WO2016077541A1 - Novel anti-infective compounds and methods using same - Google Patents

Novel anti-infective compounds and methods using same Download PDF

Info

Publication number
WO2016077541A1
WO2016077541A1 PCT/US2015/060315 US2015060315W WO2016077541A1 WO 2016077541 A1 WO2016077541 A1 WO 2016077541A1 US 2015060315 W US2015060315 W US 2015060315W WO 2016077541 A1 WO2016077541 A1 WO 2016077541A1
Authority
WO
WIPO (PCT)
Prior art keywords
imidazol
benzo
dihydro
imino
dichlorophenoxy
Prior art date
Application number
PCT/US2015/060315
Other languages
French (fr)
Inventor
Harvey Rubin
Allen Reitz
Trevor SELWOOD
Takahiro Yano
Jay Wrobel
H. Marie Loughran
Damian Weaver
Michael John COSTANZO
Original Assignee
The Trustees Of The University Of Pennsylvania
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Trustees Of The University Of Pennsylvania filed Critical The Trustees Of The University Of Pennsylvania
Publication of WO2016077541A1 publication Critical patent/WO2016077541A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • Gram-negative bacteria may be broadly classified as Gram-negative or Gram-positive, depending on the general structure of their cell wall.
  • Gram-negative bacteria present a thin peptidoglycan layer in their cell wall, and this layer is sandwiched between an inner cell membrane and an outer cell membrane, which comprises lipopolysaccharide (comprising lipid A, core polysaccharide, and O antigen) in its outer leaflet and phospholipids in the inner leaflet.
  • Gram-positive bacteria on the other hand have a thick peptidoglycan layer outside the cell membrane. Gram-positive bacteria tend to be more permeable to antibiotics than Gram- negative bacteria, because of the latter' s relatively impermeable lipid-based bacterial outer membrane.
  • the proteobacteria are a major group of Gram-negative bacteria, including Escherichia coli (E. coii), Salmonella, Shigella and other Enter obacteriaceae, Pseudomonas, Moraxella, Helicobacter, Stenotrophomonas, Bdellovibrio, acetic acid bacteria, and
  • Gram-negative bacteria include the cyanobacteria, spirochaetes, green sulfur, and green non-sulfur bacteria.
  • Medically relevant Gram-negative cocci may cause sexually transmitted diseases (Neisseria gonorrhoeae), meningitis (Neisseria meningitidis), and respiratory symptoms (Moraxella catarrhalis).
  • Medically relevant Gram- negative bacilli may cause respiratory problems (Hemophilus influenzae, Klebsiella pneumoniae, Legionella pneumophila, Pseudomonas aeruginosa), urinary problems
  • Gram-negative bacteria associated with hospital-acquired infections include Acinetobacter baumannii, which cause bacteremia, secondary meningitis, and ventilator-associated pneumonia in hospital intensive-care units.
  • Gram-negative pathogens such as Acinetobacter baumannii
  • Acinetobacter baumannii are responsible for over 30% of the most common hospital acquired infections (Hidron, et al, 2008, Infect. Control Hosp. Epidemiol., 29:996). This number rises to 70% in intensive care units, where 74% of A. baumannii isolated are multidrug-resistant (Hidron, et al, 2008, Infect. Control Hosp. Epidemiol, 29:996).
  • A. baumannii infection in burn wound patients increases the cost of treatment by almost $ 100,000 per patient (Wilson, et al, 2004, Am. J. Infect. Control 32:342).
  • baumannii is the second most commonly isolated non-fermenting bacterium in humans, and infection can result in pneumonia, skin and wound infections, bacteremia and meningitis.
  • A. baumannii biofilms have been implicated in diseases such as cystic fibrosis, periodontitis and urinary tract infections, partly because of the bacteria's ability to colonize indwelling medical devices.
  • the continual appearance oiA. baumannii strains resistant to ⁇ -lactams, cephalosporins, aminoglycosides, quinolones and lately carbapenems has compromised treatment options.
  • the OxPhos system is composed of the electron transport chain (ETC) and ATP synthase, and plays a central role in ATP synthesis in various Gram-positive and Gram- negative bacterial pathogens.
  • ETC electron transport chain
  • Biochemical studies (de Graef, et al, 1999, J. Bacteriol 181 :2351 ; Gyan, et al, 2006, J. Bacteriol. 188:7062; Yano, et al, 2006, J. Biol. Chem. 281 : 1 1456; Weinstein, et al, 2005, Proc. Natl. Acad. Sci. USA 102:4548) and genetic studies (Green & Paget, 2004, Nat. Rev. Microbiol.
  • NADH:quinone oxidoreductase (NDH-2) have no counterpart in human mitochondria, but appear to be nearly ubiquitous in aerobic bacteria.
  • bioinformatic analyses indicate structural divergence between the enzymes of the OxPhos pathway of bacteria and mitochondria.
  • the invention provides certain compounds, or salts, solvates or N-oxides thereof.
  • the invention further provides pharmaceutical compositions comprising at least one compound of the invention.
  • the invention further provides a method of treating or preventing a infection by a bacterium in a subject in need thereof.
  • the compound of the invention or a salt, solvate or N- oxide thereof, is selected from the group consisting of:
  • ring A is a fused aryl or heteroaryl ring selected from the group consisting of
  • each occurrence of R 1 is independently a bond or -CH 2 -;
  • R 2 is selected from the group consisting of -(CH 2 )i_3-0-(CH 2 )o- 2 -,
  • R 4 is -(CH 2 )o- 4 ;
  • each occurrence of R 9 is independently O, S or R 7 ;
  • each occurrence of R 3 is independently -NH 2 , -NH(Ci-C 6 alkyl), -N(Ci-C 6 alkyl)(Ci- Ce alkyl), pyrrolidin- 1 -yl, pyperidin-l -yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR 7 , SR 7 ,
  • R 4 is a bond and R 5 combines with the N atom to which it is bound to form morpholyn-l -yl, piperidin- l-yl, 4-cyano-piperidin-l -yl and piperazin-l -yl, wherein N 4 of the piperazinyl is optionally substituted with Ci-Ce alkyl;
  • each occurrence of R 3 is independently H, -NH 2 , -NH(Ci-C 6 alkyl), -N(Ci-C 6 alkyl)(Ci-C6 alkyl), pyrrolidin-l -yl, pyperidin- l-yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR 7 ,
  • the compound is not: l -(2,4-dichlorophenoxy)-3-(2- imino-3-propyl-2,3-dihydro- lH-benzo[d]imidazol-l -yl) propan-2-ol (Al); l -(3-allyl-2-imino- 2,3-dihydro- lH-benzo[d]imidazol- l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A2); l -(2,4- dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro- lH-benzo[d] imidazol- 1 -yl)propan-2-ol (1); 1 -(3 -benzyl-2-imino-2,3 -dihydro- lH-benzo[d]imida
  • the at least one compound is selected from the group consisting of:
  • (I) is not a compound of formula (lb): (lb), wherein in (lb): R A is C1-C7 alkyl, C 2 -C 7 alkenyl, - CH2CH2 Et2 or -CH ⁇ CH ⁇ 1 -morpholinyl), or benzyl optionaly substituted with at least one halogen or C1-C3 alkyl; R B is phenyl optionally substituted with at least one halogen; aryl; aroxy; naphthyl; phenoxymethyl optionally substituted with at least one halogen, C1-C3 alkyl and/or C1-C3 alkoxy; phenyl isoxazole optionally substituted with at least one halogen; or 4- methyl-3 -pheny
  • ring A is , wherein R 6 is CH or CR 10 . In other embodiments, at least one occurrence of R 6 in ring A is CR 10 . In yet other
  • ring A is selected from the group consisting of:
  • R 10 is optionally substituted phenyl or optionally substituted pyridyl.
  • R 10 is 4-fluorophenyl, p-tolyl or phenyl.
  • R 9 is O or NR 7 .
  • R 7 is methyl or 4-fluorophenyl.
  • both occurrences of R 1 are a bond.
  • R 2 is selected from the group consisting of -(CH 2 ) 1-3 -
  • R 2 is -(CH 2 )i- 3 -NR 7 -, -(CH 2 ) 1-3 -S-, and -(CH 2 )i-6-.
  • at least one of the CH 2 groups in R 2 is independently substituted with OR 7 .
  • R 2 is -
  • R 3 or R 3 is phenyl, 2-methylphenyl, 3- methylphenyl, 4-methylphenyl, 2-chlorophenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4- methoxyphenyl, 3-chlorophenyl, 4-chlorophenyl, 2-bromophenyl, 3-bromophenyl, 4- bromophenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2,4-difluorophenyl, 2,4-dichlorophenyl, 1,2-biphenyl, 1,3-biphenyl, 1 ,4-biphenyl, 4- trifluoromethylmethyl- 1 ,4-biphenyl, 4' -fluoro-2-chloro- 1 ,4-biphenyl, 2-trifluoromethyl- pyridin-5-yl, and 2-furyl.
  • R 5 or R 5 is selected from the group consisting of H, Ci-Ce alkyl, aryl, N(R 7 ) 2 , pyrrolidin- l-yl, piperidin-l -yl, N ⁇ H-piperidin ⁇ - yl, 1 -(Ci-C6 alkyl)-piperidin-4-yl, N 1 -(tBoc)-piperidin-4-yl, morpholin-l -yl, imidazol- 1 -yl, imidazol-2-yl, pyrrolidin-2-one- l-yl, pyridyl, and
  • R 5 or R 5 is N(R 7 ) 2 , wherein each occurrence of R 7 is independently H, methyl, ethyl, w-propyl or isopropyl.
  • R 6 is CR 7 . In other embodiments, each occurrence of
  • R 6 is CH.
  • the compound of formula (I) is at least one selected from the group consisting of: l-(3-(2,4-dichlorophenoxy)propyl)- lH-benzo[d]imidazol-2- amine (4); 2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol-
  • the compound of formula (II) is at least one selected from the group consisting of: l-(2,4-dichlorophenoxy)-3-(2-((3-morpholinopropyl)amino)-lH- benzo[d] imidazol- l-yl)propan-2-ol (B55); l-(2,4-dichlorophenoxy)-3-(2-(phenethylamino)- lH-benzo[d] imidazol- l-yl)propan-2-ol (B56); l-(2-((l-benzylpiperidin-4-yl)amino)-lH- benzo [d] imidazol- l-yl)-3 -(2,4-dichlorophenoxy )propan-2-ol (B57); l-(2,4-dichlorophenoxy)- 3-(2-((2-(diethylamino)ethyl)amino)-l
  • the compound of formula (III) is at least one selected from the group consisting of: N-((lH-benzo[d]imidazol-2-yl)methyl)-3-(2,4- dichlorophenoxy)propan-l -amine (B58); l-(((lH-benzo[d]imidazol-2-yl)methyl)amino)-3- (2,4-dichlorophenoxy)propan-2-ol (B59); l-(2,4-dichlorophenoxy)-3-((l-(2- (diethylamino)ethyl)-lH-benzo[d]imidazol-2-yl)oxy)propan-2-ol (B64); N-pentyl-lH- benzo[d]imidazol-2-amine (B83); N-phenethyl-lH-benzo[d]imidazol-2-amine (B84); Nl- ((lH-benzo[d]imid
  • the compound is at least one selected from the group consisting of: l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH- benzo[d] imidazol- 1-yl) propan-2-ol (Al); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-3 -(2,4-dichlorophenoxy) propan-2-ol (A2); l-(2,4-dichlorophenoxy)-
  • the pharmaceutical composition comprises at least one pharmaceutically acceptable carrier. In other embodiments, the pharmaceutical composition further comprises at least one additional antibacterial agent. In yet other embodiments, the at least one compound and the at least one additional antibacterial agent are synergistic. In yet other embodiments, the Fractional Inhibitory Concentration Index (FICI) determined by the combination of the at least one compound and the at least one additional antibacterial agent is equal to or lower than about 0.50. In yet other embodiments, the at least one additional antibacterial agent comprises a lipoprotein antibiotic. In yet other
  • the at least one additional antibacterial agent comprises a polymyxin.
  • the at least one compound comprises at least one selected from the group consisting of 5, 25, and B55-B58.
  • the pharmaceutical composition comprises at least one additional antibacterial agent and at least one compound selected from the group consisting of l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl) propan-2-ol; 1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 - yl)-3-(2,4-dichlorophenoxy) propan-2-ol; l-(2,4-dichlorophenoxy)-3-(3-(2- (diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol-l-yl)propan-2-ol; l-(3- benzyl-2-imino-2,3-dihydro-lH-benzo[d] imi
  • the method comprises administering to the subject a therapeutically effective amount of at least one compound of claim 1, or a salt, solvate, tautomer or N-oxide thereof. In other embodiments, the method comprises administering to the subject a therapeutically effective amount of at least one compound, or a salt, solvate, tautomer or N-oxide thereof, selected from the group consisting of l-(2,4-dichlorophenoxy)- 3-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl) propan-2-ol; l-(3-allyl-2-imino- 2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol; l-(2,4- dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-i
  • the bacterium is Gram-negative. In other embodiments, the bacterium is obligatory aerobic. In yet other embodiments, the bacterium belongs to the Acinetobacter genus. In yet other embodiments, the bacterium comprises A. baumannii. In yet other embodiments, the compound interferes with or inhibits the Oxidative Phosphorylation (OxPhos) pathway of the bacterium. In yet other embodiments, the compound selectively inhibits the bacterial OxPhos over mammalian mitochondrial OxPhos.
  • OxPhos Oxidative Phosphorylation
  • the compound is administered to the subject by a route selected from the group consisting of nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous.
  • the subject is further administered at least one additional antibacterial agent.
  • the at least one additional antibacterial agent is co-administered with the compound to the subject.
  • the at least one additional antibacterial agent and the compound are coformulated.
  • the subject is administered the at least one additional antibacterial agent at a lower dose or frequency as compared to the administering of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
  • the subject develops a lower or slower rate of resistance to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
  • the subject develops lower toxicity from the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
  • the subject is a mammal. In other embodiments, the mammal is human.
  • Fig. 1 illustrates the structure of compound 1.
  • Fig. 2A illustrates analogs of compound 1
  • Fig. 2B illustrates their activity against .4. baumannii.
  • Sub-mitocondrial particle (SMP, bovine) ATP synthesis, n 2, standard deviations between the duplicates were on average 9% of the mean.
  • SMP Stacondrial particle
  • bovine bovine
  • standard deviations between the duplicates were on average 9% of the mean.
  • 3 A. baumannii MICs were performed in duplicate and all reproduced to within one well. In cases where the MICs did not duplicate, the lower MIC value is reported.
  • Fig. 3A illustrates the effect of compound 5 on oxygen gas consumption.
  • FIG. 3B illustrates bacterial oxidative phosphorylation (OxPhos).
  • Fig. 3C illustrates the A.
  • Fig. 4 illustrates synthetic schemes useful within the present invention.
  • Figs. 5A-5B are a set of graph and tables illustrating MIC distribution for compound 5 and comparator agents.
  • Fig. 5B 1 MICs were determined by serial dilution of 5 in Mueller-Hinton broth at a starting concentration of bacteria of 10 5 — 10 6 CFU/mL. Values are reported after 18 h incubation at 37 °C. 2 Methicillin-resistant Staphylococcus aureus. 3 The medium contained 40% serum.
  • Figs. 6A-6C are a set of tables illustrating structure-activity relationship (SAR) studies for compounds of the invention.
  • SMP, bovine ATP synthesis 2
  • n 2
  • standard deviations between the duplicates were on average 9% of the mean.
  • 3 A. baumannii minimum inhibitory concentration, MICs were performed in duplicate and all reproduced to within one well. In cases where the MICs did not duplicate the lower MIC value is reported. 4 Cells are exposed to compounds for 1 h in the absence of serum.
  • Fig. 6C 1 A. baumannii membrane ATP synthesis. 2 Sub-mitochondrial particle (SMP, bovine) ATP synthesis. 3 A. baumannii IC50/SMP IC 50 . 4 A. baumannii MIC.
  • Fig. 7 illustrates SAR studies performed in the present investigation.
  • Fig. 8 is a table illustrating bacterial strains used within the methods of the invention.
  • Fig. 10 is an illustration of mice infected with bioluminescent AB5075 in a wound model of infection. Relative light units (RLU) are measured by the IVIS systems, where red indicates areas of the highest bacterial numbers, as opposed to blue & black with lesser numbers. In certain embodiments, blue color corresponds to about 1.0 x 10 5 CFU and red color corresponds to about 1.0 x 10 8 CFU present in the wound.
  • RLU Relative light units
  • Figs. 1 1 A- 1 IB illustrate the checkerboard assay, FICI and isobologram. Fig.
  • FIG. 11A a schematic representation of a checkerboard assay.
  • One compound is serially diluted from the right to then left in an MIC plate, and the other compound from top to bottom, to produce the checkerboard array of mixtures.
  • the * indicates MIC concentrations of x alone and y alone.
  • the gray squares indicate growth in the wells.
  • the white and green squares indicate no growth, with the green squares indicating the wells used to calculate FICI.
  • Fig. 1 IB illustrates an isobologram. In the isobologram the FIC values for the green squares are plotted. The line indicates a situation where the effect of the two drugs are additive. Points below are produced by synergy, and those above by antagonism.
  • Figs. 12A-12C illustrate isobolograms.
  • Fig. 13 is a table illustrating a representative checkerboard plot demonstrating the synergy between B57 and colistin against A. baumannii.
  • Fig. 14A is a graph illustrating cytotoxic data for 25 in HepG2 cells with added amounts of polymyxin B.
  • Fig. 14B is a graph illustrating the correlation between IC5 0 and MIC in the presence of polymyxin B.
  • Fig. 15 is a scheme illustrating synergistic interactions between OxPhos and polymyxin B/colistin via energy across the membranes.
  • Figs. 16A-16B are graphs illustrating the effects of drugs on oxygen consumption of live A. baumannii cells (about 10 7 cells/ml) determined using an oxygen electrode. Compounds 5 (75 ⁇ ), B55 (50 ⁇ ) and colistin (10 ⁇ g/ml) were added to a chamber.
  • the present invention relates to the unexpected discovery of novel antibacterial compounds.
  • the compounds of the invention are active against Gram-negative bacteria, such as but not limited to A. baumannii.
  • the compounds of the invention are active against obligatory aerobic bacteria, such as but not limited to A. baumannii.
  • the compounds of the invention impair bacterial viability.
  • the compounds of the invention comprise 2-aminobenzimidazoles, 2-iminobenzimidazoles, analogs or derivatives thereof, and salts, N-oxides or solvates thereof.
  • the compounds of the invention are synergistic with lipoprotein antibiotics, such as polymyxins, such as colistin (polymyxyn E) and/or polymyxin B, or any other cell wall-modifying antibiotic.
  • the compounds of the invention are antibacterial because they interfere and/or inhibit the Oxidative Phosphorylation (OxPhos) pathway used by bacteria to produce energy. In other embodiments, the compounds of the invention selectively inhibit bacterial OxPhos over mammalian mitochondrial OxPhos.
  • OxPhos Oxidative Phosphorylation
  • OxPhos oxidative phosphorylation
  • a library of about 10,000 compounds was screened using a membrane-based ATP synthesis assay.
  • One of the compounds identified as active in this assay was the 2-iminobenzimidazole 1, which inhibited the OxPhos oiA.baumannii with a good selectivity against mitochondrial OxPhos and also and displayed a MIC of 17 ⁇ (13 ⁇ g/mL) against the pathogen.
  • the 2-iminobenzimidazole 1 was found to inhibit the type 1 NADH-quinone oxidoreductase (NDH-1) of A. baumannii OxPhos by a biochemical approach.
  • Des-hydroxy analog 5 is also very active against bacteria, and showed low cytotoxicity against NIH3T3 and HepG2 mammalian cell lines.
  • the compounds described herein are active anti-Acinetobacter agents.
  • the articles “a” and “an” refer to one or to more than one (i.e., to at least one) of the grammatical object of the article.
  • an element means one element or more than one element.
  • use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting.
  • the term “Ab” or “Ab” refers to A. baumannii.
  • the term “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term “about” is meant to encompass variations of ⁇ 20% or ⁇ 10%, more preferably ⁇ 5%, even more preferably ⁇ 1%, and still more preferably ⁇ 0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
  • co-administered and “co-administration” as relating to a subject refer to administering to the subject a compound of the invention or salt thereof along with a compound that may also treat diseases and/or disorders contemplated herein.
  • the co-administered compounds are administered separately, or in any kind of combination as part of a single therapeutic approach.
  • the co-administered compound may be formulated in any kind of combinations as mixtures of solids and liquids under a variety of solid, gel, and liquid formulations, and as a solution.
  • the term "container” includes any receptacle for holding the pharmaceutical composition.
  • the container is the packaging that contains the pharmaceutical composition.
  • the container is not the packaging that contains the pharmaceutical composition, i.e., the container is a receptacle, such as a box or vial that contains the packaged pharmaceutical composition or unpackaged pharmaceutical composition and the instructions for use of the pharmaceutical composition.
  • packaging techniques are well known in the art. It should be understood that the instructions for use of the pharmaceutical composition may be contained on the packaging containing the pharmaceutical composition, and as such the instructions form an increased functional relationship to the packaged product. However, it should be understood that the instructions may contain information pertaining to the compound's ability to perform its intended function, e.g., treating, preventing, or reducing bacterial infection in a patient.
  • a "disease” is a state of health of a subject wherein the subject cannot maintain homeostasis, and wherein if the disease is not ameliorated then the subject's health continues to deteriorate.
  • a disorder in a subject is a state of health in which the subject is able to maintain homeostasis, but in which the subject's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the subject's state of health.
  • the terms "effective amount,” “pharmaceutically effective amount” and “therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
  • instructional material includes a publication, a recording, a diagram, or any other medium of expression that may be used to communicate the usefulness of the compounds and/or methods of the invention.
  • the instructional material may be part of a kit useful for effecting alleviating or treating the various diseases or disorders recited herein.
  • the instructional material may describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a mammal.
  • the instructional material of the kit may, for example, be affixed to a container that contains the compounds of the invention or be shipped together with a container that contains the compounds.
  • the instructional material may be shipped separately from the container with the intention that the recipient uses the instructional material and the compound cooperatively.
  • the instructional material is for use of a kit; instructions for use of the compound; or instructions for use of a formulation of the compound.
  • the term "patient,” “individual” or “subject” refers to a human or a non-human mammal.
  • Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and murine mammals.
  • the patient, subject or individual is human.
  • the phrase "pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • the phrase "pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function.
  • Such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient.
  • materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil;
  • glycols such as propylene glycol
  • polyols such as glycerin, sorbitol, mannitol and polyethylene glycol
  • esters such as ethyl oleate and ethyl laurate
  • agar buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid;
  • pyrogen-free water isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
  • the phrase "pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions.
  • the "pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound useful within the invention.
  • Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
  • pharmaceutically acceptable salt refers to a salt of the administered compounds prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids, organic acids, solvates, hydrates, or clathrates thereof.
  • Salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present invention, such as for example utility in process of synthesis, purification or formulation of compounds useful within the methods of the invention.
  • Salts may be comprised of a fraction of one, one or more than one molar equivalent of acid or base with respect to any compound of the invention.
  • Suitable pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid.
