cycloalkyl, cycloalkenyl, aryl and heteroaryl radicals attached to the main molecular moiety, i.e. the basic moiety depicted in the formulae, through an alkyl radical, typically an alkyl radical having 1-10, preferably 1-8, most preferably 1-6 carbon atoms, as will be understood by those skilled in the art. Representative examples of arylalkyl include, but not limited to, phenylmethyl, phenylethyl and naphthylmethyl. Representative examples of heteroarylalkyl groups include, but are not limited to, thiazolylmethyl, thienylmethyl, furylmethyl, imidazolylmethyl and pyridylmethyl.
The term "sulfonamide" includes moieties which contain a group of the formula - SO2NRR, where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.
The term "amide" includes moieties which contain a nitrogen atom which is bound to the carbon of a carbonyl or a thiocarbonyl group. The term includes "alkaminocarboxy" groups which include alkyl, alkenyl, or alkynyl groups bound to an amino group bound to a carboxy group. It includes arylaminocarboxy groups which include aryl or heteroaryl moieties bound to an amino group which is bound to the carbon of a carbonyl or thiocarbonyl group. The terms "alkylamino carboxy," "alkenylaminocarboxy," "alkynylaminocarboxy," and "arylaminocarboxy" include moieties wherein alkyl, alkenyl, alkynyl and aryl moieties, respectively, are bound to a nitrogen atom which is in turn bound to the carbon of a carbonyl group.
The compounds of the invention can contain one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers, or diastereomers. According to the invention, the chemical structures depicted herein, and therefore the compounds of the invention, encompass all of the corresponding compound's enantiomers and stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
A compound of the invention is considered optically active or enantiomerically pure (i.e., substantially the R-form or substantially the S-form) with respect to a chiral center when the compound is about 90% enantiomeric excess (ee) or greater, preferably, equal to or greater than 95% enantiomeric excess with respect to a particular chiral center. A compound of the invention is considered to be in enantiomerically-enriched form when the compound has an enantiomeric excess of greater than about 1% ee, preferably greater than about 5% ee, more preferably, greater than about 10% ee with respect to a particular chiral center. A compound of the invention is considered diastereomerically pure with respect to multiple chiral centers when the compound is about 90%> de (diastereomeric excess) or greater, preferably, equal to or greater than 95% de with respect to a particular chiral center. A compound of the invention is considered to be in diastereomerically-enriched form when the compound has an diastereomeric excess of greater than about 1% de, preferably greater than about 5% de, more preferably, greater than about 10% de with respect to a particular chiral center. As used herein, a racemic mixture means about 50% of one enantiomer and about 50% of is corresponding enantiomer relative to all chiral centers in the molecule. Thus, the invention encompasses all enantiomerically-pure, enantiomerically-enriched, diastereomerically pure, diastereomerically enriched, and racemic mixtures of compounds of Formulas I through III.
Enantiomeric and diastereomeric mixtures can be resolved into their component enantiomers or stereoisomers by well known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and diastereomers can also be obtained from diastereomerically- or enantiomerically-pure intermediates, reagents, and catalysts by well known asymmetric synthetic methods.
In a particularly preferred embodiment of the invention, the pantothenic acid and/or pantothenamide derivatives or analogues are 'enantiomerically pure' (i.e. according to the above definitions) derivatives or analogues of D(+) pantothenic acid and/or D(+) pantothenamide, i.e. 'enantiomerically pure' substances possessing the same stereochemical arrangement as the corresponding stereocenter in D-(+) pantothenic acid and/or D(+) pantothenamide.
The term "pharmaceutically acceptable salts, esters, amides, and prodrugs" as used herein refers to salts, amino acid addition salts, esters, amides, and prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
The term "salts" refers to inorganic and organic acid or base addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. Depending on the particular substituents found on the compounds described herein. When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present invention contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al, "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19).
Examples of suitable esters include compounds of the invention which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Examples of pharmaceutically acceptable, relatively non-toxic esters of the invention include Ci-C6 alkyl esters and C5-C7 cycloalkyl esters. Esters of the compounds of the invention can be prepared according to conventional methods. Pharmaceutically acceptable esters can be obtained through reaction of hydroxy groups of the compound with an organic acid, such as acetic acid or benzoic acid. In the case of compounds containing carboxylic acid groups, the pharmaceutically acceptable esters are prepared by reaction of said carboxylic acid group, as will be understood by those skilled in the art.
Examples of pharmaceutically acceptable, non-toxic amides of the compounds of this invention include amides derived from ammonia, primary Ci-C6 alkyl amines and secondary Ci-C6 dialkyl amines wherein the alkyl groups are straight or branched chain. In the case of secondary amines, the amine may also be in the form of a 5- or 6-membered heterocycle containing one nitrogen atom. Amides derived from ammonia. Amides of the compounds of the invention may be prepared according to conventional methods.
The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compounds of the above formula, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
Furthermore, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
The compounds of the invention can be provided as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
Another aspect of the present invention concerns the use of the compounds of the invention, the combinations thereof or the pharmaceutical compositions containing them, as defined herein before, in a therapeutic or prophylactic method of treatment, in particular in treating or preventing a disease or condition selected from the group of bacterial infections, fungal and yeast infections and intracellular protozoan infections in a human or animal subject in need thereof.
Another aspect of the present invention, a method of treating and/or preventing a disease or condition selected from the group of bacterial infections, fungal and yeast infections and intracellular protozoan infections in a human or animal subject in need thereof is provided, said method comprising administering to said human or animal subject an effective amount of a compound of the invention, a combination thereof or a pharmaceutical composition containing it, as defined herein before.
In a preferred embodiment of the invention, these uses and methods concern treatment of human subjects in need thereof. For administration to human patients, the total daily dose of antibiotic pantothenamide derivative of the invention is typically within the range 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg depending, of course, on the mode of administration and/or the severity of the disease or condition. For example, an intravenous daily dose may only require from 0.001 mg to 40 mg. Likewise, the total daily dose of pantetheinase inhibitor of the invention is typically within the range 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg depending, also, on the mode of administration and/or the severity of the condition to be treated.
The total daily dosage(s) may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein. These dosages, furthermore, are based on an average human subject having a weight of about 65kg to 70kg. The physician will readily be able to determine appropriate doses for subjects whose bodily weight differs significantly from said average, for example for children, as will be understood by those skilled in the art.
