WO2012109458A1 - Flavin derivatives - Google Patents

Flavin derivatives Download PDF

Info

Publication number
WO2012109458A1
WO2012109458A1 PCT/US2012/024507 US2012024507W WO2012109458A1 WO 2012109458 A1 WO2012109458 A1 WO 2012109458A1 US 2012024507 W US2012024507 W US 2012024507W WO 2012109458 A1 WO2012109458 A1 WO 2012109458A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
infection
methyl
compound
formula
Prior art date
Application number
PCT/US2012/024507
Other languages
French (fr)
Inventor
Philip D.G. Coish
Paul Adrian Aristoff
Brian R. Dixon
Original Assignee
Biorelix, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2011/000617 external-priority patent/WO2011126567A1/en
Application filed by Biorelix, Inc. filed Critical Biorelix, Inc.
Publication of WO2012109458A1 publication Critical patent/WO2012109458A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N43/00Biocides, pest repellants or attractants, or plant growth regulators containing heterocyclic compounds
    • A01N43/90Biocides, pest repellants or attractants, or plant growth regulators containing heterocyclic compounds having two or more relevant hetero rings, condensed among themselves or with a common carbocyclic ring system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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 relates to novel flavin derivatives, composition comprising the same, and their use and compositions for use as riboswitch ligands and/or anti-infectives.
  • RNA structures termed riboswitches regulate the expression of various genes crucial for survival or virulence.
  • members of each known class of riboswitch can fold into a distinct, three-dimensionally structured receptor that recognizes a specific organic metabolite.
  • the riboswitch receptor binds to the metabolite and induces a structural change in the nascent mRNA that prevents expression of the open reading frame (ORF), thereby altering gene expression.
  • ORF open reading frame
  • the riboswitch folds into a structure that does not interfere with the expression of the ORF.
  • Riboswitch motifs have been identified that bind to thiamine pyrophosphate (TPP), flavin mononucleotide (FMN), glycine, guanine, 3'-5'-cyclic eiguanylic acid (c-di-GMP), molybdenum cofactor, glucosamine-6-phosphate (GlcN6P), lysine, adenine, and adocobalamin (AdoCbl) riboswitches.
  • TPP thiamine pyrophosphate
  • FMN flavin mononucleotide
  • c-di-GMP 3'-5'-cyclic eiguanylic acid
  • GlcN6P glucosamine-6-phosphate
  • AdoCbl adocobalamin
  • riboswitch motifs have been identified that recognize S-adenosylmethionine (SAM) I, II and III, IV and two distinct motifs that recognize pre-queosine- 1 (PreQl).
  • SAM S-adenosylmethionine
  • PreQl pre-queosine- 1
  • PTPP pyrithiamine pyrophosphate
  • AEC L- aminoethylcysteine
  • DL-4-oxalysine which bind to lysine riboswitches and roseoflavin and FMN which bind to FMN riboswitches.
  • the riboswitch-receptors bind to their respective ligands in an interface that approaches the level of complexity and selectivity of proteins. This highly specific interaction allows riboswitches to discriminate against most intimately related analogs of ligands.
  • the receptor of a guanine- binding riboswitch from Bacillus subtilis forms a three-dimensional structure such that the ligand is almost completely enveloped.
  • the guanine is positioned between two aromatic bases and each polar functional group of the guanine hydrogen bonds with four additional riboswitch nucleotides surrounding it. This level of specificity allows the riboswitch to discriminate against most closely related purine analogs.
  • SAM- binding riboswitches comprise one subdomain that recognizes every polar functional group of the 4-amino-5-hydroxymethyl-2- methylpyrimidine (HMP) moiety, albeit not the thiazole moiety, and another subdomain that coordinates two metal ions and several water molecules to bind the negatively charged pyrophosphate moiety of the ligand.
  • HMP 4-amino-5-hydroxymethyl-2- methylpyrimidine
  • FMN riboswitches Similar to TPP, guanine and SAM riboswitches, FMN riboswitches form receptor structures that are highly specific for the natural metabolite FMN. It is by this highly specific interaction that allows for the design of small molecules for the regulation of specific genes.
  • FMN riboswitches are of particular interest of this invention because it is believed that the riboswitch binds to flavin mono-nucleotide (FMN) and represses the expression of enzymes responsible for riboflavin and FMN biosynthesis.
  • Riboflavin is a water-soluble vitamin that is converted by flavokinases and FAD synthases to co-factors FMN and FAD, which are indispensable cofactors involved in energy metabolism and metabolism of fats, ketones, carbohydrates and proteins crucial for all living organisms.
  • flavokinases and FAD synthases are indispensable cofactors involved in energy metabolism and metabolism of fats, ketones, carbohydrates and proteins crucial for all living organisms.
  • Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci- 6 alkyl (e.g., methyl) or one hydroxy or Ci
  • X is a single bond, -S- or -0-;
  • A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci ⁇ alkyl (e.g., methyl ) , Ci_ 4 alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi ⁇ alkyl (e g., CF 3 ) or -0-haloCi_ 4 alkyl (e.g.
  • Ri is H, Ci_ 4 alkyl (e.g., methyl, ethyl, n-propyl or isopropyl), or Ci_ 4 alkoxy (e.g., methoxy);
  • R 2 is H, Ci- 4 alkyl (e.g., methyl or n-propyl), Ci_ 4 alkoxy (e.g., methoxy), halo (e.g., CI), C 3 - 8 cycloalkyl-Ci_ 4 alkyl, -Ci ⁇ alkyl-N(R a )(R b ), (Ci ⁇ alkoxy)- Ci ⁇ alkyl or (2-Ci_ 4 alkoxyethoxy)-Ci ⁇ alkyl;
  • R3 is H or Ci ⁇ alkyl (e.g., methyl);
  • R 4 is H or Ci ⁇ alkyl (e.g., methyl);
  • R a and R b are independently H, Ci ⁇ alkyl (e.g., methyl) or C 3 _ 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl);
  • the invention provides a compound of Formula IF ' :
  • Ci_ 6 alkylene e.g., n-propylene, n-butylene, n-pentylene
  • Ci_ 6 alkyl e.g., methyl
  • Ci_ 4alkoxy group e.g., methyl
  • ( ⁇ ) X is a single bond, -S- or -0-;
  • A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci ⁇ alkyl (e.g., methyl ) , Ci_ 4 alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi ⁇ alkyl (e . g., CF 3 ) or -0-haloCi_ 4 alkyl (e.g., -OCF 3 );
  • Ri is H or Ci ⁇ alkyl (e.g., methyl), or Ci ⁇ alkoxy (e.g., methoxy);
  • R 2 is H or Ci ⁇ alkyl (e.g., methyl), Ci_ 4 alkoxy (e.g., methoxy), halo (e.g.,
  • R 3 is H or Ci ⁇ alkyl (e.g., methyl);
  • R 4 is H or Ci ⁇ alkyl (e.g., methyl);
  • R a and 3 ⁇ 4 are independently H, Ci ⁇ alkyl (e.g., methyl) or C3_ 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl);
  • the invention provides a compound of Formula II:
  • Ci_ 6 alkylene e.g., n-propylene, n-butylene, n-pentylene
  • Ci_ 4 alkoxy group optionally substituted with one hydroxy or Ci_ 4 alkoxy group
  • ( ⁇ ) X is a single bond, -S- or -0-;
  • A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci ⁇ alkyl (e.g., methyl), Ci ⁇ alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi ⁇ alkyl (e . g., CF 3 ) or -0-haloCi_ 4 alkyl (e.g., -OCF 3 );
  • Ri is H, Ci_ 4 alkyl (e.g., methyl) or Ci ⁇ alkoxy (e.g., methoxy);
  • R 2 is H, Ci_ 4 alkyl (e.g., methyl), Ci ⁇ alkoxy (e.g., methoxy), halo (e.g., CI), C 3 - 8 cycloalkyl-Ci ⁇ alkyl, -Ci_ 4 alkyl-N(R a )(R b ), (Ci_ 4 alkoxy)-Ci_ 4 alkyl or (2- Ci _ 4 alkoxyethoxy) -C 1 _ 4 alkyl ;
  • R3 is H or Ci ⁇ alkyl (e.g., methyl);
  • R 4 is H or Ci ⁇ alkyl (e.g., methyl);
  • R a and R b are independently H, Ci ⁇ alkyl (e.g., methyl) or C 3 _ 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl);
  • the invention provides a compound of the following formulae: a compound of Formula P, wherein Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci_ 6 alkyl
  • Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci_ 6 alkyl
  • Ci_ 4 alkoxy e.g., ethoxy
  • Alk is n-propylene substituted with one or more Ci_ 6 alkyl (e.g., methyl), for example Alk is
  • Ci_ 6 alkylene e.g., n-propylene, n-butylene, n-pentylene
  • Ci_ 6 alkylene e.g., n-propylene, n-butylene, n-pentylene
  • a compound of Formula P, IF ' or II or any of 2.1-2.6 wherein A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci_ 4 alkyl (e.g., methyl), Ci ⁇ alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi_ 4 alkyl (e.g., CF 3 ) or -O-haloCi ⁇ alkyl (e.g., -OCF 3 ); a compound of Formula P, IF ' or II or any of 2.1-2.7, wherein A is aryl (e.g., phenyl);
  • a compound of Formula P, IF ' or II or any of 2.1-2.8 wherein A is phenyl; 2.10 a compound of Formula P, IF ' or II or any of 2.1-2.7, wherein A is aryl (e.g., phenyl) substituted with one or more Ci- 4 alkyl (e.g., methyl) or halo (e.g., CI, F);
  • Ci ⁇ alkoxy e.g., methoxy
  • Ci ⁇ alkyl e.g., methyl
  • Ci ⁇ alkoxy e.g., methoxy
  • Ci ⁇ alkoxy e.g., methoxy
  • halo e.g., CI
  • Ci ⁇ alkyl e.g., methyl
  • Ci ⁇ alkoxy e.g., methoxy
  • halo e.g., CI
  • Ci ⁇ alkyl e.g., methyl
  • Ci ⁇ alkoxy e.g., methoxy
  • halo e.g., CI
  • Ci ⁇ alkyl e.g., methyl
  • Ci ⁇ alkyl e.g., methyl
  • Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene)
  • Ci_ 6 alkyl e.g., methyl
  • Ci_ 4 alkoxy e.g., ethoxy
  • A is aryl (e.g., phenyl) optionally substituted with one or more C ⁇ alkyl (e.g., methyl) or halo (e.g., CI, F);
  • Ri is H or Ci_ 4 alkyl (e.g., methyl, ethyl, n-propyl or isopropyl);
  • R 2 is H or Ci_ 4 alkyl (e.g., methyl or n-propyl);
  • Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene)
  • X is a single bond, -S- or -0-;
  • A is aryl (e.g., phenyl) optionally substituted with one or more Ci_ 4alkyl (e.g., methyl), Ci ⁇ alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi ⁇ alkyl (e.g., CF 3 ) or -O-haloCi ⁇ alkyl (e.g., -
  • Ri is Ci ⁇ alkyl (e.g., methyl);
  • R 2 is Ci ⁇ alkyl (e.g., methyl);
  • R 3 is H
  • R t is H
  • Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one hydroxy or Ci_ 4 alkoxy group;
  • X is a single bond;
  • A is aryl (e.g., phenyl) optionally substituted with one or more Ci_ 4alkyl (e.g., methyl), Ci ⁇ alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi ⁇ alkyl (e.g., CF 3 ) or -O-haloCi ⁇ alkyl (e.g., - OCF 3 );
  • Ci_ 4alkyl e.g., methyl
  • Ci ⁇ alkoxy e.g., methoxy
  • hydroxy halo
  • haloCi ⁇ alkyl e.g., CF 3
  • -O-haloCi ⁇ alkyl e.g., - OCF 3
  • Ri is Ci ⁇ alkyl (e.g., methyl);
  • R 2 is Ci ⁇ alkyl (e.g., methyl);
  • R 3 is H
  • Alk is Ci_ 6 alkylene (e.g., n-propylene, n-butylene, n-pentylene); X is a single bond;
  • A is aryl (e.g., phenyl);
  • Ri is Ci ⁇ alkyl (e.g., methyl);
  • R 2 is Ci ⁇ alkyl (e.g., methyl);
  • R 3 is H
  • MIC Minimum Inhibitory Concentration
  • the compound of the invention is a compound as hereinbefore described (e.g., Formula P, II", IF, II, or any of 2.1-2.31), wherein X is a single bond and the remaining substituent is as defined in any of the formula described therein (e.g., Formula P, ⁇ ", IF , II, or any of 2.1-2.31), in free or salt form.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form in admixture with a pharmaceutically acceptable diluent or carrier.
  • the invention provides a method for the treatment or prophylaxis of a bacterial infection (Method P) comprising administering to a subject in need thereof an effective amount of a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form.
  • Methods P as hereinbefore described are useful for the treatment or prophylaxis of a Gram-positive or Gram- negative bacterial infection (Method P-A).
  • Method P is useful for treating a bacterial infection including, but not limited to an infection by one or more of the following bacteria: Clostridium difficile (or C.
  • Method P is particularly useful for treating an infection caused by Clostridium difficile.
  • the Compounds of the Invention, particularly Examples 1-16 are also active against the FMN riboswitch.
  • Compounds which are active against FMN riboswitch are generally also active against Staphylococcus aureus and/or Clostridium difficile infections.
  • the compounds of the invention may also be useful for the treatment of a Staphylococcus aureus infection.
  • Method P as hereinbefore described is useful for the treatment or prophylaxis of a disease, infection or condition selected from a group consisting of anthrax, staphylococcal scalded skin syndrome (staph infections), pneumonia, impetigo, boils, cellulitis, folliculitis, furuncles, carbuncles, scalded skin syndrome, abscesses, meningitis, osteomyelitis endocarditis, Toxic Shock Syndrome (TSS), septicemia, acute sinusitis, otitis media, septic arthritis, endocarditis, peritonitis, pericarditis, brain abscess, tularemia, urinary tract infection, empyema, food poisoning, diarrhea, conjunctivitis and Clostridium difficile associated disease (CDAD), comprising administering to a subject in need thereof an effective amount of a compound of Formula P, II" or II, e.g.,
  • the current invention provides methods of treating a bacterial infection via a novel mechanism, e.g., by utilizing riboswitch-ligand binding to alter gene expression. Therefore in one aspect, the compounds of the invention, particularly compounds of Examples 1-16 bind to FMN riboswitches, thereby affecting downstream riboflavin biosynthesis. In another aspect, various compounds of the invention are also active against the CD3299 riboswitch, thereby affecting expression of the adjacent coding region. Compounds that are active against CD3299 and/or FMN riboswitch are particularly selective against C. difficile.
  • the Compounds of the Invention e.g., the compounds of Formula P, II" or II, e.g., any of formulae 2.1-2.31, particularly Examples 1-16, in free or pharmaceutically acceptable salt form, are effective in treating an infection wherein traditional antibiotics are rendered ineffective due to drug resistance. Therefore, in a particular embodiment, the invention provides Method P as hereinbefore described wherein the infection is by an infectious agent which is resistant to a drug that is not a riboswitch ligand (Method P-E). In a further embodiment, the infection to be treated in Method P is a C.
  • the infection is by an infectious agent which is resistant to any drug that is not a riboswitch ligand, e.g., fluoroquinolone (e.g., ciprofloxacin- and/or levofloxacin-resistant infection), metronidazole and/or vancomycin.
  • the compounds of Formula P, ⁇ " or II e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form may be useful for a Staphylococcus aureus infection which is resistant to one or more drugs selected from a group consisting of a penicillin, vancomycin, cephalosporin and methicillin.