  • inorganic acids include sulfate, hydrogen sulfate, hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids (including hydrogen phosphate and dihydrogen phosphate).
  • Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4- hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic,
  • ethanesulfonic benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2- hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, ⁇ -hydroxybutyric, salicylic, galactaric, galacturonic acid, glycerophosphonic acids and saccharin (e.g., saccharinate, saccharate).
  • saccharin e.g., saccharinate, saccharate
  • Suitable pharmaceutically acceptable base addition salts of compounds of the invention include, for example, ammonium salts, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts.
  • Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N'-dibenzylethylene- diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine
  • the phrase "pharmaceutical composition” refers to a mixture of at least one compound useful in the methods of the invention with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition facilitates administration of the compound to a patient.
  • Multiple techniques of administering a compound exist in the art including, but not limited to, nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous route administration.
  • prevent means no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease.
  • a compound of the invention is 'synergistic" with one additional antibiotic if the Fractional Inhibitory Concentration Index (FICI) determined by the combination of the compound of the invention and the one additional antibiotic is equal to or lower than about 0.50.
  • FICI Fractional Inhibitory Concentration Index
  • treatment is defined as the application or administration of a therapeutic agent, i.e., a compound of the invention (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has a condition contemplated herein, a symptom of a condition contemplated herein or the potential to develop a condition contemplated herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect a condition contemplated herein, the symptoms of a condition contemplated herein or the potential to develop a condition contemplated herein.
  • Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.
  • the condition is bacterium infection.
  • alkyl by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., C ⁇ -Ce means one to six carbon atoms) and includes straight, branched chain, or cyclic substituent groups. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, and cyclopropylmethyl.
  • substituted alkyls include, but are not limited to, 2,2-difluoropropyl, 2-carboxycyclopentyl and 3-chloropropyl.
  • heteroalkyl by itself or in combination with another term means, unless otherwise stated, a stable straight or branched chain alkyl group consisting of the stated number of carbon atoms and one or two heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may be optionally oxidized and the nitrogen heteroatom may be optionally quaternized.
  • the heteroatom(s) may be placed at any position of the heteroalkyl group, including between the rest of the heteroalkyl group and the fragment to which it is attached, as well as attached to the most distal carbon atom in the heteroalkyl group. Examples include: -0-CH 2 -CH 2 -CH 3 ,
  • -CH 2 -CH 2 -CH 2 -OH -CH 2 -CH 2 -NH-CH 3
  • Up to two heteroatoms may be consecutive, such as, for example, -CH 2 -NH-OCH 3 , or
  • alkoxy employed alone or in combination with other terms means, unless otherwise stated, an alkyl group having the designated number of carbon atoms, as defined above, connected to the rest of the molecule via an oxygen atom, such as, for example, methoxy, ethoxy, 1-propoxy, 2-propoxy (isopropoxy) and the higher homologs and isomers. Examples are (Ci-C 3 ) alkoxy, particularly ethoxy and methoxy.
  • halo or halogen alone or as part of another substituent means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom, preferably, fluorine, chlorine, or bromine, more preferably, fluorine or chlorine.
  • cycloalkyl refers to a mono cyclic or polycyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom.
  • the cycloalkyl group is saturated or partially unsaturated.
  • the cycloalkyl group is fused with an aromatic ring.
  • Cycloalkyl groups include groups having from 3 to 10 ring atoms.
  • Illustrative examples of cycloalkyl groups include, but are not limited to, the following moieties:
  • Monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Dicyclic cycloalkyls include, but are not limited to, tetrahydronaphthyl, indanyl, and tetrahydropentalene.
  • Polycyclic cycloalkyls include adamantine and norbornane.
  • cycloalkyl includes "unsaturated nonaromatic carbocyclyl” or “nonaromatic unsaturated carbocyclyl” groups, both of which refer to a nonaromatic carbocycle as defined herein, which contains at least one carbon carbon double bond or one carbon carbon triple bond.
  • heterocycloalkyl refers to a heteroalicyclic group containing one to four ring heteroatoms each selected from O, Sand N.
  • each heterocycloalkyl group has from 4 to 10 atoms in its ring system, with the proviso that the ring of said group does not contain two adjacent O or S atoms.
  • the heterocycloalkyl group is fused with an aromatic ring.
  • the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen atom may be optionally quaternized.
  • the heterocyclic system may be attached, unless otherwise stated, at any heteroatom or carbon atom that affords a stable structure.
  • a heterocycle may be aromatic or non-aromatic in nature.
  • the heterocycle is a heteroaryl.
  • An example of a 3 -membered heterocycloalkyl group includes, and is not limited to, aziridine.
  • Examples of 4-membered heterocycloalkyl groups include, and are not limited to, azetidine and a beta lactam.
  • Examples of 5-membered heterocycloalkyl groups include, and are not limited to, pyrrolidine, oxazolidine and thiazolidinedione.
  • Examples of 6- membered heterocycloalkyl groups include, and are not limited to, piperidine, morpholine and piperazine.
  • Other non-limiting examples of heterocycloalkyl groups are:
  • non-aromatic heterocycles include monocyclic groups such as aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, pyrroline, pyrazolidine, imidazoline, dioxolane, sulfolane, 2,3-dihydrofuran, 2,5-dihydrofuran, tetrahydrofuran, thiophane, piperidine, 1,2,3,6-tetrahydropyridine, 1,4-dihydropyridine, piperazine, morpholine, thiomorpholine, pyran, 2,3-dihydropyran, tetrahydropyran, 1,4-dioxane,
  • 1,3-dioxane 1,3-dioxane, homopiperazine, homopiperidine, 1,3-dioxepane, 4,7-dihydro-l,3-dioxepin, and hexamethyleneoxide.
  • aromatic refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character, i.e. having (4n + 2) delocalized ⁇ (pi) electrons, where n is an integer.
  • aryl employed alone or in combination with other terms, means, unless otherwise stated, a carbocyclic aromatic system containing one or more rings (typically one, two or three rings), wherein such rings may be attached together in a pendent manner, such as a biphenyl, or may be fused, such as naphthalene.
  • aryl groups include phenyl, anthracyl, and naphthyl. Examples are phenyl and naphthyl.
  • aryl-(Ci-C3)alkyl means a functional group wherein a one- to three-carbon alkylene chain is attached to an aryl group, e.g., -CH2CH2-phenyl. Examples are aryl-CH 2 - and aryl-CH(C]3 ⁇ 4)-.
  • substituted aryl-(Ci-C3) alkyl means an aryl-(Ci-C3) alkyl functional group in which the aryl group is substituted. An example is substituted aryl(CH 2 )-.
  • heteroaryl-(Ci-C3) alkyl means a functional group wherein a one to three carbon alkylene chain is attached to a heteroaryl group, e.g., -CH 2 CH 2 -pyridyl.
  • a heteroaryl group e.g., -CH 2 CH 2 -pyridyl.
  • An example is heteroaryl-(CH 2 )-.
  • substituted heteroaryl-(Ci-C3) alkyl means a heteroaryl-(Ci-C 3 ) alkyl functional group in which the heteroaryl group is substituted.
  • An example is substituted heteroaryl-(CH 2 )-.
  • heteroaryl or “heteroaromatic” refers to a heterocycle having aromatic character.
  • a polycyclic heteroaryl may include one or more rings that are partially saturated. Examples include the following moieties:
  • heteroaryl groups also include pyridyl, pyrazinyl, pyrimidinyl
  • 2-pyrrolyl imidazolyl, thiazolyl, oxazolyl, pyrazolyl (particularly 3- and 5-pyrazolyl), isothiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-thiadiazolyl and 1,3,4-oxadiazolyl.
  • polycyclic heterocycles and heteroaryls examples include indolyl
  • 2-benzimidazolyl benzotriazolyl, thioxanthinyl, carbazolyl, carbolinyl, acridinyl, pyrrolizidinyl, and quinolizidinyl.
  • substituted means that an atom or group of atoms has replaced hydrogen as the substituent attached to another group.
  • substituted further refers to any level of substitution, namely mono-, di-, tri-, tetra-, or penta-substitution, where such substitution is permitted.
  • the substituents are independently selected, and substitution may be at any chemically accessible position. In certain embodiments, the substituents vary in number between one and four. In other embodiments, the substituents vary in number between one and three. In yet other embodiments, the substituents vary in number between one and two.
  • the term "optionally substituted” means that the referenced group may be substituted or unsubstituted. In certain embodiments, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In other embodiments, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein.
  • the substituents are independently selected from the group consisting of Ci_6 alkyl, -OH, Ci_6 alkoxy, halo, amino, acetamido, oxo and nitro. In yet other embodiments, the substituents are independently selected from the group consisting of Ci_6 alkyl, Ci_6 alkoxy, halo, acetamido, and nitro.
  • the carbon chain may be branched, straight or cyclic, with straight being a specific example.
  • the present invention relates to the unexpected discovery of novel antibacterial compounds.
  • the compounds of the invention are active against Gram-negative bacteria, such as but not limited to A. baumannii.
  • the compounds of the invention are active against obligatory aerobic bacteria, such as but not limited to A. baumannii.
  • the compounds of the invention impair viability of bacteria.
  • the compounds of the invention comprise 2-aminobenzimidazoles, 2-iminobenzimidazoles, analogs or derivatives thereof, and salts or solvates thereof.
  • the compounds of the invention are synergistic with lipoprotein antibiotics, such as polymyxins, such as colistin (polymyxyn E) and/or polymyxin B, or any other cell wall-modifying antibiotic.
  • the compounds of the invention are antibacterial because they interfere and/or inhibit the Oxidative Phosphorylation (OxPhos) pathway used by bacteria to produce energy.
  • Oxidative Phosphorylation Oxidative Phosphorylation
  • the compounds of the invention may be synthesized using techniques well- known in the art of organic synthesis.
  • the starting materials and intermediates required for the synthesis may be obtained from commercial sources or synthesized according to methods known to those skilled in the art.
  • the compounds of the invention are prepared according to the methodology outlined in Fig. 4.
  • the compound of the invention is a compound of formula (I), or a salt, solvate or N-oxide thereof:
  • ring A is a fused aryl or heteroaryl ring selected from the group consisting of:
  • each occurrence of R 1 is independently a bond or -CH 2 -;
  • R 2 is selected from the group consisting of -(CH 2 )i-3-0-(CH 2 )o-2-,
  • R 4 is -(CH 2 )o- 4 ;
  • eeaacchh ooccccurrence of R 6 is independently CH, CR 10 or N, wherein 0-2 occurrences of R 6 in ring A are N;
  • each occurrence of R 9 is independently O, S or R 7 ;
  • the compound of the invention is a compound of formula (II), or a salt, solvate or N-oxide thereof:
  • ring A is a fused aryl or heteroaryl ring selected from the group consisting of:
  • each occurrence of R 1 is independently a bond or -CH 2 -;
  • R 2 is selected from the group consisting of -(CH 2 ) 1 - 3-0-(CH 2 )o -2 -,
  • R 4 is -(CH 2 )o- 4 ;
  • R 4 is a bond and R 5 combines with the N atom to which it is bound to form morpholyn- 1 -yl, piperidin- 1-yl, 4-cyano-piperidin-l -yl and piperazin- 1 -yl, wherein N 4 of the piperazinyl is optionally substituted with Ci-Ce alkyl;
  • each occurrence of R 6 is independently CH, CR 10 or N, wherein 0-2 occurrences of R 6 in ring A are N;
  • each occurrence of R 9 is independently O, S or R 7 ;
  • the compound of the invention is a compound of formula (III), or a salt, solvate or N-oxide thereof:
  • ring A is a fused aryl or heteroaryl ring selected from the group consisting of:
  • each occurrence of R 1 is independently a bond or -CH 2 -;
  • R 2 is selected from the group consisting of -(CH 2 ) 1 - 3-0-(CH 2 )o -2 -,
  • each of the CH 2 groups in R 2 is independently optionally substituted with a substituent selected from the group consisting of C ⁇ -Ce alkyl, C ⁇ -Ce fluoroalkyl, F, CI, Br, I,
  • R 4 is -(CH 2 )o- 4 ;
  • alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted;
  • m is 2, 3, 4 or 5; and
  • n is 0 or 1;
  • eeaacchh ooccccurrence of R 6 is independently CH, CR 10 or N, wherein 0-2 occurrences of R 6 in ring A are N;
  • each occurrence of R 9 is independently O, S or R 7 ;
  • the compound is at least one compound, or a salt, solvat r N-oxide thereof, selected from the group consisting of:
  • (I) is not a compound of formula (lb):
  • R A is Ci-C 6 alkyl, C 2 -C 7 alkenyl, -CH 2 CH 2 NEt 2 or -CH 2 CH 2 -(N 1 -morpholinyl), or benzyl optionaly substituted with at least one halogen or C1-C 3 alkyl;
  • R is phenyl optionally substituted with at least one halogen; aryl; aroxy; naphthyl; phenoxymethyl optionally substituted with at least one halogen, C1-C 3 alkyl and/or C1-C 3 alkoxy; phenyl isoxazole optionally substituted with at least one halogen; or 4-methyl-3- phenyl- 1 ,2,4-oxadiazole-5 (4H)-one.
  • (I) is not l-(2,4-dichlorophenoxy)-3-(2-imino-3- propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl) propan-2-ol (Al); l-(3-allyl-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A2); l-(2,4- dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol- l-yl)propan-2-ol (A3, also known as 1); l-(3-benzyl-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl-yl
  • ring A is , wherein R 6 is CH or CR 10 . In other embodiments, at least one occurr of R 6 in ring A is CR 10 . In yet other
  • ring A is R 1 ° or .
  • R 10 is F
  • ring A is selected from the group consisting of:
  • Hal is halogen
  • R 10 is optionally substituted phenyl or optionally substituted pyridyl. In yet other embodiments, R 10 is 4-fluorophenyl, p-tolyl or phenyl. In yet other embodiments, R 9 is O or NR 7 . In yet other embodiments, R 7 is methyl or 4- fluorophenyl.
  • one occurrence of R 1 is a bond, and the other occurrence of R 1 is -CH 2 -. In other embodiments, both occurrences of R 1 are a bond.
  • R 2 is selected from the group consisting of
  • R 2 is -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 0-, -CH 2 CH 2 CH 2 0-, or - CH 2 CH(OH)CH 2 0-.
  • R 3 or R 3 is phenyl, pyridyl, pyrimidyl, 1,2-biphi
  • R 3 or R 3 is phenyl, 2-methylphenyl, 3-methylphi 4-methylphenyl, 2-chlorophenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 3- chlorophenyl, 4-chlorophenyl, 2-bromophenyl, 3-bromophenyl, 4-bromophenyl, 2- trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2,4-difluorophenyl, 2,4-dichlorophenyl, 1,2-biphenyl, 1,3-biphenyl, 1 ,4-biphenyl, 4-trifluoromethylmethyl-l,4- biphenyl, 4'-fluoro-2-chloro-l,4-biphenyl, 2-trifluoromethyl-pyridin-5-yl, and 2-furyl.
  • R 5 or R 5 is selected from the group consisting of H,
  • Ci-Ce alkyl, aryl, N(R 7 ) 2 , pyrrolidin-l-yl, piperidin-l-yl, N 1 -H-piperidin-4-yl, N ⁇ CQ-Ce alkyl)-piperidin-4-yl, N 1 -(tBoc)-piperidin-4-yl, morpholin-l-yl, imidazol- 1 -yl, imidazol-2-yl, pyrrolidin-2-one-l-yl, pyridyl, and -(C 0)o-i-( 4 -R 7 -piperazin-l-yl).
  • R 5 or R 5 is N(R 7 ) 2 , wherein each occurrence of R 7 is independently H, methyl, ethyl, n- propyl or isopropyl.
  • each occurrence of R 6 is CR 7 . In other embodiments, each occurrence of R 6 is CH.
  • the compound of formula (I), or a salt, solvate or N- oxide thereof is selected from the group consisting of:
  • the compound of formula (I), or a salt, solvate or N- oxide thereof is selected from the group consisting of:
  • diethylethan- 1 -amine (B92 or FC #3284) ;
  • the compound of formula (II), or a salt, solvate or N- oxide thereof is selected from the group consisting of:
  • the compound of formula (III), or a salt, solvate or N- oxide thereof is selected from the group consisting of:
  • N-pentyl-1 H-benzo [d]imidazol-2-amine (B83 or FC #3004) N-phenethyl-lH-benzo[d]imidazol-2-amine (B84 or FC #3005) ;
  • the invention provides one or more of the following compounds, or a salt, solvate or N-oxide thereof:
  • N-(3-morpholinopropyl)-lH-benzo[d]imidazol-2-amine (B72 or FC #2860) Nl-(lH-benzo[d]imidazol-2-yl)-N2,N2-diethylethane-l,2-diamine (B75 or FC #2864)
  • the compounds of the invention may possess one or more stereocenters, and each stereocenter may exist independently in either the (R) or (S) configuration.
  • compounds described herein are present in optically active or racemic forms. It is to be understood that the compounds described herein encompass racemic, optically- active, regioisomeric and stereoisomeric forms, or combinations thereof that possess the therapeutically useful properties described herein. Preparation of optically active forms is achieved in any suitable manner, including by way of non-limiting example, by resolution of the racemic form with recrystallization techniques, synthesis from optically-active starting materials, chiral synthesis, or chromatographic separation using a chiral stationary phase. In certain embodiments, a mixture of one or more isomer is utilized as the therapeutic compound described herein. In other embodiments, compounds described herein contain one or more chiral centers. These compounds are prepared by any means, including
  • stereoselective synthesis, enantios elective synthesis and/or separation of a mixture of enantiomers and/ or diastereomers Resolution of compounds and isomers thereof is achieved by any means including, by way of non-limiting example, chemical processes, enzymatic processes, fractional crystallization, distillation, and chromatography.
  • the methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), solvates, amorphous phases, and/or pharmaceutically acceptable salts of compounds having the structure of any compound of the invention, as well as metabolites and active metabolites of these compounds having the same type of activity.
  • Solvates include water, ether (e.g., tetrahydrofuran, or methyl tert- butyl ether) or alcohol (e.g., ethanol) solvates, acetates and the like.
  • the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, and ethanol. In other embodiments, the compounds described herein exist in unsolvated form.
  • the compounds of the invention may exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • compounds described herein are prepared as prodrugs.
  • a "prodrug” refers to an agent that is converted into the parent drug in vivo.
  • a prodrug upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • a pro drug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.
  • sites on, for example, the aromatic ring portion of compounds of the invention are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the aromatic ring structures may reduce, minimize or eliminate this metabolic pathway. In certain embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a deuterium, a halogen, or an alkyl group.
  • Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes suitable for inclusion in the compounds described herein include and are not limited to 2 H, 3 H, n C, 13 C, 14 C, 36 C1, 18 F, 123 I, 125 I, 13 N, 15 N, 15 0, 17 0, 18 0, 32 P, and 35 S.
  • isotopically-labeled compounds are useful in drug and/or substrate tissue distribution studies.
  • substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements).
  • substitution with positron emitting isotopes such as n C, 18 F, 15 0 and 13 N, is useful in Positron Emission
  • Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
  • the compounds described herein are labeled by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • reactive functional groups such as hydroxyl, amino, imino, thio or carboxy groups
  • Protecting groups are used to block some or all of the reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed.
  • each protective group is removable by a different means.
  • Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal.
  • protective groups are removed by acid, base, reducing conditions (such as, for example, hydrogenolysis), and/or oxidative conditions.
  • reducing conditions such as, for example, hydrogenolysis
  • oxidative conditions such as, for example, hydrogenolysis
  • Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and Fmoc groups, which are base labile.
  • Carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl, in the presence of amines that are blocked with acid labile groups, such as t-butyl carbamate, or with carbamates that are both acid and base stable but hydrolytically removable.
  • base labile groups such as, but not limited to, methyl, ethyl, and acetyl
  • carboxylic acid and hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc.
  • Carboxylic acid reactive moieties are protected by conversion to simple ester compounds as exemplified herein, which include conversion to alkyl esters, or are blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while coexisting amino groups are blocked with fluoride labile silyl carbamates.
  • Allyl blocking groups are useful in the presence of acid- and base- protecting groups since the former are stable and are subsequently removed by metal or pi-acid catalysts.
  • an allyl-blocked carboxylic acid is deprotected with a palladium- catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups.
  • Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and does not react. Once released from the resin, the functional 3 ⁇ 4roup is available to react.
  • blocking/protecting groups may be selected from:
  • the invention includes a method of treating or preventing a bacterium infection in a subject in need thereof, comprising administering to the subject a
  • a pharmaceutically acceptable composition comprising a compound of the invention or a salt or solvate thereof, whereby the bacterium infection is treated or prevented in the subject.
  • the bacterium is Gram-negative. In other embodiments, the bacterium is obligatory aerobic. In yet other embodiments, the bacterium comprises A. baumannii. In yet other embodiments, the compounds of the invention interfere and/or inhibit the Oxidative Phosphorylation (OxPhos) pathway used by the bacterium to produce energy. In yet other embodiments, the compounds of the invention selectively inhibit bacterial OxPhos over mammalian mitochondrial OxPhos.
  • OxPhos Oxidative Phosphorylation
  • the subject is a mammal. In other embodiments, the subject is human.
  • the compounds of the present invention are further intended to be useful in combination with one or more additional compounds useful for treating a condition considered herein.
  • additional compounds may comprise compounds of the present invention or compounds known to treat, prevent, or reduce the symptoms or effects of the conditions considered herein.
  • Non-limiting examples of additional antibacterial agents contemplated within the invention include lipoprotein antibiotics, such as polymyxins, such as colistin
  • polymyxyn E and/or polymyxin B, or any other cell wall-modifying antibiotic.
  • the at least one compound and the at least one additional antibacterial agent are synergistic.
  • the at least one additional antibacterial agent comprises a lipoprotein antibiotic.
  • the at least one additional antibacterial agent comprises a polymyxin.
  • the at least one compound comprises at least one selected from the group consisting of 5, 25, and B55-B58.
  • the at least one additional antibacterial agent is coadministered with the compound to the subject.
  • the at least one additional antibacterial agent and the compound are coformulated.
  • the subject upon administering to the subject the compound and the at least one additional antibacterial agent, the subject is administered the at least one additional antibacterial agent at a lower dose or frequency as compared to the administering of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
  • the subject upon administering to the subject the compound and the at least one additional antibacterial agent, the subject develops a lower or slower rate of resistance to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof. In yet other embodiments, upon administering to the subject the compound and the at least one additional antibacterial agent, the subject develops lower toxicity due to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
  • a synergistic effect may be calculated, for example, using suitable methods such as, for example, the Sigmoid-E max equation (Holford & Scheiner, 19981, Clin.
  • the regimen of administration may affect what constitutes an effective amount.
  • the therapeutic formulations may be administered to the patient either prior to or after the onset of a condition considered herein. Further, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be
  • the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
  • compositions of the present invention may be carried out using known procedures, at dosages and for periods of time effective to treat a condition considered herein in the patient.
  • An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the state of the disease or disorder in the patient; the age, sex, and weight of the patient; and the ability of the therapeutic compound to treat a condition considered herein in the patient.
  • Dosage regimens 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.
  • a non-limiting example of an effective dose range for a therapeutic compound of the invention is from about 1 and 5,000 mg kg of body weight/per day.