In another, equally preferred embodiment of the invention, the above uses and methods concern the treatment of animals in need thereof. Preferably the methods and uses concern the treatment of domestic animals and/or farm animals, such as dogs, cats, horses, pigs, cattle, sheep, goats, poultry, etc. A veterinarian will be able to determine appropriate doses for these different kinds of animals depending, of course, on their average body weight, the route of administration and/or the severity of the disease or condition.
It is preferred that the compounds of the invention, the combinations thereof or the pharmaceutical or veterinary compositions containing them can be administered through any of the conventional routes. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth. Parenteral administration may involve administration directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques. The compounds of the invention may furthermore be administered topically or locally to the skin or mucosa. It is preferred that the compounds of the invention, the combinations thereof or the pharmaceutical or veterinary compositions containing them are administered orally or parenterally, preferably orally or intravenously.
Furthermore, it is preferred that the compounds of the invention, the combinations thereof or the pharmaceutical or veterinary compositions containing them are administered repeatedly, typically at least once weekly, more preferably at least once every three days, e.g. once daily. Typically, treatment lasts at least a week, more preferably at least two weeks, more preferably at least three weeks. As is generally known by those skilled in the art, repeated and continued administration of antibiotics typically reduces the risks of development of resistance towards the antibiotic and, without wishing to be bound by any particular theory, it is hypothesized that this issue might apply to compounds and combinations of the present invention. Compounds of the invention or combinations thereof may be administered to a human or animal subject either alone or as part of a pharmaceutical or veterinary composition
Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable excipients. The term "excipient" is used herein to describe any ingredient other than the compound(s) of the invention. The choice of excipient will, to a large extent, depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
In a preferred embodiment of the invention a pharmaceutical composition for use in humans is provided comprising one or more antibiotic pantothenamide derivatives of the invention, typically in an amount within the range of 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg and/or one or more pantetheinase inhibitors of the invention, typically in a total amount within the range of 0.001 mg to 5000 mg, preferably 0.01 mg to 500 mg, most preferably 0.05 mg to 250 mg; as well as one or more pharmaceutically acceptable excipients.
Likewise, in a preferred embodiment of the invention a veterinary composition for use in animals is provided comprising one or more antibiotic pantothenamide derivatives of the invention, typically in an amount within the range of 0.0001 mg to 50000 mg, preferably 0.001 mg to 5000 mg, most preferably 0.005 mg to 2500 mg and/or one or more pantetheinase inhibitors of the invention, typically in a total amount within the range of 0.0001 mg to 50000 mg, preferably 0.001 mg to 5000 mg, most preferably 0.005 mg to 2500 mg; as well as one or more veterinary acceptable excipients.
Compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in 'Remington's Pharmaceutical Sciences', 19th Edition (Mack Publishing Company, 1995).
Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including liquid- filled), chews, multi- and nano- particulates, gels, solid solution, liposome, films, ovules, sprays and liquid formulations.
Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet. The compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, 1 1 (6), 981- 986, by Liang and Chen (2001 ).
For tablet dosage forms, depending on dose, the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form. In addition to the drug, tablets generally contain a disintegrant. Examples of disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl -substituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate. Generally, the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form. Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate. Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When present, surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet. Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate. Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet. Other possible ingredients include anti-oxidants, colourants, flavouring agents, preservatives and taste- masking agents. Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant. Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting. The final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both. A syrup or elixir may contain the active compounds sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compounds may be incorporated into sustained-release preparation and formulations. The formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets, Vol. 1 , by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980). Suitable modified release formulations for the purposes of the invention are, such as high energy dispersions and osmotic and coated particles, are to be found in Pharmaceutical Technology On-line, 25(2), 1- 14, by Verma et al (2001 ). The use of chewing gum to achieve controlled release is described in WO 00/35298.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof. Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water. The preparation of parenteral formulations under sterile conditions, for example, by lyophilisation, may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art. The solubility of compounds used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility- enhancing agents. Formulations for parenteral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and poly(c//-lactic-coglycolic)acid (PGLA) microspheres.
Typical formulations for topical or local administration include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958 by Finnin and Morgan (October 1999). Formulations for topical or local administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1 , 1 , 1 ,2-tetrafluoroethane or 1,1 ,1 ,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin. The pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid. The compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate. Formulations for rectal/vaginal administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
The compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene glycol- containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration. Drug-cyclodextrin complexes, for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion complexes may be used. As an alternative to direct complexation with the drug, the cyclodextrin may be used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser.
As defined herein before, the present invention provides compositions comprising more than one antibiotic pantothenamide derivatives. According to another embodiment of the present invention, the compounds and combinations defined herein before, can also be used in combination with one or more additional antibiotic agents from a different class. Such combinations may result in an antibiotic product with an increased spectrum of action and restored efficacy against resistant bacteria. Suitable examples of such other (classes of) therapeutic agents which may be used in combination with the compounds or combinations of the invention include, but are by no means limited to: • aminoglycosides such as netilmicin, kanamycin, gentamycin, streptomycin, amikacin, and tobramycin;
• macro lides such as erythromycin and lincomycin;
• tetracyclines such as tetracycline, doxycycline, chlortetracycline, and minocycline;