  • the infection is a methicillin-resistant Staphylococcus aureus infection.
  • various compounds of the Invention have a low CC5 0 value in an assay as disclosed in Example C and therefore, may have anti-metabolite activities which may interfere with DNA biosynthesis. Therefore, in one embodiment, these compounds may be useful as an anti-cancer or anti- viral agent. In another embodiment, the compounds that have a low MIC and/or a high I max value in an assay as disclosed in Example B and A respectively, and a low CC5 0 value in an assay as disclosed in Example C are used as an antibacterial, for topical administration.
  • the invention provides a method for the treatment of an infection in a plant (Method P-F) comprising administering to such plant an effective amount of a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form.
  • the invention provides use of a compound, or use of a pharmaceutical composition comprising a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form, (in the manufacture of a medicament) for the treatment or prophylaxis of an infection, e.g., a bacterial infection as described in Methods P, or any of Methods P-A through P-F.
  • a compound of Formula P, II" or II e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form, (in the manufacture of a medicament) for the treatment or prophylaxis of an infection, e.g., a bacterial infection as described in Methods P, or any of Methods P-A through P-F.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form, for use in the treatment of any disease or condition as hereinbefore described, e.g., in any of Methods P or Methods P-A through P-F.
  • riboswitch or "riboswitches” is an art recognized term and refers to an mRNA which comprises a natural aptamer that binds target metabolite and an expression platform which changes in the RNA structure to regulate genes.
  • riboswitch ligand refers to any compound such as a compound of Formula P, II” or II, e.g., any of formulae 2.1-2.31 ,, in free or salt form, that binds to that particular riboswitch.
  • FMN riboswitch refers to a riboswitch that binds a metabolite such as flavin mono-nucleotide (FMN) or other ligands such as the compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or salt form, and which affects downstream FMN biosynthesis and transport proteins.
  • FMN flavin mono-nucleotide
  • the binding of the ligand to its riboswitch induces a conformational change in the bacterial mRNA such that the expression of the ORF is altered, for example, such that the expression of enzymes responsible for, e.g., riboflavin and FMN biosynthesis is repressed or overexpressed.
  • CD3299 riboswitch refers to a riboswitch found in C. difficile, controlling the gene designated CD3299.
  • accession number AM180355 is as follows:
  • ORF start site in the above sequence is downstream from the riboswitch and is depicted in italics and is:
  • the putative terminator hairpin is in bold italics and is:
  • the hairpin can form a loop having a structure as depicted in Formula 1 :
  • a possible antiterminator has a structure as depicted in Formula 2:
  • infection encompasses an infection by a Gram-positive or Gram- negative bacteria.
  • the infection is by a Gram-positive bacteria.
  • the infection is by a Gram-negative bacteria.
  • the infection is an infection by one or more bacteria selected from a group consisting of Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii,
  • Escherichia coli Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes, Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borrelia burgdorferi.
  • the infection is a Clostridium difficile and/or Staphylococcus aureus infection.
  • the infection is an infection which is resistant to a drug which is not a riboswitch ligand.
  • the infection is an infection which is resistant to one or more drugs selected from a group consisting of penicillin, vancomycin, cephalosporin, methicillin and fluoroquinolone (e.g., ciprofloxacin- and/or levofloxacin).
  • the infection is a methicillin-resistant Staphylococcus aureus (MRSA) infection.
  • the infection is a fluoroquinolone-resistant (e.g., ciprofloxacin- and/or levofloxacin-resistant), metronidazole and/or vancomycin-resistant C. difficile infection.
  • bacteria or "bacterial” include, but are not limited to Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes,
  • Salmonella enterica Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borre lia burgdorferi .
  • Alkyl as used herein is a saturated or unsaturated hydrocarbon moiety, preferably saturated, e.g., one to eight, e.g., one to six, e.g., one to four carbon atoms in length, which may be linear or branched (e.g., n-butyl or tert-butyl) unless otherwise specified, and may be optionally substituted, e.g., mono-, di-, or tri-substituted on any one of the carbon atoms, e.g., with Ci_ 4 alkyl (e.g., methyl), Ci- 4 alkoxy, halogen (e.g., chloro or fluoro), haloCi ⁇ alkyl (e.g., trifluoromethyl), hydroxy, and carboxy.
  • Ci_ 4 alkyl e.g., methyl
  • Ci- 4 alkoxy e.g., halogen (e.g., chloro or fluoro), halo
  • C1-C8 alkyl denotes alkyl having 1 to 8 carbon atoms.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, i- butyl, sec -butyl, t-butyl, 3-methylpentyl, 4-methylpentyl, n-pentyl, n-hexyl and n-heptyl.
  • alkylene is intended to denote an alkyl group bridging between two substituents (e.g., between the flavin core structure and another substituent, for example -X-A). Therefore Ci_ 4alkylene, e.g., methylene, ethylene, n-propylene and n-butylene are intended to represent -CH 2 - -CH 2 CH 2 -, -CH 2 CH 2 CH 2 - and - CH 2 CH 2 CH 2 CH 2 - respectively. Wherein the alkylene group is unsaturated or partially saturated, it is denoted as "alkenylene" or "alkynylene".
  • Aryl as used herein is a monocyclic or polycyclic aromatic hydrocarbon, preferably phenyl, optionally substituted, e.g., with Ci_ 4 alkyl (e.g., methyl), Ci ⁇ alkoxy, halogen (e.g., chloro or fluoro), haloCi ⁇ alkyl (e.g., trifluoromethyl), hydroxy, carboxy, or an additional aryl or heteroaryl.
  • Ci_ 4 alkyl e.g., methyl
  • Ci ⁇ alkoxy e.g., methyl
  • halogen e.g., chloro or fluoro
  • haloCi ⁇ alkyl e.g., trifluoromethyl
  • Heterocycloalkyl refers to a cycloalkyl as defined above wherein at least one of the carbon atoms is replaced with a heteroatom selected from N, O, S. Therefore, “C3_ 8 heterocycloalkyl” or “heteroC 3 _ 8 cycloalkyl” refers to a 3- to 8-membered non-aromatic ring system containing at least one heteroatom selected from N, O and S.
  • the substituent is connected via an alkyl group, e.g., -Co- 4 alkyl-C 3 _ gcycloalkyl or aryl-Ci ⁇ alkyl, it is understood that the alkyl group may be saturated or unsaturated or linear or branched.
  • the substituent is connected via the Co-alkyl, it is understood that the alkyl is not present and the connectivity is directly to the next substituent.
  • the substituent is -Coalkyl-C3_ 8 cycloalkyl, it is understood that the alkyl group is not present and the cycloalkyl (e.g., cyclopropyl) is directly connected.
  • An acid-addition salt of a compound of the invention which is sufficiently basic for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulphuric, phosphoric, acid acetic, trifluoroacetic, citric, maleic acid, toluene sulfonic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, hydroxy maleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disul
  • an inorganic or organic acid for example hydrochloric, hydrobromic, sulphuric, phosphoric, acid acetic, trifluoroacetic, citric, maleic acid, toluene sulfonic
  • a salt of a compound of the invention which is sufficiently acidic is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically-acceptable cation, for example a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • the salt of the compound of the invention is a trifluoroacetic or hydrochloric acid addition salt.
  • the salt of the compound of the invention is an acetic acid addition salt.
  • Compounds of the Invention is to be understood as embracing the compounds disclosed herein, such as a compound of Formula P, II" or II, e.g., any of formulae 2.1 -2.31 , in any form, for example free or acid addition salt or prodrug form, or where the compounds contain acidic substituents, in base addition salt form.
  • the Compounds of the Invention are intended for use as pharmaceuticals, therefore pharmaceutically acceptable salts are preferred. Salts which are unsuitable for pharmaceutical uses may be useful, for example, for the isolation or purification of free Compounds of the Invention, and are therefore also included.
  • the Compounds of the Invention may comprise one or more chiral carbon atoms.
  • the compounds thus exist in individual isomeric, e.g., enantiomeric or diasteriomeric form or as mixtures of individual forms, e.g., racemic/diastereomeric mixtures. Any isomer may be present in which the asymmetric center is in the (R)-, (S)-, or (R,S)- configuration.
  • the invention is to be understood as embracing both individual optically active isomers as well as mixtures (e.g., racemic/diasteromeric mixtures) thereof.
  • the Compound of the Invention may be a racemic mixture or it may be predominantly, e.g., in pure, or substantially pure, isomeric form, e.g., greater than 70% enantiomeric excess ("ee"), preferably greater than 80% ee, more preferably greater than 90% ee, most preferably greater than 95% ee.
  • ee enantiomeric excess
  • the purification of said isomers and the separation of said isomeric mixtures may be accomplished by standard techniques known in the art (e.g., column chromatography, preparative TLC, preparative HPLC, simulated moving bed and the like).
  • the Compounds of the Invention encompass their stable isotopes.
  • the hydrogen atom at a certain position on the Compounds of the Invention may be replaced with deuterium. It is expected that the activity of compounds comprising such isotopes would be retained and/or it may have altered pharmacokinetic or pharmacodynamic properties.
  • pharmacodynamic properties would also have utility for measuring pharmacokinetics of the non-isotopic analogs.
  • Compounds of the Invention may in some cases also exist in prodrug form.
  • prodrug is an art recognized term and refers to a drug precursors prior to administration, but generate or release the active metabolite in vivo following
  • the Compounds of the Invention e.g., a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31
  • these substituents may be esterified to form physiologically hydrolysable and acceptable esters (e.g., acyl esters, e.g., CH 3 C(0)-0- Compound).
  • physiologically hydrolysable and acceptable esters means esters of Compounds of the Invention which are hydrolysable under physiological conditions to yield hydroxy on the one hand and acid, e.g., carboxylic acid on the other (e.g., Drug-0-C(0)-CH 3 -» Drug-OH + CH 3 COOH), which are themselves
  • amide prodrugs may also exist wherein the prodrug is cleaved to release the active amine metabolite in vivo following administration. Further details of amine prodrugs may be found in Jeffrey P. Krise and Reza Oliyai, Biotechnology: Pharmaceutical Aspects, Prodrugs, Volume 5, Part 3, pages 801-831, the contents of which are herein incorporated by reference in their entirety. As will be appreciated, the term thus embraces conventional pharmaceutical prodrug forms.
  • the Compounds of the Invention are useful for the treatment of an infection, particularly an infection by bacteria including but not limited to Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes, Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borrelia burgdorferi bacteria.
  • the bacteria is Clostridium difficile.
  • the invention therefore provides methods of treatment of any one or more of the following conditions: anthrax infection, staphylococcal scalded skin syndrome (staph infections), pneumonia, impetigo, boils, cellulitis, folliculitis, furuncles, carbuncles, scalded skin syndrome, abscesses, meningitis, osteomyelitis endocarditis, Toxic Shock Syndrome (TSS), septicemia, acute sinusitis, otitis media, septic arthritis, endocarditis, peritonitis, pericarditis, brain abscess, tularemia, urinary tract infection, empyema, food poisoning, diarrhea, conjunctivitis and Clostridium difficile associated disease (CD AD); comprising administering an effective amount of a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form, to a subject in need thereof.
  • treatment and “treating” are to be understood accordingly as embracing prophylaxis and treatment or amelioration of symptoms of disease as well as treatment of the cause of the disease.
  • the invention encompasses prophylaxis of symptoms of disease or cause of the disease.
  • the invention encompasses treatment or amelioration of symptoms of disease or cause of the disease.
  • subject encompasses human and/or non-human
  • a therapeutically active amount of the therapeutic compositions is defined as an amount effective, at dosages and for periods of time necessary to achieve the desired result.
  • a therapeutically effective amount of a Compound of the Invention reactive with at least a portion of the FMN or the CD3299 riboswitch may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual.
  • Dosage regiment may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. In general, satisfactory results, e.g.
  • Unit dosage forms for oral administration thus for example may comprise from about 0.2 to 75 mg, 250 mg,1000 mg, e.g. from about 0.2 or 2.0 to 50, 75, 100, 250, 500, 750 or 1000 mg of a Compound of the Invention, together with a pharmaceutically acceptable diluent or carrier therefor.
  • compositions comprising the Compounds of the Invention may be prepared using conventional diluents or excipients and techniques known in the galenic art.
  • oral dosage forms may include tablets, capsules, solutions, suspensions, spray-dried dispersions [e.g. Eudragit L100] and the like.
  • pharmaceutically acceptable carrier as used herein is intended to include diluents such as saline and aqueous buffer solutions.
  • the Compounds of the Invention may be administered in a convenient manner such as by injection such as subcutaneous, intravenous, by oral administration, inhalation, transdermal application, intravaginal application, topical application, intranasal, sublingual or rectal administration.
  • the active compound may be coated in a material to protect the compound from the degradation by enzymes, acids and other natural conditions that may inactivate the compound.