  • One of ordinary skill in the art would be able to study the relevant factors and make the
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound employed, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds or materials used in combination with the compound, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well, known in the medical arts.
  • a medical doctor e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the patients to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
  • the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of
  • compositions of the invention are formulated using one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions of the invention comprise a therapeutically effective amount of a compound of the invention and a pharmaceutically acceptable carrier.
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity may be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition.
  • Prolonged absorption of the injectable compositions may be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • compositions of the invention are administered to the patient in dosages that range from one to five times per day or more. In other words, the compositions of the invention are administered to the patient in dosages that range from one to five times per day or more. In other words,
  • the compositions of the invention are administered to the patient in range of dosages that include, but are not limited to, once every day, every two, days, every three days to once a week, and once every two weeks. It will be readily apparent to one skilled in the art that the frequency of administration of the various combination compositions of the invention will vary from individual to individual depending on many factors including, but not limited to, age, disease or disorder to be treated, gender, overall health, and other factors. Thus, the invention should not be construed to be limited to any particular dosage regime and the precise dosage and composition to be administered to any patient will be determined by the attending physical taking all other factors about the patient into account.
  • Compounds of the invention for administration may be in the range of from about 1 ⁇ g to about 10,000 mg, about 20 ⁇ g to about 9,500 mg, about 40 ⁇ g to about 9,000 mg, about 75 ⁇ g to about 8,500 mg, about 150 ⁇ g to about 7,500 mg, about 200 ⁇ g to about 7,000 mg, about 3050 ⁇ g to about 6,000 mg, about 500 ⁇ g to about 5,000 mg, about 750 ⁇ g to about 4,000 mg, about 1 mg to about 3,000 mg, about 10 mg to about 2,500 mg, about 20 mg to about 2,000 mg, about 25 mg to about 1,500 mg, about 30 mg to about 1,000 mg, about 40 mg to about 900 mg, about 50 mg to about 800 mg, about 60 mg to about 750 mg, about 70 mg to about 600 mg, about 80 mg to about 500 mg, and any and all whole or partial increments therebetween.
  • the dose of a compound of the invention is from about 1 mg and about 2,500 mg. In some embodiments, a dose of a compound of the invention used in compositions described herein is less than about 10,000 mg, or less than about 8,000 mg, or less than about 6,000 mg, or less than about 5,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1,000 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg.
  • a dose of a second compound is less than about 1,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof.
  • the present invention is directed to a packaged pharmaceutical composition
  • a packaged pharmaceutical composition comprising a container holding a therapeutically effective amount of a compound of the invention, alone or in combination with a second
  • Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for oral, parenteral, nasal, intravenous, subcutaneous, enteral, or any other suitable mode of administration, known to the art.
  • the pharmaceutical preparations may be sterilized and if desired mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or aromatic substances and the like. They may also be combined where desired with other active agents, e.g., other analgesic agents.
  • compositions of the invention include nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous.
  • the compounds for use in the invention may be formulated for administration by any suitable route, such as for oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and
  • compositions and dosage forms include, for example, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for intravesical administration and the like. It should be understood that the formulations and compositions that would be useful in the present invention are not limited to the particular formulations and compositions that are described herein.
  • compositions intended for oral use may be prepared according to any method known in the art and such compositions may contain one or more agents selected from the group consisting of inert, non-toxic pharmaceutically excipients that are suitable for the manufacture of tablets.
  • excipients include, for example an inert diluent such as lactose; granulating and disintegrating agents such as cornstarch; binding agents such as starch; and lubricating agents such as magnesium stearate.
  • the tablets may be uncoated or they may be coated by known techniques for elegance or to delay the release of the active ingredients.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert diluent.
  • the compounds of the invention may be in the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropylmethylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate).
  • the tablets may be coated using suitable methods and coating materials such as OPADRYTM film coating systems available from Colorcon, West Point, Pa.
  • Liquid preparation for oral administration may be in the form of solutions, syrups or suspensions.
  • the liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxy benzoates or sorbic acid).
  • suspending agents e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats
  • emulsifying agent e.g., lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters or ethyl alcohol
  • preservatives e.g., methyl or propyl p-hydroxy benzoates or sorbic acid
  • Granulating techniques are well known in the pharmaceutical art for modifying starting powders or other particulate materials of an active ingredient.
  • the powders are typically mixed with a binder material into larger permanent free-flowing agglomerates or granules referred to as a "granulation.”
  • solvent-using "wet" granulation processes are generally characterized in that the powders are combined with a binder material and moistened with water or an organic solvent under conditions resulting in the formation of a wet granulated mass from which the solvent must then be evaporated.
  • Melt granulation generally consists in the use of materials that are solid or semi-solid at room temperature (i.e.
  • melt granulation improves the dissolution rate and bioavailability of an active (i.e. drug) by forming a solid dispersion or solid solution.
  • U.S. Patent No. 5, 169,645 discloses directly compressible wax-containing granules having improved flow properties.
  • the granules are obtained when waxes are admixed in the melt with certain flow improving additives, followed by cooling and granulation of the admixture.
  • certain flow improving additives such as sodium bicarbonate
  • the present invention also includes a multi-layer tablet comprising a layer providing for the delayed release of one or more compounds of the invention, and a further layer providing for the immediate release of a medication for treatment of Parkinson's Disease.
  • a multi-layer tablet comprising a layer providing for the delayed release of one or more compounds of the invention, and a further layer providing for the immediate release of a medication for treatment of Parkinson's Disease.
  • a wax/pH-sensitive polymer mix a gastric insoluble composition may be obtained in which the active ingredient is entrapped, ensuring its delayed release.
  • the compounds of the invention may be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose and/or continuous infusion.
  • Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing and/or dispersing agents may be used. Additional Administration Forms
  • Additional dosage forms of this invention include dosage forms as described in U.S. Patents Nos. 6,340,475; 6,488,962; 6,451,808; 5,972,389; 5,582,837; and 5,007,790. Additional dosage forms of this invention also include dosage forms as described in U.S. Patent Applications Nos. 20030147952; 20030104062; 20030104053; 20030044466;
  • Additional dosage forms of this invention also include dosage forms as described in PCT Applications Nos. WO 03/35041 ; WO 03/35040; WO 03/35029; WO 03/35177; WO 03/35039; WO 02/96404; WO 02/32416; WO 01/97783; WO 01/56544; WO 01/32217; WO 98/55107; WO 98/11879; WO 97/47285; WO 93/18755; and WO 90/11757.
  • the formulations of the present invention may be, but are not limited to, short-term, rapid-offset, as well as controlled, for example, sustained release, delayed release and pulsatile release formulations.
  • sustained release is used in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that may, although not necessarily, result in substantially constant blood levels of a drug over an extended time period.
  • the period of time may be as long as a month or more and should be a release which is longer that the same amount of agent administered in bolus form.
  • the compounds may be formulated with a suitable polymer or hydrophobic material which provides sustained release properties to the compounds.
  • the compounds for use the method of the invention may be administered in the form of microparticles, for example, by injection or in the form of wafers or discs by implantation.
  • the compounds of the invention are administered to a patient, alone or in combination with another pharmaceutical agent, using a sustained release formulation.
  • delayed release is used herein in its conventional sense to refer to a drug formulation that provides for an initial release of the drug after some delay following drug administration and that mat, although not necessarily, includes a delay of from about 10 minutes up to about 12 hours.
  • pulsatile release is used herein in its conventional sense to refer to a drug formulation that provides release of the drug in such a way as to produce pulsed plasma profiles of the drug after drug administration.
  • immediate release is used in its conventional sense to refer to a drug formulation that provides for release of the drug immediately after drug administration.
  • short-term refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes and any or all whole or partial increments thereof after drug administration after drug administration.
  • rapid-offset refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes, and any and all whole or partial increments thereof after drug administration.
  • the therapeutically effective amount or dose of a compound of the present invention will depend on the age, sex and weight of the patient, the current medical condition of the patient and the progression of a condition considered herein in the patient being treated. The skilled artisan will be able to determine appropriate dosages depending on these and other factors.
  • a suitable dose of a compound of the present invention may be in the range of from about 0.01 mg to about 5,000 mg per day, such as from about 0.1 mg to about 1,000 mg, for example, from about 1 mg to about 500 mg, such as about 5 mg to about 250 mg per day.
  • the dose may be administered in a single dosage or in multiple dosages, for example from 1 to 4 or more times per day. When multiple dosages are used, the amount of each dosage may be the same or different. For example, a dose of 1 mg per day may be administered as two 0.5 mg doses, with about a 12-hour interval between doses.
  • the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days.
  • a 5 mg per day dose may be initiated on Monday with a first subsequent 5 mg per day dose administered on Wednesday, a second subsequent 5 mg per day dose administered on Friday, and so on.
  • the length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.
  • the dose reduction during a drug holiday includes from 10%- 100%, including, by way of example only, 10%, 15%,20%,25%,30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.
  • patients require intermittent treatment on a long-term basis upon any recurrence of symptoms and/or infection.
  • the compounds for use in the method of the invention may be formulated in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosage for patients undergoing treatment, with each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier.
  • the unit dosage form may be for a single daily dose or one of multiple daily doses (e.g., about 1 to 4 or more times per day). When multiple daily doses are used, the unit dosage form may be the same or different for each dose.
  • Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the determination of the LD5 0 (the dose lethal to 50% of the population) and the ED5 0 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD 50 and ED5 0 .
  • Capsid assembly inhibitors exhibiting high therapeutic indices are a specific example.
  • the data obtained from cell culture assays and animal studies is optionally used in formulating a range of dosage for use in human.
  • the dosage of such capsid assembly inhibitors lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity.
  • the dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.
  • reaction conditions including but not limited to reaction times, reaction size/volume, and experimental reagents, such as solvents, catalysts, pressures, atmospheric conditions, e.g., nitrogen atmosphere, and reducing/oxidizing agents, are within the scope of the present application.
  • Method A LC/MS data were determined with a Waters Alliance 2695 HPLC/MS (Waters Symmetry CI 8 column, 4.6 x 75 mm, 3.5 ⁇ ) with a 2996 diode array detector from 210-400 nm; the solvent system was 5-95% acetonitrile in water (with 0.1% TFA) over nine minutes using a linear gradient, and retention times were in minutes. Mass spectrometry was performed on a Waters ZQ using electrospray in positive mode.
  • Method B Preparative reversed phase HPLC was performed on a Waters Sunfire column (19 x 150 mm, CI 8, 10 ⁇ ) with a 20 min adjustable mobile phase gradient of acetonitrile/water with 0.1% TFA as buffer using 214 and 254 nm as detection
  • Method C LC/MS data were determined on a Shimadzu LC 20AD instrument with a Phenomenex Luna column (CI 8, 3.0 x 50 mm column, 3 ⁇ ). Mobile phase consisted of water and acetonitrile with 0.1% formic acid buffer. Gradient was 10-90% acetonitrile over three minutes and held at 90% acetonitrile for two minutes. Detection was performed on diode array detector from 210-400 nm and retention times are in minutes. Mass specs were determined on an Applied Biosystems MDS Sciex API 2000 instrument using electrospray ionization positive or negative mode.
  • Bacteria (10 7 -10 9 colony forming units, CFU) are spread onto the surface of agar plates containing 2x, 4x or lOx MIC of each organism/compound combination. Plates are incubated for 48 hr at the appropriate temperature. Colonies that appear are purified on agar containing the appropriate concentration of compound, and resistance is confirmed by MIC determination. Total DNA is prepared from colonies resistant to the different compounds and their parent strains and the A baumannii NDH-1 operon consisting of 14 structural genes (nuoA-N) is amplified by PCR. DNA is sequenced, and DNA sequences of parent and mutant genes are compared to identify any changes.
  • the mutant gene is PCR amplified and cloned into an appropriate vector and expressed in the wild type strain.
  • a wild type gene is expressed in isolated mutants to determine whether the expression of wild type protein is able to reverse resistance to the compound tested. These tests confirm if NDH-1 is the target for the compound. If a compound does not appear to target NDH-1, the genome is sequenced in an attempt to identify the target protein whose mutation conferred resistance to the organism.
  • the MIC is defined as the lowest concentration of compound that inhibits visible growth of the organism. Determinations are performed according to the Clinical and Laboratory Standards Institute recommended procedures for aerobic bacteria (Performanc e Standards for Antimicrobial Susceptibility Testing; 18 th Informational Supplement CSLI document M100-S18, Wayne, PA: Clinical and Laboratory Standards Institute, 2008). A comparator antibiotics is used. A positive growth control well containing medium and the test isolate is included on each plate.
  • WRAIR The Walter Reed Army Institute of Research
  • PFGE pulsed- field gel electrophoresis
  • ICC International Clonal Complex
  • XDR extremely drug-resistant
  • Bovine heart mitochondria is prepared (Smith, 1967, Methods Enzymol. 10:81-88), and SMPs are prepared (Matsuno-Yagi & Hatefi, 1985, J. Biol. Chem. 260: 14424- 14427) and IC5 0 values for ATP synthesis are determined in the same way as bacterial membranes.
  • mice are rendered neutropenic via intraperitoneal (IP) administration of 150 mg/kg and 100 mg/kg cyclophosphamide in sterile saline on Day -4 and Day -1 prior to inoculation (Day 0), respectively.
  • IP intraperitoneal
  • AB5075 is more virulent than other representative isolates and genetically tractable, which allows for larger therapeutic windows and subsequent studies.
  • Both models require AB5075 grown overnight in LB broth and then sub-cultured to mid-exponential phase. The inoculating culture is then washed and re-suspended in PBS with OD 6 oo values corresponding to 2.0 x 10 8 CFU/mL.
  • Murine pulmonary model of A. baumannii infection Murine pulmonary model of A. baumannii infection:
  • mice are anesthetized with oxygenated isofluorane immediately prior to intranasal inoculation with 25 ⁇ L of AB5075 corresponding to 5.0 x 10 6 CFU. Subsequently, mice are injected IP with the selected derivatives diluted in sterile saline, starting at 4 hours post-infection. Four hours postinfection is chosen because by this time point AB5075 has grown 1-1.5 log CFU. Other time points and dosing schedule are determined based on pilot efficacy experiments. In certain embodiments, the schedule is no more than 2-3 times per day where the effective dose is no more than 25 mg/kg in no more than 200 ⁇ L volumes injected IP.
  • a high dose (25 mg/kg) and a low dose (10 mg/kg) are tested in this model.
  • Safety is also assessed in these experiments as well where no bacteria are inoculated as a control).
  • the dosing regimen such as timing and amount are determined based upon the PK properties of the test compounds. A subset of mice is used for CFU/g wound tissue calculations, histopathology, and PK/PD evaluation. Formulation and vehicle are selected upon the physical properties of the test compounds.
  • mice Animal morbidity is scored twice daily for six days using a system evaluating mobility, coat condition, and conjunctivitis (Yano, et al, 2006, J. Biol. Chem. 281 : 11456- 11463). As mice become exceedingly moribund based on clinical score, they are humanely euthanized. To assess CFU burden in the lungs, mice are humanely euthanized according to protocol on Days 2 and 3 post- infection via an injection of ketamine (100 mg/kg) and xylazine (10 mg/kg). To quantify the pulmonary CFU burden, lungs are homogenized in 1 mL PBS and serial dilutions are plated using the Autoplate® Spiral Plating System
  • Murine wound model of A. baumannii infection Murine wound model of A. baumannii infection:
  • the wound model of infection is carried out as previously described
  • mice are pretreated with cyclophosphamide as described above, and after anesthesia, the dorsum of the mouse is shaved with an electric razor. The surgery area is sterilized with iodine and 70% alcohol. A full-thickness, excisional dermal wound is made on the back of each mouse with a 6 mm sterile biopsy punch, and 30 iL of AB5075 is inoculated in the wound with a 5.0 x 10 4 CFU dose (Fig. 10). Other groups of mice are wounded, but not inoculated with bacteria, and these mice serve as a negative control for efficacy and an evaluation of safety in this model of infection. Another negative control may include bacterial inoculum but no treatment.
  • mice are then re-caged, and analgesics are subcutaneous ly given with buprenorphine at 0.05 mg/kg twice per day for 1-2 days (depending on the clinical condition).
  • analgesics are subcutaneous ly given with buprenorphine at 0.05 mg/kg twice per day for 1-2 days (depending on the clinical condition).
  • mice are treated by IP dose of BDI derivative with a dosing schedule as determined by the murine pulmonary model experiments. A subset of mice is used for CFU/g wound tissue calculations, histopathology, and PK/PD evaluation.
  • mice For bacteria enumeration to calculate efficacy, the mice are monitored using an in vivo imaging system (IVIS Lumina XR, Caliper Life Sciences) every day during the first week, and every other day after the first week.
  • IVIS Lumina XR in vivo imaging system
  • mice For IVIS monitoring, mice are anesthetized by using isoflurane gas at 2 ⁇ 5% for induction, then 2% for maintenance anesthesia inside IVIS equipment.
  • the bacterial load is also enumerated by counting the bacteria from the grounded wound tissues of the excisional dermal area (central cores from punch biopsy made with 4 mm sterile biopsy punch) or from other tissues if needed (spleens, lungs, livers, kidneys or draining lymphoid nodes, and so on) of euthanized mice at 3 and 5 days; 1, 2 and 3 weeks post-infection, comparing the efficiency with both IVIS and classical CFU enumeration. All tissue is weighed and suspended in 1 mL of PBS and individually homogenized in tissue grinders or mesh bags. Serial 10-fold saline dilutions of homogenates are cultured on LB plate for bacterial enumeration. The number of bacterial colonies are accounted and expressed as CFU per gram tissue.
  • PCR RT 2 ProfilerTM PCR Array (Qiagen Inc.) according to manufacturer's instructions. This PCR array was designed specifically for mice. Separately and to confirm the presence or absence of certain cytokines, ELISA-based assays are performed.
  • cytokines and chemokines associated with the innate immune response are monitored such as: IL- ⁇ , IL-6, IL-8, and TNFa, as these are indicators of sepsis and an inflammatory response.
  • some animals are sacrificed at different time points and skin biopsies are fixed for histopathology analysis.
  • This analysis includes the characterization of the cell types at the wound site, inflammatory response, and immunohistochemistry staining to evaluate the presence of predatory bacteria in the wound. This may include hematoxylin and eosin as well as Brown and Brenn, and Brown and Hopps, methods for bacterial staining.
  • One section per block is evaluated via light microscopy by a Veterinary Pathologist blinded as to treatment group.
  • Epithelial Maturation the epithelial maturation is noted and scored based on the number of cell layers present (stratum basale, stratum spinosum, stratum granulosum, stratum corneum);
  • Granulation tissue the amount of granulation tissue is scored;
  • White cell infiltrate the amount of subepithelial mixed leukocytic infiltrates is scored as an indicator of the degree of inflammation;
  • Wound closure is measured over time via a Silhouette (Aranz Medical Ltd., New Zealand). At each time point a picture is taken, and wound area is determined by this instrument to provide a time to close metric of efficacy, which is really an evaluation of both the antimicrobial activity as well as wound healing. Gross pathology, impact on biofilm (scanning electron microscopy of the wound bed and dressing), and histology of the wound are evaluated (Thompson, et al, 2014, Antimicrob. Agents Chemother. 58: 1332-42).
  • Cytochrome P450 (CYP) inhibition In order to assess cytotoxicity, activity against actively dividing cells of the mammalian cell lines NIH3T3, HepG2 and A549 is analyzed, tested at half log dilutions. Standard antibiotics are used as comparators. Cytochrome P450 (CYP) inhibition:
  • Inhibition of CYP metabolite formation is assessed for the isoforms CYP1A2, CYP2D6, and CYP3A4 in human and mouse liver microsomes using isoform-specific probe substrates. Appropriate positive control inhibitors are used.
  • mice A total of 6 male mice, aged approximately 6-8 weeks, aree used for each test compound assessed.
  • Animals in the oral gavage dose group are bled via jugular vein cannula at the following time points: pre-dose, 30 minutes, and 1, 3, 6, 12, and 24 hours.
  • the blood is processed to plasma for analysis of the test compound via LC-MS/MS.
  • Isolated A. baumannii (Ab) membranes were screened against a small- molecule library of about 10,000 compounds.
  • the primary screen identified several drug-like scaffolds such as compound 1, which inhibited A. baumannii ATP synthesis.
  • Compound 1 belongs to the 1H- benzo[d]imidazol-2(3H)-imine compound class, and was found to have IC5 0 of 1.9 ⁇ for the inhibition of ATP synthesis, MIC of 17 ⁇ against .4. baumannii, and good selectivity for A baumannii OxPhos over mammalian (SMP) OxPhos.
  • SMP mammalian
  • the compounds of the invention do not inhibit mammalian NDH-1 up to about 100 ⁇ .
  • Example 2 Biochemical Investigations
  • the isolated membranes used for screening contain a fully functional OxPhos system and are amenable to biochemical investigation of the mechanism of action of compounds.
  • NDH-1 the type 1 NADH-quinone oxidoreductase was identified as a target of compound 1.
  • Fig. 3A illustrates the result of monitoring O2 consumption by A.
  • Trace (1) shows initial O2 consumption with NADH (ImM) as the substrate, which is abruptly terminated upon addition of compound 5 to give a concentration of 50 ⁇ .
  • Fig. 3B depicts the sequence of proteins that shuttle electrons through the OxPhos system of A.
  • Fig. 3A shows initial O2 consumption with succinate (50 mM) as the substrate, and no inhibition of O2 consumption upon addition of compound 5 to a 50 ⁇ concentration or upon a second addition to give a 100 ⁇ concentration.
  • succinate pathway via succinate dehydrogenase (SDH), is not affected by inhibition of NDH-1.
  • addition of the 4- F group to the phenyl ring blocks certain metabolism there.
  • Replacement of the 2,4-dichlorophenoxy of 5 with 4-CF 3 -biphenyl as in B43 yielded an improvement of MIC from 13.0 ⁇ to 3.1 ⁇ .
  • increased MIC potency upon substitution on the benzimidazole ring may be related to increased hydrophobicity (increased LogD) and thus increased cell penetration.
  • the 4-F group added to the phenyl ring blocks possible metabolism at that site.
  • the cytotoxicity evaluation is illustrated in Table 2 as an average between two cell lines (3T3/HepG2, EC5 0 S), with the ratios given relative to MIC.
  • the two cell lines were mouse 3T3 fibroblasts (ATCC, CRL-1658) and human transformed liver HepG2 cells (ATCC HB-8065), using an MTS viability assay (CellTiter 96® AQueous Non-Radioactive Cell Proliferation Assay) from Promega. Cytotoxicity selectivity was improved by incorporation of the 4-FPh to the benzimidazole, and improved by changing diethylamino to pyrrolidino (viz. B41).
  • Compound 5 was further evaluated in a panel of 23 clinical isolates of A. baumannii including multi-drug resistant (MDR) strains from various regions around the US, Europe and Latin America. The results are summarized in Figs. 5A-5B.
  • the MIC 90 for compound 5 was 8 ⁇ g/mL (13 ⁇ ) and the range was tight (4-16 ⁇ g/mL).
  • Compound 5 showed low metabolic stability in mouse and human liver microsomes (t 2 of 13 min and 9.2 min, respectively), which translates to the potential for high hepatic clearance rates in vivo.