· oxalidinones such as linezoloid; and fusidic acid; and chloramphenicol.
• beta-lactam penicillins such as penicillin, dicloxacillin, and ampicillin;
• beta lactam cephalsporins such as cefepime, ceftazidime, cefotaxime, cefuroxime, cefaclor, and cetriaxone;
• beta lactam carbapenems such as imipenem and meropenem;
· quinolones such as ciprofloxacin, moxifloxacin, and levofloxacin;
• sulfonamides such as sulfanilamide and sulfamethoxazole; metronidazole; rifampin; vancomycin; and nitrofurantoin
• bacteriocins, such as agrocin alveicin carnocin colicin curvaticin divercin enterocin enterolysin epidermin erwiniocin glycinecin halocin lactococin lacticin leucoccin mesentericin nisin pediocin plantaricin sakacin subtilin sulfolobicin vibriocin warnerin
Another class of therapeutic agents which may suitably be used in conjunction with the antibiotic compounds or combinations of the invention include the so-called resistance modifying agents. Resistance modifying agents may target and inhibit multiple drug resistance (MDR) mechanisms, rendering the bacteria susceptible to antibiotics to which they were previously resistant. These compounds include among others efflux inhibitors and Beta Lactamase inhibitors.
As used herein, the terms "co-administration", "co-administered" and "in combination with", referring to the compounds of the invention and, optionally, the one or more other therapeutic agents, is intended to mean, and does refer to and include the following:
· simultaneous administration of such combination of compounds, when such components are formulated together into a single dosage form which releases said components at substantially the same time following administration,
• substantially simultaneous administration of such combination of compounds, when such components are formulated apart from each other into separate dosage forms which are administered at substantially the same time, where after said components are released at substantially the same time,
• sequential administration of such combination of compounds, when such components are formulated apart from each other into separate dosage forms which are administered at consecutive times with a significant time interval between each administration; and It is within the scope of the present invention that two or more pharmaceutical compositions, at least one of which contains an anitbiotic pantothenamide derivative in accordance with the invention, may conveniently be combined in the form of a kit suitable for co-administration of the compositions. Thus a kit of the invention typically comprises two or more separate pharmaceutical or veterinary composition, at least one of which contains an antibiotic pantothenamide derivative in accordance with the invention as well as means for separately retaining said composition, such as a container, divided bottle, or divided foil packet. An example of such a kit is the familiar blister pack used for the packaging of tablets, capsules and the like. In a particularly preferred embodiment of the invention the kit comprises two or more separate pharmaceutical compositions, at least one of which contains an antibiotic pantothenamide derivative in accordance with the invention while another one contains a pantetheinase activity reducing/inhibiting agant in accordance with the invention, as well as means for separately retaining said compositions.
In this document and in its claims, the verb "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition, reference to an element by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements. The indefinite article "a" or "an" thus usually means "at least one".
The invention will be illustrated in more detail in the following examples, which are in no way intended to limit the scope of the invention.
Example 1: Synthesis of pantetheine analogues of the invention All reactions were performed under an argon atmosphere, unless stated otherwise.
Solvents were distilled from appropriate drying agents prior to use. Et3N was distilled and stored over KOH. All other chemicals were purchased from commercial suppliers and were used without further purification, unless stated otherwise. Reactions were followed, and Revalues are obtained using thin layer chromatography (TLC) on silica gel-coated plates (Merck 60 F254) with the indicated eluent and compounds were detected with UV-light and/or by charring at ca. 150 °C after dipping into a solution of potassium permanganate, or ninhydrin. Column or flash chromatography was carried out using ACROS silica gel (0.035-0.070 mm, pore diameter ca. 6 mm). IR spectra were recorded on an ATI Mattson Genesis Series FTIR spectrometer. High-resolution mass spectra were recorded on a JEOL AccuTOF (ESI) or a MAT900 (EI, CI, and ESI). Melting points were analyzed with a Biichi melting point B-545 and are not corrected. NMR spectra were recorded at 298 K on a Bruker DMX 300 (300 MHz) and a Varian 400 (400 MHz) spectrometer in the solvent indicated. Chemical shifts are given in parts per million (ppm) with respect to tetramethylsilane (0.00 ppm), or CHD2OD (3.31 ppm) as internal standard for 1H-NMR; and CDC13 (77.16 ppm), or CD3OD (49.00 ppm) as internal standard for 13C-NMR. Coupling constants are reported as J values in hertz (Hz).
(R)-3-(2,2,5,5-tetramethyl-l,3-dioxane-4-carboxamido)propanoic acid (1)
To a solution of pantothenic acid (4.47 g, 20.4 mmol) in a mixture of dry CH2Cl2/acetone (200 mL, 1 : 1 v/v) at 0 °C, were added 2-
Figure imgf000026_0001
methoxyprop-l-ene (3.77 mL, 2.0 equiv) and /?TsOH (119 mg, 3 mol
%). After 15 min the reaction mixture was allowed to warm to rt. After 2 h the reaction was quenched with saturated aqueous NaHC03 (2 mL), dried (Na2S04), and concentrated in vacuo. The crude product was purified by flash column chromatography (MeOH/CH2Cl2, 0: 1→2:3) to afford 1 (4.98 g, 94% yield) as a colorless oil. R/ 0.15 (MeOH/CH2Cl2, 1 :9). 1H NMR (CDC13, 400 MHz): δ 7.62 (t, J = 5.3 Hz, 1H), 4.12 (s, 1H), 3.73 (d, J = 11.7 Hz, 1H), 3.56-3.38 (m, 2H), 3.27 (d, J = 11.7 Ηζ,ΙΗ), 2.52 (t, J = 6.7 Hz, 2H), 1.44 (s, 6H), 0.99 (s, 3H), 0.97 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 175.2, 171.9, 100.2, 78.2, 72.2, 35.5, 34.5, 33.8, 29.7, 22.4, 19.3, 19.1.
(R)-N- {3- [methoxy(methyl)amino] -3-oxopropyl}-2,2,5,5-tetr amethyl- 1 ,3-dioxane-4- carboxamide (2)
To a solution of 1 (1.80 g, 6.92 mmol) in dry CH2C12 (65 mL) at rt were added, EDC (2.09 g, 1.5 equiv), N,0-dimethylhydroxylamine
Figure imgf000026_0002
hydrochloride (1.04 g, 1.5 equiv) and DIPEA (3.43 mL, 3.0 equiv), followed by DMAP (483 mg, 0.5 equiv). The reaction mixture was stirred over night at rt, quenched with saturated aqueous NH4C1 (40 mL), extracted with CH2C12.( 3 x 50 mL), dried (Na2S04), and concentrated in vacuo. The product was purified by column chromatography (MeOH/CH2Cl2, 0: 1→1 :4) to afford 2 (1.90 g, 91% yield) as a colorless oil. Rf 0.56 (MeOH/CH2Cl2, 1 :9). [<x]∞+44.5 (c 1.32, CH2C12). IR (ATR) 3417, 3334, 2980, 2940, 2871,
1661, 1520, 1378, 1196, 1095, 873 cm-1. 1H NMR (CDC13, 400 MHz): δ 7.13 (t, J = 5.7 Hz, 1H), 4.07 (s, 1H), 3.68 (d, J= 11.7 Hz, 1H), 3.67 (s, 3H), 3.64-3.48 (m, 2H), 3.27 (d, J= 11.7 Hz, 1H), 3.18 (s, 3H), 2.76-2.59 (m, 2H), 1.46 (s, 3H), 1.42 (s, 3H), 1.03 (s, 3H), 0.96 (s, 3H). 13C NMR (CDCls, 75 MHz): δ 169.9, 99.1, 77.3, 71.6, 61.4, 34.2, 33.1, 32.3, 31.9, 29.6, 22.3, 19.0, 18.8. Ci4H26N205 (M+Na)+: 325.1739, found: 325.1746. (R)-2,2,5,5-tetramethyl- V-(3-oxohept-6-en-l-yl)-l,3-dioxane-4-carboxamide (3)
To a solution of 2 (2.10 g, 6.95 mmol) in a mixture of dry Et20/THF (70 mL, 1 : 1 v/v) at 0 °C, was added dropwise 3-
Figure imgf000027_0001
butenylmagnesium bromide (28.0 mL of a 0.5 M solution in THF,