  • the compound may be orally administered.
  • the compound is administered via topical application.
  • the Compounds of the Invention may be administered alone or in conjunction, e.g., at or about the same time or simultaneously and separately or simultaneously in an admixture, with another agent, e.g., an agent to facilitate entry or permeability of the Compounds of the Invention into the cell, e.g., an antimicrobial cationic peptide.
  • Antimicrobial cationic peptides include peptides which contain (1) a disulfide-bonded ⁇ -sheet peptides; (2) amphipathic a-helical peptides; (3) extended peptides; or (4) loop-structured peptides.
  • cationic peptide examples include but are not limited to defensins, cecropins, melittins, magainins, indolicidins, bactenecin and protegrins.
  • antimicrobial cationic peptides include but are not limited to human neutrophil defensin-1 (HNP-1), platelet microbicidal protein- 1 (tPMP), inhibitors of DNA gyrase or protein synthesis, CP26, CP29, CP11CN, CPIOA, Bac2A- NH 2 as disclosed in Friedrich et al., Antimicrob. Agents Chemother. (2000) 44(8):2086, the contents of which are hereby incorporated by reference in its entirety.
  • Further examples of antibacterial cationic peptides include but are not limited to polymyxin e.g., polymixin B, polymyxin E or polymyxin nonapeptide. Therefore, in another embodiment, the
  • Compounds of the Invention may be administered in conjunction with polymyxin, e.g., polymyxin B, polymyxin E or polymyxin nonapeptide, preferably polymyxin B.
  • polymyxin e.g., polymyxin B, polymyxin E or polymyxin nonapeptide, preferably polymyxin B.
  • the Compounds of the Invention may be administered alone or in conjunction, e.g., at or about the same time, simultaneously and separately, or simultaneously in an admixture, with other antimicrobial agents, e.g., other antifungal or other systemic antibacterial (bactericidal or bacteriostatic) agents.
  • other antimicrobial agents e.g., other antifungal or other systemic antibacterial (bactericidal or bacteriostatic) agents.
  • bacterial agents include agents which inhibit bacterial cell wall synthesis (e.g., penicillins, cephalosporins, carbapenems, vancomycin), agents which damage cytoplasmic membrane (e.g., polymixins as discussed above), agents which modify the synthesis or metabolism of nucleic acids (e.g., quinolones, rifampin, nitrofurantoin), agents which inhibit protein synthesis (aminoglycosides, tetracyclines, chloramphenicol, erythomycin, clindamycin), agents which interfer with the folate synthesis (e.g., folate-inhibitors), agents which modify energy metabolism (e.g., sulfonamides, trimethoprim) and/or other antibiotics (beta-lactam antibiotic, beta-lactamase inhibitors).
  • agents which inhibit bacterial cell wall synthesis e.g., penicillins, cephalosporins, carbapenems, vancomycin
  • the compounds of the Invention e.g., compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31 , in free or salt form may be made using the methods as described and exemplified herein and by methods similar thereto and by methods known in the chemical art. Such methods include, but not limited to, those described below.
  • synthetic methods include, but not limited to, those described below.
  • all proposed reaction conditions including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, are chosen to be the conditions standard for that reaction, which should be readily recognized by one skilled in the art. Therefore, at times, the reaction may require to be run at elevated temperature or for a longer or shorter period of time.
  • 2.1-2.31 may be prepared by reacting Intermediate-5 (Int-5) with ammonia in a pressure tube.
  • Int-5 may be prepared by reacting Intermediate-4 (Int-4) with diethyl 2-bromo-3- oxopentanedioate in the presence of a base, e.g., cesium carbonate, in a solvent, for example, a mixture of dimethylformamide (DMF) and methylene chloride (CH 2 CI 2 ).
  • a base e.g., cesium carbonate
  • Int- 4 may be may be prepared by converting Intermediate-3 (Int-3) to Int-4, for example, by catalytic hydrogenation, e.g., by reacting Int-3 with a metal, e.g., Raney-Nickel, and hydrogen gas in a solvent such as ethanol.
  • Int-3 may be prepared by reacting Intermediate-1 (Int-1) with NH 2 -Alk-X-A (Int-2), wherein Alk, X and A are defined in Formula P, IF ' or II or any of 2.1-2.31 to yield Int-3.
  • Int-1 is either commercially available or may be prepared by methods known in the art.
  • R 2 of compounds of Formula P, IF ' or II is alkoxy
  • this compound may be prepared by reacting a compound of Formula P, IF ' or II, wherein R 2 is halo, e.g., chloro, with R 2 -H, e.g., methanol, in the presence of a base.
  • R 2 is halo, e.g., chloro
  • R 2 -H e.g., methanol
  • R 2 of the compounds of Formula P, IF ' or II is (Ci ⁇ alkoxy)- methyl
  • these compounds may be prepared by first halogenating the compound of Formula P, IF ' or II, wherein R 2 is methyl, for example by reacting such compound with a halogen, e.g., bromine, e.g., optionally in the presence of a catalyst such as
  • azobisisobutyronitrile AIBN
  • the resulting intermediate, Int-6 may then react with a R 2 -H, wherein R 2 -H is e.g. methanol, in the presence of a base to provide the
  • R 2 of the compounds of Formula P, IF ' or II is -methyl-N(R a )(Rb), e.g., -CH 2 -N(CH 3 ) 2
  • this compound may be prepared by halogenating the compounds of Formula P, IF ' or II, wherein R 2 is e.g., a methyl group, for example by reacting bromine with the compounds of Formula P, IF ' or II, wherein R 2 is methyl, optionally in the presence of a catalyst such as azobisisobutyronitrile (AIBN).
  • AIBN azobisisobutyronitrile
  • the resulting intermediate, Int-6 may then react with an amine, HN(R a )(Rb), e.g.
  • R 2 of the compounds of Formula P, II" or II is hydrogen
  • these compounds may be prepared by heating Intermediate- 10 (Int-10) with acid [e.g.
  • Int-10 may be prepared by heating Intermediate-9 (Int- 9) with sodium dithionite in a mixture of ethanol and water.
  • Intermediate-9 (Int-9) may be prepared by treating Intermediate-8 (Int-8) with sodium nitrite in acetic acid with cooling.
  • Int-8 may be prepared by condensing Intermediate-7 (Int-7) with methyl 5-[(4- methoxybenzyl)amino]-3,5-dioxopentanoate in the presence of acid [e.g. p-toluenesulfonic acid monohydrate] in toluene.
  • Intermediate-7 (Int-7) may be prepared with Br- Alk-X-Ar , wherein Alk, X, and A are defined in any of Formulae P, II", or II. This preparation may be summarized in the following reaction scheme:
  • Precursor mRNA leader molecules are prepared by in vitro transcription from templates generated by PCR and [5'- 32 P] -labeling using methods described previously (Regulski and Breaker, In-line probing analysis of riboswitches (2008), Methods in Molecular Biology Vol 419, pp 53-67).
  • RNA precursor Approximately 5 nM of labeled RNA precursor is incubated for 41 hours at 25°C in 20 mM MgCl 2 , 50 mM Tris/HCl (pH 8.3 at 25°C) in the presence or absence of a fixed concentration of each ligand. Binding to the FMN and CD3299 riboswitches are measured at 20 ⁇ and 100 ⁇ , respectively. In-line cleavage products are separated on 10% polyacrylamide gel electrophoresis (PAGE), and the resulting gel is visualized using a Molecular Dynamics Phosphorimager. The location of products bands corresponding to cleavage are identified by comparison to a partial digest of the RNA with RNase Tl (G- specific cleavage) or alkali (nonspecific cleavage).
  • RNA In-line probing exploits the natural ability of RNA to self-cleave at elevated pH and metal ion concentrations (pH ⁇ 8.3, 25 mM MgCl 2 ) in a conformation-dependent manner.
  • the 2'-hydroxyl of the ribose For self-cleavage to occur, the 2'-hydroxyl of the ribose must be "in-line" with the phosphate-oxygen bond of the internucleotide linkage, facilitating a S 2P nucleophilic transesterification and strand cleavage.
  • single-stranded regions of the riboswitch are dynamic in the absence of an active ligand, and the internucleotide linkages in these regions can frequently access the required in-line conformation.
  • Binding of an active ligand to the riboswitch generally reduces the dynamics of these regions, thereby reducing the accessibility to the in-line conformation, resulting in fewer in-line cleavage events within those regions.
  • These ligand-dependent changes in RNA cleavage can be readily detected by denaturing gel electrophoresis.
  • the relative binding affinity of each ligand is expressed as I max , wherein I max represents the percent inhibition of in-line cleavage at selected internucleotide ligands in the presence of a fixed ligand concentration (20 ⁇ for the FMN riboswitch and 100 ⁇ for the CD3299 riboswitch) normalized to the percent inhibition in the absence of ligand and the percent inhibition in the presence of a saturation concentration of a control ligand.
  • 100 ⁇ FMN is used as a control ligand for estimating binding to the FMN riboswitch and 100 ⁇ of 7,8-dimethyl-10-(3- phenylpropyl)benzo[g]pteridine-2,4(3H,10H)-dione (which is a compound which has a high affinity against the CD3299 riboswitch) is used as a control ligand for estimating binding to the CD3299 riboswitch.
  • Example B [0048] The MIC assays are carried out in a final volume of 100 in 96-well clear round-bottom plates according to methods established by the Clinical Laboratory
  • test compound suspended in 100 % DMSO or another suitable solubilizing buffer
  • 100 % DMSO or another suitable solubilizing buffer
  • This solution is serially diluted by 2-fold into successive tubes of the same media to give a range of test compound concentrations appropriate to the assay.
  • 50 ⁇ of a bacterial suspension from an overnight culture growth in media appropriate to a given pathogen is added 50 ⁇ of a bacterial suspension from an overnight culture growth in media appropriate to a given pathogen.
  • Final bacterial inoculum is approximately 10 5 -10 6 CFU/well.
  • the MIC is defined as the lowest concentration of antimicrobial agent that completely inhibits growth of the organism as detected by the unaided eye, relative to control for bacterial growth in the absence of added antibiotic. Ciprofloxacin is used as an antibiotic-positive control in each screening assay.
  • Each of the bacterial cultures that are available from the American Type Culture Collection (ATCC, www.atcc.org) is identified by its ATCC number.
  • the exemplified compounds of the invention e.g., the compounds of Examples 1-16 have a minimum inhibitory concentration (MIC) of less than 2 ⁇ g/mL against at least one of the bacteria selected from C. difficile MMX3581 (clinical), C. difficile ATCC 43596, C. difficile ATCC 700057 (MMX 4381), C. difficile ATCC BAA-1805 (NAP1), C. difficile ATCC BAA-1382 (MMX4820), C. difficile 43255 (MMX4821), C. difficile ATCC BAA-1803 (NAP1) and C. difficile ATCC BAA-1870 (NAP1).
  • MIC minimum inhibitory concentration
  • All of the exemplified compounds of the invention have either an I max value of greater than 20% in an assay as described in Example A (compared to at least one of the two controls) and/or a MIC of less than or equal to 2 ⁇ g/mL against at least one of the bacterial strains as decribed in Example B.
  • certain compounds of the invention have either an I max value of greater than 50% in an assay as described in Example A (compared to at least one of the two controls) and/or a MIC of less than or equal to 2 ⁇ g/mL against at least one of the bacterial strains as decribed in Example B.
  • Example C The cytotoxic effects of test compounds on HepG2 are measured with a commercially available cell viability assay kit from Promega. On day 1, HepG2 cells ( ⁇ 1 x 10 4 cells) are seeded into each well in 96-well plate and cultured for approximately 24 h at 37°C in a 5% C0 2 atmosphere under saturating humidity. On day 2, test compounds and DMSO controls are added to appropriate wells to give a range of test compound concentrations appropriate to the assay. Terfenadine is also added to each plate as a positive cytotoxic control. Control wells containing medium without cell are prepared to obtain a value for background luminescence.
  • Assay plates are then cultured for approximately 24 h at 37°C in a 5% C02 atmosphere under saturating humidity. On day 3, assay plates are removed from 37°C incubator and equilibrated to 22°C. Once equilibrated, CellTiter-Glo ® reagent is added to each well containing cell culture medium, followed by mixing to allow cell lysis. The CellTiter-Glo ® Assay measures the number of viable cells in culture based on quantitation of the ATP present, an indicator of metabolically active cells. This assay generates a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly proportional to the number of cells present in culture. After the assay plate is incubated at room temperature for
  • CC5 0 is defined as the concentration of test compounds in ⁇ to result in 50% reduction in luminescence signal relative to the signal for untreated cells.
  • the experiments show that the exemplified compounds of the invention have a CC5 0 value of greater than or equal to 19 ⁇ .
  • the exemplified compounds of the invention generally have a MIC to cytotox ratio of at least 1:20.
  • Method D Agilent 1100 HPLC, Agilent XDB C18 50 x 4.6 mm 1.8 micron column, 1.5 mL/min., Solvent A-Water (0.1% TFA), Solvent B -Acetonitrile (0.07% TFA), Gradient -5 min. 95%A to 95%B; lmin. hold; then recycle, UV Detection @ 210 and 254nm.
  • Method G Agilent 1100 HPLC, Agilent XDB C18 50 x 4.6 mm 5 micron column, 1.5 mL/min., Solvent A- Water (0.1% TFA), Solvent B -Acetonitrile (0.07% TFA), Gradient -6 min. 95%A to 95%B; 1 min. hold; then recycle, UV Detection @ 210 and 250nm.
  • NaHCC sodium bicarbonate
  • Na 2 S04 sodium sulfate
  • NH 3 ammonia gas
  • RNA ribonucleic acid
  • RNase Tl an endoribonuc lease that specifically degrades single-stranded RNA at G residues
  • 3,4-dihydroquinoxaline-2-carboxylate (0.300 g, 0.690 mmol) is taken up in 20 mL of MeOH and the solution is cooled in an ice water bath. Ammonia gas is bubbled through the solution for 5 minutes in a pressure tube. The solution is stirred at rt overnight in the capped pressure tube. The pressure tube is opened slowly to allow NH 3 to evolve. The remaining solution is evaporated to give 0.2 g of a dark solid. The solid is adsorbed onto silica gel and chromatographed on 50 g of silica gel. The column is eluted with 1% MeOH