  • the major metabolite generated in mouse liver microsomes was N-dealkylation of the diethyl amine, and minor amounts of hydroxy lated products were also observed.
  • other compounds not possessing the dialkylamino side chain were evaluated in mouse liver microsome stability assays including compounds 15, 25 and 26 (Figs. 6A-6B). Each of these three compounds showed considerably greater stability than 5 (48.2 min, 25.2 min and >90 min, respectively).
  • Metabolic stability in liver microsomes was indeed improved by changing the amine group and adding substitution onto the phenyl portion of the benzimidazole ring.
  • Compound B41 with the 4-FPh group on the middle ring has a metabolic half-life of 12.2 min, improved from 2.8 min for 18.
  • Benzimidazoles 2, 4-7, 13-29, B55 and B59-B62 were prepared using the synthetic schemes shown in Fig. 4. Compounds 2, 4-7 and 13-29 were prepared by sequential alkylation of 2-aminobenzimidazole (compound A) under relatively mild conditions to generate compound B, followed by a second alkylation under more rigorous conditions. 2-Aminobenzimidazoles B55 and B59-B62 were prepared by sequential reaction of 2-chlorobenzimidazole (compound C) to afford 2-aminobenzimidazoles D followed by alkylation on an internal nitrogen. Aminoalcohol B59 was prepared by reaction of 2- aminomethyl benzimidazole E with epoxide F derived upon reaction of epichlorohydrin and 2,4-dichlorophenol.
  • the crude was purified by silica chromatography using a gradient of 0-100% (90:9: 1 dichloromethane / methanol / ammonium hydroxide) to dichloromethane to generate N,N-diethyl-2-(2-imino-2,3-dihydro-lHbenzo[d]imidazol-yl)ethanamine (240 mg, 1.03 mmol, 39% yield).
  • Step 1 l-[2-(4-Bromo ⁇ henyl)-ethyl]-3-(2-diethylamino-ethyl)-2-imino-2,3-dihydro-lH- benzoimidazole
  • Example 12 the title compound was prepared (290 mg, 73% yield) by using l-(2-diethylamino-ethyl)-lH4jenzoimidazol-2-ylamine (Example 12, Step 1) as the starting material with 4-bromophenethyl bromide (Acros).
  • FC3317 was prepared by two procedures A and B below. Using procedure A. regiochemistry of the final product is unambiguous, as the procedure starts from starting material 4-chloro-2-fluoro-l -nitrobenzene. The same final product was obtained using both procedures A and B, indicating that chlorination step 2 of procedure B yields the chlorine atom in the position described. Procedure A
  • Step 4 l-(3-Diethylamino-propyl)-6-phenyl-lH-benzoimidazol-2-ylamine, 5-Chloro-l-(3- diethylamino-propyl)-6-phenyl-lH-benzoimidazol-2-ylamine, l-(3-Diethylamino-propyl)-
  • Step 5 l-[3-(2,4-Dichloro ⁇ henoxy) ⁇ ropyl]-3-(3-diethylamino-propyl)-2-imino-5-phenyl- 2,3-dihydro-lH-benzoimidazole
  • Example 12 l-[3-(2,4-Dichloro-phenoxy)-propyl]-3-(2-diethylamino-ethyl)-2-imino-5- phenyl-2,3-dihydro-lH-b B37)
  • Step 2 l-[3-(2,4-Dichloro ⁇ henoxy) ⁇ ropyl]-3-(2-diethylamino-ethyl)-2-imino-5-phenyl- 2,3-dihydro-lH-benzoimidazole
  • Example 13 l-[3-(2,4-Dichloro-phenoxy)-propyl]-5-(4-fluoro-phenyl)-3-(2-pyrrolidin- l-yl-ethyl)-l,3-dihydro-b or Compound B41)
  • Zinc dust (19.2 g, 296 mmol) was added in three portions 1.5 hours apart, while the reaction was at 45 °C.
  • the reaction was heated at 45 °C overnight then cooled, filtered through a thin pack of Celite, while rinsing with methanol, and concentrated in vacuo.
  • the residue was partitioned between ethyl acetate (222 mL) and IN HC1 (139 mL), and the mixture was stirred vigorously for one hour.
  • the layers were separated and the aqueous phase was made basic with -30% aq NaOH (30 mL).
  • the solid white zinc oxide that precipitated out of the solution was filtered off.
  • the product was extracted using ethyl acetate (3 x 100 mL).
  • Step 5 l-[3-(2,4-Dichloro-phenoxy)-propyl]-5-(4-fluoro-phenyl)-3-(2-pyrrolidin-l-yl-
  • Example 13 6-(4-fluoro-phenyl)-l-(2- pyrrolidin-1 -yl-ethyl)- 1, 3 -dihydro-benzoimidazol-2-ylideneamine (Example 13, Step 4) was used as a starting material to afford the bis TFA salt of the title compound (47 % yield).
  • the aqueous phase was extracted with 2 more portions of ethyl acetate.
  • the combined ethyl acetate extracts were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the crude product, which was purified by silica chromatography using a gradient of 0-100% (90:9: 1 DCM/methanol/ammonium hydroxide) to DCM to obtain N,N-diethyl-2-(2-imino-2,3-dihydro-lH-benzo[i/]imidazol-yl)ethanamine (240 mg, 1.03 mmol, 39% yield).
  • polymyxin E (colistin) is used clinically but suffers from dose-limiting toxicity.
  • Polymyxin B and colistin are lipopeptide antibiotics that are very similar, differing by one amino acid (D-Phe changed to D-Leu).
  • D-Phe changed to D-Leu
  • polymyxin B and E behave in the same way, and polymyxin B/colistin herein is used to indicate either polymyxin B or colistin.
  • Polymyxin B/colistin is believed to target bacterial membrane lipopolysaccharides and phospholipids, and to disrupt their integrity by increasing permeability and disturbing membrane surface charges. Polymyxin B/colistin also affects numerous membrane-associated cellular functions such as OxPhos and elevates drug penetration into bacterial cells. In certain embodiments, combination of the compounds of the present invention with polymyxin B/colistin allows for lower toxicity by using reduced levels of both compounds and/or reduces the frequency of resistance encountered with monotherapy.
  • FICx X/MICx
  • FICy Y/MICy
  • X is the concentration of compound x
  • MICx is the MIC of compound x alone
  • Y is the concentration of compound y
  • MICy is the MIC for compound y alone.
  • the FICI was displayed as an isobologram (Fig. 12B).
  • Colistin while fairly toxic, is prescribed clinically as the last line of defense against MDR- baumannii.
  • the isobolograms for B41, 25 and B45 and polymyxin B against .4. baumannii are shown in Figs. 12A-12B. Synergy with polymyxin B was observed for both 25 and B45, and more modestly for B41.
  • Compound 25 demonstrated a 1 ⁇ ICso when tested for its ability to inhibit ATP synthesis in A. baumannii membranes (Table 9).
  • FICI values for B41, 25, B45, 15 and 26 with A baumannii are shown in Table 10.
  • the synergy of 25 with polymyxin B was also examined against Klebsiella pneumonia (ATCC 4352) as shown in Fig. 12C and Table 11. Based on this analysis, there is at least comparable synergy in the action of 25 and polymyxin B on K. pneumonia, to that observed with A baumannii.
  • Related analogs B55-B58 also showed significant synergy with colistin (Table 12), but they have poor MICs as a single agent against A. baumannii (MICs > 100 ⁇ ). However, in the presence of sub-MIC concentrations of colistin, their MIC values dropped substantially, similar to the effect seen for 25, B45, 15 and 26.
  • Table 13 illustrates MIC values for compounds of the invention against colistin resistant ⁇ /? strains. MIC values of these strains for colistin were >128 ⁇ g/ml. Their growth are much slower than wild type, suggesting impaired fitness.
  • Synergistic inhibitory effects can be observed when two or more inhibitors interact at separate sites of an enzyme or a pathway and produce an effect greater than the sum of the individual effects.
  • the strong synergy of polymyxin B/colistin and compounds of the invention may be due to the fact that both polymyxin B, colistin and BDIs target bacterial membranes. Polymyxin B and colistin are believed to increase membrane permeability thereby permitting higher influx of compounds into bacterial cells.
  • permeablized membranes may lose the ability to maintain the chemical potential across the membrane that is generated by OxPhos. ATP synthesis activity depends on this potential and thus may be significantly compromised in the presence of low concentrations of polymyxin B or colistin.
  • drugs are often removed from cells by a wide array of efflux pumps (1) ABC transporters, (2) major facilitator superfamily (MFS) proteins, and (3) small multidrug resistance (SMR) family.
  • ABC transporters utilize ATP whereas the MFS and SMR families utilize H + /Na + motive forces. All efflux pumps depend on the OxPhos activity (Fig. 15), thus compounds of the invention, even in low concentrations, render bacteria more susceptible to treatment with polymyxin B and/or colistin.
  • B55 exerts bactericidal effects only with colistin.
  • colistin may enhance drug delivery to the target site and inhibition of respiration decreases PMF and intracellular ATP, thus attenuating efflux pump activities.
  • Combination doses and ratios of compounds of the invention and colistin for in vivo testing may be determined using a rapid, low-cost, invertebrate wax worm model of infection, followed by evaluation in the mouse A. baumannii wound model.
  • the Galleria mellonella (wax worm) infection model can be used to assess the safety and efficacy of novel therapeutics against bacterial pathogens including ⁇ , baumannii.
  • evaluation in G. mellonella can be used to evaluate combination therapy with colistin, specifically against A. baumannii and is useful as a low cost prescreen prior to going into rodent models.
  • the Galleria mellonella model is relatively easy to implement and does not require animal protocol approvals.

Abstract

The present invention includes a method of treating or preventing a bacterial infection in a subject in need thereof. The method comprises administering to the subject a therapeutically effective amount of at least one compound of the invention. In certain embodiments, the bacterium is Gram-negative. In other embodiments, the bacterium is obligatory aerobic.

Description

TITLE OF THE INVENTION
Novel Anti-Infective Compounds and Methods Using Same
CROSS-REFERENCE TO RELATED APPLICATION This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application No. 62/078,726, filed November 12, 2014, which application is hereby incorporated herein by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT
This invention was made with government support under R41 AI 108196-01 awarded by National Institutes of Health. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
Bacteria may be broadly classified as Gram-negative or Gram-positive, depending on the general structure of their cell wall. Gram-negative bacteria present a thin peptidoglycan layer in their cell wall, and this layer is sandwiched between an inner cell membrane and an outer cell membrane, which comprises lipopolysaccharide (comprising lipid A, core polysaccharide, and O antigen) in its outer leaflet and phospholipids in the inner leaflet. Gram-positive bacteria on the other hand have a thick peptidoglycan layer outside the cell membrane. Gram-positive bacteria tend to be more permeable to antibiotics than Gram- negative bacteria, because of the latter' s relatively impermeable lipid-based bacterial outer membrane.
The proteobacteria are a major group of Gram-negative bacteria, including Escherichia coli (E. coii), Salmonella, Shigella and other Enter obacteriaceae, Pseudomonas, Moraxella, Helicobacter, Stenotrophomonas, Bdellovibrio, acetic acid bacteria, and
Legionella. Other notable groups of Gram-negative bacteria include the cyanobacteria, spirochaetes, green sulfur, and green non-sulfur bacteria. Medically relevant Gram-negative cocci may cause sexually transmitted diseases (Neisseria gonorrhoeae), meningitis (Neisseria meningitidis), and respiratory symptoms (Moraxella catarrhalis). Medically relevant Gram- negative bacilli may cause respiratory problems (Hemophilus influenzae, Klebsiella pneumoniae, Legionella pneumophila, Pseudomonas aeruginosa), urinary problems
(Escherichia coli, Proteus mirabilis, Enterobacter cloacae, Serratia marcescens), and gastrointestinal problems (Helicobacter pylori, Salmonella enteritidi , Salmonella typhi). Gram-negative bacteria associated with hospital-acquired infections include Acinetobacter baumannii, which cause bacteremia, secondary meningitis, and ventilator-associated pneumonia in hospital intensive-care units.
Gram-negative pathogens, such as Acinetobacter baumannii, are responsible for over 30% of the most common hospital acquired infections (Hidron, et al, 2008, Infect. Control Hosp. Epidemiol., 29:996). This number rises to 70% in intensive care units, where 74% of A. baumannii isolated are multidrug-resistant (Hidron, et al, 2008, Infect. Control Hosp. Epidemiol, 29:996). A. baumannii infection in burn wound patients increases the cost of treatment by almost $ 100,000 per patient (Wilson, et al, 2004, Am. J. Infect. Control 32:342). A. baumannii is the second most commonly isolated non-fermenting bacterium in humans, and infection can result in pneumonia, skin and wound infections, bacteremia and meningitis. In addition, A. baumannii biofilms have been implicated in diseases such as cystic fibrosis, periodontitis and urinary tract infections, partly because of the bacteria's ability to colonize indwelling medical devices. The continual appearance oiA. baumannii strains resistant to β-lactams, cephalosporins, aminoglycosides, quinolones and lately carbapenems has compromised treatment options.
The OxPhos system is composed of the electron transport chain (ETC) and ATP synthase, and plays a central role in ATP synthesis in various Gram-positive and Gram- negative bacterial pathogens. Biochemical studies (de Graef, et al, 1999, J. Bacteriol 181 :2351 ; Gyan, et al, 2006, J. Bacteriol. 188:7062; Yano, et al, 2006, J. Biol. Chem. 281 : 1 1456; Weinstein, et al, 2005, Proc. Natl. Acad. Sci. USA 102:4548) and genetic studies (Green & Paget, 2004, Nat. Rev. Microbiol. 2:954; Partridge, et ah, 2006, J. Biol. Chem. 281 :27806) on the OxPhos pathway in bacteria indicate that these coupled processes (re- oxidation of NADH, transfer of electrons along the ETC and the establishment, maintenance and utilization of the electrochemical proton gradient to drive ATP synthesis) are essential for bacterial survival under a wide range of conditions. While sequence overlap exists between some bacterial and mammalian mitochondrial OxPhos components, there is evidence that marked differences exist between the components of the bacterial ETC/ATP synthase and those of mammalian mitochondria. For example, cytochrome bd oxidases and type II
NADH:quinone oxidoreductase (NDH-2) have no counterpart in human mitochondria, but appear to be nearly ubiquitous in aerobic bacteria. In addition, bioinformatic analyses indicate structural divergence between the enzymes of the OxPhos pathway of bacteria and mitochondria. There is a need in the art to identify novel compounds useful in the treatment of Gram-negative infections, such as infections caused by A. baumannii. The present invention addresses this need.
BRIEF SUMMARY OF THE INVENTION
The invention provides certain compounds, or salts, solvates or N-oxides thereof. The invention further provides pharmaceutical compositions comprising at least one compound of the invention. The invention further provides a method of treating or preventing a infection by a bacterium in a subject in need thereof.
In certain embodiments, the compound of the invention, or a salt, solvate or N- oxide thereof, is selected from the group consisting of:
Figure imgf000004_0001
wherein in (I)-(III):
ring A is a fused aryl or heteroaryl ring selected from the group consisting
Figure imgf000004_0002
each occurrence of R1 is independently a bond or -CH2-;
R2 is selected from the group consisting of -(CH2)i_3-0-(CH2)o-2-,
Figure imgf000004_0003
and -(CH2)!_6, wherein each of the CH2 groups in R2 is independently optionally substituted with a substituent selected from the group consisting of C1-C6 alkyl, C1-C6 fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R4 is -(CH2)o-4;
each occurrence of R6 is independently CH, CR10 or N, wherein 0-2 occurrences of R6 in ring A are N; each occurrence of R7 is independently H, Ci-Ce alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted; each occurrence of R8 is independently Ci-Ce alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted;
each occurrence of R9 is independently O, S or R7;
each occurrence of R10 is independently selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
wherein in (I):
X1 and X2 are independently selected from the group consisting of H and optionally substituted Ci-C6 alkyl, X1 and X2 combine to form =NH, =N(Ci-C6 alkyl), =CH-CN, =N- CN, or =CH-N02, or X1 is absent and X2 is -N=CH-NMe2;
wherein in (I) and (II):
each occurrence of R3 is independently -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci- Ce alkyl), pyrrolidin- 1 -yl, pyperidin-l -yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7, SR7,
S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
wherein in (I) and (III):
R5 is selected from the group consisting of H, prop-2-en- l-yl, Ci-Ce alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2, -(C=O)0-i-(OCH2CH2)1_
3-OR ,
Figure imgf000005_0001
; wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1 ;
wherein in (II):
R5 is selected from the group consisting of H, prop-2-en- 1 -yl, Ci-Ce alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2,
Figure imgf000006_0001
; wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1 ;
or R4 is a bond and R5 combines with the N atom to which it is bound to form morpholyn-l -yl, piperidin- l-yl, 4-cyano-piperidin-l -yl and piperazin-l -yl, wherein N4 of the piperazinyl is optionally substituted with Ci-Ce alkyl;
wherein in (III):
each occurrence of R3 is independently H, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), pyrrolidin-l -yl, pyperidin- l-yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7,
SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7; and any mixtures thereof.
In certain embodiments, the compound is not: l -(2,4-dichlorophenoxy)-3-(2- imino-3-propyl-2,3-dihydro- lH-benzo[d]imidazol-l -yl) propan-2-ol (Al); l -(3-allyl-2-imino- 2,3-dihydro- lH-benzo[d]imidazol- l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A2); l -(2,4- dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro- lH-benzo[d] imidazol- 1 -yl)propan-2-ol (1); 1 -(3 -benzyl-2-imino-2,3 -dihydro- lH-benzo[d]imidazol- 1 -yl)-3 -(2,4- dichlorophenoxy) propan-2-ol (A4); or l -(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin- l-yl)ethyl)-2,3-dihydro- lH-benzo[d] imidazol- l-yl)propan-2-ol (A5).
In certain embodiments, the at least one compound is selected from the group consisting of:
Figure imgf000006_0002
(Ilia).
In certain embodiments, in (I), when X1 and X2 combine to form =NH and ring A is a six-membered ring, then at least one occurrence of R6 is CR10 or N. In other embodiments, (I) is not a compound of formula (lb):
Figure imgf000007_0001
(lb), wherein in (lb): RA is C1-C7 alkyl, C2-C7 alkenyl, - CH2CH2 Et2 or -CH^CH^ 1 -morpholinyl), or benzyl optionaly substituted with at least one halogen or C1-C3 alkyl; RB is phenyl optionally substituted with at least one halogen; aryl; aroxy; naphthyl; phenoxymethyl optionally substituted with at least one halogen, C1-C3 alkyl and/or C1-C3 alkoxy; phenyl isoxazole optionally substituted with at least one halogen; or 4- methyl-3 -phenyl- l,2,4-oxadiazole-5(4H)-one.
In certain embodiments, ring A is
Figure imgf000007_0002
, wherein R6 is CH or CR10. In other embodiments, at least one occurrence of R6 in ring A is CR10. In yet other
em s F, CI, Br, sisting
of:
Figure imgf000007_0003
. In yet other embodiments, ring A is selected from the group consisting of:
Figure imgf000007_0004
wherein Hal is halogen. In yet other embodiments, R10 is optionally substituted phenyl or optionally substituted pyridyl. In yet other embodiments, R10 is 4-fluorophenyl, p-tolyl or phenyl. In yet other embodiments, R9 is O or NR7. In yet other embodiments, R7 is methyl or 4-fluorophenyl.
In certain embodiments, both occurrences of R1 are a bond.
In certain embodiments, R2 is selected from the group consisting of -(CH2)1-3-
0- , -(CH2)i-3-NR7-, -(CH2)1-3-S-, and -(CH2)i-6-. In other embodiments, at least one of the CH2 groups in R2 is independently substituted with OR7. In yet other embodiments, R2 is -
CH2CH2-, -CH2CH2CH2-, -CH2CH20-, -CH2CH2CH20-, or -CH2CH(OH)CH20-. In yet other embodiments, R3 or R3 is phenyl, pyridyl, pyrimidyl, 1 ,2-biphenyl, 1 ,3-biphenyl, 1,4- biphenyl, or naphthyl, wherein the aryl or heteroaryl group is optionally substituted with at least one selected from the group consisting of Ci-Ce alkyl, Ci-Ce fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7,
OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7. In yet other embodiments, R3 or R3 is phenyl, 2-methylphenyl, 3- methylphenyl, 4-methylphenyl, 2-chlorophenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4- methoxyphenyl, 3-chlorophenyl, 4-chlorophenyl, 2-bromophenyl, 3-bromophenyl, 4- bromophenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2,4-difluorophenyl, 2,4-dichlorophenyl, 1,2-biphenyl, 1,3-biphenyl, 1 ,4-biphenyl, 4- trifluoromethylmethyl- 1 ,4-biphenyl, 4' -fluoro-2-chloro- 1 ,4-biphenyl, 2-trifluoromethyl- pyridin-5-yl, and 2-furyl. In yet other embodiments, R5 or R5 is selected from the group consisting of H, Ci-Ce alkyl, aryl, N(R7)2, pyrrolidin- l-yl, piperidin-l -yl, N^H-piperidin^- yl, 1-(Ci-C6 alkyl)-piperidin-4-yl, N1-(tBoc)-piperidin-4-yl, morpholin-l -yl, imidazol- 1 -yl, imidazol-2-yl, pyrrolidin-2-one- l-yl, pyridyl, and
Figure imgf000008_0001
In yet other embodiments, R5 or R5 is N(R7)2, wherein each occurrence of R7 is independently H, methyl, ethyl, w-propyl or isopropyl. In yet other embodiments, R5 or R5 is selected from the group consisting of pyrrolidin- l -yl, piperidin- l-yl, N1-H-piperidin-4-yl,
Figure imgf000008_0002
alkyl)- piperidin-4-yl, N1-(tBoc)-piperidin-4-yl, morpholin- l-yl, imidazol- 1 -yl, imidazol-2-yl, pyrrolidin-2-one-l -yl, pyridyl, 3,5-dimethylisoxazol-4-yl, 6-trifluoromethyl-pyridin-3-yl, - (C=0)o-i-(OCH2CH2)1_3-0(Ci-C6 alkyl), and -(C=O)0-i-( 4-phenyl-piperazin- l-yl).
In certain embodiments, R6 is CR7. In other embodiments, each occurrence of
R6 is CH.