13.9 mmol, 2.0 equiv). After 15 min the reaction mixture was allowed to warm to rt and stirred for 4 h. Next, saturated aqueous NH4C1 was added to quench the reaction, followed by extraction with CH2C12 (3 x 70 mL). The organic layers were combined, dried (Na2S04), and concentrated in vacuo. The product was purified by column chromatography (EtO Ac/heptane, 0: 1→1 :2) to afford 3 (1.23 g, 59% yield) as a colorless oil. R/ 0.86 (MeOH/CH2Cl2, 1 :9). [a] ∞ +44.1 (c 1.38, CH2C12). IR (ATR) 3421, 2915, 2849, 1709, 1666, 1519, 1377, 1092, 871, 701 cm-1. 1H NMR (CDC13, 400 MHz): δ 6.96-6.87 (m, 1H), 5.79 (ddt, J= 6.5, 10.2, 16.7 Hz, 1H), 5.05-4.96 (m, 2H), 4.05 (s, 1H), 3.68 (d, J = 1 1.7 Hz, 1H), 3.58-3.42 (m, 2H), 3.27 (d, J = 11.7 Hz, 1H), 2.68 (t, J = 6.0 Hz, 2H), 2.53-2.49 (m, 2H), 2.36-2.30 (m, 2H), 1.46 (s, 3H), 1.41 (s, 3H), 1.04 (s, 3H), 0.95 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 209.2, 169.9, 136.9, 115.6, 99.1, 77.3, 71.6, 42.3, 42.1, 33.4, 33.1, 29.6, 27.8, 22.3, 19.0, 18.8. HRMS (ESI) m/z calcd for Ci6H27Ni04Na (M+Na)+: 320.1838, found: 320.1837.
(R)-2,2,5,5-tetramethykV- [3-oxo-3-(pentylamino)pr opyl] - 1 ,3-dioxane-4-carboxamide (4)
Prepared as described for 2, starting from 1 (3.40 g, 13.1 mmol) and n-amylamine (2.30 mL, 1.5 equiv). Column
Figure imgf000027_0002
chromatography (EtO Ac/heptane, 0: 1— >4: 1) afforded 4 (1.89 g, 44% yield) as a white solid. Rf 0.56 (MeOH/CH2Cl2, 1 :9). Mp 81.5 °C. [<x]∞+41.6 (c 1.01,
CH2C12). IR (ATR) 3430, 3317, 3300, 2954, 2931, 2868, 1649, 1526, 1463, 1377, 1197, 1098, 873 cm-1. 1H NMR (CDCI3, 400 MHz): δ 7.02 (t, J = 5.2 Hz, 1H), 5.88-5.84 (m, 1H), 4.07 (s, 1H), 3.68 (d, J = 11.7 Hz, 1H), 3.64-3.46 (m, 2H), 3.28 (d, J = 11.7 Hz, 1H), 3.26-3.21 (m, 2H), 2.43 (t, J = 6.2 Hz, 2H), 1.49 (dt, J = 7.3, 14.6 Hz, 2H), 1.46 (s, 3H), 1.42 (s, 3H), 1.38- 1.24 (m, 4H), 1.04 (s, 3H), 0.97 (s, 3H), 0.90 (t, J= 6.8 Hz, 3H). 13C NMR (CDC13, 75 MHz): δ 170.9, 170.3, 99.2, 77.3, 71.6, 39.7, 36.4, 35.1 , 33.1, 29.6, 29.4, 29.2, 22.5, 22.3, 19.0, 18.8, 14.1. HRMS (ESI) m/z calcd for Ci7H33N204 (M+H)+: 329.2440, found: 329.2426. (R)- V-[3-(heptylamino)-3-oxopropyl]-2,2,5,5-tetramethyl-l,3-dioxane-4-carboxamide (5)
Prepared as described for 2, starting from 1 (4.50 g, 17.4 mmol) and n-heptylamine (3.90 mL, 1.5 equiv). Column
Figure imgf000028_0001
chromatography (EtO Ac/heptane, 1 :2— 1 :0) afforded 5 (3.15 g, 51% yield) as a colorless oil. Rf 0.56 (MeOH/CH2Cl2, 1 :9). [o¾° +39.4 (c 1.00,
CH2C12). IR (ATR) 3425, 3321, 2927, 2863, 1650, 1526, 1459, 1377, 1196, 1098, 875 cnT1. 1H NMR (CDCls, 400 MHz): δ 7.04 (t, J = 5.9 Hz, 1H), 6.03 (t, J = 4.9 Hz, 1H), 4.06 (s, 1H), 3.68 (d, J = 11.7 Hz, 1H), 3.63-3.46 (m, 2H), 3.28 (d, J = 11.7 Hz, 1H), 3.26-3.20 (m, 2H), 2.42 (t, J= 6.2 Hz, 2H), 1.53-1.47 (m, 2H), 1.46 (s, 3H), 1.41 (s, 3H), 1.32-1.24 (m, 8H), 1.03 (s, 3H), 0.97 (s, 3H), .88 (t, J= 6.9 Hz, 3H). 13C NMR (CDC13, 75 MHz): δ 170.8, 170.2, 99.1, 77.1, 71.5, 39.6, 36.2, 35.0, 33.0, 31.7, 29.6, 29.5, 29.0, 26.9, 22.6, 22.2, 18.9, 18.7, 14.1. HRMS (ESI) m/z calcd for Ci9H37N204 (M+H)+: 357.2753, found: 357.2747.
(R)-2,4-dihydroxy-3,3-dimethyl- V-(3-oxohept-6-en-l-yl)butanamide (RR2, 6)
To a solution of 3 (30 mg, 0.10 mmol) in MeCN (1.0 mL) was added, B1CI3 (6.5 mg, 20 mol%), followed by distilled H20 (36
Figure imgf000028_0002
μί, 20 equiv). The reaction was stirred at rt for 4 h, then filtered and concentrated in vacuo. After dilution with EtO Ac (10 mL), the reaction mixture was washed with saturated aqueous NaHC03 (2 x 8 mL) and the aqueous layer was extracted with EtOAc (3 >< 8 mL). The organic layers were combined, dried (Na2S04), and concentrated in vacuo. The product was purified by column chromatography (EtO Ac/heptane, 1 : 1— 1 :0) to afford 6 (24 mg, 92% yield) as a colorless oil. R/ 0.52 (MeOH/CH2Cl2, 1 :9). [<x]∞ +32.6 (c 1.18, CH2C12). IR (ATR)
3347, 2955, 2872, 1706, 1646, 1532, 1079, 1041, 919 cm-1. 1H NMR (CDC13, 400 MHz): δ 7.25 (t, J = 6.0 Hz, 1H), 5.78 (ddt, J = 6.5, 10.2, 16.7 Hz, 1H), 5.03 (ddd, J = 1.3, 3.4, 16.7 Hz, 1H), 4.99 (ddd, J = 1.3, 3.4, 10.2 Hz, 1H), 4.16 (d, J = 4.5 Hz, 1H), 3.98 (d, J = 4.3 Hz, 1H), 3.66 (bs, 1H), 3.59-3.45 (m, 4H), 2.70 (t, J = 5.9 Hz, 2H), 2.53 (t, J = 7.4 Hz, 2H), 2.35- 2.30 (m, 2H), 0.98 (s, 3H) 0.89 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 209.6, 173.4, 136.8, 115.6, 77.6, 71.2, 42.2, 42.0, 39.4, 33.8, 27.7, 21.4, 20.4. Ci3H24Ni04 (M+H)+: 258.1705, found: 258.1699.
(R)-2,4-dihydroxy-3,3-dimethyl- V-[3-oxo-3-(pentylamino)propyl]butanamide ( V5Pan, 7)
Prepared as described for 6, starting from 4 (1.80 g, 5.65 mmol). Column chromatography (MeOH/CH2Cl2, 0: 1→1 :9)
Figure imgf000028_0003
afforded 7 (1.22 g, 75% yield) as a white solid. R/0.46 (MeOH/CH2Cl2, 1 :9). Mp 89.4 °C. [a] ∞ +29.7 (c 1.00, MeOH). IR (ATR) 3330, 3280, 3088, 2937, 2872, 1642, 1546, 1089, 1033,
691 cm-1. 1H NMR (CD3OD, 400 MHz): δ 3.88 (s, 1H), 3.54-3.37 (m, 4H), 3.17-3.13 (m, 2H), 2.41 (t, J = 6.7 Hz, 2H), 1.53-1.46 (m, 2H), 1.40-1.27 (m, 4H), 0.94-0.90 (m, 9H). 13C NMR (CDC13, 75 MHz): δ 174.1, 171.6, 77.5, 70.9, 39.8, 39.4, 35.9, 35.4, 29.2, 22.4, 21.4, 20.6, 14.1 HRMS (ESI) m/z calcd for Ci4H28N204Na (M+Na)+: 311.1947, found: 311.1933.
(R)- V-[3-(heptylamino)-3-oxopropyl]-2,4-dihydroxy-3,3-dimethylbutanamide ( V7Pan, 8)
Prepared as described for 6, starting from 5 (2.90 g, 8.13 i in H 10 mmol). Column chromatography (MeOH/CH2Cl2,
O H
0: 1→1 :9) afforded 8 (2.21 g, 86% yield) as a white solid. R/ 0.47 (MeOH/CH2Cl2, 1 :9). Mp 78.2 °C. [a]∞+26.9 (c 1.01, MeOH). IR (ATR) 3352, 2483, 2068, 1119, 973 cm-1. 1H NMR
(CD3OD, 400 MHz): δ 3.89 (s, 1H), 3.54-3.37 (m, 4H), 3.15 (dt, J = 1.2, 6.9 Hz, 2H), 2.41 (t, J= 6.7 Hz, 2H), 1.53-1.46 (m, 2H), 1.32-1.31 (m, 8H), 0.92-0.89 (m, 9H). 13C NMR (CD3OD, 75 MHz): δ 176.1, 173.6, 77.3, 70.4, 40.5, 40.4, 36.4, 32.9, 30.4, 30.1, 28.0, 23.7, 21.3, 20.9, 14.4. HRMS (ESI) m/z calcd for Ci6H33N204 (M+H)+: 317.2440, found: 317.2429. -2,2,5,5-tetramethyl- V-(3-oxo-5-phenylbutyl)-l,3-dioxane-4-carboxamide (9)
Figure imgf000029_0001
0: 1→1 : 1) afforded 9 (827 mg, 40% yield) as a colorless oil. R/ 0.69 (EtOAc). [<x]∞+41.1 (c I . I5. CH2CI2). IR (ATR) 3430, 2993, 2950, 2868, 2358, 1713, 1670, 1522, 1377, 1 196, 1095 cm"1. 1H NMR (CDCI3, 300 MHz): δ 7.35-7.17 (m, 5H), 6.88 (m, 1H), 4.02 (s, 1H), 3.68 (s, 2H), 3.65 (d, J = 11.7 Hz, 1H), 3.55-3.38 (m, 2H), 3.25 (d, J = 11.7 Hz, 1H), 2.73-2.69 (m, 2H), 1.45 (s, 3H), 1.40 (s, 3H), 1.01 (s, 3H), 0.88 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 207.4, 169.8, 133.8, 129.4, 128.9, 127.3, 99.1, 71.5, 50.3, 41.5, 33.3, 33.0, 29.5, 22.2, 18.9, 18.8. HRMS (ESI) m/z calcd for Ci9H28N04 (M +H)+, 334.2018; found, 334.2017. (R)-2,4-dihydroxy-3,3-dimethyl- V-(3-oxo-5-phenylbutyl)butanamide (RR6, 10)
Prepared as described for 6, starting from 9 (367 mg, 1.10 mmol). Column chromatography (EtO Ac/heptane, 0: 1— »1 :0)
Figure imgf000029_0002
afforded 10 (197 mg, 61% yield) as a colorless oil. Rf 0.30 (EtOAc). [o¾° +28.7 (c 1.17, CH2CI2). IR (ATR) 3372, 3058, 3036, 2960, 2868, 1710, 1643, 1530, 1496, 1453, 1366, 1287, 1076, 1041 cm \ 1H NMR (CDCI3, 300 MHz): δ 7.36-7.18 (m, 5H), 7.07 (m, 1H), 3.93 (s, 1H), 3.69 (s, 2H), 3.52-3.45 (m, 2H), 3.42 (m, 2H), 3.05 (br s, 1H), 2.73 (t, J = 5.6 Hz, 2H), 1.72 (br s, 1H), 0.95 (s, 3H), 0.83 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 207.9, 173.2, 133.7, 129.5, 129.0, 127.4, 77.6, 71.3, 50.3, 41.4, 39.4, 33.8, 21.5, 20.3. HRMS (ESI) m/z calcd for Ci6H24N04 (M+H)+, 294.1705; found 294.1694.
(R)-2,2,5,5-tetramethyl-/V-(3-oxo-5-phenylpentyl)-l,3-dioxane-4-carboxamide (11)
Prepared as described for compound 3, starting from 2 (1.03 g, 3.40 mmol) and phenethylmagnesium chloride (6.96 rnL of a 1.0
Figure imgf000030_0001
M solution in THF, 6.96 mmol, 2.05 equiv). After stirring for 2 h, the reaction mixrure was worked up to yield compound 11 (0.754 g, 64%) as a colorless oil. R/ 0.71 (EtOAc). [<x]∞ +37.7 (c 1.07, CH2C12). IR (ATR): 3426, 3334, 2989, 2952, 2870, 2362, 2332, 1712, 1671, 1521 , 1377, 1197, 1095 cm"1. 1H NMR (CDC13, 300 MHz): δ 7.30 - 6.94 (m, 5H), 6.94-6.90 (m, 1H), 4.05 (s, 1H), 3.67 (d, J = 11.7 Hz, 1H), 3.58-3.39 (m, 2H), 3.27 (d, J = 11.7 Hz, 1H), 2.90 (m, 2H), 2.76-2.70 (m, 2H), 2.65 (t, J = 6.0 Hz, 2H), 1.47 (s, 3H), 1.41 (s, 3H), 1.03 (s, 3H), 0.93 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 209.0, 169.9, 140.9, 128.7, 128.4, 126.3, 99.2, 77.3, 71.6, 44.5, 42.4, 33.4, 33.1, 29.8, 29.6, 28.5, 22.3, 19.0, 18.8. HRMS (ESI) m/z calcd for C20H30NO4 (M+H)+, 348.2175; found 348.2175.
(R)-2,4-dihydroxy-3,3-dimethyl-/V-(3-oxo-5-phenylpentyl)butanamide (RR7, 12)
Prepared as described for 6, starting from compound 11 (366 mg, 1.053 mmol), compound 12 (260 mg, 80%) was obtained
Figure imgf000030_0002