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pest Control & Pesticides (AREA)
  • Agronomy & Crop Science (AREA)
  • Plant Pathology (AREA)
  • Engineering & Computer Science (AREA)
  • Dentistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Environmental Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates novel flavin derivatives, composition comprising the same, their use and compositions for use as riboswitch ligands and/or anti-infectives.

Description

FLAVIN DERIVATIVES
This application claims priority from U.S. Provisional Application Number
61/441,160 filed February 9, 2011, International Application Number
PCT/US2011/000617 file April 6, 2011, and U.S. provisional application 61/516,651 filed April 6, 2011, the contents of each of which are incorporated by reference in their entirety.
TECHNICAL FIELD
[0001] The present invention relates to novel flavin derivatives, composition comprising the same, and their use and compositions for use as riboswitch ligands and/or anti-infectives.
BACKGROUND OF THE INVENTION
[0002] The fast growing rate of antibiotic resistance over the past decades has raised serious concerns that the antibiotic treatment options currently available will soon be ineffective. With the widespread usage of antibiotics in combination with the rapid growing rate of bacterial resistance in stark contrast with the decade-old chemical scaffolds available for their treatment, it is imperative that new drugs are developed in the battle against bacterial pathogens.
[0003] In many bacteria and fungi, RNA structures termed riboswitches regulate the expression of various genes crucial for survival or virulence. Typically located within the 5 '-untranslated region (5'-UTR) of certain mRNAs, members of each known class of riboswitch can fold into a distinct, three-dimensionally structured receptor that recognizes a specific organic metabolite. When the cognate metabolite is present at sufficiently high concentrations during transcription of the mRNA, the riboswitch receptor binds to the metabolite and induces a structural change in the nascent mRNA that prevents expression of the open reading frame (ORF), thereby altering gene expression. In the absence of the cognate metabolite, the riboswitch folds into a structure that does not interfere with the expression of the ORF.
[0004] Sixteen different classes of riboswitches have been reported. Members of each class of riboswitch bind to the same metabolite and share a highly conserved sequence and secondary structure. Riboswitch motifs have been identified that bind to thiamine pyrophosphate (TPP), flavin mononucleotide (FMN), glycine, guanine, 3'-5'-cyclic eiguanylic acid (c-di-GMP), molybdenum cofactor, glucosamine-6-phosphate (GlcN6P), lysine, adenine, and adocobalamin (AdoCbl) riboswitches. Additionally, four dinstinct riboswitch motifs have been identified that recognize S-adenosylmethionine (SAM) I, II and III, IV and two distinct motifs that recognize pre-queosine- 1 (PreQl). Several antimetabolite ligands have also been identified that bind to known riboswitch classes, including pyrithiamine pyrophosphate (PTPP) which binds TPP riboswitches, L- aminoethylcysteine (AEC) and DL-4-oxalysine which bind to lysine riboswitches and roseoflavin and FMN which bind to FMN riboswitches. The riboswitch-receptors bind to their respective ligands in an interface that approaches the level of complexity and selectivity of proteins. This highly specific interaction allows riboswitches to discriminate against most intimately related analogs of ligands. For instance, the receptor of a guanine- binding riboswitch from Bacillus subtilis forms a three-dimensional structure such that the ligand is almost completely enveloped. The guanine is positioned between two aromatic bases and each polar functional group of the guanine hydrogen bonds with four additional riboswitch nucleotides surrounding it. This level of specificity allows the riboswitch to discriminate against most closely related purine analogs. Similarly, studies of the SAM- binding riboswitches reveal that nearly every functional group of SAM is critical in binding the ligands, allowing it to discriminate highly similar compounds such as S- adenosylhomocysteine (SAH) and S-adenosylmethionine (SAM), which only differ by a single methyl group. Likewise, TPP riboswitches comprise one subdomain that recognizes every polar functional group of the 4-amino-5-hydroxymethyl-2- methylpyrimidine (HMP) moiety, albeit not the thiazole moiety, and another subdomain that coordinates two metal ions and several water molecules to bind the negatively charged pyrophosphate moiety of the ligand. Similar to TPP, guanine and SAM riboswitches, FMN riboswitches form receptor structures that are highly specific for the natural metabolite FMN. It is by this highly specific interaction that allows for the design of small molecules for the regulation of specific genes.
[0005] FMN riboswitches are of particular interest of this invention because it is believed that the riboswitch binds to flavin mono-nucleotide (FMN) and represses the expression of enzymes responsible for riboflavin and FMN biosynthesis. Riboflavin is a water-soluble vitamin that is converted by flavokinases and FAD synthases to co-factors FMN and FAD, which are indispensable cofactors involved in energy metabolism and metabolism of fats, ketones, carbohydrates and proteins crucial for all living organisms. Although vertebrates rely on uptake of vitamin from their gut for riboflavin sources, most prokaryotes, fungi and plants synthesize the necessary riboflavin for survival. It is therefore suggested that compounds that are selective for FMN riboswitches may be useful against bacterial pathogens by causing disregulation of biosynthesis of riboflavin crucial for survival or virulence. In addition, no examples of the FMN, TPP, nor any other riboswitch class have presently been identified in humans. Therefore, riboswitches appear to offer the potential for the discovery of selective antipathogenic drugs. It is therefore the objective of this invention to provide novel flavin derivatives for targeting FMN riboswitches and methods of treating infections comprising administering flavin derivatives. Various flavin derivatives that target FMN riboswitch are disclosed in PCT/US2009/004576, PCT/US2010/001876 and PCT/US2011/000617, the contents of which are incorporated by reference in their entirety. The current application provides further flavin derivatives that target the FMN and/or the CD3299 riboswitch and/or are active against various bacterial strains.
SUMMARY OF THE INVENTION
[0006] The invention P:
Figure imgf000004_0001
wherein:
(i) Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci-6alkyl (e.g., methyl) or one hydroxy or Ci
4alkoxy (e.g., ethoxy) group;
X is a single bond, -S- or -0-;
A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci^alkyl (e.g., methyl), Ci_4alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi^alkyl (e g., CF3) or -0-haloCi_4alkyl (e.g., -OCF3); (iv) Ri is H, Ci_4alkyl (e.g., methyl, ethyl, n-propyl or isopropyl), or Ci_4alkoxy (e.g., methoxy);
(v) R2 is H, Ci-4alkyl (e.g., methyl or n-propyl), Ci_4alkoxy (e.g., methoxy), halo (e.g., CI), C3-8cycloalkyl-Ci_4alkyl, -Ci^alkyl-N(Ra)(Rb), (Ci^alkoxy)- Ci^alkyl or (2-Ci_4alkoxyethoxy)-Ci^alkyl;
(vi) R3 is H or Ci^alkyl (e.g., methyl);
(vii) R4 is H or Ci^alkyl (e.g., methyl);
(viii) Ra and Rb are independently H, Ci^alkyl (e.g., methyl) or C3_8cycloalkyl (e.g., cyclopropyl, cyclopentyl);
in free or salt form.
[0007] In a further embodiment, the invention provides a compound of Formula IF ' :
Figure imgf000005_0001
ormu a wherein:
(0 Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci_6alkyl (e.g., methyl) or one hydroxy or Ci_ 4alkoxy group;
(ϋ) X is a single bond, -S- or -0-;
(iii) A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci^alkyl (e.g., methyl), Ci_4alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi^alkyl (e.g., CF3) or -0-haloCi_4alkyl (e.g., -OCF3);
(iv) Ri is H or Ci^alkyl (e.g., methyl), or Ci^alkoxy (e.g., methoxy);
(v) R2 is H or Ci^alkyl (e.g., methyl), Ci_4alkoxy (e.g., methoxy), halo (e.g.,
CI), C3-8cycloalkyl-Ci_4alkyl, -Ci^alkyl-N(Ra)(Rb), (Ci^alkoxy)-Ci_4alkyl or (2 - C 1 _4alkoxy ethoxy ) - C 1 _4alky 1 ;
(vi) R3 is H or Ci^alkyl (e.g., methyl); (vii) R4 is H or Ci^alkyl (e.g., methyl);
(viii) Ra and ¾ are independently H, Ci^alkyl (e.g., methyl) or C3_8cycloalkyl (e.g., cyclopropyl, cyclopentyl);
in free or salt form.
[0008] In still a further embodiment, the invention provides a compound of Formula II:
Figure imgf000006_0001
ormu a wherein:
(0 Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one hydroxy or Ci_4alkoxy group;
(ϋ) X is a single bond, -S- or -0-;
(iii) A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci^alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi^alkyl (e.g., CF3) or -0-haloCi_4alkyl (e.g., -OCF3);
(iv) Ri is H, Ci_4alkyl (e.g., methyl) or Ci^alkoxy (e.g., methoxy);
(v) R2 is H, Ci_4alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), halo (e.g., CI), C3-8cycloalkyl-Ci^alkyl, -Ci_4alkyl-N(Ra)(Rb), (Ci_4alkoxy)-Ci_4alkyl or (2- Ci _4alkoxyethoxy) -C 1 _4alkyl ;
(vi) R3 is H or Ci^alkyl (e.g., methyl);
(vii) R4 is H or Ci^alkyl (e.g., methyl);
(viii) Ra and Rb are independently H, Ci^alkyl (e.g., methyl) or C3_8cycloalkyl (e.g., cyclopropyl, cyclopentyl);
in free or salt form.
[0009] In yet a further embodiment, the invention provides a compound of the following formulae: a compound of Formula P, wherein Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci_6alkyl
(e.g., methyl) or one hydroxy or Ci_4alkoxy (e.g., ethoxy) group;
a compound of Formula P or 2.1 , wherein Alk is n-propylene substituted with one or more Ci_6alkyl (e.g., methyl), for example Alk is
-CH2CH2CH(CH3)-;
a compound of Formula P, IF ' or II or 2.1, wherein Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one hydroxy or Ci^alkoxy group;
a compound of Formula P, IF ' or II or 2.1, wherein Alk is n-propylene; a compound of Formula P, IF ' or II or any of 2.1-2.4, wherein X is a single bond, -S- or -0-;
a compound of Formula P, IF ' or II or any of 2.1-2.5, wherein X is a single bond, wherein said compound is represented by a compound of formula IF ;
Figure imgf000007_0001
a compound of Formula P, IF ' or II or any of 2.1-2.6, wherein A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci_4alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi_4alkyl (e.g., CF3) or -O-haloCi^alkyl (e.g., -OCF3); a compound of Formula P, IF ' or II or any of 2.1-2.7, wherein A is aryl (e.g., phenyl);
a compound of Formula P, IF ' or II or any of 2.1-2.8, wherein A is phenyl; 2.10 a compound of Formula P, IF ' or II or any of 2.1-2.7, wherein A is aryl (e.g., phenyl) substituted with one or more Ci-4alkyl (e.g., methyl) or halo (e.g., CI, F);
2.11 a compound of Formula P or any of 2.1 -2.10, wherein Ri is H, Ci^alkyl
(e.g., methyl, ethyl, n-propyl or isopropyl) or Ci^alkoxy (e.g., methoxy);
2.12 a compound of Formula P or any of 2.1 -2.11 , wherein R] is Ci_4alkyl
(e.g., methyl, ethyl, n-propyl or isopropyl);
2.13 a compound of Formula P, II" or II or any of 2.1-2.10, wherein Ri is H,
Ci^alkyl (e.g., methyl), or Ci^alkoxy (e.g., methoxy);
2.14 a compound of Formula P, II" or II or any of 2.1-2.13, wherein Ri is methyl;
2.15 a compound of Formula P or any of 2.1-2.14, wherein R2 is H, Ci^alkyl
(e.g., methyl or n-propyl), Ci^alkoxy (e.g., methoxy), halo (e.g., CI), C3- 8cycloalkyl-Ci_4alkyl, -Ci_4alkyl-N(Ra)(Rb), (Ci^alkoxy)-Ci^alkyl or (2- Ci^alkoxyethoxy)-Ci_4alkyl;
2.16 a compound of Formula P, IF ' or II or any of 2.1 -2.14, wherein R2 is H,
Ci^alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), halo (e.g., CI), C3- 8cycloalkyl-Ci_4alkyl, -Ci_4alkyl-N(Ra)(Rb), (Ci^alkoxy)-Ci^alkyl or (2- C 1 ^alkoxyethoxy) -C 1 _4alkyl ;
2.17 a compound of Formula P, II" or II or any of 2.1-2.14, wherein R2 is H,
Ci^alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), halo (e.g., CI), C3- 8cycloalkyl-Ci_4alkyl, -Cialkyl-N(Ra)(Rb), (Ci^alkoxy)-Ci^alkyl or (2-Ci. 4alkoxyethoxy)-Ci^alkyl;
2.18 a compound of Formula P, II" or II or any of 2.1 -2.16, wherein R2 is methyl;
2.19 a compound of Formula P or any of 2.1 -2.15 , wherein R2 is n-propyl;
2.20 a compound of Formula P, II" or II or any of 2.1-2.19, wherein R3 is H or
Ci^alkyl (e.g., methyl);
2.21 a compound of Formula P, IF ' or II or any of 2.1-2.20, wherein R3 is H; 2.22 a compound of Formula P, IF ' or II or any of 2.1 -2.21 , wherein R4 is H or
Ci^alkyl (e.g., methyl);
2.23 a compound of Formula P, IF ' or II or any of 2.1-2.22, wherein R4 is H; a compound of Formula P or any of 2.1-2.23, wherein X is a single bond, wherein a compound of formula IF :
Figure imgf000009_0001
Formula ΙΓ and
(i) Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene)
optionally substituted with one or more Ci_6alkyl (e.g., methyl) or one Ci_4alkoxy (e.g., ethoxy) group;
(ii) A is aryl (e.g., phenyl) optionally substituted with one or more C^alkyl (e.g., methyl) or halo (e.g., CI, F);
(iii) Ri is H or Ci_4alkyl (e.g., methyl, ethyl, n-propyl or isopropyl);
(iv) R2 is H or Ci_4alkyl (e.g., methyl or n-propyl);
(v) R3 is H;
(vi) P is H;
a compound of Formula P, IF ' or II or or any of 2.1-2.23, wherein:
Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene)
optionally substituted with one hydroxy or Ci_4alkoxy group;
X is a single bond, -S- or -0-;
A is aryl (e.g., phenyl) optionally substituted with one or more Ci_ 4alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi^alkyl (e.g., CF3) or -O-haloCi^alkyl (e.g., -
OCF3);
Ri is Ci^alkyl (e.g., methyl);
R2 is Ci^alkyl (e.g., methyl);
R3 is H;
Rt is H;
a compound of Formula P, IF ' or II or any of 2.1-2.23, wherein: Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one hydroxy or Ci_4alkoxy group; X is a single bond;
A is aryl (e.g., phenyl) optionally substituted with one or more Ci_ 4alkyl (e.g., methyl), Ci^alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi^alkyl (e.g., CF3) or -O-haloCi^alkyl (e.g., - OCF3);
Ri is Ci^alkyl (e.g., methyl);
R2 is Ci^alkyl (e.g., methyl);
R3 is H;
R4 IS H;
2.27 a compound of Formula P, IF ' or II or any of 2.1-2.23, wherein:
Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene); X is a single bond;
A is aryl (e.g., phenyl);
Ri is Ci^alkyl (e.g., methyl);
R2 is Ci^alkyl (e.g., methyl);
R3 is H;
R4 IS H;
2.28 any of the preceding formulae, wherein the compound of Formula P, IF '
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
2.31 any of the preceding formulae, wherein the compound of Formula P, IF ' or II binds to FMN and/or CD3299 riboswitch, e.g., with an Imax of greater than 20%, preferably greater than 30%, more preferably greater than 40%, still more preferably greater than 50% in an assay, for example, as described in Example A, and/or has a Minimum Inhibitory Concentration (MIC) of less than or equal to 64μg/mL, more preferably less than or equal to 32μg/mL, still more preferably less than or equal to 16μg/mL, most preferably less than or equal to 2μg/mL, for example, in an assay as described in Example B;
in free or salt form.
[0010] In a particular embodiment, the compound of the invention is a compound as hereinbefore described (e.g., Formula P, II", IF, II, or any of 2.1-2.31), wherein X is a single bond and the remaining substituent is as defined in any of the formula described therein (e.g., Formula P, Π", IF , II, or any of 2.1-2.31), in free or salt form.
[0011] In the second aspect, the invention provides a pharmaceutical composition comprising a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form in admixture with a pharmaceutically acceptable diluent or carrier.
[0012] In the third aspect, the invention provides a method for the treatment or prophylaxis of a bacterial infection (Method P) comprising administering to a subject in need thereof an effective amount of a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form.
[0013] In a further embodiment of the third aspect, Methods P as hereinbefore described are useful for the treatment or prophylaxis of a Gram-positive or Gram- negative bacterial infection (Method P-A). In another specific embodiment, Method P, is useful for treating a bacterial infection including, but not limited to an infection by one or more of the following bacteria: Clostridium difficile (or C. difficile), Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes, Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borrelia burgdorferi bacteria (Method P-B). Patients taking antibiotics, particularly those with a broad spectrum activity, are particularly vulnerable to C. difficile infection as a result of the use of antibiotics which disrupts the normal intestinal flora, leading to an overgrowth of C. difficile, causing an infection ranging from asymptomatic to severe and life-threatening condition. The exemplified Compounds of the Invention, e.g., compounds of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31, particularly compounds of Examples 1-16 selectively inhibit C.