In certain embodiments, the compound of formula (I) is at least one selected from the group consisting of: l-(3-(2,4-dichlorophenoxy)propyl)- lH-benzo[d]imidazol-2- amine (4); 2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol-
1- yl)-N,N-diethylethan- l -amine (5); 2-(3-(2-(2,4-dichlorophenoxy)ethyl)-2-imino-2,3- dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (6); 2-(3 -(3 -(2,4- dichlorophenyl)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 - amine (7); l-(3-(2,4-dichlorophenoxy)propyl)-3-ethyl-l,3-dihydro-2H-benzo[d]imidazol-2- imine (13); l-butyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (14); l-(3-(2,4-dichlorophenoxy)propyl)-3-(3-phenylpropyl)-l,3-dihydro-2H- benzo [d]imidazol-2-imine (15); N-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl) ethyl)-N-propylpropan- 1 -amine (16); N-(2-(3-(3-(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)ethyl)-N- isopropylpropan-2-amine (17); l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyrrolidin-l- yl)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (18); l-(3-(2,4-dichlorophenoxy)propyl)- 3-(2-(piperidin-l-yl)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (19); l-(3-(2,4- dichlorophenoxy)propyl)-3-(2-moφholinoethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (20); l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyridin-2-yl)ethyl)-l,3-dihydro-2H-benzo[d] imidazol-2-imine (21); tert-butyl 4-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3- dihydro-lH-benzo[d] imidazol- l-yl)ethyl)piperidine-l-carboxylate (22); l-(2-(lH-imidazol- 1 -yl)ethyl)-3 -(3 -(2,4-dichlorophenoxy)propyl)- 1 ,3 -dihydro-2H-benzo [d] imidazol-2-imine (23); l-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethyl)pyrrolidin-2-one (24); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(4-phenylpiperazin- 1 -yl)ethan- 1 -one (25); 1 -(3 -(2,4- dichlorophenoxy)propyl)-3-((6-(trifluoromethyl)pyridin-3-yl)methyl)-l,3-dihydro-2H- benzo [d]imidazol-2-imine (26); l-(3-(2,4-dichlorophenoxy)propyl)-3-((3,5-dimethylisoxazol- 4-yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (27); 2-(3-(3-(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N- diethylacetamide (28); 1 -(3 -(2,4-dichlorophenoxy)propyl)-3-(2-(2-ethoxyethoxy)ethyl)- 1,3- dihydro-2H-benzo[d]imidazol-2-imine (29); 3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylpropan-l-amine (33); 3-(5-chloro-3-(3- (2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)-N,N- diethylpropan-1 -amine (34); l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B4); (R)-l-(2,4-dichlorophenoxy)-3-(3-(2- (diethylamino)ethyl)-2-imino-2, 3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol (B5); ((S)- l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B6); l-(2-(2,4-dichlorophenoxy)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B13); l-(3-(2,4-dichlorophenyl)propyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B14); 2-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B15); l-(cyclopropylmethyl)-3-(3- (2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B17); l-(3-(2,4- dichlorophenoxy)propyl)-3-(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B18); 3-(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)-N,N-diethylpropan- 1 -amine (B23); 3 -(3 -(3 -(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N- dipropylpropan- 1 -amine (B25); N-benzyl-2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3- dihydro- lH-benzo[d]imidazol- 1 -yl)-N-ethylethan- 1 -amine (B26); 2-(3-(3 -(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N-ethylethan-l- amine (B27); N-benzyl-2-(3-(3-(2-chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N-ethylethan-l -amine (B28); N,N-diethyl-2-(2-imino-3-(3-(2- (trifluoromethyl)phenoxy)propyl)-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B29); 3-(2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N,N-dipropylpropan-l -amine (B30); N,N-diethyl-3-(2-imino-5,6- diphenyl-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan- 1 -amine (B31); 3 -(3 -(3 -(2,4- dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylpropan-1 -amine (B32); 3-(5-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6- phenyl-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)-N,N-diethylpropan- 1 -amine (B33); 3 -(3 -(3 - (2,4-dichlorophenoxy)propyl)-2-imino-5,6-diphenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)- N,N-diethylpropan-l -amine (B34); 2-(3-(2-([l, l'-biphenyl]-4-yl)ethyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B35); 2-(6-chloro-3-(3-(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B36); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B37); 2-(3 -(3 -(2,4- dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3 -dihydro- lH-benzo[d]imidazol-l- yl)-N,N-diethylethan-l -amine (B38); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4- trifluoromethylphenyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylethan-l- amine (B39); 1 -(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(piperidin- 1 - yl)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B40); l-(3-(2,4- dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(pyrrolidin-l-yl)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B41); 6-(4-fluorophenyl)-l-(2-(pyrrolidin-l-yl)ethyl)-lH- benzo[d]imidazol-2-amine (B42); N,N-diethyl-2-(2-imino-3-(2-(4'-(trifluoromethyl)-[l,r- biphenyl] -4-yl)ethyl)-2, 3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B43); N-(2-(3 - (3 -(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3 -dihydro- 1 H- benzo[d] imidazol- l-yl)ethyl)-N-isopropylpropan-2-amine (B44); N-(2-(6-(4-fluorophenyl)-2- imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethyl)-N-isopropylpropan-2-amine (B45); N,N-diethyl-2-(2-imino-3-(3-((4'- (trifluoromethyl)-[l,r-biphenyl]-4-yl)oxy)propyl)-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethan- 1 -amine (B46); 2-(l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-l,2-dihydro-3H- imidazo[4,5-b]pyridin-3-yl)-N,N-diethylethan-l-amine (B47); l-(3-(2,4- dichlorophenoxy)propyl)-6-(4-fluorophenyl)- 1 H-benzo [d] imidazol-2-amine (B48); 1 -(3 -((3 - chloro-4'-fluoro-[l,r-biphenyl]-4-yl)oxy)propyl)-6-(4-fluorophenyl)-lH-benzo[d]imidazol-2- amine (B49); 3-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-l-(2-(pyrrolidin-l- yl)ethyl)- 1 ,3 -dihydro-2H-benzo [d] imidazol-2-imine (B50); 3 -(3 -((3 -chloro-4'-fluoro-[ 1,1'- biphenyl]-4-yl)oxy)propyl)-5-(4-fluorophenyl)-l-(2-(pyrrolidin-l-yl)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B51); methyl 2-amino-l-(2-(pyrrolidin-l-yl)ethyl)-lH- benzo[d]imidazole-6-carboxylate (B52); methyl 3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 1 -(2-(pyrrolidin- 1 -yl)ethyl)-2,3 -dihydro- 1 H-benzo [d] imidazole-5 -carboxylate (B53); methyl l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-3-(2-(pyrrolidin-l-yl)ethyl)-2,3-dihydro-lH- benzo[d]imidazole-5-carboxylate (B54); l-(3-((2,4-dichlorophenyl)thio)propyl)-lH- benzo[d]imidazol-2-amine (B76); 2-(2-amino-lH-benzo[d]imidazol-l-yl)-N-(2,4- dichlorophenyl)acetamide (B77); N-(2,4-dichlorophenyl)-2-(3-(2-(diethylamino)ethyl)-2- imino-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)acetamide (B78); (E)-N, 1 -dibutyl-3 -(3 -(2,4- dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B79); (E)-l-(3-(2,4- dichlorophenoxy)propyl)-N,3-bis(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B81); (E)-N'-(l-(3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-yl)-N,N- dimethylformimidamide (B80); l-(2-(dipropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B87); l-(2-(diisopropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B88); l-(2-(pyrrolidin- l-yl)ethyl)-lH-benzo[d]imidazol-2-amine (B89); 3-(3-(3-(2,4-dichlorophenoxy)propyl)-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylpropan-l-amine (B90); 2-(3-(3-(2- chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N-ethylethan-l- amine (B91); 2-(3-(3-(2,4-difluorophenoxy)propyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N,N-diethylethan-l -amine (B92); l-(3-(diethylamino)propyl)-6- phenyl-lH-benzo[d]imidazol-2-amine (B93); 5-chloro-l-(3-(diethylamino)propyl)-6-phenyl- lH-benzo[d]imidazol-2-amine (B94); 2-(3-(4-bromophenethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N,N-diethylethan-l -amine (B95); l-(2-(diethylamino)ethyl)-6- phenyl-lH-benzo[d]imidazol-2-amine (B97); 6-chloro-l-(3-(2,4-dichlorophenoxy)propyl)- lH-benzo[d]imidazol-2-amine (B98); l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)- lH-benzo[d]imidazol-2-amine (B99); l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)- 3-((6-(trifluoromethyl)pyridin-3-yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-im^
(B100); 5-(4-fluorophenyl)-l-((6-(trifluoromethyl)pyridin-3-yl)methyl)-lH- benzo[d]imidazol-2-amine (B101); l-([l, l'-biphenyl]-4-yl)-2-(3-allyl-2-imino-2,3-dihydro- lH-benzo[d]imidazol-l-yl)ethan-l-ol (B102); l-(3-(2,4-dichlorophenoxy)propyl)-3-propyl- l,3-dihydro-2H-benzo[d]imidazol-2-imine (B103); 2-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d]imidazol- 1 -yl)- 1 -(p-tolyl)ethan- 1 -ol (B104); 2-(3-allyl-2-imino-2,3 -dihydro- 1 H- benzo[d]imidazol-l-yl)-l-(4-bromophenyl)ethan-l-ol (B105); 2-(3-allyl-2-imino-2,3- dihydro- lH-benzo[d]imidazol- 1 -yl)-l -(3-bromophenyl)ethan- 1 -ol (B106); 2-(3-allyl-2- imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)- 1 -(furan-2-yl)ethan- 1 -ol (B107); 2-(3 -allyl-2- imino-2,3 -dihydro- lH-benzo[d]imidazol-l-yl)-l-(4-methoxyphenyl)ethan-l-ol (B108); l-(4- chlorophenoxy)-3-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B109); l-(3-propyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4- chlorophenoxy)propan-2-ol (B110); l-(4-chlorophenoxy)-3-(2-imino-3-methyl-2,3-dihydro- lH-benzo[d]imidazol-l-yl)propan-2-ol (Bill); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy)propan-2-ol (B112); l-(3-butyl-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol (B113); l-(3-ethyl-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B114); l-(3-allyl-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol (B115); l-(2- imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B116); l-(2- imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B117); l-(2- imino-3 -propyl-2,3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-3 -(2-methoxyphenoxy)propan-2-ol (B118); l-(2-chlorophenoxy)-3-(3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2- ol (B119); l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B120); 1 -(3 -butyl-2-imino-2,3 -dihydro- 1 H- benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B121); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B122); l-(4-chlorophenoxy)-3-(3-(2- (diethylamino)ethyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol (B123); 1 - (2-imino-3 -methyl-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3-(p-tolyloxy)propan-2-ol
(B124); l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2- methoxyphenoxy)propan-2-ol (B125); l-(2-chlorophenoxy)-3-(2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B126); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-3-(4-methoxyphenoxy)propan-2-ol (B127); l-(2-imino-3-methyl-2,3- dihydro-lH-benzo[d]imidazol-l-yl)-3-(m-tolyloxy)propan-2-ol (B128); and l-(2-imino-3- methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(o-tolyloxy)propan-2-ol (B129). In certain embodiments, the compound of formula (II) is at least one selected from the group consisting of: l-(2,4-dichlorophenoxy)-3-(2-((3-morpholinopropyl)amino)-lH- benzo[d] imidazol- l-yl)propan-2-ol (B55); l-(2,4-dichlorophenoxy)-3-(2-(phenethylamino)- lH-benzo[d] imidazol- l-yl)propan-2-ol (B56); l-(2-((l-benzylpiperidin-4-yl)amino)-lH- benzo [d] imidazol- l-yl)-3 -(2,4-dichlorophenoxy )propan-2-ol (B57); l-(2,4-dichlorophenoxy)- 3-(2-((2-(diethylamino)ethyl)amino)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B60); l-(2,4- dichlorophenoxy)-3 -(2-((3 -(diethylamino)propyl)amino)- 1 H-benzo[d] imidazol- 1 -yl)propan-
2- ol (B61); 1 -( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo[d] imidazol-2-yl)-N3 ,N3 - diethylpropane-1 ,3-diamine (B62); 1 -(2,4-dichlorophenoxy)-3-(2-(4-ethylpiperazin- 1 -yl)- 1H- benzo [d] imidazol- l-yl)propan-2-ol (B66); l-(2,4-dichlorophenoxy)-3-(2-((5-
(diethylamino)pentan-2-yl)amino)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B73); and 1-(1- (3-(2,4-dichlorophenoxy)-2-hydroxypropyl)-lH-benzo[d]imidazol-2-yl)piperidine-4- carbonitrile (B74).
In certain embodiments, the compound of formula (III) is at least one selected from the group consisting of: N-((lH-benzo[d]imidazol-2-yl)methyl)-3-(2,4- dichlorophenoxy)propan-l -amine (B58); l-(((lH-benzo[d]imidazol-2-yl)methyl)amino)-3- (2,4-dichlorophenoxy)propan-2-ol (B59); l-(2,4-dichlorophenoxy)-3-((l-(2- (diethylamino)ethyl)-lH-benzo[d]imidazol-2-yl)oxy)propan-2-ol (B64); N-pentyl-lH- benzo[d]imidazol-2-amine (B83); N-phenethyl-lH-benzo[d]imidazol-2-amine (B84); Nl- ((lH-benzo[d]imidazol-2-yl)methyl)-Nl-(3-(2,4-dichlorophenoxy)propyl)-N2,N2- diethylethane-l,2-diamine (B85); and Nl-(3-(2,4-dichlorophenoxy)propyl)-Nl-((l-(2- (diethylamino)ethyl)- lH-benzo[d]imidazol-2-yl)methyl)-N2,N2-diethylethane- 1 ,2-diamine (B86).
In certain embodiments, the compound is at least one selected from the group consisting of: l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH- benzo[d] imidazol- 1-yl) propan-2-ol (Al); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-3 -(2,4-dichlorophenoxy) propan-2-ol (A2); l-(2,4-dichlorophenoxy)-
3 - (3 -(2-(diethylamino)ethyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol (1); l-(3-benzyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A4); l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin-l-yl)ethyl)-2,3- dihydro-lH-benzo[d] imidazol- 1 -yl)propan-2-ol (A5); 1 -(3 -(2,4-dichlorophenoxy )propyl)- lH-benzo[d]imidazol-2-amine (4); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3- dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (5); 2-(3 -(2-(2,4- dichlorophenoxy)ethyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylethan- 1-amine (6); 2-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (7); 1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -ethyl- 1 ,3 -dihydro- 2H-benzo[d]imidazol-2-imine (13); l-butyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro- 2H-benzo[d]imidazol-2-imine (14); l-(3-(2,4-dichlorophenoxy)propyl)-3-(3-phenylpropyl)- l,3-dihydro-2H-benzo[d]imidazol-2-imine (15); N-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl) ethyl)-N-propylpropan- 1-amine (16); N-(2-(3- (3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)ethyl)-N- isopropylpropan-2-amine (17); l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyrrolidin-l- yl)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (18); l-(3-(2,4-dichlorophenoxy)propyl)- 3-(2-(piperidin-l-yl)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (19); l-(3-(2,4- dichlorophenoxy)propyl)-3-(2-moφholinoethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (20); l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyridin-2-yl)ethyl)-l,3-dihydro-2H-benzo[d] imidazol-2-imine (21); tert-butyl 4-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3- dihydro-lH-benzo[d] imidazol-l-yl)ethyl)piperidine-l-carboxylate (22); l-(2-(lH-imidazol- 1 -yl)ethyl)-3 -(3 -(2,4-dichlorophenoxy)propyl)- 1 ,3 -dihydro-2H-benzo [d] imidazol-2-imine (23); l-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethyl)pyrrolidin-2-one (24); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(4-phenylpiperazin- 1 -yl)ethan- 1 -one (25); 1 -(3 -(2,4- dichlorophenoxy)propyl)-3-((6-(trifluoromethyl)pyridin-3-yl)methyl)-l,3-dihydro-2H- benzo [d]imidazol-2-imine (26); l-(3-(2,4-dichlorophenoxy)propyl)-3-((3,5-dimethylisoxazol- 4-yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (27); 2-(3-(3-(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N- diethylacetamide (28); 1 -(3 -(2,4-dichlorophenoxy)propyl)-3-(2-(2-ethoxyethoxy)ethyl)- 1,3- dihydro-2H-benzo[d]imidazol-2-imine (29); 3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylpropan-l-amine (33); 3-(5-chloro-3-(3- (2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)-N,N- diethylpropan- 1-amine (34); l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B4); (R)-l-(2,4-dichlorophenoxy)-3-(3-(2- (diethylamino)ethyl)-2-imino-2, 3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol (B5); ((S)- l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)propan-2-ol (B6); l-(2-(2,4-dichlorophenoxy)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B13); l-(3-(2,4-dichlorophenyl)propyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B14); 2-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1-amine (B15); l-(cyclopropylmethyl)-3-(3- (2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B17); l-(3-(2,4- dichlorophenoxy)propyl)-3-(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B18); 3-(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)-N,N-diethylpropan- 1 -amine (B23); 3 -(3 -(3 -(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N- dipropylpropan- 1 -amine (B25); N-benzyl-2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3- dihydro- lH-benzo[d]imidazol- 1 -yl)-N-ethylethan- 1 -amine (B26); 2-(3-(3 -(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N-ethylethan-l- amine (B27); N-benzyl-2-(3-(3-(2-chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N-ethylethan-l -amine (B28); N,N-diethyl-2-(2-imino-3-(3-(2- (trifluoromethyl)phenoxy)propyl)-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B29); 3-(2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N,N-dipropylpropan-l -amine (B30); N,N-diethyl-3-(2-imino-5,6- diphenyl-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan- 1 -amine (B31); 3 -(3 -(3 -(2,4- dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylpropan-1 -amine (B32); 3-(5-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6- phenyl-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)-N,N-diethylpropan- 1 -amine (B33); 3 -(3 -(3 - (2,4-dichlorophenoxy)propyl)-2-imino-5,6-diphenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)- N,N-diethylpropan-l -amine (B34); 2-(3-(2-([l, l'-biphenyl]-4-yl)ethyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B35); 2-(6-chloro-3-(3-(2,4- dichlorophenoxy)propyl)-2-imino-2,3-dihydro- lH-benzo[d]imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B36); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B37); 2-(3 -(3 -(2,4- dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3 -dihydro- lH-benzo[d]imidazol-l- yl)-N,N-diethylethan-l -amine (B38); 2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4- trifluoromethylphenyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylethan-l- amine (B39); 1 -(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(piperidin- 1 - yl)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B40); l-(3-(2,4- dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(pyrrolidin-l-yl)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B41); 6-(4-fluorophenyl)-l-(2-(pyrrolidin-l-yl)ethyl)-lH- benzo[d]imidazol-2-amine (B42); N,N-diethyl-2-(2-imino-3-(2-(4'-(trifluoromethyl)-[l, r- biphenyl] -4-yl)ethyl)-2, 3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B43); N-(2-(3 - (3 -(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3 -dihydro- 1 H- benzo[d] imidazol- l-yl)ethyl)-N-isopropylpropan-2-amine (B44); N-(2-(6-(4-fluorophenyl)-2- imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethyl)-N-isopropylpropan-2-amine (B45); N,N-diethyl-2-(2-imino-3-(3-((4'- (trifluoromethyl)-[l,r-biphenyl]-4-yl)oxy)propyl)-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethan- 1 -amine (B46); 2-(l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-l,2-dihydro-3H- imidazo[4,5-b]pyridin-3-yl)-N,N-diethylethan-l-amine (B47); l-(3-(2,4- dichlorophenoxy)propyl)-6-(4-fluorophenyl)- 1 H-benzo [d] imidazol-2-amine (B48); 1 -(3 -((3 - chloro-4'-fluoro-[l,r-biphenyl]-4-yl)oxy)propyl)-6-(4-fluorophenyl)-lH-benzo[d]imidazol-2- amine (B49); 3-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-l-(2-(pyrrolidin-l- yl)ethyl)- 1 ,3 -dihydro-2H-benzo [d] imidazol-2-imine (B50); 3 -(3 -((3 -chloro-4'-fluoro-[ 1,1'- biphenyl]-4-yl)oxy)propyl)-5-(4-fluorophenyl)-l-(2-(pyrrolidin-l-yl)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B51); methyl 2-amino-l-(2-(pyrrolidin-l-yl)ethyl)-lH- benzo[d]imidazole-6-carboxylate (B52); methyl 3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 1 -(2-(pyrrolidin- 1 -yl)ethyl)-2,3 -dihydro- 1 H-benzo [d] imidazole-5 -carboxylate (B53); methyl l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-3-(2-(pyrrolidin-l-yl)ethyl)-2,3-dihydro-lH- benzo[d]imidazole-5-carboxylate (B54); l-(3-((2,4-dichlorophenyl)thio)propyl)-lH- benzo[d]imidazol-2-amine (B76); 2-(2-amino-lH-benzo[d]imidazol-l-yl)-N-(2,4- dichlorophenyl)acetamide (B77); N-(2,4-dichlorophenyl)-2-(3-(2-(diethylamino)ethyl)-2- imino-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)acetamide (B78); (E)-N, 1 -dibutyl-3 -(3 -(2,4- dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B79); (E)-l-(3-(2,4- dichlorophenoxy)propyl)-N,3-bis(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B81); (E)-N'-(l-(3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-yl)-N,N- dimethylformimidamide (B80); l-(2-(dipropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B87); l-(2-(diisopropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B88); l-(2-(pyrrolidin- l-yl)ethyl)-lH-benzo[d]imidazol-2-amine (B89); 3-(3-(3-(2,4-dichlorophenoxy)propyl)-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylpropan-l-amine (B90); 2-(3-(3-(2- chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N-ethylethan-l- amine (B91); 2-(3-(3-(2,4-difluorophenoxy)propyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N,N-diethylethan-l -amine (B92); l-(3-(diethylamino)propyl)-6- phenyl-lH-benzo[d]imidazol-2-amine (B93); 5-chloro-l-(3-(diethylamino)propyl)-6-phenyl- lH-benzo[d]imidazol-2-amine (B94); 2-(3-(4-bromophenethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-N,N-diethylethan-l -amine (B95); l-(2-(diethylamino)ethyl)-6- phenyl-lH-benzo[d]imidazol-2-amine (B97); 6-chloro-l-(3-(2,4-dichlorophenoxy)propyl)- lH-benzo[d]imidazol-2-amine (B98); l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)- lH-benzo[d]imidazol-2-amine (B99); l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)- 3-((6-(trifluoromethyl)pyridin-3-yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-im^
(B100); 5-(4-fluorophenyl)-l-((6-(trifluoromethyl)pyridin-3-yl)methyl)-lH- benzo[d]imidazol-2-amine (B101); l-([l, l'-biphenyl]-4-yl)-2-(3-allyl-2-imino-2,3-dihydro- lH-benzo[d]imidazol-l-yl)ethan-l-ol (B102); l-(3-(2,4-dichlorophenoxy)propyl)-3-propyl- l,3-dihydro-2H-benzo[d]imidazol-2-imine (B103); 2-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d]imidazol- 1 -yl)- 1 -(p-tolyl)ethan- 1 -ol (B104); 2-(3-allyl-2-imino-2,3 -dihydro- 1 H- benzo[d]imidazol-l-yl)-l-(4-bromophenyl)ethan-l-ol (B105); 2-(3-allyl-2-imino-2,3- dihydro- lH-benzo[d]imidazol- 1 -yl)-l -(3-bromophenyl)ethan- 1 -ol (B106); 2-(3-allyl-2- imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)- 1 -(furan-2-yl)ethan- 1 -ol (B107); 2-(3 -allyl-2- imino-2,3 -dihydro- lH-benzo[d]imidazol-l-yl)-l-(4-methoxyphenyl)ethan-l-ol (B108); l-(4- chlorophenoxy)-3-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B109); l-(3-propyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4- chlorophenoxy)propan-2-ol (B110); l-(4-chlorophenoxy)-3-(2-imino-3-methyl-2,3-dihydro- lH-benzo[d]imidazol-l-yl)propan-2-ol (Bill); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy)propan-2-ol (B112); l-(3-butyl-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol (B113); l-(3-ethyl-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B114); l-(3-allyl-2- imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol (B115); l-(2- imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B116); l-(2- imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B117); l-(2- imino-3 -propyl-2,3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-3 -(2-methoxyphenoxy)propan-2-ol (B118); l-(2-chlorophenoxy)-3-(3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2- ol (B119); l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B120); 1 -(3 -butyl-2-imino-2,3 -dihydro- 1 H- benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B121); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B122); l-(4-chlorophenoxy)-3-(3-(2- (diethylamino)ethyl)-2-imino-2,3-dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol (B123); 1 - (2-imino-3 -methyl-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3-(p-tolyloxy)propan-2-ol
(B124); l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2- methoxyphenoxy)propan-2-ol (B125); l-(2-chlorophenoxy)-3-(2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B126); l-(3-allyl-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-3-(4-methoxyphenoxy)propan-2-ol (B127); l-(2-imino-3-methyl-2,3- dihydro-lH-benzo[d]imidazol-l-yl)-3-(m-tolyloxy)propan-2-ol (B128); l-(2-imino-3-methyl- 2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(o-tolyloxy)propan-2-ol (B129); l-(2,4- dichlorophenoxy)-3 -(2-((3 -morpholinopropyl)amino)- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol (B55); l-(2,4-dichlorophenoxy)-3-(2-(phenethylamino)-lH-benzo[d]imidazol-l-yl)propan-2- ol (B56); 1 -(2-(( 1 -benzylpiperidin-4-yl)amino)- 1 H-benzo[d]imidazol- 1 -yl)-3 -(2,4- dichlorophenoxy)propan-2-ol (B57); l-(2,4-dichlorophenoxy)-3-(2-((2- (diethylamino)ethyl)amino)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B60); l-(2,4- dichlorophenoxy)-3 -(2-((3 -(diethylamino)propyl)amino)- 1 H-benzo[d] imidazol- 1 -yl)propan- 2-ol (B61); 1 -( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo[d]imidazol-2-yl)-N3,N3- diethylpropane-1 ,3-diamine (B62); 1 -(2,4-dichlorophenoxy)-3-(2-(4-ethylpiperazin- 1 -yl)- 1H- benzo[d] imidazol- l-yl)propan-2-ol (B66); l-(2,4-dichlorophenoxy)-3-(2-((5- (diethylamino)pentan-2-yl)amino)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B73); l-(l-(3-
(2,4-dichlorophenoxy)-2-hydroxypropyl)-lH-benzo[d]imidazol-2-yl)piperidine-4-carbonitrile (B74); N-((lH-benzo[d]imidazol-2-yl)methyl)-3-(2,4-dichlorophenoxy)propan-l-amine (B58); l-(((lH-benzo[d]imidazol-2-yl)methyl)amino)-3-(2,4-dichlorophenoxy)propan-2-ol (B59); l-(2,4-dichlorophenoxy)-3-((l-(2-(diethylamino)ethyl)-lH-benzo[d]imidazol-2- yl)oxy)propan-2-ol (B64); N-pentyl-1 H-benzo [d]imidazol-2-amine (B83); N-phenethyl-lH- benzo [d] imidazol-2-amine (B84); 1 -(( 1 H-benzo[d] imidazol-2-yl)methyl)-N 1 -(3 -(2,4- dichlorophenoxy)propyl)-N2,N2-diethylethane- 1,2 -diamine (B85); and Nl-(3-(2,4- dichlorophenoxy)propyl)-N 1 -(( 1 -(2-(diethylamino)ethyl)- lH-benzo[d]imidazol-2-yl)methyl)- N2,N2-diethylethane-l,2-diamine (B86).