as a colorless oil. R/0.33 (EtOAc). [o¾° +28.6 (c 1.61, CH2C12). IR (ATR): 3354, 2933, 2859, 1708, 1644, 1529, 1452, 1369, 1287, 1075, 1041 cm"1. 1H NMR (CDC13, 300 MHz): δ 7.31- 7.15 (m, 5H), 7.09 (m, 1H), 3.96 (s, 1H), 3.58 (br s, 1H), 3.54-3.42 (m, 4H), 3.12 (br s, 1H), 2.92-2.87 (m, 2H), 2.78-2.72 (m, 2H), 2.66 (t, J= 5.8 Hz, 2H), 0.99 (s, 3H), 0.87 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 209.4, 173.1, 140.8, 128.7, 128.4, 126.4, 77.7, 71.3, 44.4, 42.3, 39.5, 33.9, 29.7, 21.6, 20.3. HRMS (ESI) m/z calcd for Ci7H26N04 (M+H)+, 308.1861; found 308.1851.
(R)-2,2,5,5-tetramethyl-N-(3-oxo-6-phenylhexyl-l,3-dioxane-4-carboxamide (13) Three drops of 1 ,2-dibromoethane were added to a suspension of magnesium turnings (1.00 g, 41.2 mmol, 12.0 equiv) in
Figure imgf000031_0001
diethyl ether (20 mL) and the misture was briefly heated. Next, a solution of (3-bromopropyl)benzene (3.08 mL, 20.2 mmol, 6.0 equiv) in diethyl ether (20 mL) was added at such a rate that the mixture kept refluxing, after which the clouded solution was stirred for 2 h. The resulting Grignard reagent was added dropwise to a cooled (0 °C) solution of compound 2 (1.02 g, 3.37 mmol) in THF (20 mL) and stirred at 0 °C for 3.5 h. The reaction mixture was quenched with saturated aqueous NH4C1 (100 mL), extracted with dichloromethane (3 x 70 mL), dried over Na2S04 and concentrated. The crude product was purified by flash column chromatography (heptane/EtOAc, 1 :0→ 1 : 1), yielding compound 13 (0.95 g, 78%) as a colorless oil. R/0 0 (EtOAc), [<x]∞ +34.4 (c 1.23, CH2C12). IR (ATR):
3421 , 2997, 2947, 2872, 1710, 1672, 1522, 1454, 1377, 1260, 1222, 1 197, 1 159, 1096 cm"1. 1H NMR (CDCls, 300 MHz): δ 7.31-7.14 (m, 5H), 6.92 (m, 1H), 4.05 (s, 1H), 3.67 (d, J = 1 1.7 Hz, 1H), 3.56-3.41 (m, 2H), 3.27 (d, J = 1 1.7 Hz, 1H), 2.64 (t, J = 6.1 Hz, 2H), 2.61 (t, J = 6.0 Hz, 2H), 2.41 (t, J = 7.3 Hz, 2H), 1.91 (quintet, J = 7.7 Hz, 2H), 1.45 (s, 3H), 1.40 (s, 3H), 1.03 (s, 3H), 0.94 (s, 3H). 13C NMR (CDC13, 75 MHz): δ 209.8, 169.9, 141.5, 128.6, 126.2, 99.2, 77.3, 71.6, 42.2, 42.1 , 35.1 , 33.5, 33.1 29.6, 25.3, 22.3, 19.0, 18.8. HRMS (ESI) m/z calcd for C2iH32N04 (M+H)+, 362.2331 ; found 362.2352. (R)-2,4-dihydroxy-3,3-dimethyl- V-(3-oxo-6-phenylhexyl)butanamide (RR8, 14)
Prepared as described for 6, starting from compound 13 (380 mg, 1.05 mmol), compound 14 (268 mg, 79%) was
Figure imgf000031_0002
obtained as a colorless oil. R/ 0.36 (EtOAc). [<x]∞ +26.6 (c 1.16, CH2C12). IR (ATR): 3358,
3028, 2946, 2868, 1708, 1643, 1530, 1453, 1368, 1269 cm"1. 1H NMR (CDC13, 300 MHz): δ 7.13-7.1 1 (m, 5H), 3.98 (s, 1H), 3.54-3.43 (m, 5H), 3.04 (br s, 1H), 3.66-3.59 (m, 4H), 2.42 (t, J = 7.4 Hz, 2H), 1.91 (quintet, J = 7.5 Hz, 2H), 1.00 (s, 3H), 0.88 (s, 3H). 13C NMR (CDCI3, 75 MHz): δ 210.2, 173.1 , 141.5, 128.6, 126.2, 77.7, 71.3, 42.2, 42.1 , 39.5, 35.1 , 33.9, 25.2, 21.5, 20.3. HRMS (ESI) m/z calcd for Ci8H28N04 (M+H)+, 322.2018; found 322.2009.
Example 2: antibiotic and pantetheinase inhibitory activity of the substances and composition of the invention Pantheteinase activity
Pantetheinase activity was measured by the amount of free aminomethylcoumarin (AMC) released by the hydrolysis of the pantetheine-analogue pantothenate- AMC. Pantothenate- AMC was incubated in phosphate buffer (100 mM potassium phosphate buffer pH 8.0) in the presence of serum or plasma as pantetheinase source with or without a potential pantetheinase inhibitor. In time, samples were taken and the reaction was terminated by addition of 100 mM CaC03 pH 10.5. Fluorescence was measured using a luminescence spectrometer (LS55, Perkin Elmer, EX 350 ± 2.5 nm, EM 450 ± 2.5 nm) against samples without serum or plasma as negative control.
Mass spectrometric analysis
The pantothenamide N7Pan was incubated in phosphate buffer (500 μΜ potassium phosphate buffer pH 8.0) for 24 hours at 37 °C with or without potential pantetheinase inhibitors in the presence of 1% human or fetal bovine serum (FBS, HyClone, Logan, UT) as pantetheinase source. Prior to analysis samples were diluted 100-fold in methanol (HPLC grade, Fisher Scientific). Mass spectrometry was performed on a JEOL JMS-T100CS (AccuTOF CS) connected to a Agilent 1100 series HPLC system. Analysis was performed in infusion mode, 3 μΐ of sample was injected into a stream of methanol (HPLC grade, Fisher Scientific) containing 0.1% formic acid (puriss. Pa for mass spectrometry, Fluka) and 50 nanomolar PPG425 (poly[propylene glycol] average M.W. 425, Sigma-Aldrich Chemie GmbH) for use as an internal mass drift compensator. Total analysis time with a flow rate of ΙΟΟμΙ/min was 2.5 minutes per sample. Sample information elutes between 0.3 and 1.0 minutes. Data between 0 and 0.3 minutes was used to mass drift compensate the calibration against PPG425 peaks resulting in a mass precision better than 5 ppm.
Bacterial growth assays
Bacterial strains of Escherichia coli BL21(DE3) and Staphylococcus aureus (ATCC 6538) were grown overnight in 1% Bacto Tryptone media (BD, Sparks, MD). Cultures were then diluted 1 : 1000 in fresh assay media and added to 96 well sterile ELISA plate (100 ul per well). The compounds to be tested were diluted in the same media and added to the bacteria (100 ul per well). Plates were incubated at 37 °C and bacterial growth was followed in time by reading the optical density at 490 nm using a microplate-reader (Model 450, Bio-Rad Laboratories Inc., Hercules, CA). A bacterial strain of a bio luminescent Staphylococcus aureus (Xen8.1) was cultured in Mueller Hinton medium as described above in a 96-well plate with varying concentrations of the pantothenamide N7Pan and the pantetheinase inhibitor R 6, as indicated. Bio luminescence was recorded by IVIS optical imaging technology (Caliper Life Sciences). The emitted light is proportional to the number of bacteria in the wells.
In vivo administration of pantetheinase inhibitors
Adult C57BL/6 mice (Harlan, IN) received RR2 dissolved in PBS orally or intraperitoneally. In each group, two mice received approximately 20 mg per kg and 3 mice received approximately 100 mg per kg. In time, 50 μΐ blood samples were collected using a tail cut and immediately mixed with 10 μΐ EDTA (50 mM in PBS) to obtain plasma. After centrifugation (10 min 3000 rpm), pantetheinase activity was measured in plasma according to the method described above. Plasma pantetheinase activity was plotted as a fraction of the activity at t=0 (100% activity).
Adult Wistar rats (Harlan, IN) received R 6 dissolved in PBS orally. Dosing was 2,
10 and 50 mg/kg, 3 rats per group. At several time points, approximately 200 microliter blood samples were collected using a tail cut and immediately mixed with 10 μΐ EDTA (50 mM in PBS) to obtain plasma. After centrifugation (10 min 3000 rpm), pantetheinase activity was measured in plasma according to the method described above. Plasma pantetheinase activity was plotted as a fraction of the activity at t=0 (100% activity).
Pantothenamide antibiotics are inactive in the presence of serum
Pantothenamides, of which N5Pan and N7Pan are the prototypes, are active against gram-negative and gram-positive bacteria. Pantothenamides were found to be substrates of the key rate-controlling enzyme pantothenate kinase, and the rate of conversion of N5Pan was found to be more rapid than that of pantothenate itself. This led to the conclusion that the mechanism for toxicity towards E.coli is due to the formation of Co A analogs that lead to the transfer of an inactive 4'-phosphopantothenamide moiety to acyl carrier protein (ACP) which is the first step in the bacterial type II route of fatty acid synthesis (Zhang et al).
In this study it was discovered that pantothenamides as such cannot be used as a viable antibiotic strategy in mammals because these compounds are hydrolyzed in serum. Incubation of a prototypic pantothenamide N7Pan in serum readily generated the pantothenate and heptylamine degradation products as determined by mass spectrometry (see figures la and 2b). Both pantetheinase inhibitors RR2 (figure la-b) and R 6 (figure 2a-c) inhibited the serum-mediated breakdown of N7Pan. In this case N7Pan was chosen rather than N5Pan for reasons of easy detection of the free amide.