difficile bacteria in a minimum inhibitory concentration assay as described in Example B below. Further, various compounds of the invention, e.g., various compounds of Formula P, e.g., various compounds of Examples 1-16 are also active against the
CD3299 riboswitch. Therefore, in a particular embodiment, Method P is particularly useful for treating an infection caused by Clostridium difficile. In addition to having MIC activity against C. Difficile and some having CD3299 riboswitch activity, the Compounds of the Invention, particularly Examples 1-16 are also active against the FMN riboswitch. Compounds which are active against FMN riboswitch are generally also active against Staphylococcus aureus and/or Clostridium difficile infections.
Therefore, in particular embodiment, the compounds of the invention, particular compounds of Examples 1-16 may also be useful for the treatment of a Staphylococcus aureus infection.
[0014] In still another embodiment of the third aspect, Method P as hereinbefore described is useful for the treatment or prophylaxis of a disease, infection or condition selected from a group consisting of anthrax, staphylococcal scalded skin syndrome (staph infections), pneumonia, impetigo, boils, cellulitis, folliculitis, furuncles, carbuncles, scalded skin syndrome, abscesses, meningitis, osteomyelitis endocarditis, Toxic Shock Syndrome (TSS), septicemia, acute sinusitis, otitis media, septic arthritis, endocarditis, peritonitis, pericarditis, brain abscess, tularemia, urinary tract infection, empyema, food poisoning, diarrhea, conjunctivitis and Clostridium difficile associated disease (CDAD), comprising administering to a subject in need thereof an effective amount of a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form (Method P-D).
[0015] Without being bound to any particular theory, it is believed that the current invention provides methods of treating a bacterial infection via a novel mechanism, e.g., by utilizing riboswitch-ligand binding to alter gene expression. Therefore in one aspect, the compounds of the invention, particularly compounds of Examples 1-16 bind to FMN riboswitches, thereby affecting downstream riboflavin biosynthesis. In another aspect, various compounds of the invention are also active against the CD3299 riboswitch, thereby affecting expression of the adjacent coding region. Compounds that are active against CD3299 and/or FMN riboswitch are particularly selective against C. difficile. As such, the Compounds of the Invention, e.g., the compounds of Formula P, II" or II, e.g., any of formulae 2.1-2.31, particularly Examples 1-16, in free or pharmaceutically acceptable salt form, are effective in treating an infection wherein traditional antibiotics are rendered ineffective due to drug resistance. Therefore, in a particular embodiment, the invention provides Method P as hereinbefore described wherein the infection is by an infectious agent which is resistant to a drug that is not a riboswitch ligand (Method P-E). In a further embodiment, the infection to be treated in Method P is a C. difficile infection, wherein the infection is by an infectious agent which is resistant to any drug that is not a riboswitch ligand, e.g., fluoroquinolone (e.g., ciprofloxacin- and/or levofloxacin-resistant infection), metronidazole and/or vancomycin. In another embodiment, the compounds of Formula P, Π" or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form may be useful for a Staphylococcus aureus infection which is resistant to one or more drugs selected from a group consisting of a penicillin, vancomycin, cephalosporin and methicillin. In a particular embodiment, the infection is a methicillin-resistant Staphylococcus aureus infection.
[0016] It will be noted that various compounds of the Invention have a low CC50 value in an assay as disclosed in Example C and therefore, may have anti-metabolite activities which may interfere with DNA biosynthesis. Therefore, in one embodiment, these compounds may be useful as an anti-cancer or anti- viral agent. In another embodiment, the compounds that have a low MIC and/or a high Imax value in an assay as disclosed in Example B and A respectively, and a low CC50 value in an assay as disclosed in Example C are used as an antibacterial, for topical administration.
[0017] In the fourth aspect, the invention provides a method for the treatment of an infection in a plant (Method P-F) comprising administering to such plant an effective amount of a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form.
[0018] In the fifth aspect, the invention provides use of a compound, or use of a pharmaceutical composition comprising a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form, (in the manufacture of a medicament) for the treatment or prophylaxis of an infection, e.g., a bacterial infection as described in Methods P, or any of Methods P-A through P-F.
[0019] In the sixth aspect, the invention provides a pharmaceutical composition comprising a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31 , in free or pharmaceutically acceptable salt form, for use in the treatment of any disease or condition as hereinbefore described, e.g., in any of Methods P or Methods P-A through P-F.
DETAILED DESCRIPTION OF THE INVENTION
[0020] The term "riboswitch" or "riboswitches" is an art recognized term and refers to an mRNA which comprises a natural aptamer that binds target metabolite and an expression platform which changes in the RNA structure to regulate genes. The term "riboswitch ligand" refers to any compound such as a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31 ,, in free or salt form, that binds to that particular riboswitch. For example, "FMN riboswitch" refers to a riboswitch that binds a metabolite such as flavin mono-nucleotide (FMN) or other ligands such as the compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or salt form, and which affects downstream FMN biosynthesis and transport proteins. Without intended to be bound by any particular theory, it is believed the binding of the ligand to its riboswitch induces a conformational change in the bacterial mRNA such that the expression of the ORF is altered, for example, such that the expression of enzymes responsible for, e.g., riboflavin and FMN biosynthesis is repressed or overexpressed.
[0021] "CD3299 riboswitch" refers to a riboswitch found in C. difficile, controlling the gene designated CD3299. The 5'UTR and beginning of ORF from CD3299 gene of C. difficile 630, accession number AM180355 is as follows:
SEQ ID NO: 1 :
TTACAGCTTTCTGATTTTGATAAATTTAAAACTTACCATCTAATACTAAT AACAGGTTAATTTTATCTAATTATTATAGATTCTCATACTGTGCCTTATT CTATCTATAAATACAATTTAAGTGTCCATATTGAAATATTTGTATTGTA ATACAGCTGGATATTACTTAAATCCAATTGTTTCCATTATAATTTTATGT TAAAATAATATTACAAAATACATCTGTTTTTCTTCATAAACGGGTGAA ATTCCCTATCGGCGGTAAAAGCCCGCGAGCCTTATGGCATAATTTG GTCATATTCCAAAGCCAACAGTAAAATCTGGATGGTAGAAGAAAAT AGTATATGAGTACCTTTATGTAATTTTACATGAGTAATCTATACAAATC CTTCAACTACCGTATTTATTCATGAAATTAGACACATTCAAGG7ACC7A ATATACAGGTGCTTTTTTTGTTGTTTATTTTACAATTATATCGTACTTATA AAATCTATTAAGATTGGAGTGTTATCATGAAACAAAAATGGATAGTATT GATTATCATCTGTATTGGTGTATTTATGTCTACTCTTGATGGAAGTATAC T AA AT ATC GC A A A
In the above depiction of the sequence, the riboswitch is highlighted in bold, and is
SEQ ID NO: 2
GTTTTTCTTCATAAACGGGTGAAATTCCCTATCGGCGGTAAAAGCC CGCGAGCCTTATGGCATAATTTGGTCATATTCCAAAGCCAACAGTA AAATCTGGATGGTAGAAGAAAATA The ORF start site in the above sequence is downstream from the riboswitch and is depicted in italics and is:
SEQ ID NO: 3
ATGAAACAAA
The putative terminator hairpin is in bold italics and is:
SEQ ID NO: 4
GTACCTAATATACAGGTGC
The hairpin can form a loop having a structure as depicted in Formula 1 :
Figure imgf000017_0001
_ 40
A possible antiterminator has a structure as depicted in Formula 2:
Figure imgf000017_0002
Various Compounds of the Invention, e.g., Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or salt form, bind well to the CD3299 riboswitch and have antibacterial activity against C. difficile, provided these compounds possess physicochemical characteristics amenable to uptake into the bacteria.
[0022] The term "infection" encompasses an infection by a Gram-positive or Gram- negative bacteria. In one embodiment, the infection is by a Gram-positive bacteria. In another embodiment, the infection is by a Gram-negative bacteria. In still another embodiment, the infection is an infection by one or more bacteria selected from a group consisting of Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii,
Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes, Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borrelia burgdorferi. In a further embodiment, the infection is a Clostridium difficile and/or Staphylococcus aureus infection. In a particular embodiment, the infection is an infection which is resistant to a drug which is not a riboswitch ligand. In a further aspect of this particular embodiment, the infection is an infection which is resistant to one or more drugs selected from a group consisting of penicillin, vancomycin, cephalosporin, methicillin and fluoroquinolone (e.g., ciprofloxacin- and/or levofloxacin). In a particular embodiment, the infection is a methicillin-resistant Staphylococcus aureus (MRSA) infection. In another particular embodiment, the infection is a fluoroquinolone-resistant (e.g., ciprofloxacin- and/or levofloxacin-resistant), metronidazole and/or vancomycin-resistant C. difficile infection.
[0023] The term "bacteria" or "bacterial" include, but are not limited to Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes,
Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borre lia burgdorferi .
[0024] If not otherwise specified or clear from context, the following terms as used herein have the following meetings:
a. "Alkyl" as used herein is a saturated or unsaturated hydrocarbon moiety, preferably saturated, e.g., one to eight, e.g., one to six, e.g., one to four carbon atoms in length, which may be linear or branched (e.g., n-butyl or tert-butyl) unless otherwise specified, and may be optionally substituted, e.g., mono-, di-, or tri-substituted on any one of the carbon atoms, e.g., with Ci_4alkyl (e.g., methyl), Ci-4alkoxy, halogen (e.g., chloro or fluoro), haloCi^alkyl (e.g., trifluoromethyl), hydroxy, and carboxy. For example,
"C1-C8 alkyl" denotes alkyl having 1 to 8 carbon atoms. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, i- butyl, sec -butyl, t-butyl, 3-methylpentyl, 4-methylpentyl, n-pentyl, n-hexyl and n-heptyl. Wherein the alkyl group is unsaturated or partially saturated, it is denoted as "alkenyl" or "alkynyl". Therefore, n-prop-2-en-l-yl is intended to be -CH2-CH=CH2. b. For the avoidance of doubt, the term "alkylene" is intended to denote an alkyl group bridging between two substituents (e.g., between the flavin core structure and another substituent, for example -X-A). Therefore Ci_ 4alkylene, e.g., methylene, ethylene, n-propylene and n-butylene are intended to represent -CH2- -CH2CH2-, -CH2CH2CH2- and - CH2CH2CH2CH2- respectively. Wherein the alkylene group is unsaturated or partially saturated, it is denoted as "alkenylene" or "alkynylene".
Therefore, n-but-2-enylene is intended to be -CH2-CH=CHCH2-.
c. "Aryl" as used herein is a monocyclic or polycyclic aromatic hydrocarbon, preferably phenyl, optionally substituted, e.g., with Ci_4alkyl (e.g., methyl), Ci^alkoxy, halogen (e.g., chloro or fluoro), haloCi^alkyl (e.g., trifluoromethyl), hydroxy, carboxy, or an additional aryl or heteroaryl. d. "Cycloalkyl" refers to a saturated or unsaturated nonaromatic hydrocarbon moiety, preferably saturated, preferably comprising three to eight carbon atoms, at least some of which form a nonaromatic mono- or bicyclic, or bridged cyclic structure.
e. "Heterocycloalkyl" refers to a cycloalkyl as defined above wherein at least one of the carbon atoms is replaced with a heteroatom selected from N, O, S. Therefore, "C3_8heterocycloalkyl" or "heteroC3_8cycloalkyl" refers to a 3- to 8-membered non-aromatic ring system containing at least one heteroatom selected from N, O and S.
f. Wherein the substituent is connected via an alkyl group, e.g., -Co-4alkyl-C3_ gcycloalkyl or aryl-Ci^alkyl, it is understood that the alkyl group may be saturated or unsaturated or linear or branched. Wherein the substituent is connected via the Co-alkyl, it is understood that the alkyl is not present and the connectivity is directly to the next substituent. For example, wherein the substituent is -Coalkyl-C3_8cycloalkyl, it is understood that the alkyl group is not present and the cycloalkyl (e.g., cyclopropyl) is directly connected.
[0025] The Compounds of the Invention or any of the compounds disclosed herein
(e.g. a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31), may exist in free or salt, e.g., as acid addition salts, or prodrug form. An acid-addition salt of a compound of the invention which is sufficiently basic, for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulphuric, phosphoric, acid acetic, trifluoroacetic, citric, maleic acid, toluene sulfonic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, hydroxy maleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic acid, and the like. In addition a salt of a compound of the invention which is sufficiently acidic is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically-acceptable cation, for example a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine. In a particular embodiment, the salt of the compound of the invention is a trifluoroacetic or hydrochloric acid addition salt. In another embodiment, the salt of the compound of the invention is an acetic acid addition salt.
[0026] In this specification, unless otherwise indicated, language such as
Compounds of the Invention is to be understood as embracing the compounds disclosed herein, such as a compound of Formula P, II" or II, e.g., any of formulae 2.1 -2.31 , in any form, for example free or acid addition salt or prodrug form, or where the compounds contain acidic substituents, in base addition salt form. The Compounds of the Invention are intended for use as pharmaceuticals, therefore pharmaceutically acceptable salts are preferred. Salts which are unsuitable for pharmaceutical uses may be useful, for example, for the isolation or purification of free Compounds of the Invention, and are therefore also included.
[0027] The Compounds of the Invention may comprise one or more chiral carbon atoms. The compounds thus exist in individual isomeric, e.g., enantiomeric or diasteriomeric form or as mixtures of individual forms, e.g., racemic/diastereomeric mixtures. Any isomer may be present in which the asymmetric center is in the (R)-, (S)-, or (R,S)- configuration. The invention is to be understood as embracing both individual optically active isomers as well as mixtures (e.g., racemic/diasteromeric mixtures) thereof. Accordingly, the Compound of the Invention may be a racemic mixture or it may be predominantly, e.g., in pure, or substantially pure, isomeric form, e.g., greater than 70% enantiomeric excess ("ee"), preferably greater than 80% ee, more preferably greater than 90% ee, most preferably greater than 95% ee. The purification of said isomers and the separation of said isomeric mixtures may be accomplished by standard techniques known in the art (e.g., column chromatography, preparative TLC, preparative HPLC, simulated moving bed and the like).
[0028] Geometric isomers by nature of substituents about a double bond or a ring may be present in cis (=Z-) or trans (=£"-) form, and both isomeric forms are
encompassed within the scope of this invention.
[0029] As will be appreciated by those skilled in the art, the Compounds of the
Invention may exhibit keto-enol tautomerization. Therefore, the invention as defined in the present invention is to be understood as embracing both the structures as setforth herewith and their tautomeric forms.
[0030] It is also intended that the Compounds of the Invention encompass their stable isotopes. For example, the hydrogen atom at a certain position on the Compounds of the Invention may be replaced with deuterium. It is expected that the activity of compounds comprising such isotopes would be retained and/or it may have altered pharmacokinetic or pharmacodynamic properties. In addition to therapeutic use, compounds comprising such isotopes and having altered pharmacokinetic or