In certain embodiments, the pharmaceutical composition comprises at least one pharmaceutically acceptable carrier. In other embodiments, the pharmaceutical composition further comprises at least one additional antibacterial agent. In yet other embodiments, the at least one compound and the at least one additional antibacterial agent are synergistic. In yet other embodiments, the Fractional Inhibitory Concentration Index (FICI) determined by the combination of the at least one compound and the at least one additional antibacterial agent is equal to or lower than about 0.50. In yet other embodiments, the at least one additional antibacterial agent comprises a lipoprotein antibiotic. In yet other
embodiments, the at least one additional antibacterial agent comprises a polymyxin. In yet other embodiments, the at least one compound comprises at least one selected from the group consisting of 5, 25, and B55-B58.
In certain embodiments, the pharmaceutical composition comprises at least one additional antibacterial agent and at least one compound selected from the group consisting of l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl) propan-2-ol; 1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 - yl)-3-(2,4-dichlorophenoxy) propan-2-ol; l-(2,4-dichlorophenoxy)-3-(3-(2- (diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol-l-yl)propan-2-ol; l-(3- benzyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2- ol; l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin-l-yl)ethyl)-2,3-dihydro-lH-benzo[d] imidazol-l-yl)propan-2-ol.
In certain embodiments, the method comprises administering to the subject a therapeutically effective amount of at least one compound of claim 1, or a salt, solvate, tautomer or N-oxide thereof. In other embodiments, the method comprises administering to the subject a therapeutically effective amount of at least one compound, or a salt, solvate, tautomer or N-oxide thereof, selected from the group consisting of l-(2,4-dichlorophenoxy)- 3-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl) propan-2-ol; l-(3-allyl-2-imino- 2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol; l-(2,4- dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol- l-yl)propan-2-ol; l-(3-benzyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4- dichlorophenoxy) propan-2-ol; l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin-l- yl)ethyl)-2,3-dihydro-lH-benzo[d] imidazol-l-yl)propan-2-ol. In yet other embodiments, the infection by the bacterium in the subject is treated or prevented.
In certain embodiments, the bacterium is Gram-negative. In other embodiments, the bacterium is obligatory aerobic. In yet other embodiments, the bacterium belongs to the Acinetobacter genus. In yet other embodiments, the bacterium comprises A. baumannii. In yet other embodiments, the compound interferes with or inhibits the Oxidative Phosphorylation (OxPhos) pathway of the bacterium. In yet other embodiments, the compound selectively inhibits the bacterial OxPhos over mammalian mitochondrial OxPhos.
In certain embodiments, the compound is administered to the subject by a route selected from the group consisting of nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous. In other embodiments, the subject is further administered at least one additional antibacterial agent. In yet other embodiments, the at least one additional antibacterial agent is co-administered with the compound to the subject. In yet other embodiments, the at least one additional antibacterial agent and the compound are coformulated.
In certain embodiments, the subject is administered the at least one additional antibacterial agent at a lower dose or frequency as compared to the administering of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
In certain embodiments, the subject develops a lower or slower rate of resistance to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
In certain embodiments, the subject develops lower toxicity from the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
In certain embodiments, the subject is a mammal. In other embodiments, the mammal is human.
BRIEF DESCRIPTION OF THE DRAWINGS
For the purpose of illustrating the invention, there are depicted in the drawings certain embodiments of the invention. However, the invention is not limited to the precise arrangements and instrumentalities of the embodiments depicted in the drawings.
Fig. 1 illustrates the structure of compound 1.
Fig. 2A illustrates analogs of compound 1, and Fig. 2B illustrates their activity against .4. baumannii. Fig. 2B: 1 A. baumannii membrane ATP synthesis, n=2, standard deviations between the duplicates were on average 9% of the mean. 2 Sub-mitocondrial particle (SMP, bovine) ATP synthesis, n=2, standard deviations between the duplicates were on average 9% of the mean. 3 A. baumannii MICs were performed in duplicate and all reproduced to within one well. In cases where the MICs did not duplicate, the lower MIC value is reported.
Fig. 3A illustrates the effect of compound 5 on oxygen gas consumption. Fig.
3B illustrates bacterial oxidative phosphorylation (OxPhos). Fig. 3C illustrates the A.
baumannii (Ab) OxPhos pathway.
Fig. 4 illustrates synthetic schemes useful within the present invention.
Figs. 5A-5B are a set of graph and tables illustrating MIC distribution for compound 5 and comparator agents. Fig. 5B: 1 MICs were determined by serial dilution of 5 in Mueller-Hinton broth at a starting concentration of bacteria of 105— 106 CFU/mL. Values are reported after 18 h incubation at 37 °C. 2 Methicillin-resistant Staphylococcus aureus. 3 The medium contained 40% serum.
Figs. 6A-6C are a set of tables illustrating structure-activity relationship (SAR) studies for compounds of the invention. Figs. 6A-6B: 1 A. baumannii membrane ATP synthesis, n = 2, standard deviations between the duplicates were on average 9% of the mean. 2 Sub-mitochondrial particle (SMP, bovine) ATP synthesis, n = 2, standard deviations between the duplicates were on average 9% of the mean. 3 A. baumannii minimum inhibitory concentration, MICs were performed in duplicate and all reproduced to within one well. In cases where the MICs did not duplicate the lower MIC value is reported. 4 Cells are exposed to compounds for 1 h in the absence of serum. Compounds are removed and metabolically active cells are measured using MTS reduction, N = 3. Fig. 6C: 1 A. baumannii membrane ATP synthesis. 2 Sub-mitochondrial particle (SMP, bovine) ATP synthesis. 3 A. baumannii IC50/SMP IC50. 4 A. baumannii MIC.
Fig. 7 illustrates SAR studies performed in the present investigation.
Fig. 8 is a table illustrating bacterial strains used within the methods of the invention.
Fig. 9 is a table illustrating microsomal stability data (FC2837 = compound 5). Fig. 10 is an illustration of mice infected with bioluminescent AB5075 in a wound model of infection. Relative light units (RLU) are measured by the IVIS systems, where red indicates areas of the highest bacterial numbers, as opposed to blue & black with lesser numbers. In certain embodiments, blue color corresponds to about 1.0 x 105 CFU and red color corresponds to about 1.0 x 108 CFU present in the wound.
Figs. 1 1 A- 1 IB illustrate the checkerboard assay, FICI and isobologram. Fig.
11A: a schematic representation of a checkerboard assay. One compound is serially diluted from the right to then left in an MIC plate, and the other compound from top to bottom, to produce the checkerboard array of mixtures. The * indicates MIC concentrations of x alone and y alone. The gray squares indicate growth in the wells. The white and green squares indicate no growth, with the green squares indicating the wells used to calculate FICI. Fig. 1 IB illustrates an isobologram. In the isobologram the FIC values for the green squares are plotted. The line indicates a situation where the effect of the two drugs are additive. Points below are produced by synergy, and those above by antagonism.
Figs. 12A-12C illustrate isobolograms. Fig. 12A: compounds B41 (MIC alone = 0.8 μΜ) and 25 (MIC alone = 400 μΜ) and polymyxin B (MIC alone = 0.25 μg/mL) with .4. baumannii. Fig. 12B: compound B45 (MIC alone = 13 μΜ) and polymyxin B (MIC alone = 0.25 μg/mL ) with A baumannii. Fig. 12C: compound 25 (MIC alone = 400 μΜ) and polymyxin B (MIC alone = 0.125 μg/mL) with K. pneumonia.
Fig. 13 is a table illustrating a representative checkerboard plot demonstrating the synergy between B57 and colistin against A. baumannii.
Fig. 14A is a graph illustrating cytotoxic data for 25 in HepG2 cells with added amounts of polymyxin B. Fig. 14B is a graph illustrating the correlation between IC50 and MIC in the presence of polymyxin B.
Fig. 15 is a scheme illustrating synergistic interactions between OxPhos and polymyxin B/colistin via energy across the membranes.
Figs. 16A-16B are graphs illustrating the effects of drugs on oxygen consumption of live A. baumannii cells (about 107 cells/ml) determined using an oxygen electrode. Compounds 5 (75 μΜ), B55 (50 μΜ) and colistin (10 μg/ml) were added to a chamber.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to the unexpected discovery of novel antibacterial compounds. In certain embodiments, the compounds of the invention are active against Gram-negative bacteria, such as but not limited to A. baumannii. In other embodiments, the compounds of the invention are active against obligatory aerobic bacteria, such as but not limited to A. baumannii. In yet other embodiments, the compounds of the invention impair bacterial viability. In yet other embodiments, the compounds of the invention comprise 2-aminobenzimidazoles, 2-iminobenzimidazoles, analogs or derivatives thereof, and salts, N-oxides or solvates thereof. In yet other embodiments, the compounds of the invention are synergistic with lipoprotein antibiotics, such as polymyxins, such as colistin (polymyxyn E) and/or polymyxin B, or any other cell wall-modifying antibiotic.
In certain embodiments, the compounds of the invention are antibacterial because they interfere and/or inhibit the Oxidative Phosphorylation (OxPhos) pathway used by bacteria to produce energy. In other embodiments, the compounds of the invention selectively inhibit bacterial OxPhos over mammalian mitochondrial OxPhos.
As demonstrated herein, oxidative phosphorylation (OxPhos) was identified as a novel target for novel antibacterial agents. Accordingly, a library of about 10,000 compounds was screened using a membrane-based ATP synthesis assay. One of the compounds identified as active in this assay was the 2-iminobenzimidazole 1, which inhibited the OxPhos oiA.baumannii with a good selectivity against mitochondrial OxPhos and also and displayed a MIC of 17 μΜ (13 μg/mL) against the pathogen. The 2-iminobenzimidazole 1 was found to inhibit the type 1 NADH-quinone oxidoreductase (NDH-1) of A. baumannii OxPhos by a biochemical approach. Des-hydroxy analog 5 is also very active against bacteria, and showed low cytotoxicity against NIH3T3 and HepG2 mammalian cell lines. The compounds described herein are active anti-Acinetobacter agents.
Definitions
As used herein, each of the following terms has the meaning associated with it in this section.
Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Generally, the nomenclature used herein and the laboratory procedures in cell culture, microbiology, pharmacology and organic chemistry are those well- known and commonly employed in the art.
As used herein, the articles "a" and "an" refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element. Furthermore, use of the term "including" as well as other forms, such as "include", "includes," and "included," is not limiting.
As used herein, the term "Ab" or "Ab" refers to A. baumannii.
As used herein, the term "about" will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term "about" is meant to encompass variations of ±20% or ±10%, more preferably ±5%, even more preferably ±1%, and still more preferably ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
In one aspect, the terms "co-administered" and "co-administration" as relating to a subject refer to administering to the subject a compound of the invention or salt thereof along with a compound that may also treat diseases and/or disorders contemplated herein. In certain embodiments, the co-administered compounds are administered separately, or in any kind of combination as part of a single therapeutic approach. The co-administered compound may be formulated in any kind of combinations as mixtures of solids and liquids under a variety of solid, gel, and liquid formulations, and as a solution.
As used herein, the term "container" includes any receptacle for holding the pharmaceutical composition. For example, in certain embodiments, the container is the packaging that contains the pharmaceutical composition. In other embodiments, the container is not the packaging that contains the pharmaceutical composition, i.e., the container is a receptacle, such as a box or vial that contains the packaged pharmaceutical composition or unpackaged pharmaceutical composition and the instructions for use of the pharmaceutical composition. Moreover, packaging techniques are well known in the art. It should be understood that the instructions for use of the pharmaceutical composition may be contained on the packaging containing the pharmaceutical composition, and as such the instructions form an increased functional relationship to the packaged product. However, it should be understood that the instructions may contain information pertaining to the compound's ability to perform its intended function, e.g., treating, preventing, or reducing bacterial infection in a patient.
As used herein, a "disease" is a state of health of a subject wherein the subject cannot maintain homeostasis, and wherein if the disease is not ameliorated then the subject's health continues to deteriorate.
As used herein, a "disorder" in a subject is a state of health in which the subject is able to maintain homeostasis, but in which the subject's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the subject's state of health.
As used herein, the terms "effective amount," "pharmaceutically effective amount" and "therapeutically effective amount" refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
"Instructional material," as that phrase is used herein, includes a publication, a recording, a diagram, or any other medium of expression that may be used to communicate the usefulness of the compounds and/or methods of the invention. In some instances, the instructional material may be part of a kit useful for effecting alleviating or treating the various diseases or disorders recited herein. Optionally, or alternately, the instructional material may describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a mammal. The instructional material of the kit may, for example, be affixed to a container that contains the compounds of the invention or be shipped together with a container that contains the compounds. Alternatively, the instructional material may be shipped separately from the container with the intention that the recipient uses the instructional material and the compound cooperatively. For example, the instructional material is for use of a kit; instructions for use of the compound; or instructions for use of a formulation of the compound. As used herein, the term "patient," "individual" or "subject" refers to a human or a non-human mammal. Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and murine mammals. Preferably, the patient, subject or individual is human.
As used herein, the phrase "pharmaceutically acceptable" refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
As used herein, the phrase "pharmaceutically acceptable carrier" means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient. Some examples of materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil;
glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid;
pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
As used herein, the phrase "pharmaceutically acceptable carrier" also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions. The "pharmaceutically acceptable carrier" may further include a pharmaceutically acceptable salt of the compound useful within the invention. Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
As used herein, the phrase "pharmaceutically acceptable salt" refers to a salt of the administered compounds prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids, organic acids, solvates, hydrates, or clathrates thereof.
Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present invention, such as for example utility in process of synthesis, purification or formulation of compounds useful within the methods of the invention. Salts may be comprised of a fraction of one, one or more than one molar equivalent of acid or base with respect to any compound of the invention.
Suitable pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include sulfate, hydrogen sulfate, hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids (including hydrogen phosphate and dihydrogen phosphate). Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4- hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic,
ethanesulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2- hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, β-hydroxybutyric, salicylic, galactaric, galacturonic acid, glycerophosphonic acids and saccharin (e.g., saccharinate, saccharate).
Suitable pharmaceutically acceptable base addition salts of compounds of the invention include, for example, ammonium salts, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N'-dibenzylethylene- diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine
(N-methylglucamine) and procaine. All of these salts may be prepared from the corresponding compound by reacting, for example, the appropriate acid or base with the compound.
As used herein, the phrase "pharmaceutical composition" refers to a mixture of at least one compound useful in the methods of the invention with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient. Multiple techniques of administering a compound exist in the art including, but not limited to, nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous route administration.
As used herein, the term "prevent" or "prevention" means no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease.
As used herein, a compound of the invention is 'synergistic" with one additional antibiotic if the Fractional Inhibitory Concentration Index (FICI) determined by the combination of the compound of the invention and the one additional antibiotic is equal to or lower than about 0.50.
As used herein, the term "treatment" or "treating" is defined as the application or administration of a therapeutic agent, i.e., a compound of the invention (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has a condition contemplated herein, a symptom of a condition contemplated herein or the potential to develop a condition contemplated herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect a condition contemplated herein, the symptoms of a condition contemplated herein or the potential to develop a condition contemplated herein. Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics. In certain embodiments, the condition is bacterium infection.
As used herein, the term "alkyl" by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., C\-Ce means one to six carbon atoms) and includes straight, branched chain, or cyclic substituent groups. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, and cyclopropylmethyl.
Specific examples are C\-Ce alkyl, particularly ethyl, methyl, isopropyl, isobutyl, w-pentyl, n- hexyl and cyclopropylmethyl. As used herein, the term "substituted alkyl" means alkyl as defined above, substituted by one, two or three substituents selected from the group consisting of halogen, - OH, alkoxy, -NH2, -N(CH3)2, -C(=0)OH, trifluoromethyl, -ON, -C(=0)0(Ci-C4)alkyl, - C(=0)NH2, -SO2NH2, -C(=NH)NH2, and -N02; in some embodiments containing one or two substituents selected from the group consisting of halogen, -OH, alkoxy, -NH2,
trifluoromethyl, -N(CH3)2, and -C(=0)OH; in other embodiments selected from the group consisting of halogen, alkoxy and -OH. Examples of substituted alkyls include, but are not limited to, 2,2-difluoropropyl, 2-carboxycyclopentyl and 3-chloropropyl.
As used herein, the term "heteroalkyl" by itself or in combination with another term means, unless otherwise stated, a stable straight or branched chain alkyl group consisting of the stated number of carbon atoms and one or two heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may be optionally oxidized and the nitrogen heteroatom may be optionally quaternized. The heteroatom(s) may be placed at any position of the heteroalkyl group, including between the rest of the heteroalkyl group and the fragment to which it is attached, as well as attached to the most distal carbon atom in the heteroalkyl group. Examples include: -0-CH2-CH2-CH3,
-CH2-CH2-CH2-OH, -CH2-CH2-NH-CH3, -CH2-S-CH2-CH3, and -CH2CH2-S(=0)-CH3. Up to two heteroatoms may be consecutive, such as, for example, -CH2-NH-OCH3, or
-CH2-CH2-S-S-CH3
As used herein, the term "alkoxy" employed alone or in combination with other terms means, unless otherwise stated, an alkyl group having the designated number of carbon atoms, as defined above, connected to the rest of the molecule via an oxygen atom, such as, for example, methoxy, ethoxy, 1-propoxy, 2-propoxy (isopropoxy) and the higher homologs and isomers. Examples are (Ci-C3) alkoxy, particularly ethoxy and methoxy.
As used herein, the term "halo" or "halogen" alone or as part of another substituent means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom, preferably, fluorine, chlorine, or bromine, more preferably, fluorine or chlorine.
As used herein, the term "cycloalkyl" refers to a mono cyclic or polycyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. In certain embodiments, the cycloalkyl group is saturated or partially unsaturated. In other embodiments, the cycloalkyl group is fused with an aromatic ring. Cycloalkyl groups include groups having from 3 to 10 ring atoms. Illustrative examples of cycloalkyl groups include, but are not limited to, the following moieties:
Figure imgf000029_0001
Monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Dicyclic cycloalkyls include, but are not limited to, tetrahydronaphthyl, indanyl, and tetrahydropentalene. Polycyclic cycloalkyls include adamantine and norbornane. The term cycloalkyl includes "unsaturated nonaromatic carbocyclyl" or "nonaromatic unsaturated carbocyclyl" groups, both of which refer to a nonaromatic carbocycle as defined herein, which contains at least one carbon carbon double bond or one carbon carbon triple bond.
As used herein, the term "heterocycloalkyl" or "heterocyclyl" refers to a heteroalicyclic group containing one to four ring heteroatoms each selected from O, Sand N. In certain embodiments, each heterocycloalkyl group has from 4 to 10 atoms in its ring system, with the proviso that the ring of said group does not contain two adjacent O or S atoms. In other embodiments, the heterocycloalkyl group is fused with an aromatic ring. In certain embodiments, the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen atom may be optionally quaternized. The heterocyclic system may be attached, unless otherwise stated, at any heteroatom or carbon atom that affords a stable structure. A heterocycle may be aromatic or non-aromatic in nature. In certain embodiments, the heterocycle is a heteroaryl.
An example of a 3 -membered heterocycloalkyl group includes, and is not limited to, aziridine. Examples of 4-membered heterocycloalkyl groups include, and are not limited to, azetidine and a beta lactam. Examples of 5-membered heterocycloalkyl groups include, and are not limited to, pyrrolidine, oxazolidine and thiazolidinedione. Examples of 6- membered heterocycloalkyl groups include, and are not limited to, piperidine, morpholine and piperazine. Other non-limiting examples of heterocycloalkyl groups are:
Figure imgf000030_0001
Examples of non-aromatic heterocycles include monocyclic groups such as aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, pyrroline, pyrazolidine, imidazoline, dioxolane, sulfolane, 2,3-dihydrofuran, 2,5-dihydrofuran, tetrahydrofuran, thiophane, piperidine, 1,2,3,6-tetrahydropyridine, 1,4-dihydropyridine, piperazine, morpholine, thiomorpholine, pyran, 2,3-dihydropyran, tetrahydropyran, 1,4-dioxane,
1,3-dioxane, homopiperazine, homopiperidine, 1,3-dioxepane, 4,7-dihydro-l,3-dioxepin, and hexamethyleneoxide.