As a result of pantothenamide breakdown, the antibiotic activity is lost. Figure 3 shows that N5Pan is active as an antibiotic against E.coli in 1% tryptone medium but addition of 10% serum completely abolishes antibiotic activity.
Inhibition of pantetheinase activity preserves antibiotic activity of pantothenamides
Inhibition of serum pantetheinase activity could prevent breakdown of pantothenamides, and could preserve their antibiotic activity under physiological conditions in mammals.
Several of our pantetheine analogues that could not be hydrolyzed by pantetheinase, such as RR2 and RR6, inhibited serum pantetheinase activity, with an IC50 of approximately 50 μΜ (RR2) to 0.5 μΜ (RR6) as measured with a fluorogenic substrate (developed in collaboration with Dr F.Pinnen, Universita G. D'Annunzio, Chieti, Italy) represented by the structure:
Figure imgf000034_0001
Figure 4 shows the potent inhibition of pantheteinase activity by the pantothenones RR2 and RR6. The triazole analogue of pantetheine (denominated RR1), (R)-2,4-dihydroxy-N-((l-(2- hydroxyethyl)-lH-l,2,3-triazol-4-yl)methyl)-3,3-dimethylbutanamide) was found to be a poor pantetheinase inhibitor (IC50 > 1 mM).
RR2 was then used to study the effect on the antimicrobial activity of N5Pan or N7Pan in vitro, using these compounds in the presence of human serum, in a 96-well plate format.
It was found that growth of E.coli, S.aureus and S.pyogenes was inhibited by a combination of pantothenamides (20-200 μΜ) and RR2 (200-1000 μΜ) even in the presence of 10% serum. Figure 5a-b shows an example of E.coli and S.aureus growth inhibition by combining RR2 with N5Pan or N7Pan, in a medium consisting of 1% tryptone and 10% complement-inactivated human serum.
This experiment provided proof of concept that the combination of an antimicrobial pantothenamide with an inhibitor of host (mammalian) pantetheinase activity, could be a novel antibiotic strategy. As shown in figure 5, RR2 itself is not antibiotic. This may be caused by the loss of the amide bond which is likely to be a necessary structural requirement for entering the first step of CoA synthesis leading to CoA antimetabolites, as recently suggested in a study by Mercer et al.
A second example (figure 6) serves to illustrate the effect of another pantetheinase inhibitor (RR6) to increase the potency of a pantothenamide antibiotic (N7Pan). Figure 6 shows the effect of concentration ranges of RR6 and N7Pan on growth of a bio luminescent S.aureus strain (Xen 8.1). From this experiment it can be concluded that inhibition of pantetheinase activity will increase the potency of N7Pan, in the presence of serum, 30-60 fold, thereby bringing the MIC value in the range of currently used antibiotics.
Effect of pantetheinase inhibitors on mammalian cells in vitro and in vivo
As the putative mode of action of pantothenamides is at the level of CoA-dependent biochemical pathways, this raised the question of selectivity for microbial versus host targets. Using high concentrations of RR2 and N5Pan, which were inhibitory for E.coli, no toxicity or significant inhibition of cell growth in the human kidney cell line 293T nor in primary epidermal keratinocytes was observed (data not shown). Although the proposed mechanism of pantothenamides is at the level of bacterial fatty acid synthesis, which would also offer a good explanation for their selectivity, other mechanisms should be considered12.
To examine its potential for in vivo inhibition of pantetheinase activity a pilot study was performed by administration of RR2 (20 or 100 mg/kg) to mice, either orally or intraperitoneally to monitor the kinetics of inhibition of pantetheinase activity in blood. These doses of RR2 were well tolerated by the mice and the highest dose caused a significant drop of plasma panthetheinase activity (figure 7a).
An even better pharmacodynamic profile was observed in rats as shown in figure 7b. A single oral administration of 50 mg/kg caused a 100% drop of plasma pantetheinase activity up to 12 hours.
Description of the Figures Figure 1 (a+b): mass spectra of RR2 and N7Pan following incubation in 1% bovine serum. Figure 1(a) shows that N7Pan is completely degraded as witnessed by the absence of the peaks at 339 and 355, and the appearance of the heptylamine peak at 116. Figure 1(b) shows that addition of RR2 prevents breakdown of N7Pan as witnessed by the presence of peaks at 339 and 355, and the smaller heptylamine peak.
Figure 2a-c:mass spectra of R 6 and N7Pan following incubation in 1% human serum. Figure 2a: spectrum of N7Pan (m/z of the parent compound: 339). Figure 2b; spectrum of N7Pan following incubation with human serum resulting in the appearance of the heptylamine hydrolysis product (m/z: 116). Figure 2c: Spectrum of N7Pan incubated with human serum in the presence of pantetheinase inhibitor R 6. Degradation of N7Pan is prevented as witnessed by the absence of the heptylamine peak. The R 6 peak is present at m/z = 316.
Figure 3: Addition of serum abolishes the antibiotic effect of N5Pan on E.coli growth. Figure 4: Inhibition of plasma pantetheinase activity by compounds R 1, R 2 and R 6.
Figure 5 (a+b): Graphs showing antibacterial activity of N5Pan and RR2 on E.coli in the presence of human serum (5a); the antibacterial activity of N7Pan and RR2 on S. aureus in the presence of human serum (5b).
Figure 6: Antibacterial activity of N7Pan and R 6 against a bio luminescent S. aureus strain. Wells with bacterial growth are dark to light grey here (e.g. right hand column); wells without growth are transparent (e.g. left hand column). As shown in the upper row, at the highest concentration of the pantetheinase inhibitor R 6 (128 μg/ml), a concentration of 0.5 μg/ml N7Pan is sufficient to cause complete inhibition of growth (well marked with asterisk)
Figure 7a+b: Graphs showing decrease of plasma pantetheinase activity in vivo following administration of RR2 to mice (7a) or RR6 to rats (7b). On the Y-axis: the plasma pantetheinase activity, on the X-axis: time in minutes. A dose of 50 mg/kg in rats causes complete inhibition of plasma pantetehinase activity, which lasts up to 12 hours.
References
1. Clifton G, Bryant SR, Skinner CG (1970). N'-(substituted) pantothenamides,
antimetabolites of pantothenic acid. Arch Biochem Biophys 137: 523-528.
2. Choudhry AE, Mandichak TL, Broskey JP, Egolf RW, Kinsland C, Begley TP,
Seefeld MA, Ku TW, Brown JR, Zalacain M, Ratnam K (2003). Inhibitors of pantothenate kinase: novel antibiotics for staphylococcal infections. Antimicrob Agents Chemother 47: 2051-2055.
Mercer AC, Meier JL, Hur GH, Smith AR, Burkart MD (2008). Antibiotic evaluation and in vivo analysis of alkynyl Coenzyme A antimetabolites in Escherichia coli. Bioorg Med Chem Lett 18: 5991-5994.
Virga KG, Zhang YM, Leonardi R, Ivey RA, Hevener K, Park HW, Jackowski S, Rock CO, Lee RE (2006). Structure-activity relationships and enzyme inhibition of pantothenamide-type pantothenate kinase inhibitors. Bioorg Med Chem 14: 1007- 1020.
Zhang YM, Frank MW, Virga KG, Lee RE, Rock CO, Jackowski S (2004). Acyl carrier protein is a cellular target for the antibacterial action of the pantothenamide class of pantothenate antimetabolites. J Biol Chem 279: 50969-50975.
Fischbach MA, Walsh CT (2009). Antibiotics for emerging pathogens. Science 325: 1089-1093.
Maras B, Barra D, Dupre S, Pitari G (1999). Is pantetheinase the actual identity of mouse and human vanin-1 proteins? FEBS Lett 461 : 149-152.
Martin F, Malergue F, Pitari G, Philippe JM, Philips S, Chabret C, Granjeaud S, Mattei MG, Mungall AJ, Naquet P, Galland F (2001). Vanin genes are clustered (human 6q22-24 and mouse 10A2B1) and encode isoforms of pantetheinase ectoenzymes. Immunogenetics 53: 296-306.
Jansen PA, kamsteeg M, Rodijk-Olthuis D, van Vlijmen-Willems IM, de Jongh GJ, Bergers M, Tjabringa GS, Zeeuwen PL, Schalkwijk J (2009). Expression of the vanin gene family in normal and inflamed human skin: induction by proinflammatory cytokines. J Invest Dermatol 129: 2167-2174.
Ruan BH, Cole DC, Wu P, Quazi A, Page K, Wright JF, Huang N, Stock JR, Nocka K, Aulabaugh A, Krykbaev R, Fitz LJ, Wolfman NM, Fleming ML (2009). A fluorescent assay suitable for inhibitor screening and vanin tissue quantification. Anal Biochem.