pharmacodynamic properties would also have utility for measuring pharmacokinetics of the non-isotopic analogs.
[0031] Compounds of the Invention may in some cases also exist in prodrug form.
The term "prodrug" is an art recognized term and refers to a drug precursors prior to administration, but generate or release the active metabolite in vivo following
administration, via some chemical or physiological process. For example, when the Compounds of the Invention (e.g., a compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31) contain a hydroxy group, these substituents may be esterified to form physiologically hydrolysable and acceptable esters (e.g., acyl esters, e.g., CH3C(0)-0- Compound). As used herein, "physiologically hydrolysable and acceptable esters" means esters of Compounds of the Invention which are hydrolysable under physiological conditions to yield hydroxy on the one hand and acid, e.g., carboxylic acid on the other (e.g., Drug-0-C(0)-CH3 -» Drug-OH + CH3COOH), which are themselves
physiologically tolerable at doses to be administered. Similarly, wherein the compounds of the invention contain an amine group, prodrug of such amine, e.g., amino
acid, carbamic acid ester, amide prodrugs may also exist wherein the prodrug is cleaved to release the active amine metabolite in vivo following administration. Further details of amine prodrugs may may be found in Jeffrey P. Krise and Reza Oliyai, Biotechnology: Pharmaceutical Aspects, Prodrugs, Volume 5, Part 3, pages 801-831, the contents of which are herein incorporated by reference in their entirety. As will be appreciated, the term thus embraces conventional pharmaceutical prodrug forms. Methods of using Compounds of the Invention
[0032] The Compounds of the Invention are useful for the treatment of an infection, particularly an infection by bacteria including but not limited to Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes, Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and/or Borrelia burgdorferi bacteria. In one particular embodiment, the bacteria is Clostridium difficile. In another particular embodiment, the bacteria ^Staphylococcus aureus.
[0033] The invention therefore provides methods of treatment of any one or more of the following conditions: anthrax infection, staphylococcal scalded skin syndrome (staph infections), pneumonia, impetigo, boils, cellulitis, folliculitis, furuncles, carbuncles, scalded skin syndrome, abscesses, meningitis, osteomyelitis endocarditis, Toxic Shock Syndrome (TSS), septicemia, acute sinusitis, otitis media, septic arthritis, endocarditis, peritonitis, pericarditis, brain abscess, tularemia, urinary tract infection, empyema, food poisoning, diarrhea, conjunctivitis and Clostridium difficile associated disease (CD AD); comprising administering an effective amount of a compound of Formula P, II" or II, e.g., any of formulae 2.1-2.31, in free or pharmaceutically acceptable salt form, to a subject in need thereof.
[0034] The words "treatment" and "treating" are to be understood accordingly as embracing prophylaxis and treatment or amelioration of symptoms of disease as well as treatment of the cause of the disease. In one particular embodiment, the invention encompasses prophylaxis of symptoms of disease or cause of the disease. In another particular embodiment, the invention encompasses treatment or amelioration of symptoms of disease or cause of the disease.
[0035] The term "subject" as used herein encompasses human and/or non-human
(e.g., animal). [031] Dosages employed in practicing the present invention will of course vary depending, e.g. on the particular disease or condition to be treated, the particular
Compound of the Invention used, the mode of administration, and the therapy desired. Administration of a therapeutically active amount of the therapeutic compositions is defined as an amount effective, at dosages and for periods of time necessary to achieve the desired result. For example, a therapeutically effective amount of a Compound of the Invention reactive with at least a portion of the FMN or the CD3299 riboswitch may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regiment may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. In general, satisfactory results, e.g. for the treatment of diseases as hereinbefore set forth are indicated to be obtained on oral administration at dosages of the order from about 0.01 to 2.0 mg/kg. In larger mammals, for example humans, an indicated daily dosage for oral administration will accordingly be in the range of from about 0.75 to 1000 mg, conveniently administered once, or in divided doses 2 to 4 times, daily or in sustained release form. Unit dosage forms for oral administration thus for example may comprise from about 0.2 to 75 mg, 250 mg,1000 mg, e.g. from about 0.2 or 2.0 to 50, 75, 100, 250, 500, 750 or 1000 mg of a Compound of the Invention, together with a pharmaceutically acceptable diluent or carrier therefor.
[0036] Pharmaceutical compositions comprising the Compounds of the Invention may be prepared using conventional diluents or excipients and techniques known in the galenic art. Thus oral dosage forms may include tablets, capsules, solutions, suspensions, spray-dried dispersions [e.g. Eudragit L100] and the like. The term "pharmaceutically acceptable carrier" as used herein is intended to include diluents such as saline and aqueous buffer solutions. The Compounds of the Invention may be administered in a convenient manner such as by injection such as subcutaneous, intravenous, by oral administration, inhalation, transdermal application, intravaginal application, topical application, intranasal, sublingual or rectal administration. Depending on the route of administration, the active compound may be coated in a material to protect the compound from the degradation by enzymes, acids and other natural conditions that may inactivate the compound. In one embodiment, the compound may be orally administered. In another embodiment, the compound is administered via topical application.
[0037] In certain embodiment, the Compounds of the Invention may be administered alone or in conjunction, e.g., at or about the same time or simultaneously and separately or simultaneously in an admixture, with another agent, e.g., an agent to facilitate entry or permeability of the Compounds of the Invention into the cell, e.g., an antimicrobial cationic peptide. Antimicrobial cationic peptides include peptides which contain (1) a disulfide-bonded β-sheet peptides; (2) amphipathic a-helical peptides; (3) extended peptides; or (4) loop-structured peptides. Examples of cationic peptide include but are not limited to defensins, cecropins, melittins, magainins, indolicidins, bactenecin and protegrins. Other examples of antimicrobial cationic peptides include but are not limited to human neutrophil defensin-1 (HNP-1), platelet microbicidal protein- 1 (tPMP), inhibitors of DNA gyrase or protein synthesis, CP26, CP29, CP11CN, CPIOA, Bac2A- NH2 as disclosed in Friedrich et al., Antimicrob. Agents Chemother. (2000) 44(8):2086, the contents of which are hereby incorporated by reference in its entirety. Further examples of antibacterial cationic peptides include but are not limited to polymyxin e.g., polymixin B, polymyxin E or polymyxin nonapeptide. Therefore, in another embodiment, the
Compounds of the Invention may be administered in conjunction with polymyxin, e.g., polymyxin B, polymyxin E or polymyxin nonapeptide, preferably polymyxin B.
[0038] In still another embodiment, the Compounds of the Invention may be administered alone or in conjunction, e.g., at or about the same time, simultaneously and separately, or simultaneously in an admixture, with other antimicrobial agents, e.g., other antifungal or other systemic antibacterial (bactericidal or bacteriostatic) agents. Examples of bacterial agents include agents which inhibit bacterial cell wall synthesis (e.g., penicillins, cephalosporins, carbapenems, vancomycin), agents which damage cytoplasmic membrane (e.g., polymixins as discussed above), agents which modify the synthesis or metabolism of nucleic acids (e.g., quinolones, rifampin, nitrofurantoin), agents which inhibit protein synthesis (aminoglycosides, tetracyclines, chloramphenicol, erythomycin, clindamycin), agents which interfer with the folate synthesis (e.g., folate-inhibitors), agents which modify energy metabolism (e.g., sulfonamides, trimethoprim) and/or other antibiotics (beta-lactam antibiotic, beta-lactamase inhibitors). Specific anti-infective agents, particularly antibacterial and antifungal agents, are discussed in Remington: The Science and Practice of Pharmacy, Chapter 90, pp. 1626-1684 (21s Ed., Lippincott Williams & Wilkins 2005), the contents of which are hereby incorporated by reference.
Methods of making the Compounds of the Invention:
[0039] The compounds of the Invention, e.g., compound of Formula P, IF ' or II, e.g., any of formulae 2.1-2.31 , in free or salt form may be made using the methods as described and exemplified herein and by methods similar thereto and by methods known in the chemical art. Such methods include, but not limited to, those described below. In the description of the synthetic methods described herein, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, are chosen to be the conditions standard for that reaction, which should be readily recognized by one skilled in the art. Therefore, at times, the reaction may require to be run at elevated temperature or for a longer or shorter period of time. It is understood by one skilled in the art of organic synthesis that functionality present on various portions of the molecule must be compatible with the reagents and reactions proposed. If not commercially available, starting materials for these processes may be made by procedures, which are selected from the chemical art using techniques which are similar or analogous to the synthesis of known compounds. All references cited herein are hereby incorporated by reference in their entirety.
[0040] The synthetic methods for the Compounds of the Invention are illustrated below either in the generic synthetic scheme and/or in the specific Examples, which methods are claimed individually and/or collectively. The significances for the substituents are as set forth above in Formula P, II" or II, e.g., any of formulae 2.1-2.31, unless otherwise indicated.
[0041] Generally, the compounds of Formula P, II" or II, e.g., any of formulae
2.1-2.31 may be prepared by reacting Intermediate-5 (Int-5) with ammonia in a pressure tube. Int-5 may be prepared by reacting Intermediate-4 (Int-4) with diethyl 2-bromo-3- oxopentanedioate in the presence of a base, e.g., cesium carbonate, in a solvent, for example, a mixture of dimethylformamide (DMF) and methylene chloride (CH2CI2). Int- 4 may be may be prepared by converting Intermediate-3 (Int-3) to Int-4, for example, by catalytic hydrogenation, e.g., by reacting Int-3 with a metal, e.g., Raney-Nickel, and hydrogen gas in a solvent such as ethanol. In turn, Int-3 may be prepared by reacting Intermediate-1 (Int-1) with NH2-Alk-X-A (Int-2), wherein Alk, X and A are defined in Formula P, IF ' or II or any of 2.1-2.31 to yield Int-3. Int-1 is either commercially available or may be prepared by methods known in the art. Wherein R2 of compounds of Formula P, IF ' or II is alkoxy, this compound may be prepared by reacting a compound of Formula P, IF ' or II, wherein R2 is halo, e.g., chloro, with R2-H, e.g., methanol, in the presence of a base. The methods for preparing a compound of Formula P, IF ' or II may be described in the reaction scheme below, wherein all substituents are defined in Formula P II" or II or any of 2.1-2.31:
Figure imgf000026_0001
[0042] Wherein R2 of the compounds of Formula P, IF ' or II is (Ci^alkoxy)- methyl, these compounds may be prepared by first halogenating the compound of Formula P, IF ' or II, wherein R2 is methyl, for example by reacting such compound with a halogen, e.g., bromine, e.g., optionally in the presence of a catalyst such as
azobisisobutyronitrile (AIBN). The resulting intermediate, Int-6, may then react with a R2-H, wherein R2-H is e.g. methanol, in the presence of a base to provide the
corresponding alkoxy-methyl product.
[0043] Wherein R2 of the compounds of Formula P, IF ' or II is -methyl-N(Ra)(Rb), e.g., -CH2-N(CH3)2, this compound may be prepared by halogenating the compounds of Formula P, IF ' or II, wherein R2 is e.g., a methyl group, for example by reacting bromine with the compounds of Formula P, IF ' or II, wherein R2 is methyl, optionally in the presence of a catalyst such as azobisisobutyronitrile (AIBN). The resulting intermediate, Int-6, may then react with an amine, HN(Ra)(Rb), e.g. HN(CH3)2, to provide a Compound of Formula P, IF ' or II wherein R2 is -methyl-N(Ra)(Rb), e.g., -CH2-N(CH3)2. This reparation may be summarized in the following reaction scheme:
Figure imgf000027_0001
Formula P, II" or II
[0044] Wherein R2 of the compounds of Formula P, II" or II is hydrogen, these compounds may be prepared by heating Intermediate- 10 (Int-10) with acid [e.g.
methanesulfonic acid] in toluene. Int-10 may be prepared by heating Intermediate-9 (Int- 9) with sodium dithionite in a mixture of ethanol and water. Intermediate-9 (Int-9) may be prepared by treating Intermediate-8 (Int-8) with sodium nitrite in acetic acid with cooling. Int-8 may be prepared by condensing Intermediate-7 (Int-7) with methyl 5-[(4- methoxybenzyl)amino]-3,5-dioxopentanoate in the presence of acid [e.g. p-toluenesulfonic acid monohydrate] in toluene. In turn, Intermediate-7 (Int-7) may be prepared with Br- Alk-X-Ar , wherein Alk, X, and A are defined in any of Formulae P, II", or II. This preparation may be summarized in the following reaction scheme:
Figure imgf000028_0001
lnt-7 I
Ar
Figure imgf000028_0002
r lnt-i Ar
Figure imgf000028_0003
Formula P, II" or II
Examples: Binding of ligand to riboswitch
Example A:
[0045] An in-line probing assay, as described in Regulski and Breaker, "In-line probing analysis of riboswitches", (2008), Methods in Molecular Biology, Vol 419, pp 53- 67, the contents of which are incorporated by reference in their entirety, is used to estimate the dissociation binding constants for the interaction of each of the ligands described herein with either an FMN riboswitch amplified from the genome of Bacillus subtilis or a CD3299 riboswitch amplified from Clostridium difficile. Precursor mRNA leader molecules are prepared by in vitro transcription from templates generated by PCR and [5'- 32P] -labeling using methods described previously (Regulski and Breaker, In-line probing analysis of riboswitches (2008), Methods in Molecular Biology Vol 419, pp 53-67).
Approximately 5 nM of labeled RNA precursor is incubated for 41 hours at 25°C in 20 mM MgCl2, 50 mM Tris/HCl (pH 8.3 at 25°C) in the presence or absence of a fixed concentration of each ligand. Binding to the FMN and CD3299 riboswitches are measured at 20 μΜ and 100 μΜ, respectively. In-line cleavage products are separated on 10% polyacrylamide gel electrophoresis (PAGE), and the resulting gel is visualized using a Molecular Dynamics Phosphorimager. The location of products bands corresponding to cleavage are identified by comparison to a partial digest of the RNA with RNase Tl (G- specific cleavage) or alkali (nonspecific cleavage).
[0046] In-line probing exploits the natural ability of RNA to self-cleave at elevated pH and metal ion concentrations (pH ~ 8.3, 25 mM MgCl2) in a conformation-dependent manner. For self-cleavage to occur, the 2'-hydroxyl of the ribose must be "in-line" with the phosphate-oxygen bond of the internucleotide linkage, facilitating a S 2P nucleophilic transesterification and strand cleavage. Typically, single-stranded regions of the riboswitch are dynamic in the absence of an active ligand, and the internucleotide linkages in these regions can frequently access the required in-line conformation. Binding of an active ligand to the riboswitch generally reduces the dynamics of these regions, thereby reducing the accessibility to the in-line conformation, resulting in fewer in-line cleavage events within those regions. These ligand-dependent changes in RNA cleavage can be readily detected by denaturing gel electrophoresis. The relative binding affinity of each ligand is expressed as Imax, wherein Imax represents the percent inhibition of in-line cleavage at selected internucleotide ligands in the presence of a fixed ligand concentration (20 μΜ for the FMN riboswitch and 100 μΜ for the CD3299 riboswitch) normalized to the percent inhibition in the absence of ligand and the percent inhibition in the presence of a saturation concentration of a control ligand. 100 μΜ FMN is used as a control ligand for estimating binding to the FMN riboswitch and 100 μΜ of 7,8-dimethyl-10-(3- phenylpropyl)benzo[g]pteridine-2,4(3H,10H)-dione (which is a compound which has a high affinity against the CD3299 riboswitch) is used as a control ligand for estimating binding to the CD3299 riboswitch.