As used herein, the term "aromatic" refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character, i.e. having (4n + 2) delocalized π (pi) electrons, where n is an integer.
As used herein, the term "aryl," employed alone or in combination with other terms, means, unless otherwise stated, a carbocyclic aromatic system containing one or more rings (typically one, two or three rings), wherein such rings may be attached together in a pendent manner, such as a biphenyl, or may be fused, such as naphthalene. Examples of aryl groups include phenyl, anthracyl, and naphthyl. Examples are phenyl and naphthyl.
As used herein, the term "aryl-(Ci-C3)alkyl" means a functional group wherein a one- to three-carbon alkylene chain is attached to an aryl group, e.g., -CH2CH2-phenyl. Examples are aryl-CH2- and aryl-CH(C]¾)-. The term "substituted aryl-(Ci-C3) alkyl" means an aryl-(Ci-C3) alkyl functional group in which the aryl group is substituted. An example is substituted aryl(CH2)-. Similarly, the term "heteroaryl-(Ci-C3) alkyl" means a functional group wherein a one to three carbon alkylene chain is attached to a heteroaryl group, e.g., -CH2CH2-pyridyl. An example is heteroaryl-(CH2)-. The term "substituted heteroaryl-(Ci-C3) alkyl" means a heteroaryl-(Ci-C3) alkyl functional group in which the heteroaryl group is substituted. An example is substituted heteroaryl-(CH2)-.
As used herein, the term "heteroaryl" or "heteroaromatic" refers to a heterocycle having aromatic character. A polycyclic heteroaryl may include one or more rings that are partially saturated. Examples include the following moieties:
Figure imgf000031_0001
Examples of heteroaryl groups also include pyridyl, pyrazinyl, pyrimidinyl
(particularly 2- and 4-pyrimidinyl), pyridazinyl, thienyl, furyl, pyrrolyl (particularly
2-pyrrolyl), imidazolyl, thiazolyl, oxazolyl, pyrazolyl (particularly 3- and 5-pyrazolyl), isothiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-thiadiazolyl and 1,3,4-oxadiazolyl.
Examples of polycyclic heterocycles and heteroaryls include indolyl
(particularly 3-, 4-, 5-, 6- and 7-indolyl), indolinyl, quinolyl, tetrahydroquinolyl, isoquinolyl (particularly 1- and 5-isoquinolyl), 1,2,3,4-tetrahydroisoquinolyl, cinnolinyl, quinoxalinyl (particularly 2- and 5-quinoxalinyl), quinazolinyl, phthalazinyl, 1,8-naphthyridinyl,
1,4-benzodioxanyl, coumarin, dihydrocoumarin, 1,5-naphthyridinyl, benzofuryl (particularly 3-, 4-, 5-, 6- and 7-benzofuryl), 2,3-dihydrobenzofuryl, 1 ,2-benzisoxazolyl, benzothienyl (particularly 3-, 4-, 5-, 6-, and 7-benzothienyl), benzoxazolyl, benzothiazolyl (particularly 2-benzothiazolyl and 5-benzothiazolyl), purinyl, benzimidazolyl (particularly
2-benzimidazolyl), benzotriazolyl, thioxanthinyl, carbazolyl, carbolinyl, acridinyl, pyrrolizidinyl, and quinolizidinyl.
As used herein, the term "substituted" means that an atom or group of atoms has replaced hydrogen as the substituent attached to another group. The term "substituted" further refers to any level of substitution, namely mono-, di-, tri-, tetra-, or penta-substitution, where such substitution is permitted. The substituents are independently selected, and substitution may be at any chemically accessible position. In certain embodiments, the substituents vary in number between one and four. In other embodiments, the substituents vary in number between one and three. In yet other embodiments, the substituents vary in number between one and two.
As used herein, the term "optionally substituted" means that the referenced group may be substituted or unsubstituted. In certain embodiments, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In other embodiments, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein.
In certain embodiments, the substituents are independently selected from the group consisting of oxo, halogen, -CN, -NH2, -OH, -NH(CH3), -N(CH3)2, alkyl (including straight chain, branched and/or unsaturated alkyl), substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, fluoro alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted alkoxy, fluoroalkoxy, -S-alkyl, S(=0)2alkyl, - C(=0)NH[substituted or unsubstituted alkyl, or substituted or unsubstituted phenyl], - C(=0)N[H or alkyl]2, -OC(=0)N[substituted or unsubstituted alkyl]2, - NHC(=0)NH [substituted or unsubstituted alkyl, or substituted or unsubstituted phenyl], - NHC(=0)alkyl, - [substituted or unsubstituted alkyl]C(=0)[substituted or unsubstituted alkyl], -NHC(=0) [substituted or unsubstituted alkyl], -C(OH) [substituted or unsubstituted alkyl]2, and -C(NH2)[substituted or unsubstituted alkyl]2. In other embodiments, by way of example, an optional substituent is selected from oxo, fluorine, chlorine, bromine, iodine, - CN, -NH2, -OH, -NH(CH3), -N(CH3)2, -CH3, -CH2CH3, -CH(CH3)2, -CF3, -CH2CF3, -OCH3, - OCH2CH3, -OCH(CH3)2, -OCF3, -OCH2CF3, -S(=0)2-CH3, -C(=0)NH2, -C(=0)-NHCH3, - NHC(=0)NHCH3, -C(=0)CH3, and -C(=0)OH. In yet one embodiment, the substituents are independently selected from the group consisting of Ci_6 alkyl, -OH, Ci_6 alkoxy, halo, amino, acetamido, oxo and nitro. In yet other embodiments, the substituents are independently selected from the group consisting of Ci_6 alkyl, Ci_6 alkoxy, halo, acetamido, and nitro. As used herein, where a substituent is an alkyl or alkoxy group, the carbon chain may be branched, straight or cyclic, with straight being a specific example.
Throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.1, 5.3, 5.5, and 6. This applies regardless of the breadth of the range. Description
The present invention relates to the unexpected discovery of novel antibacterial compounds. In certain embodiments, the compounds of the invention are active against Gram-negative bacteria, such as but not limited to A. baumannii. In other embodiments, the compounds of the invention are active against obligatory aerobic bacteria, such as but not limited to A. baumannii. In yet other embodiments, the compounds of the invention impair viability of bacteria. In yet other embodiments, the compounds of the invention comprise 2-aminobenzimidazoles, 2-iminobenzimidazoles, analogs or derivatives thereof, and salts or solvates thereof. In yet other embodiments, the compounds of the invention are synergistic with lipoprotein antibiotics, such as polymyxins, such as colistin (polymyxyn E) and/or polymyxin B, or any other cell wall-modifying antibiotic.
In certain embodiments, the compounds of the invention are antibacterial because they interfere and/or inhibit the Oxidative Phosphorylation (OxPhos) pathway used by bacteria to produce energy.
As described herein, a novel mechanistic approach to target Gram-negative bacteria, such as A. baumannii, by inhibition of bacterial OxPhos, specifically NDH-1, was demonstrated. Selectively targeting OxPhos in bacteria, without effect upon host OxPhos, represents a new approach to attack the growing problem of resistant Gram-negative bacteria, such as A. baumannii. In certain embodiments, this novel mechanism of action affords standalone agents or agents that are useful in combination with other agents to minimize potential drug resistance over time.
As described herein, a validated and robust assay for identifying inhibitors of the OxPhos pathway was developed. In certain embodiments, biochemical and genetic characterization of the interactions of test compounds with target membranes allow for the characterization of the role of NDH-1 in the antibacterial activity and identification of additional potential OxPhos targets. As an example of the potency of the novel compounds identified herein, compounds 4 and 5 were found to be active against a battery of more than 20 multi-drug resistant baumannii clinical isolates. Compounds
The compounds of the invention may be synthesized using techniques well- known in the art of organic synthesis. The starting materials and intermediates required for the synthesis may be obtained from commercial sources or synthesized according to methods known to those skilled in the art. In certain embodiments, the compounds of the invention are prepared according to the methodology outlined in Fig. 4.
In one aspect, the compound of the invention is a compound of formula (I), or a salt, solvate or N-oxide thereof:
Figure imgf000034_0001
(I), wherein in (I):
X1 and X2 are independently selected from the group consisting of H and optionally substituted Ci-C6 alkyl, X1 and X2 combine to form =NH, =N(Ci-C6 alkyl), =CH-CN, =N- CN, or =CH-N02, or X1 is absent and X2 is -N=CH-NMe2;
ring A is a fused aryl or heteroaryl ring selected from the group consisting of:
Figure imgf000034_0002
each occurrence of R1 is independently a bond or -CH2-;
R2 is selected from the group consisting of -(CH2)i-3-0-(CH2)o-2-,
Figure imgf000034_0003
and -(CH2)!_6, wherein each of the CH2 groups in R2 is independently optionally substituted with a substituent selected from the group consisting of C1-C6 alkyl, C1-C6 fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7,
OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R3 is -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), pyrrolidin-l-yl, pyperidin-l-yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted C1-C6 alkyl, C1-C6 fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2,
C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R4 is -(CH2)o-4;
R5 is selected from the group consisting of H, prop-2-en- l-yl, Ci-Ce alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2, -(C=O)0-i-(OCH2CH2)1_
Figure imgf000035_0001
; wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1 ;
eeaacchh ooccccurrence of R6 is independently CH, CR10 or N, wherein 0-2 occurrences of R6 in ring A are N;
each occurrence of R7 is independently H, C1-C6 alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted; each occurrence of R8 is independently C1-C6 alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted;
each occurrence of R9 is independently O, S or R7; and,
each occurrence of R10 is independently selected from the group consisting of optionally substituted C1-C6 alkyl, C1-C6 fluoroalkyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7.
In certain embodiments, the compound of formula (I), or a salt, solvate or N-
oxide thereof, is a compound of formula (la):
Figure imgf000035_0002
(la).
In one aspect, the compound of the invention is a compound of formula (II), or a salt, solvate or N-oxide thereof:
Figure imgf000035_0003
(II), wherein in (II): ring A is a fused aryl or heteroaryl ring selected from the group consisting of:
Figure imgf000036_0001
each occurrence of R1 is independently a bond or -CH2-;
R2 is selected from the group consisting of -(CH2)1 -3-0-(CH2)o-2-,
Figure imgf000036_0002
-(CH2)1_3-NR7-(CH2)o-2-, -(CH2)1.3-S(=0)o-2-(CH2)o-2-, and -(CH2)!_6, wherein each of the CH2 groups in R2 is independently optionally substituted with a substituent selected from the group consisting of Ci-Ce alkyl, Ci-Ce fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R3 is-NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), pyrrolidin- 1 -yl, pyperidin- l-yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2,
C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R4 is -(CH2)o-4;
R5 is selected from the group consisting of H, prop-2-en- 1 -yl, Ci-Ce alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2,
-(C=0)o-i-(OCH2CH2)i_3-OR7,
Figure imgf000036_0003
; wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1 ;
or R4 is a bond and R5 combines with the N atom to which it is bound to form morpholyn- 1 -yl, piperidin- 1-yl, 4-cyano-piperidin-l -yl and piperazin- 1 -yl, wherein N4 of the piperazinyl is optionally substituted with Ci-Ce alkyl;
each occurrence of R6 is independently CH, CR10 or N, wherein 0-2 occurrences of R6 in ring A are N;
each occurrence of R7 is independently H, Ci-Ce alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted; each occurrence of R8 is independently Ci-Ce alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted;
each occurrence of R9 is independently O, S or R7; and,
each occurrence of R10 is independently selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7.
In certain embodiments, the (II), or a salt, solvate or N-
oxide thereof, is a compound of formula (Il
Figure imgf000037_0001
(Ha)-
In one aspect, the compound of the invention is a compound of formula (III), or a salt, solvate or N-oxide thereof:
Figure imgf000037_0002
(III), wherein in (III):
ring A is a fused aryl or heteroaryl ring selected from the group consisting of:
Figure imgf000037_0003
each occurrence of R1 is independently a bond or -CH2-;
R2 is selected from the group consisting of -(CH2)1 -3-0-(CH2)o-2-,
Figure imgf000037_0004
and -(CH2)!_6, wherein each of the CH2 groups in R2 is independently optionally substituted with a substituent selected from the group consisting of C\-Ce alkyl, C\-Ce fluoroalkyl, F, CI, Br, I,
CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7,
OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R3 is H, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), pyrrolidin-l-yl, pyperidin-l-yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted C1-C6 alkyl, C1-C6 fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2,
C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R4 is -(CH2)o-4;
R5 is selected from the group consisting of H, prop-2-en-l-yl, C1-C6 alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2, -(C=O)0-i-(OCH2CH2)1_
Figure imgf000038_0001
3' ; wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1;
eeaacchh ooccccurrence of R6 is independently CH, CR10 or N, wherein 0-2 occurrences of R6 in ring A are N;
each occurrence of R7 is independently H, C1-C6 alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted; each occurrence of R8 is independently C1-C6 alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted;
each occurrence of R9 is independently O, S or R7; and,
each occurrence of R10 is independently selected from the group consisting of optionally substituted C1-C6 alkyl, C1-C6 fluoroalkyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7.
In certain embodiments, the compound of formula (III), or a salt, solvate or N-
Figure imgf000038_0002
oxide thereof, is a compound of formula (Ilia): (Ilia).
In certain embodiments, the compound is at least one compound, or a salt, solvat r N-oxide thereof, selected from the group consisting of:
Figure imgf000039_0001
as defined elsewhere herein.
In certain embodiments, when X1 and X2 combine to form =NH and ring A is a six-membered ring, then at least one occurrence of R6 is CR10 or .
In certain embodiments, (I) is not a compound of formula (lb):
Figure imgf000039_0002
(lb), wherein in (lb):
RA is Ci-C6 alkyl, C2-C7 alkenyl, -CH2CH2NEt2 or -CH2CH2-(N1-morpholinyl), or benzyl optionaly substituted with at least one halogen or C1-C3 alkyl;
R is phenyl optionally substituted with at least one halogen; aryl; aroxy; naphthyl; phenoxymethyl optionally substituted with at least one halogen, C1-C3 alkyl and/or C1-C3 alkoxy; phenyl isoxazole optionally substituted with at least one halogen; or 4-methyl-3- phenyl- 1 ,2,4-oxadiazole-5 (4H)-one.
In certain embodiments, (I) is not l-(2,4-dichlorophenoxy)-3-(2-imino-3- propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl) propan-2-ol (Al); l-(3-allyl-2-imino-2,3- dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A2); l-(2,4- dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol- l-yl)propan-2-ol (A3, also known as 1); l-(3-benzyl-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A4); l-(2,4-dichlorophenoxy)- 3-(2-imino-3-(2-(piperidin-l-yl)ethyl)-2,3-dihydro-lH-benzo[d] imidazol- l-yl)propan-2-ol (A5).
In certain embodiments, ring A is
Figure imgf000039_0003
, wherein R6 is CH or CR10. In other embodiments, at least one occurr of R6 in ring A is CR10. In yet other
embodiments, ring A is R
Figure imgf000040_0001
1° or . In yet other embodiments, R10 is F,
Br or I. In yet other embodiments, R10 is C(=0)OR7.
In certain embodiments, ring A is selected from the group consisting of:
Figure imgf000040_0002
wherein Hal is halogen.
In certain embodiments, R10 is optionally substituted phenyl or optionally substituted pyridyl. In yet other embodiments, R10 is 4-fluorophenyl, p-tolyl or phenyl. In yet other embodiments, R9 is O or NR7. In yet other embodiments, R7 is methyl or 4- fluorophenyl.
In certain embodiments, one occurrence of R1 is a bond, and the other occurrence of R1 is -CH2-. In other embodiments, both occurrences of R1 are a bond.
In certain embodiments, R2 is selected from the group consisting of
-(CH2)i-3-0-, -(CH2)i-3-NR7-, -(CH2)i-3-S-, and -(CH2)1-6-. In other embodiments, at least one of the CH2 groups in R2 is independently substituted with one substituent selected from the group consisting of Ci-C6 alkyl, Ci-C6 fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2,
C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NH(R7), NHC(=0)R7, and NHC(=0)OR7. In other embodiments, at least one of the CH2 groups in R2 is independently substituted with OR7. In yet other embodiments, R2 is -CH2CH2-, -CH2CH2CH2-, -CH2CH20-, -CH2CH2CH20-, or - CH2CH(OH)CH20-.
In certain embodiments, R3 or R3 is phenyl, pyridyl, pyrimidyl, 1,2-biphi
1,3-biphenyl, 1 ,4-biphenyl, or naphthyl, wherein the aryl or heteroaryl group is optionally substituted with at least one selected from the group consisting of Ci-Ce alkyl, Ci-Ce fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2,
NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7.
In certain embodiments, R3 or R3 is phenyl, 2-methylphenyl, 3-methylphi 4-methylphenyl, 2-chlorophenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 3- chlorophenyl, 4-chlorophenyl, 2-bromophenyl, 3-bromophenyl, 4-bromophenyl, 2- trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2,4-difluorophenyl, 2,4-dichlorophenyl, 1,2-biphenyl, 1,3-biphenyl, 1 ,4-biphenyl, 4-trifluoromethylmethyl-l,4- biphenyl, 4'-fluoro-2-chloro-l,4-biphenyl, 2-trifluoromethyl-pyridin-5-yl, and 2-furyl.
In certain embodiments, R5 or R5 is selected from the group consisting of H,
Ci-Ce alkyl, aryl, N(R7)2, pyrrolidin-l-yl, piperidin-l-yl, N1-H-piperidin-4-yl, N^CQ-Ce alkyl)-piperidin-4-yl, N1-(tBoc)-piperidin-4-yl, morpholin-l-yl, imidazol- 1 -yl, imidazol-2-yl, pyrrolidin-2-one-l-yl, pyridyl, and -(C=0)o-i-( 4-R7-piperazin-l-yl). In other embodiments, R5 or R5 is N(R7)2, wherein each occurrence of R7 is independently H, methyl, ethyl, n- propyl or isopropyl. In yet other embodiments, R5 or R5 is selected from the group consisting of pyrrolidin-l-yl, piperidin-l-yl, N1-H-piperidin-4-yl, N1-(Ci-C6 alkyl)-piperidin- 4-yl, N1-(tBoc)-piperidin-4-yl, morpholin-l-yl, imidazol- 1 -yl, imidazol-2-yl, pyrrolidin-2- one-l-yl, pyridyl, 3,5-dimethylisoxazol-4-yl, 6-trifluoromethyl-pyridin-3-yl, -(C=0)o-i- (OCH2CH2)1_3-0(Ci-C6 alkyl), and -(C=O)0-i-( 4-phenyl-piperazin-l-yl).
In certain embodiments, each occurrence of R6 is CR7. In other embodiments, each occurrence of R6 is CH.
In certain embodiments, the compound of formula (I), or a salt, solvate or N- oxide thereof, is selected from the group consisting of:
l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)
Figure imgf000041_0001
allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-
Figure imgf000042_0001
l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d
imidazol- 1 -yl)propan-2-ol (A3, also known as 1)
Figure imgf000042_0002
1 -(3 -benzyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophei
propan-2-ol (A4)
Figure imgf000042_0003
; and
l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin-l-yl)ethyl)-2,3-dihydro-lH-benzo[d]
imidazol- 1 -yl)propan-2-ol (A5)
Figure imgf000042_0004
In certain embodiments, the compound of formula (I), or a salt, solvate or N- oxide thereof, is selected from the group consisting of:
l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)
propan-2-ol (Al)
Figure imgf000042_0005
1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-
Figure imgf000042_0006
l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d]
imidazol- 1 -yl)propan-2-ol (A3, also known as 1)
Figure imgf000042_0007
1 -(3 -benzyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-2-ol (A4)
Figure imgf000043_0001
l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-( iperidin-l-yl)ethyl)-2,3-dihydro-lH-benzo[d
imidazol-l-yl)propan-2-ol (A5)
Figure imgf000043_0002
l-(3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-amine (4)
Figure imgf000043_0003
2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
d
Figure imgf000043_0004
2-(3-(2-(2,4-dichlorophenoxy)ethyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
diethylethan- 1 -am
Figure imgf000043_0005
2-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
diethylethan- 1 -amine (7)
Figure imgf000043_0006
l-(3-(2,4-dichlorophenoxy)propyl)-3-ethyl-l,3-dihydro-2H-benzo[d]imidazol-2-imine (13)
Figure imgf000043_0007
l-butyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (14)
Figure imgf000044_0001
l-(3-(2,4-dichlorophenoxy)propyl)-3-(3-phenylpropyl)-l,3-dihydro-2H-benzo[d]imidazol-2-
imine (15)
Figure imgf000044_0002
;
N-(2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)
ethyl)-N-propylpropan- 1 -amine (16)
Figure imgf000044_0003
N-(2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -
yl)ethyl)-N-isopropylpropan-2-amine (17)
Figure imgf000044_0004
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -(2-(pyrrolidin- 1 -yl)ethyl)- 1 ,3 -dihydro-2H-
benzo[d]imidazol-2-imine (18)
Figure imgf000044_0005
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -(2-(piperidin- 1 -yl)propyl)- 1 ,3 -dihydro-2H-
benzo[d]imidazol-2-imine (19)
Figure imgf000044_0006
;
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-morpholinoethyl)-l,3-dihydro-2H-benzo[d]imidazol-
2-imine (20)
Figure imgf000044_0007
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyridin-2-yl)ethyl)-l,3-dihydro-2H-benzo[d]
imidazol-2-imine (21)
Figure imgf000045_0001
tert-butyl 4-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]
imidazol- 1 -yl)ethyl)piperidine- 1 -carboxylate (22)
Figure imgf000045_0002
1 -(2-( 1 H-imidazol- 1 -yl)ethyl)-3 -(3 -(2,4-dichlorophenoxy)propyl)- 1 ,3 -dihydro-2H-
benzo[d]imidazol-2-imine (23)
Figure imgf000045_0003
l-(2-(3-(3-(2,4-dichlorophenoxy)prop -2-imino-2,3-dihydro-lH-benzo[d]imidazol-l
yl)ethyl)pyrrolidin-2-one (24)
Figure imgf000045_0004
;
2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-l-(4-
phenylpiperazin-l-yl)ethan-l-one (25)
Figure imgf000045_0005
;
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -((6-(trifluoromethyl)pyridin-3 -yl)methyl)- 1 ,3 -dihydro-
2H-benzo[d]imidazol-2-imine (26)
Figure imgf000045_0006
;
l-(3-(2,4-dichlorophenoxy)propyl)-3-((3,5-dimethylisoxazol-4-yl)methyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (27)
Figure imgf000046_0001
2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
diethylacetamide (28)
Figure imgf000046_0002
;
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(2-ethoxyethoxy)ethyl)-l,3-dihydro-2H-
benzo[d]imidazol-2-imine (29)
Figure imgf000046_0003
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
diethylpropan- 1 -amine (33)
Figure imgf000046_0004
3 -(5 -chloro-3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol-
l-yl)-N,N-diethylpropan-l -amine (34)
Figure imgf000046_0005
l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-
benzo[d] imidazol- l-yl)propan-2-ol (B4)
Figure imgf000046_0006
(R)-l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-
benzo [d] imidazol- 1 -yl)propan-2-ol (B5)
Figure imgf000046_0007
;
((S)-l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B6)
Figure imgf000047_0001
l-(2-(2,4-dichlorophenoxy)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B13)
Figure imgf000047_0002
.