Claims

Claims
1. Pharmaceutical or veterinary composition comprising:
one or more antibiotic pantothenamide derivatives; and
one or more pantetheinase inhibitors.
2. Pharmaceutical or veterinary composition according to claim 1, wherein the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (I) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000038_0001
wherein
Pv1 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide; Pv2 and R3 independently represent a hydrogen atom or a radical selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
X1 and X2 independently represent hydrogen or a group selected from hydroxyl, thiol, cyanide, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, sulfonamide, amide, pyrazole or imidazole; and
X3 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide.
3. Pharmaceutical or veterinary composition according to claim 2, wherein the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (la) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000039_0001
wherein R1 and X3 have the same meaning as defined in claim 2.
4. Pharmaceutical or veterinary composition according to claim 3, wherein the antibiotic pantothenamide derivative is selected from the group of derivatives represented by formula (lb) and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000039_0002
wherein R1 represents a group selected from alkyl, alkenyl and alkynyl.
5. Pharmaceutical or veterinary composition according to any one of the preceding claims, wherein the pantetheinase inhibitor is selected from the group consisting of VNN antagonistic agents, including VNN-1 antagonistic agents, VNN-2 antagonistic agents and VNN-3 antagonistic agents.
6. Pharmaceutical or veterinary composition according to any one of the preceding claims, wherein the pantetheinase inhibitors is selected from the group consisting of competitive enzyme inhibitors, reactive enzyme substrates capable of covalently modifying a residue of the enzyme catalytic site, antibodies capable of specifically interacting with VNN and inhibiting its activity, nucleic acids blocking transcription of VNN gene and/or translation of VNN mRNA.
7. Pharmaceutical or veterinary composition according to any one of the preceding claims wherein the pantetheinase inhibitors is selected from the group of non-hydro lysable pantothenic acid analogues and reactive enzyme substrates capable of covalently or non- covalently binding a residue of the enzyme catalytic site.
8. Pharmaceutical or veterinary composition according to any one of the preceding claims, wherein the pantetheinase inhibitor is selected from the group of substances represented by formula Ila, Ilia or IV, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000040_0001
wherein
R4, R5 and R6 independently represent hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
R7 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide; X4 and X5 independently represent hydrogen , hydroxyl, thiol, cyanide, halogen, or a groups selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, sulfonamide, amide, pyrazole and imidazole;
X6 represents sulfur, oxygen or nitrogen, said nitrogen optionally being substituted by a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; and m and n are integers within the range of 0-6, wherein m is not 0.
9. Pharmaceutical or veterinary composition according to claim 8, wherein m is an integer within the range of 1-6.
10. Pharmaceutical or veterinary composition according to claim 8 or 9, wherein the one or more pantetheinase inhibitors are selected from substances represented by formula lie, and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000041_0001
11. Pharmaceutical or veterinary composition according to claim 1, comprising a combination of N5Pan and RR2; a combination of N7Pan and RR2; a combination of N5Pan, N7Pan and RR2; a combination of N5Pan and RR6; a combination of N7Pan and RR6; or a combination of N5Pan, N7Pan and RR6; or pharmaceutically acceptable salts, esters, amides, and prodrugs of any of the respective compounds.
12. Substance represented by any one of the formulas (Ila) (Ilia), and (IV) or a pharmaceutically acceptable salt, ester, amide, or prodrug thereof, wherein:
R4, R5 and R6 independently represent hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
R7 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted with hydroxyl, thiol, halogen and/or cyanide; X4 and X5 independently represent hydrogen, hydroxyl, thiol, cyanide, halogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, sulfonamide, amide, pyrazole and imidazole; X6 represents sulfur, oxygen, carbon or nitrogen, said nitrogen optionally being substituted by a radical selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;; and
m and n are integers within the range of 0-6, wherein m is not 0.
13. Substance according to claim 12, wherein m is an integer within the range of 1-6.
14. Substance according to any one of claims 12 or 13 selected from RR2, RR6, RR7, RR8 and pharmaceutically acceptable salts, esters, amides, and prodrugs thereof:
Figure imgf000042_0001
Figure imgf000042_0002
15. Substance represented by any one of the formulas (I), (la) and (lb) or a
pharmaceutically acceptable salt, ester, amide, or prodrug thereof, wherein: R1 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide;
R2 and R3 independently represent hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl;
X1 and X2 independently represent hydrogen , hydroxyl, thiol, cyanide, halogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, sulfonamide, amide, pyrazole or imidazole; and
X3 represents hydrogen or a group selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each optionally substituted by hydroxyl, thiol, halogen and/or cyanide.
16. Composition according to claims 1-11 or a substance according to claims 12-15, for use in a therapeutic method of treating and/or preventing a disease or condition selected from bacterial infections; fungal infections; yeast infections and intracellular protozoan infections; in a human or animal subject in need thereof.
17. Method of treating and/or preventing a disease or condition selected from bacterial infections; fungal infections; yeast infections and intracellular protozoan infections; in a human or animal subject in need thereof, said method comprising administering to said subject an effective amount of a pharmaceutical composition as defined in any one of claims 1-11 or a substance as defined in claims 12-15.
PCT/NL2011/050384 2010-05-31 2011-05-31 Pantothenic acid derivatives and their use in the treatment of microbial infections WO2011152720A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP10164455.7 2010-05-31
EP10164455 2010-05-31
EP10171435.0 2010-07-30
EP10171435 2010-07-30

Publications (1)

Publication Number Publication Date
WO2011152720A1 true WO2011152720A1 (en) 2011-12-08

Family

ID=44202147

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/NL2011/050385 WO2011152721A1 (en) 2010-05-31 2011-05-31 Derivatives of pantothenic acid and their use for the treatment of malaria
PCT/NL2011/050384 WO2011152720A1 (en) 2010-05-31 2011-05-31 Pantothenic acid derivatives and their use in the treatment of microbial infections

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/NL2011/050385 WO2011152721A1 (en) 2010-05-31 2011-05-31 Derivatives of pantothenic acid and their use for the treatment of malaria

Country Status (1)

Country Link
WO (2) WO2011152721A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014048547A1 (en) * 2012-09-25 2014-04-03 Merck Patent Gmbh Alpha hydroxy amides
WO2016072854A2 (en) 2014-11-06 2016-05-12 Radboud Universitair Medisch Centrum Pantothenamide analogues
EP3674288A1 (en) 2018-12-31 2020-07-01 MMV Medicines for Malaria Venture Pantothenamide analogues
WO2023112440A1 (en) * 2021-12-17 2023-06-22 学校法人大阪医科薬科大学 Vanin-1 inhibitor

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014080309A2 (en) * 2012-11-21 2014-05-30 Mahesh Kandula Compositions and methods for the treatment of inflammation and lipid disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000035298A1 (en) 1996-11-27 2000-06-22 Wm. Wrigley Jr. Company Chewing gum containing medicament active agents
US20040247524A1 (en) 2003-06-05 2004-12-09 Philippe Naquet Method of treatment of an inflammatory disorder with a Vanin-1 antagonist

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000035298A1 (en) 1996-11-27 2000-06-22 Wm. Wrigley Jr. Company Chewing gum containing medicament active agents
US20040247524A1 (en) 2003-06-05 2004-12-09 Philippe Naquet Method of treatment of an inflammatory disorder with a Vanin-1 antagonist