[0047] The experiments show that the exemplified Compounds of the Invention, e.g., compounds of Examples 1-16 have a binding affinity to the FMN riboswitch with an Imax value of greater than 20%, some up to80% compared to the control at 20 μΜ. In other instances, various compounds of the invention have a binding affinity to the CD3299 switch with an Imax value of greater than 20% compared to the control at ΙΟΟμΜ.
MIC Assay
Example B: [0048] The MIC assays are carried out in a final volume of 100 in 96-well clear round-bottom plates according to methods established by the Clinical Laboratory
Standards Institute (CLSI). Briefly, test compound suspended in 100 % DMSO (or another suitable solubilizing buffer) is added to an aliquot of media appropriate for a given pathogen to a total volume of 50 μL·. This solution is serially diluted by 2-fold into successive tubes of the same media to give a range of test compound concentrations appropriate to the assay. To each dilution of test compound in media is added 50 μΐ of a bacterial suspension from an overnight culture growth in media appropriate to a given pathogen. Final bacterial inoculum is approximately 105-106 CFU/well. After growth for 18-24 hours at 37° C, the MIC is defined as the lowest concentration of antimicrobial agent that completely inhibits growth of the organism as detected by the unaided eye, relative to control for bacterial growth in the absence of added antibiotic. Ciprofloxacin is used as an antibiotic-positive control in each screening assay. Each of the bacterial cultures that are available from the American Type Culture Collection (ATCC, www.atcc.org) is identified by its ATCC number.
[0049] The experiments show that the exemplified compounds of the invention, e.g., the compounds of Examples 1-16 have a minimum inhibitory concentration (MIC) of less than 2 μg/mL against at least one of the bacteria selected from C. difficile MMX3581 (clinical), C. difficile ATCC 43596, C. difficile ATCC 700057 (MMX 4381), C. difficile ATCC BAA-1805 (NAP1), C. difficile ATCC BAA-1382 (MMX4820), C. difficile 43255 (MMX4821), C. difficile ATCC BAA-1803 (NAP1) and C. difficile ATCC BAA-1870 (NAP1).
[0050] All of the exemplified compounds of the invention have either an Imax value of greater than 20% in an assay as described in Example A (compared to at least one of the two controls) and/or a MIC of less than or equal to 2μg/mL against at least one of the bacterial strains as decribed in Example B. In certain embodiment, certain compounds of the invention have either an Imax value of greater than 50% in an assay as described in Example A (compared to at least one of the two controls) and/or a MIC of less than or equal to 2μg/mL against at least one of the bacterial strains as decribed in Example B.
Cytotoxic Assay
Example C: [0051] The cytotoxic effects of test compounds on HepG2 are measured with a commercially available cell viability assay kit from Promega. On day 1, HepG2 cells (~1 x 104 cells) are seeded into each well in 96-well plate and cultured for approximately 24 h at 37°C in a 5% C02 atmosphere under saturating humidity. On day 2, test compounds and DMSO controls are added to appropriate wells to give a range of test compound concentrations appropriate to the assay. Terfenadine is also added to each plate as a positive cytotoxic control. Control wells containing medium without cell are prepared to obtain a value for background luminescence. Assay plates are then cultured for approximately 24 h at 37°C in a 5% C02 atmosphere under saturating humidity. On day 3, assay plates are removed from 37°C incubator and equilibrated to 22°C. Once equilibrated, CellTiter-Glo® reagent is added to each well containing cell culture medium, followed by mixing to allow cell lysis. The CellTiter-Glo® Assay measures the number of viable cells in culture based on quantitation of the ATP present, an indicator of metabolically active cells. This assay generates a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly proportional to the number of cells present in culture. After the assay plate is incubated at room temperature for
approximately 10 min to stabilize luminescent signal, luminescence is recorded on PerkinElmer luminometer. CC50 is defined as the concentration of test compounds in μΜ to result in 50% reduction in luminescence signal relative to the signal for untreated cells.
[0052] The experiments show that the exemplified compounds of the invention have a CC50 value of greater than or equal to 19 μΜ. The exemplified compounds of the invention generally have a MIC to cytotox ratio of at least 1:20.
Synthesis of the Compounds of the Invention:
[0053] Temperatures are given in degrees Celsius (°C); unless otherwise stated, operations are carried out at room or ambient temperature, that is, at a temperature in the range of 18-25 °C. Chromatography means flash chromatography on silica gel; thin layer chromatography (TLC) is carried out on silica gel plates. NMR data is in the delta values of major diagnostic protons, given in parts per million (ppm) relative to the deuterium lock signal of the deuterated solvent utilized. Conventional abbreviations for signal shape are used. For mass spectra (MS), the lowest mass major ion is reported for molecules where isotope splitting results in multiple mass spectral peaks. Solvent mixture compositions are given as volume percentages or volume ratios. In cases where the NMR spectra are complex, only diagnostic signals are reported.
HPLC Methods:
[0054] Method D: Agilent 1100 HPLC, Agilent XDB C18 50 x 4.6 mm 1.8 micron column, 1.5 mL/min., Solvent A-Water (0.1% TFA), Solvent B -Acetonitrile (0.07% TFA), Gradient -5 min. 95%A to 95%B; lmin. hold; then recycle, UV Detection @ 210 and 254nm.
[0055] Method G: Agilent 1100 HPLC, Agilent XDB C18 50 x 4.6 mm 5 micron column, 1.5 mL/min., Solvent A- Water (0.1% TFA), Solvent B -Acetonitrile (0.07% TFA), Gradient -6 min. 95%A to 95%B; 1 min. hold; then recycle, UV Detection @ 210 and 250nm.
Terms and abbreviations: br = broad,
CH3CN = acetonitrile,
d = doublet,
CH2C12 or DCM = dichloromethane,
DMF = NN-dimethylforamide,
DCM = dichloromethane
DMSO = dimethyl sulfoxide,
Et20 = diethyl ether,
EtOAc = ethyl acetate,
h = hour(s),
hep = heptet,
Hex = hexane,
HPLC = high performance liquid chromatography,
m = multiplet,
min. = minute(s)
MeOH = methanol,
NaHCC = sodium bicarbonate,
Na2S04 = sodium sulfate, NH3 = ammonia gas,
NMR = nuclear magnetic resonance,
p = pentet,
PMB = para-methoxybenzyl
rt = room temperature,
RNA = ribonucleic acid,
RNase Tl = an endoribonuc lease that specifically degrades single-stranded RNA at G residues,
s = singlet,
t = triplet,
THF = tetrahydrofuran,
Example 1
7,8-Dimethyl-5-(3-phenylpropyl)pyridor3,4-b1auinoxaline-l,3(2H,5H)-dione
Figure imgf000033_0001
Step 1 Preparation of ethyl (3Z)-3-(2-ethoxy-2-oxoethylidene)-6 -dimethyl-4-(3-
Figure imgf000033_0002
[0056] Cesium carbonate (5.38 g, 16.5 mmol) is added to a solution of 4,5- dimethyl-N-(3-phenylpropyl)benzene-l,2-diamine (0.600 g, 2.36 mmol) in 40 mL of 1:1 DMF / CH2CI2 followed by diethyl 2-bromo-3-oxopentanedioate (4.64 g, 16.5 mmol) and the mixture is stirred at rt under N2 overnight. The mixture is evaporated to dryness and the residue is partitioned between 50 mL of CH2CI2 and 50 mL of water. The layers are separated and the aqueous phase is extracted with 2 x 50 mL of CH2CI2. The combined organic layers are extracted with 3 x 50 mL of water. Drying over Na2S04 and evaporation gives 3.5 g of a red oil. Chromatography on 150 g of silica gel in 30% EtOAc / hexane gives desired product (0.57 g, 56%) as a red solid. ]H NMR (400 MHz, CDC13) δ ppm 7.53 (s, 1 H), 7.38 (d, 1 H), 7.41 (d, 1 H), 7.27 - 7.34 (m, 3 H), 6.49 (s, 1 H), 5.06 (s, 1 H), 4.40 (q, 2 H), 4.15 (q, 2 H), 3.80 (q, 2 H), 2.84 (q, 2 H), 2.24 (m, 6 H), 2.15 (m, 2 H), 1.43 (t, 3 H), 1.31 (t, 3 H); MS (ESI+) for C26H30N2O4 m/z 435 (M+H)+.
Step 2 Preparation of 7,8-dimethyl-5-(3-phenylpropyl)pyrido[3,4-blauinoxaline- l,3(2H,5H)-dione
Figure imgf000034_0001
[0057] Ethyl (3Z)-3-(2-ethoxy-2-oxoethylidene)-6,7-dimethyl-4-(3-phenylpropyl)-
3,4-dihydroquinoxaline-2-carboxylate (0.300 g, 0.690 mmol) is taken up in 20 mL of MeOH and the solution is cooled in an ice water bath. Ammonia gas is bubbled through the solution for 5 minutes in a pressure tube. The solution is stirred at rt overnight in the capped pressure tube. The pressure tube is opened slowly to allow NH3 to evolve. The remaining solution is evaporated to give 0.2 g of a dark solid. The solid is adsorbed onto silica gel and chromatographed on 50 g of silica gel. The column is eluted with 1% MeOH
/DCM (1L) followed by 1.5 % MeOH / CH2C12 (1.5L). The product elutes in the 1.5% MeOH / DCM. Evaporation of the fractions containing product gives 0.07 g of a purple solid. Crystallization from CH3CN (25 mL) gives 12 mg of the product as a purple solid. 1H NMR (400 MHz, DMSO- ) δ ρριη 11.17 (s, 1 H), 7.63 (s, 1 H), 7.20 - 7.36 (m, 6 H), 5.27 (d, 1 H), 4.01 (br s, 2 H), 2.82 (t, 2 H), 2.35 (s, 3 H), 2.29 (s, 3 H), 1.93 (br s, 2 H); MS (ESI+) for C22H21N3O2 m/z 360 (M+H)+, HPLC retention time: 3.84 min. (Method D) Example 2
5-[3-(4-fluorophenyl)butyll-8-propylpyrido[3,4-/>lauinoxaline-l,3(2H,5H)-dione
Figure imgf000035_0001
Step 1 Preparation of A^3- -Fluorophenyl)butyl1-4-propylaniline
Figure imgf000035_0002
[0058] A solution of 4-propylaniline (0.18 rriL, 1.2 mmol) and l-(3-bromo-l- methylpropyl)-4-fluorobenzene (0.25 g, 1.1 mmol) in toluene (4.8 rriL) is treated with sodium bicarbonate (0.18 g, 2.1 mmol) and tetra-n-butylammonium iodide (0.040 g, 0.11 mmol). The pale yellow reaction mixture is heated at 80 °C for 24.5 h and at reflux (113 °C) for 4 h. The reaction mixture is cooled to rt and partitioned between water (10 rriL) and EtOAc (10 rriL). The separated aqueous layer is extracted with EtOAc (3 x 10 rriL) and the combined organics are washed with brine (1 x 25 iriL), dried (Na2S04), and concentrated under reduced pressure to afford a tan oil. Purification by column chromatography (2 x 22 cm silica; Hex, 1: 1 :98 Et20/CH2C12/Hex, 2:2:96
Et20/CH2C12/Hex) afforded the title compound as a yellow oil (0.21 g, 68%). ]H NMR (400 MHz, CDC13) δ 7.22-7.15 (m, 2H), 7.05-6.94 (m, 4H), 6.51-6.45 (m, 2H), 3.40 (br s, 1H), 3.08-2.94 (m, 2H), 2.93-2.81 (m, 1H), 2.52-2.44 (m, 2H), 1.97-1.80 (m, 2H), 1.66- 1.51 (m, 2H), 1.29 (d, 3H), 0.93 (t, 3H); MS (ESI+) for Ci9H24FN m/z 286.2 (M+H)+; HPLC retention time: 3.79 min (Method D).
Step 2 Preparation of 4-{[3-(4-Fluorophenyl)butyll(4-propylphenyl)amino}-l-(4- methoxybenzyl)pyridine-2,6(lH,3H)-dione
Figure imgf000036_0001
[0059] A slurry of N-[3-(4-fluorophenyl)butyl]-4-propylaniline (0.16 g, 0.56 mmol) and methyl 5-[(4-methoxybenzyl)amino]-3,5-dioxopentanoate (0.19 g, 0.67 mmol) in toluene (60 mL) is treated with p-toluenesulfonic acid monohydrate (9.3 mg, 0.049 mmol). The reaction mixture is heated at reflux (131 °C) for 18 h and allowed to cool to rt. The reaction mixture is concentrated under reduced pressure and purified by column chromatography (2 x 22 cm silica; Hex, 5%, 10%, 20%, 30% EtOAc/Hex) to afford the title compound as a light tan oil (0.21 g, 73%); ]H NMR (400 MHz, CDC13) δ 7.41-7.35 (m, 2H), 7.23-7.17 (m, 2H), 7.07-7.00 (m, 2H), 6.99-6.90 (m, 4H), 6.83-6.77 (m, 2H), 5.05 (s, 1H), 4.92 (s, 2H), 3.76 (s, 3H), 3.47-3.36 (m, 1H), 3.34-3.23 (m, 1H), 3.19-3.05 (m, 2H), 2.70-2.56 (series of m, 3H), 1.95-1.79 (m, 2H), 1.71-1.59 (m, 2H), 1.21 (d, 3H), 0.97 (t, 3H); MS (ESI+) for C32H35FN2O3 m/z 515.2 (M+H)+; MS (ESI-) for C32H35FN2O3 m/z 513.3 (M-H) ; HPLC retention time: 5.50 min. (Method D).
Step 3 Preparation of 5-[3-(4-Fluorophenyl)butyll-2-(4-methoxybenzyl)-8- propylpyridor3,4-/>1auinoxaline-l,3(2H,5H)-dione 10-oxide
Figure imgf000036_0002
[0060] A solution of 4-{ [3-(4-fluorophenyl)butyl](4-propylphenyl)amino}-l-(4- methoxybenzyl)pyridine-2,6(lH,3H)-dione (0.21 g, 0.41 mmol) in acetic acid (2.4 mL) is cooled to 15 °C (ice water bath). Sodium nitrite (34 mg, 0.49 mmol) is added in one portion and the cold bath is removed. The bright red-orange reaction mixture is stirred for 20 min. to afford a dark purple mixture. At 30 min., the reaction mixture is poured into ice water (20 mL) to afford a finely dispersed purple solid. The solid remaining in the reaction flask is taken up in CH2CI2 (30 mL) and added to the aqueous mixture. The separated aqueous layer is extracted with CH2CI2 (1 x 20 mL), neutralized with NaHCC>3 (solid), and extracted with CH2CI2 (3 x 10 mL). The combined organic layers are washed with saturated aqueous NaHCC>3 (1 x 30 mL), brine (1 x 30 mL), dried (Na2S04) and concentrated under reduced pressure to afford a dark purple, glassy residue. Purification by column chromatography (5 x 17 cm silica; Hex, 10%, 20%, 40%, 50%, 60%
EtOAc/Hex) afforded the title compound as a deep violet oil (60 mg, 30%); ]H NMR (400 MHz, CDCI3) δ 8.18-8.13 (m, 1H), 7.55-7.49 (m, 2H), 7.36-7.30 (m, 1H), 7.30-7.23 (m, 3H), 7.14-7.06 (m, 2H), 6.84-6.78 (m, 2H), 6.74-6.60 (m, 1H), 5.27 (s, 1H), 5.14 (s, 2H), 3.88-3.73 (m, 1H), 3.76 (s, 3H), 3.70-3.58 (m, 1H), 2.95-2.84 (m, 1H), 2.64 (t, 2H), 2.16- 1.87 (series of m, 2H), 1.70-1.57 (m, 2H), 1.39 (d, 3H), 0.93 (t, 3H); MS (ESI+) for C32H32FN304 m/z 542.3 (M+H)+; MS (ESI-) for C32H32FN304 m/z 586.3 (M+HC02) ; HPLC retention time: 5.08 min. (Method D).
Step 4 Preparation of 5-r3-(4-fluorophenyl)butyl1-2-(4-methoxybenzyl)-8- propylpyrido[3,4-/>lauinoxaline-l,3(2H,5H)-dione
Figure imgf000037_0001
[0061] A solution of 5-[3-(4-fluorophenyl)butyl]-2-(4-methoxybenzyl)-8- propylpyrido[3,4-^]quinoxaline-l,3(2H,5H)-dione 10-oxide (60 mg, 0.11 mmol) in ethanol (2 mL)/THF (4 mL)/water (2 mL) is heated to 50 °C to afford a homogeneous solution. Sodium dithionite (0.038 g, 0.22 mmol) is added to afford a red-purple reaction mixture, which is heated at 55 °C for 1 h 10 min. and allowed to cool to rt. The reaction mixture is partitioned between water (20 mL) and CH2CI2 (25 mL). The separated aqueous layer is extracted with CH2CI2 (2 x 10 mL) and the combined organic layers are washed with brine (10 mL), dried (Na2S04), and concentrated under reduced pressure to afford the title compound as a red-purple foam, which is carried on without further purification; ]H NMR (400 MHz, CDC13) δ 7.85-7.80 (m, 1H), 7.55-7.48 (m, 2H), 7.36-7.30 (m, 1H),
7.28-7.22 (m, 2H), 7.14-7.06 (m, 2H), 6.85-6.78 (m, 2H), 6.78-6.70 (m, 1H), 5.30 (s, 1H), 5.16 (s, 2H), 3.84-3.58 (series of m, 2H), 3.76 (s, 3H), 2.95-2.83 (m, 1H), 2.63 (t, 2H), 2.11-1.88 (series of m, 2H), 1.72-1.60 (m, 2H), 1.36 (d, 3H), 0.93 (t, 3H); MS (ESI+) for
C32H32FN3O3 m/z 526.2 (M+H)+; HPLC retention time: 5.21 min. (Method D).
Step 5 Preparation of 5-[3-(4-fluorophenyl)butyll-8-propylpyrido[3,4-/>lauinoxaline- l,3(2H,5H)-dione
Figure imgf000038_0001
[0062] A solution of the 5-[3-(4-fluorophenyl)butyl]-2-(4-methoxybenzyl)-8- propylpyrido[3,4-Z?]quinoxaline-l,3(2H,5H)-dione in methanesulfonic acid (2.2 mL, 33 mmol) is diluted with toluene (5.7 mL) and the biphasic mixture is heated at 50 °C for 5 h. The reaction mixture is diluted with water (30 mL) and CH2CI2 (70 mL) and the mixture is poured over ice and adjusted to pH 8 with solid NaHCC>3. The separated aqueous layer is extracted with CH2CI2 (3 x 20 mL) and the combined organics are dried (Na2S04) and concentrated under reduced pressure to afford a red-purple solid. Purification by column chromatography (4 x 15 cm silica; Hex, 20%, 40%, 50%, 60%, 70%, 80%, 90%
EtO Ac/Hex doped with 0.1% i-PrOH) afforded the title compound as a dark purple amorphous solid (44 mg, 98%); ]H NMR (400 MHz, DMSO-i¾ δ 11.22 (s, IH), 7.68-7.63 (m, IH), 7.54-7.48 (m, IH), 7.41-7.32 (m, 2H), 7.31-7.20 (m, IH), 7.20-7.11 (m, 2H), 5.05 (br s, IH), 4.12-3.98 (m, IH), 3.67-3.54 (m, IH), 3.09-2.99 (m, IH), 2.64 (t, 2H), 1.96- 1.80 (m, 2H), 1.68-1.56 (m, 2H), 1.29 (d, 3H), 0.90 (t, 3H); MS (ESI+) for C24H24FN302 m/z 406.1 (M+H)+; MS (ESI-) for C24H24FN302 m/z 404.2 (M-H) , 450.1 (M+HC02) ; HPLC retention time: 4.29 min. (Method D).
[0063] The compounds of the invention particularly those compounds as set forth in Table 1 below which are disclosed and claimed either individually and/or collectively may generally be prepared using similar procedures as set forth in Examples 1-2 above. It is to be understood that the appropriate reagents, solvents and reaction condition for those reactions are used as apparent to one skilled in the art.
Table 1
Figure imgf000039_0001
dione
Figure imgf000040_0001
Figure imgf000041_0001