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
BERGE ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
CHOUDHRY AE, MANDICHAK TL, BROSKEY JP, EGOLFRW, KINSLAND C, BEGLEY TP, SEEFELD MA, KU TW, BROWN JR, ZALACAIN M: "Inhibitors of pantothenate kinase: novel antibiotics for staphylococcal infections", ANTIMICROB AGENTS CHEMOTHER, vol. 47, 2003, pages 2051 - 2055, XP055271027, DOI: doi:10.1128/AAC.47.6.2051-2055.2003
CLIFTON G, BRYANT SR, SKINNER CG: "N'-(substituted) pantothenamides, antimetabolites of pantothenic acid", ARCH BIOCHEM BIOPHYS, vol. 137, 1970, pages 523 - 528, XP024807120, DOI: doi:10.1016/0003-9861(70)90470-4
EDWARD B. ROCHE: "Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
FINNIN, MORGAN, J PHARM SCI, vol. 88, no. 10, October 1999 (1999-10-01), pages 955 - 958
FISCHBACH MA, WALSH CT: "Antibiotics for emerging pathogens", SCIENCE, vol. 325, 2009, pages 1089 - 1093
H. LIEBERMAN, L. LACHMAN, PHARMACEUTICAL DOSAGE FORMS: TABLETS, vol. 1, 1980
JANSEN PA, KAMSTEEG M, RODIJK-OLTHUIS D, VAN VLIJMEN-WILLEMS IM, JONGH GJ, BERGERS M, TJABRINGA GS, ZEEUWEN PL, SCHALKWIJK J: "Expression of the vanin gene family in normal and inflamed human skin: induction by proinflammatory cytokines", J INVEST DERMATOL, vol. 129, 2009, pages 2167 - 2174
LEONARDI R ET AL: "A pantothenate kinase from Staphylococcus aureus refractory to feedback regulation by coenzyme A", JOURNAL OF BIOLOGICAL CHEMISTRY, THE AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, INC., BALTIMORE, MD, US, vol. 280, no. 5, 4 February 2005 (2005-02-04), pages 3314 - 3322, XP003002291, ISSN: 0021-9258, DOI: DOI:10.1074/JBC.M411608200 *
LIANG, CHEN, EXPERT OPINION IN THERAPEUTIC PATENTS, vol. 1 1, no. 6, 2001, pages 981 - 986
MARAS B, BARRA D, DUPRE S, PITARI G: "Is pantetheinase the actual identity of mouse and human vanin-1 proteins?", FEBS LETT, vol. 461, 1999, pages 149 - 152, XP004260539, DOI: doi:10.1016/S0014-5793(99)01439-8
MARTIN F, MALERGUE F, PITARI G, PHILIPPE JM, PHILIPS S, CHABRET C, GRANJEAUD S, MATTEI MG, MUNGALL AJ, NAQUET P: "Vanin genes are clustered (human 6q22-24 and mouse 10A2B 1) and encode isoforms of pantetheinase ectoenzymes", IMMUNOGENETICS, vol. 53, 2001, pages 296 - 306
MERCER AC, MEIER JL, HUR GH, SMITH AR, BURKART MD: "Antibiotic evaluation and in vivo analysis of alkynyl Coenzyme A antimetabolites in Escherichia coli", BIOORG MED CHEM LETT, vol. 18, 2008, pages 5991 - 5994, XP025627188, DOI: doi:10.1016/j.bmcl.2008.07.078
RUAN BH, COLE DC, WU P, QUAZI A, PAGE K, WRIGHT JF, HUANG N, STOCK JR, NOCKA K, AULABAUGH A: "A fluorescent assay suitable for inhibitor screening and vanin tissue quantification", ANAL BIOCHEM., 2009
SPRY CHRISTINA ET AL: "Coenzyme A biosynthesis: an antimicrobial drug target.", FEMS MICROBIOLOGY REVIEWS JAN 2008 LNKD- PUBMED:18173393, vol. 32, no. 1, January 2008 (2008-01-01), pages 56 - 106, XP002605325, ISSN: 0168-6445 *
T. HIGUCHI, V. STELLA: "A.C.S. Symposium Series", vol. 14, article "Pro-drugs as Novel Delivery Systems"
VERMA ET AL., PHARMACEUTICAL TECHNOLOGY ON-LINE, vol. 25, no. 2, 2001, pages 1 - 14
VIRGA KG, ZHANG YM, LEONARDI R, IVEY RA, HEVENER K, PARK HW, JACKOWSKI S, ROCK CO, LEE RE: "Structure-activity relationships and enzyme inhibition of pantothenamide-type pantothenate kinase inhibitors", BIOORG MED CHEM, vol. 14, 2006, pages 1007 - 1020, XP025132982, DOI: doi:10.1016/j.bmc.2005.09.021
ZHANG YM, FRANK MW, VIRGA KG, LEE RE, ROCK CO, JACKOWSKI S: "Acyl carrier protein is a cellular target for the antibacterial action of the pantothenamide class of pantothenate antimetabolites", J BIOL CHEM, vol. 279, 2004, pages 50969 - 50975, XP002605324, DOI: doi:10.1074/JBC.M409607200
ZHANG YONG-MEI ET AL: "Acyl carrier protein is a cellular target for the antibacterial action of the pantothenamide class of pantothenate antimetabolites.", THE JOURNAL OF BIOLOGICAL CHEMISTRY 3 DEC 2004 LNKD- PUBMED:15459190, vol. 279, no. 49, 3 December 2004 (2004-12-03), pages 50969 - 50975, XP002605324, ISSN: 0021-9258 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014048547A1 (en) * 2012-09-25 2014-04-03 Merck Patent Gmbh Alpha hydroxy amides
JP2015535832A (en) * 2012-09-25 2015-12-17 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Alpha hydroxyamide
US9585891B2 (en) 2012-09-25 2017-03-07 Merck Patent Gmbh Alpha hydroxy amides
AU2013324696B2 (en) * 2012-09-25 2017-09-14 Merck Patent Gmbh Alpha hydroxy amides
WO2016072854A2 (en) 2014-11-06 2016-05-12 Radboud Universitair Medisch Centrum Pantothenamide analogues
WO2016072854A3 (en) * 2014-11-06 2016-07-07 Radboud Universitair Medisch Centrum Pantothenamide analogues
EP3674288A1 (en) 2018-12-31 2020-07-01 MMV Medicines for Malaria Venture Pantothenamide analogues
WO2020141155A1 (en) 2018-12-31 2020-07-09 Mmv Medicines For Malaria Venture Pantothenamide analogues
CN113286778A (en) * 2018-12-31 2021-08-20 Mmv疟疾药物投资公司 Panamide analogs
US11572336B2 (en) 2018-12-31 2023-02-07 Mmv Medicines For Malaria Venture Pantothenamide analogues
CN113286778B (en) * 2018-12-31 2023-09-22 Mmv疟疾药物投资公司 Ubiquitin analogs
US11834391B2 (en) 2018-12-31 2023-12-05 Mmv Medicines For Malaria Venture Pantothenamide analogues
WO2023112440A1 (en) * 2021-12-17 2023-06-22 学校法人大阪医科薬科大学 Vanin-1 inhibitor
JP7506415B2 (en) 2021-12-17 2024-06-26 学校法人大阪医科薬科大学 Vanin-1 inhibitors

Also Published As

Publication number Publication date
WO2011152721A1 (en) 2011-12-08

Similar Documents

Publication Publication Date Title
WO2011152720A1 (en) Pantothenic acid derivatives and their use in the treatment of microbial infections
US6946474B2 (en) Nitrogen-containing compounds and their use as glycine transport inhibitors
JP2008508287A (en) Inhibitors of 5&#39;-methylthioadenosine phosphorylase and 5&#39;-methylthioadenosine / S-adenosylhomocysteine nucleosidase
US10829440B2 (en) Antibacterial compounds and methods of making and using same
JP2017533187A (en) Non-β-lactam antibiotics
US12060330B2 (en) Therapeutic indazoles
JP2014513670A (en) Use of polyaminoisoprenyl derivatives in antibiotic or preservative treatment
EP3215496B1 (en) Pantothenamide analogues
US11834391B2 (en) Pantothenamide analogues
EP3126333B1 (en) Pqsr modulators
KR101900574B1 (en) Novel N-hydroxybenzamide and Use Thereof
KR101900575B1 (en) Novel Hydroxamic Acids and Uses Thereof
US11491146B2 (en) Therapeutic methods and combinations
DK2285802T3 (en) Oxo-imidazolyl compounds with antibacterial activity
WO2023039027A1 (en) Colibactin derivatives and methods of treating, ameliorating, and/or preventing cancer
JP2004508312A (en) Substituted hydrazine derivatives
WO2018162670A1 (en) 2- or 3-imidazolines as carbapenemases inhibitors
WO2008105392A1 (en) 3-amino-2-indolylbutyrolactam derivative, process for production of the same, and pharmaceutical agent comprising the same as active ingredient
Rutjes et al. Combination of Pantothenamides with

Legal Events

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

Ref document number: 11725211

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11725211

Country of ref document: EP

Kind code of ref document: A1