Claims

A compound of
Figure imgf000042_0001
ormu a wherein:
(i) Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene) optionally substituted with one or more Ci_6alkyl (e.g., methyl) or one hydroxy or Ci_ 4alkoxy (e.g., ethoxy) group;
(ϋ) X is a single bond, -S- or -0-;
(iii) A is aryl (e.g., phenyl) or heteroaryl (e.g. pyridinyl) optionally substituted with one or more Ci^alkyl (e.g., methyl), Ci_4alkoxy (e.g., methoxy), hydroxy, halo (e.g., CI, F), haloCi^alkyl (e.g., CF3) or -0-haloCi_4alkyl (e.g., -OCF3);
(iv) Ri is H, Ci_4alkyl (e.g., methyl, ethyl, n-propyl or isopropyl), or Ci_4alkoxy (e.g., methoxy);
(v) P 2 is H, Ci_4alkyl (e.g., methyl or n-propyl), Ci_4alkoxy (e.g., methoxy), halo (e.g., CI), C3-8cycloalkyl-Ci_4alkyl, -Ci^alkyl-N(Ra)(Rb), (Ci^alkoxy)- Ci^alkyl or (2-Ci_4alkoxyethoxy)-Ci^alkyl;
(vi) R3 is H or Ci^alkyl (e.g., methyl);
(vii) R4 is H or Ci^alkyl (e.g., methyl);
(viii) Ra and Rb are independently H, Ci^alkyl (e.g., methyl) or C3_8cycloalkyl (e.g., cyclopropyl, cyclopentyl);
in free or salt form.
2. The compound according to claim 1, wherein X is a single bond which compound is represented by a compound of formula ΙΓ:
Figure imgf000043_0001
Formula ΙΓ wherein:
(i) Alk is Ci_6alkylene (e.g., n-propylene, n-butylene, n-pentylene)
optionally substituted with one or more Ci_6alkyl (e.g., methyl) or one Ci-4alkoxy (e.g., ethoxy) group;
(ϋ) A is aryl (e.g., phenyl) optionally substituted with one or more Ci^alkyl (e.g., methyl) or halo (e.g., CI, F);
) Ri is H or Ci^alkyl (e.g., methyl, ethyl, n-propyl or isopropyl);
(iv) R2 is H or Ci-4alkyl (e.g., methyl or n-propyl);
(v) R3 is H;
(vi) Rt is H;
in free or salt form.
3. The compound according to claim 1 or 2 selected from any one of the following:
Figure imgf000043_0002
Figure imgf000044_0001
Figure imgf000045_0001
A pharmaceutical composition comprising the compound according to any one of claims 1-3, in free or pharmaceutically acceptable salt form, in admixture with a pharmaceutically acceptable diluent or carrier.
A method for the treatment or prophylaxis of a bacterial infection comprising administering to a patient in need of such treatment an effective amount of a compound according to any one of claims 1-3, in free or pharmaceutically acceptable salt form.
The method according to any one of claims 5, wherein the infection is a Gram- positive or Gram-negative bacterial infection.
The method according to any one of claims 5-6, wherein the bacterial infection is selected from a group consisting of Clostridium difficile, Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Haemophilus influenzae, Enterococcus faecalis, Streptococcus pyogenes, Listeria monocytogenes, Salmonella enterica, Vibrio cholerae, Brucella melitensis, Bacillus anthracis, Francisella tularensis, Moraxella catarrhalis, Klebsiella pneumoniae, Yersinia pestis, Streptococcus viridans, Enterococcus faecium, and Borrelia burgdorferi .
The method according to any one of claims 5-7, wherein the bacterial infection is a C. difficile infection.
The method according to any one of claims 5-7, wherein the bacterial infection is a Staphylococcus aureus infection.
The method according to any of claims 5-9, wherein said infection is by an infectious agent which is resistant to a drug that is not a riboswitch ligand.
The method according to claim 9 or 10, wherein the infection is an infection which is resistant to one or more drugs selected from a group consisting of a penicillin, vancomycin, cephalosporin and methicillin.
The method according to claim 11, wherein the infection is a methicillin-resistant Staphylococcus aureus infection.
The method according to claim 8 or 10, wherein the infection is by an infectious agent which is resistant to fluoroquinolone (e.g., ciprofloxacin- and/or
levofloxacin-resistant infection), metronidazole and/or vancomycin.
The method according to any one of claims 5-13, wherein such method is effective for the treatment or prophylaxis of a disease, condition or infection selected from a group consisting of anthrax, staphylococcal scalded skin syndrome (staph infections), pneumonia, impetigo, boils, cellulitis, folliculitis, furuncles, carbuncles, scalded skin syndrome, abscesses, meningitis, osteomyelitis endocarditis, Toxic Shock Syndrome (TSS), septicemia, acute sinusitis, otitis media, septic arthritis, endocarditis, peritonitis, pericarditis, brain abscess, tularemia, urinary tract infection, empyema, food poisoning, diarrhea, conjunctivitis and Clostridium difficile associated disease (CD AD).
A method for the treatment or prophylaxis of a bacterial infection in a plant comprising administering to said plant an effective amount of a compound of any one of claims 1-3, in free or pharmaceutically acceptable salt form. Use of a compound according to any one of claims 1-3, in free or pharmaceutically acceptable salt form in the manufacture of a medicament for the treatment or prophylaxis of a bacterial infection as described in any one of claims 5-15.
A pharmaceutical composition according to any one of claims 1-3, in free or pharmaceutically acceptable salt form for use in the manufacture of a medicament for the treatment or prophylaxis of a bacterial infection as described in any one of claims 5-15.
PCT/US2012/024507 2011-02-09 2012-02-09 Flavin derivatives WO2012109458A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201161441160P 2011-02-09 2011-02-09
US61/441,160 2011-02-09
US201161516651P 2011-04-06 2011-04-06
PCT/US2011/000617 WO2011126567A1 (en) 2010-04-06 2011-04-06 Flavin derivatives
USPCT/US2011/000617 2011-04-06
US61/516,651 2011-04-06

Publications (1)

Publication Number Publication Date
WO2012109458A1 true WO2012109458A1 (en) 2012-08-16

Family

ID=46638958

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/024507 WO2012109458A1 (en) 2011-02-09 2012-02-09 Flavin derivatives

Country Status (1)

Country Link
WO (1) WO2012109458A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3920650A (en) * 1973-09-19 1975-11-18 Morton Norwich Products Inc Isoalloxazines
WO2010019208A1 (en) * 2008-08-11 2010-02-18 Biorelix Pharmaceuticals, Inc. Flavin derivatives

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3920650A (en) * 1973-09-19 1975-11-18 Morton Norwich Products Inc Isoalloxazines
WO2010019208A1 (en) * 2008-08-11 2010-02-18 Biorelix Pharmaceuticals, Inc. Flavin derivatives

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CARLSON ET AL.: "Improved Chemical Syntheses of 1- and 5-Deazariboflavin", J. ORG. CHEM., vol. 69, 2004, pages 2614 - 2617 *
KITTLEMAN ET AL.: "Characterization and Mechanistic Studies of Type II Isopentenyl Diphosphate: Dimethylallyl Diphosphate Isomerase from Staphylococcus aureus.", BIOCHEMISTRY, vol. 46, no. 28, 2007, pages 8401 - 8413, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pmGarticIes/PMC2515275/pdf/nihms60389.pdf> *
OTT ET AL.: "The RFN riboswitch of Bacillus subtilis is a target for the antibiotic roseoflavin produced by Streptomyces davawensis.", RNA BIOLOGY, vol. 6, no. 3, 2009, pages 276 - 280, Retrieved from the Internet <URL:http://www.landesbioscience.com> *

Similar Documents

Publication Publication Date Title
KR102430144B1 (en) 1H-PYRROLO[2,3-c]PYRIDIN-7(6H)-ONES AND PYRAZOLO[3,4-c]PYRIDIN-7(6H)-ONES AS INHIBITORS OF BET PROTEINS
US20120077781A1 (en) Flavin derivatives
Wang et al. Novel 3-arylfuran-2 (5H)-one-fluoroquinolone hybrid: design, synthesis and evaluation as antibacterial agent
JP2021504481A (en) Heterocyclic compounds as PRMT5 inhibitors
KR20160012195A (en) Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
EP2969000B1 (en) New compounds
EP3679031A1 (en) Inhibitors of wdr5 protein-protein binding
JP2012522807A (en) Hydroxythienoquinolones and related compounds as antiinfectives
CN111655692A (en) Nitrogen-containing heterocyclic amide compound and medical application thereof
BG108548A (en) Heterocyclic compounds and their use as d-alanyl-d-alanine ligase inhibitors
EP2448926A1 (en) Flavin derivatives
US20130029980A1 (en) Flavin derivatives
WO2012109458A1 (en) Flavin derivatives
WO2021219813A1 (en) Antibacterial and antifungal pleuromutilin conjugates
KR20110137332A (en) Diaminopteridine derivatives
JP5756096B2 (en) Guanine riboswitch binding compounds and their use as antibiotics
RU2527459C1 (en) Novel heteroarene anthracenedione-based g-quadruplex ligands inhibiting tumour cell growth
WO2017046603A1 (en) Antibacterial compounds and new uses thereof
EP2600872A1 (en) Fused-imidazoyl compounds useful as antimicrobial agents
WO2024079623A1 (en) Tricyclic compounds and their uses
CN117597337A (en) Process for the preparation of antibacterial compounds
WO2024105553A1 (en) Bicyclic heterocycles and their use as wrn inhibitors
Qiu Synthesis and Evaluation of New Antimicrobial Agents and Novel Organic Fluorophores
CN116041369A (en) Pyrimidine ring derivatives, preparation method and medical application thereof
Nessaib et al. Synthesis, crystal structure and antibacterial evaluation of N-substituted perhydro-1, 3-oxazin-2-ones containing N-phenylsulfonamide

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: 12744407

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12744407

Country of ref document: EP

Kind code of ref document: A1