l-(3-(2 -dichlorophenyl)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B14)
Figure imgf000047_0003
-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
diethylethan- 1 -amine (B15)
Figure imgf000047_0004
1 -(cyclopropylmethyl)-3 -(3 -(2,4-dichlorophenoxy)propyl)- 1 ,3 -dihydro-2H-
benzo [d]imidazol-2-imine (B17)
Figure imgf000047_0005
l-(3-(2,4-dichlorophenoxy)propyl)-3-(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2-
imine (B18)
Figure imgf000047_0006
3-(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N,N-diethylpropan- 1 -amine (B23)
Figure imgf000048_0001
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
dipropylpropan-1 -amine (B25)
Figure imgf000048_0002
N-benzyl-2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N-ethylethan-l -amine (B26)
Figure imgf000048_0003
2-(3-(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2, 3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N-
ethylethan-1 -amine (B27)
Figure imgf000048_0004
N-benzyl-2-(3-(3-(2-chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-
yl)-N-ethylethan-l -amine
Figure imgf000048_0005
N,N-diethyl-2-(2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH-
benzo [d] imidazol- l-yl)ethan-l -amine (B29)
Figure imgf000048_0006
;
3 -(2-imino-3 -(3 -(2-(trifluoromethyl)phenoxy)propyl)-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -
yl)-N,N-dipropylpropan- 1 -amine (B30)
Figure imgf000048_0007
N,N-diethyl- -(2-imino-5,6-diphenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-l-ami
Figure imgf000049_0001
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- -phenyl-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N,N-diethylpropan- 1 -amine (B32)
Figure imgf000049_0002
3-(5-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH- lpropan-l -amine (B33)
Figure imgf000049_0003
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-5,6-diphenyl-2,3-dihydro-lH- lpropan- 1 -amine (B34)
Figure imgf000049_0004
2-(3 -(2-([ 1 , 1 '-biphenyl] -4-yl)ethyl)- -imino-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)-N,N-
diethylethan- 1 -amine (B35)
Figure imgf000049_0005
2-(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N,N-diethylethan-l -amine (B36)
Figure imgf000050_0001
-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- -phenyl-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N,N-diethylethan- 1 -amine (B37)
Figure imgf000050_0002
-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3-dihydro-lH- lethan- 1 -amine (B38)
Figure imgf000050_0003
-(3 -(3 -(2,4-dichlorophenoxy)propyl)-6-(4-trifluoromethylphenyl)-2-imino-2,3 -dihydro- 1 H- benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B39)
Figure imgf000050_0004
1 -(3 -(2,4-dichlorophenoxy)propyl)-5-(4-fluoropheny 1,3-
dihydro-2H-benzo[d]imidazol-2-imine (B40)
Figure imgf000050_0005
l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(pyrrolidin-l-yl)ethyl)-l,3- dihydro-2H-benzo[d]imidazol-2-imine (B41)
Figure imgf000051_0001
6-(4-fluorophenyl)- l-(2-(pyrrolidin- 1 -yl)ethyl)- lH-benzo[d]imidazol-2-amine (B42)
Figure imgf000051_0002
N,N-diethyl-2-(2 mino-3-(2-(4'-(trifluoromethyl)-[l,l'-biphenyl]-4-yl)ethyl)-2,3-dihydro- -benzo[d]imidazol- 1 -yl)ethan- 1 -amine (B43)
Figure imgf000051_0003
N-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3
benzo[d]imidazol-l-yl)ethyl)-N-isopropylpropan-2-amine (B44)
Figure imgf000051_0004
N-(2-(6-(4-fluorophenyl)-2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo[d]imidazol-l-yl)ethyl)-N-isopropylpropan-2-amine (B45)
Figure imgf000051_0005
N,N-diethyl-2-(2-imino-3-(3-((4'-(trifluoromethyl)-[l, l'-biphenyl]-4-yl)oxy)propyl)-2,3- dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B46)
Figure imgf000052_0001
2-(l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-l,2-dihydro-3H-imidazo[4,5-b]pyridin-3-yl)-
N,N-diethylethan-l -amine (B47)
Figure imgf000052_0002
l-(3-(2 -dichlorophenoxy)propyl)-6-(4-fluorophenyl)-lH-benzo[d]imidazol-2-amine (B48)
Figure imgf000052_0003
1 -(3 -((3 -chloro-4'-fluoro-[ 1 , 1 '-biphenyl]-4-yl)oxy)propyl)-6-(4-fluorophenyl)- 1H-
benzo[d]imidazol-2-amine (B49)
Figure imgf000052_0004
3 -(3 -(2,4-dichlorophenoxy)propyl)-5-(4-fluoropheny - 1 -(2-(pyrrolidin- 1 -yl)ethyl)- 1,3-
dihydro-2H-benzo[d]imidazol-2-imine (B50)
Figure imgf000052_0005
3-(3-((3-chloro-4'-fluoro-[l, -biphenyl]-4-yl)oxy)propyl)-5-(4-fluorophenyl)-l-(2- (pyrrolidin-l-yl)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B51)
Figure imgf000053_0001
methyl 2-amino-l-(2-(pyrrolidin-l-yl)ethyl)-lH-benzo[d]imidazole-6-carboxylate (B52)
Figure imgf000053_0002
methyl 3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 1 -(2-(pyrrolidin- 1 -yl)ethyl)-2,3-dihydro-
lH-benzo[d]imidazole-5-carboxylate (B53)
Figure imgf000053_0003
methyl l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-3-(2-(pyrrolidin-l-yl)ethyl)-2,3-dihydro-
lH-benzo[d]imidazole-5-carboxylate (B54)
Figure imgf000053_0004
l-(3-((2,4-dichlorophenyl)thio)propyl)-lH-benzo[d]imidazol-2-amine (B76 or FC #2935)
Figure imgf000053_0005
2-(2-amino-lH-benzo d imidazol-l-yl)-N-(2,4-dichlorophenyl)acetamide (B77 or FC #2939)
Figure imgf000053_0006
N-(2,4-dichlorophenyl)-2-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)acetamide (B78 or FC #2941)
Figure imgf000054_0001
(E)-N, l-dibutyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-
imine (B79 or FC #2985)
Figure imgf000054_0002
(E)- 1 -(3 -(2,4-dichlorophenoxy)propy 1)-N,3 -bis(4-met
benzo [d] imidazol-2-imine (B81 or FC #2989)
Figure imgf000054_0003
(E)-N'-( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo imidazol-2-yl)-N,N-
Figure imgf000054_0004
— H
dimethylformimidamide (B80 or FC #2986) ^ ;
l-(2-(dipropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B87 or FC #3128)
Figure imgf000054_0005
.
l-(2-(diisopropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B88 or FC #3130)
Figure imgf000055_0001
l-(2-(pyrrolidin-l-yl)ethyl)-lH-benzo[d]imidazol-2-amine (B89 or FC #3137)
Figure imgf000055_0002
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
Figure imgf000055_0003
diethylpropan- 1 -amine (B90 or FC #3149)
-(3 -(3 -(2-chlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N-
ethylethan- 1 -amine (B91 or FC #3192)
Figure imgf000055_0004
;
-(3-(3-(2,4-difluorophenoxy)propyl)-2-imino-2, -dihydro-lH-benzo[d]imidazol-l-yl)-N,N-
diethylethan- 1 -amine (B92 or FC #3284)
Figure imgf000055_0005
;
l-(3-(diethylamino)propyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B93 or FC #3286)
Figure imgf000055_0006
5-chloro-l-(3-(diethylamino)propyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B94 or FC
Figure imgf000056_0001
-(3-(4-bromophenethyl)-2-imino-2,3-dihyd
l-amine (B95 or FC #3308)
Figure imgf000056_0002
;
l-(2-(diethylamino)ethyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B97 or FC
Figure imgf000056_0003
6-chlor -l-(3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-amine (B98 or FC
Figure imgf000056_0004
l-(3-( -dichlorophenoxy)propyl)-5-(4-fluorophenyl)-lH-benzo[d]imidazol-2-amine (B99
Figure imgf000056_0005
l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-((6-(trifluoromethyl)pyridin-3- yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B100 or FC #3388)
Figure imgf000057_0001
5-(4-fluorophenyl)-l -((6-(trifluoromethyl)pyridin-3-yl)methyl)-lH-benzo[d]imidazol-2
amine (B101 or FC #3389)
Figure imgf000057_0002
1 -([ 1 , 1 '-biphenyl] -4-y l)-2-(3 -ally l-2-imino-2 ,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -ol
Figure imgf000057_0003
1 -(3 -(2 -dichlorophenoxy)propyl)-3 -propyl- 1,3 -dihy dro-2H-benzo[d]imidazol-2-imine
Figure imgf000057_0004
2-(3 -ally l-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(p-tolyl)ethan- 1 -ol (B104 or
RMH-0066629)
Figure imgf000057_0005
;
2-(3 -ally l-2-imino-2,3 -dihy dro- 1 H-benzo [d] imidazol- 1 -y 1)- 1 -(4-bromophenyl)ethan- 1 -ol
Figure imgf000057_0006
2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(3 -bromophenyl)ethan- 1 -ol
Figure imgf000058_0001
-(3 -allyl-2-imino-2,3 -dihydro- lH-benzo[d]imidazol- 1 -yl)- 1 -(furan-2-yl)ethan- 1 -ol (B107 or
Figure imgf000058_0002
allyl-2-imino-2,3-dihydro- 1 H-benzo[d]imidazol- 1 -yl)- 1 -(4-methoxyphenyl)ethan- 1 -ol
Figure imgf000058_0003
l-(4-chlorophenoxy)-3-(3-ethyl-2 imidazol-l-yl)propan-2-ol
(B109 or RM 1 1-0066634)
Figure imgf000058_0004
1 -(3 -propyl-2-imino-2,3 -dihydro- lH-benzo[d]imidazol- 1 -yl)-3 -(4-chlorophenoxy)propan-2-
Figure imgf000058_0005
l-(4-chlorophenoxy)-3-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-
ol (Bill or RMH-0066636)
Figure imgf000058_0006
■allyl-2-imino-2,3-dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy)propan
2-ol (B112 or RMH-0066637)
Figure imgf000058_0007
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol
Figure imgf000059_0001
l-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l -yl)-3-phenoxypropan-2-ol (B114 or
Figure imgf000059_0002
■all -2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(4-chlorophenoxy)propan-2-ol
Figure imgf000059_0003
1 -(2-imino-3 -propy 1-2,3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-3 -phenoxypropan-2-ol (Bl 16 or
NH
OH
RMH-0066641) ;
l-(2-imino-3-methyl-2,3-dihydro- lH-benzo[d]imidazol-l -yl)-3-phenoxypropan-2-ol (B117
or RMH-0066642)
Figure imgf000059_0004
;
-imino-3 -propyl-2, 3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)-3 -(2-methoxyphenoxy)propan
2-ol (B118 or RMH-0066643)
Figure imgf000059_0005
;
1 -(2-chlorophenoxy)-3 -(3 -methyl-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)propan-2-ol (B119
Figure imgf000059_0006
l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro- lH- benzo[d] imidazol- l-yl)propan-2-ol (B120 or RMH-0066645)
Figure imgf000060_0001
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B121 or
Figure imgf000060_0002
l-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B122 or
Figure imgf000060_0003
l-(4-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)propan-2-ol (B123 or RMH-0066648)
Figure imgf000060_0004
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-( -tolyloxy)propan-2-ol
Figure imgf000060_0005
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2-methoxyphenoxy)propan-2-
H3co...
HO,
HN V""
!i j ol (B125 or RMH-0066650) x— ·' ;
l-(2-chlorophenoxy)-3-(2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B126
or RMH-0066651)
Figure imgf000060_0006
l-(3-allyl-2-imino-2,3-dihy xyphenoxy)propan-2-
ol (B127 or RMI 1-0
Figure imgf000061_0001
-imino-3 -methyl-2,3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-3 -(m-tolyloxy)propan-2-ol
NH
OH
N N .
(B128 or RMH-0066653) %— ;
l-(2-imino-3-meth l-2 3-dih dro-lH-benzo d imidazol-l- l -3-(o-tolyloxy)propan-2-ol
Figure imgf000061_0002
In certain embodiments, the compound of formula (II), or a salt, solvate or N- oxide thereof, is selected from the group consisting of:
1 -(2,4-dichlorophenoxy)-3 -(2 -((3 -morpholinopropyl)amino)- 1 H-benzo [d] imidazol- 1 -
yl)propan-2-ol (B55)
Figure imgf000061_0003
l-(2,4-dichlorophenoxy)-3-(2-(phenethylamino)-lH-benzo[d]imidazol-l-yl)propan-2-ol
Figure imgf000061_0004
1 -(2-(( 1 -benzylpiperidin-4-yl)amino)- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4- dichlorophenoxy)propan-2-ol (B57)
Figure imgf000062_0001
;
l-(2,4-dichlorophenoxy)-3-(2-((2-(diethylamino)ethyl)amino)- lH-benzo[d]imidazol- l-
yl)propan-2-ol (B60)
Figure imgf000062_0002
1 -(2,4-dichlorophenoxy)-3 -(2-((3 -(diethylamino)propyl)amino)- 1 H-benzo [d] imidazol- 1 -
yl)propan-2-ol (B61)
Figure imgf000062_0003
Nl-(l-(3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-yl)-N3,N3-diethylpropane-
1, 3 -diamine (B62)
Figure imgf000062_0004
l-(2,4-dichlorophenoxy)-3-(2-(4-ethylpiperazin- l-yl)-lH-benzo[d]imidazol-l-yl)propan-2-ol
Figure imgf000062_0005
l-(2,4-dichlorophenoxy)-3-(2-((5-(diethylamino)pentan-2-yl)amino)-lH-benzo[d]imidazol-l- yl)propan-2-ol (B73 or FC #2862)
Figure imgf000063_0001
; and
l-(l-(3-(2,4-dichlorophenoxy)-2-hydroxypropyl)-lH-benzo[d]imidazol-2-yl)piperidine-4-
carbonitrile (B74 or FC #2863)
Figure imgf000063_0002
In certain embodiments, the compound of formula (III), or a salt, solvate or N- oxide thereof, is selected from the group consisting of:
N-(( 1 H-benzo [d] imidazol-2-yl)methyl)-3 -(2,4-dichlorophenoxy)propan- 1 -amine (B58)
Figure imgf000063_0003
l-(((lH-benzo[d]imidazol-2-yl)methyl)amino)-3-(2,4-dichlorophenoxy)propan-2-ol (B59)
Figure imgf000063_0004
l-(2,4-dichlorophenoxy)-3-((l-(2-(diethylamino)ethyl)-lH-benzo[d]imidazol-2-
Figure imgf000063_0005
yl)oxy)propan-2-ol (B64 or FC #2729)
N-pentyl-1 H-benzo [d]imidazol-2-amine (B83 or FC #3004)
Figure imgf000063_0006
N-phenethyl-lH-benzo[d]imidazol-2-amine (B84 or FC #3005)
Figure imgf000064_0001
;
Nl-((lH-benzo[d]imidazol-2-yl)methyl)-Nl-(3-(2,4-dichlorophenoxy)propyl)-N2,N2- cr "cs
HN ' N ^¾ diethylethane-l,2-diamine (B85 or FC #3040) \~- ; and
Nl-(3-(2,4-dichlorophenoxy)propyl)-Nl-((l-(2-(diethylamino)ethyl)-lH-benzo[d]imidazol- -yl)methyl)-N2,N2-diethylethane-l,2-diamine (B86 or FC # 3041)
Figure imgf000064_0002
In certain embodiments, the invention provides one or more of the following compounds, or a salt, solvate or N-oxide thereof:
3-(2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-l-amine (B63 or FC #35)
Figure imgf000064_0003
2-(4-ethylpiperazin-l-yl)-lH-benzo[d]imidazole (B65 or FC #2817)
lH-benz
Figure imgf000064_0004
o[d]imidazol-2-yl)methanamine (B67 or FC #2820) ;
Nl-(lH-benzo[d]imidazol-2-yl)-N3,N3-diethylpropane-l,3-diamine (B68 or FC #2830) H
■N
-NH
"N
l-(2-(diethylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B69 or FC #2831)
Figure imgf000065_0001
N4-(l -benzo[d]imidazol-2-yl)-Nl,Nl-diethylpentane-l,4-diamine (B70 or FC #2835)
l-(lH- -4-carbonitrile (B71 or FC #2836)
Figure imgf000065_0002
N-(3-morpholinopropyl)-lH-benzo[d]imidazol-2-amine (B72 or FC #2860)
Figure imgf000065_0003
Nl-(lH-benzo[d]imidazol-2-yl)-N2,N2-diethylethane-l,2-diamine (B75 or FC #2864)
Figure imgf000065_0004
N-(l-benzylpiperidin-4-yl)-lH-benzo[d]imidazol-2-amine (B82 or FC #3002)
HN' N N
6-chloro-l-(2-(diethylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B96 or FC #3316)
Figure imgf000066_0001
The compounds of the invention may possess one or more stereocenters, and each stereocenter may exist independently in either the (R) or (S) configuration. In certain embodiments, compounds described herein are present in optically active or racemic forms. It is to be understood that the compounds described herein encompass racemic, optically- active, regioisomeric and stereoisomeric forms, or combinations thereof that possess the therapeutically useful properties described herein. Preparation of optically active forms is achieved in any suitable manner, including by way of non-limiting example, by resolution of the racemic form with recrystallization techniques, synthesis from optically-active starting materials, chiral synthesis, or chromatographic separation using a chiral stationary phase. In certain embodiments, a mixture of one or more isomer is utilized as the therapeutic compound described herein. In other embodiments, compounds described herein contain one or more chiral centers. These compounds are prepared by any means, including
stereoselective synthesis, enantios elective synthesis and/or separation of a mixture of enantiomers and/ or diastereomers. Resolution of compounds and isomers thereof is achieved by any means including, by way of non-limiting example, chemical processes, enzymatic processes, fractional crystallization, distillation, and chromatography.
The methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), solvates, amorphous phases, and/or pharmaceutically acceptable salts of compounds having the structure of any compound of the invention, as well as metabolites and active metabolites of these compounds having the same type of activity. Solvates include water, ether (e.g., tetrahydrofuran, or methyl tert- butyl ether) or alcohol (e.g., ethanol) solvates, acetates and the like. In certain embodiments, the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, and ethanol. In other embodiments, the compounds described herein exist in unsolvated form.
In certain embodiments, the compounds of the invention may exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
In certain embodiments, compounds described herein are prepared as prodrugs. A "prodrug" refers to an agent that is converted into the parent drug in vivo. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound. In other embodiments, a pro drug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.
In certain embodiments, sites on, for example, the aromatic ring portion of compounds of the invention are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the aromatic ring structures may reduce, minimize or eliminate this metabolic pathway. In certain embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a deuterium, a halogen, or an alkyl group.
Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds described herein include and are not limited to 2H, 3H, nC, 13C, 14C, 36C1, 18F, 123I, 125I, 13N, 15N, 150, 170, 180, 32P, and 35S. In certain embodiments, isotopically-labeled compounds are useful in drug and/or substrate tissue distribution studies. In other embodiments, substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements). In yet other embodiments, substitution with positron emitting isotopes, such as nC, 18F, 150 and 13N, is useful in Positron Emission
Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
In certain embodiments, the compounds described herein are labeled by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
The compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein and as described, for example, in Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplemental (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, Advanced Organic Chemistry 4th Ed., (Wiley 1992); Carey and Sundberg, Advanced Organic Chemistry 4th Ed., Vols. A and B (Plenum 2000, 2001), and Green and Wuts, Protective Groups in Organic Synthesis 3rd Ed., (Wiley 1999) (all of which are incorporated by reference for such disclosure). General methods for the preparation of compound as described herein are modified by the use of appropriate reagents and conditions, for the introduction of the various moieties found in the formula as provided herein.
In certain embodiments, reactive functional groups, such as hydroxyl, amino, imino, thio or carboxy groups, are protected in order to avoid their unwanted participation in reactions. Protecting groups are used to block some or all of the reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. In other embodiments, each protective group is removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal.
In certain embodiments, protective groups are removed by acid, base, reducing conditions (such as, for example, hydrogenolysis), and/or oxidative conditions. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and Fmoc groups, which are base labile. Carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl, in the presence of amines that are blocked with acid labile groups, such as t-butyl carbamate, or with carbamates that are both acid and base stable but hydrolytically removable.
In certain embodiments, carboxylic acid and hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc. Carboxylic acid reactive moieties are protected by conversion to simple ester compounds as exemplified herein, which include conversion to alkyl esters, or are blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while coexisting amino groups are blocked with fluoride labile silyl carbamates.
Allyl blocking groups are useful in the presence of acid- and base- protecting groups since the former are stable and are subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked carboxylic acid is deprotected with a palladium- catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and does not react. Once released from the resin, the functional ¾roup is available to react.
Typically blocking/protecting groups may be selected from:
Figure imgf000069_0001
ally! ©n Cb* ailoe Me
Figure imgf000069_0002
oc PKIB trityi seetyl F»c
Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure. Methods
The invention includes a method of treating or preventing a bacterium infection in a subject in need thereof, comprising administering to the subject a
therapeutically effective amount of a pharmaceutically acceptable composition comprising a compound of the invention or a salt or solvate thereof, whereby the bacterium infection is treated or prevented in the subject.
In certain embodiments, the bacterium is Gram-negative. In other embodiments, the bacterium is obligatory aerobic. In yet other embodiments, the bacterium comprises A. baumannii. In yet other embodiments, the compounds of the invention interfere and/or inhibit the Oxidative Phosphorylation (OxPhos) pathway used by the bacterium to produce energy. In yet other embodiments, the compounds of the invention selectively inhibit bacterial OxPhos over mammalian mitochondrial OxPhos.
In certain embodiments, the subject is a mammal. In other embodiments, the subject is human. Combinations
In certain embodiments, the compounds of the present invention are further intended to be useful in combination with one or more additional compounds useful for treating a condition considered herein. These additional compounds may comprise compounds of the present invention or compounds known to treat, prevent, or reduce the symptoms or effects of the conditions considered herein.
Non-limiting examples of additional antibacterial agents contemplated within the invention include lipoprotein antibiotics, such as polymyxins, such as colistin
(polymyxyn E) and/or polymyxin B, or any other cell wall-modifying antibiotic.
In certain embodiments, the at least one compound and the at least one additional antibacterial agent are synergistic. In other embodiments, the at least one additional antibacterial agent comprises a lipoprotein antibiotic. In yet other embodiments, the at least one additional antibacterial agent comprises a polymyxin. In yet other embodiments, the at least one compound comprises at least one selected from the group consisting of 5, 25, and B55-B58.
In certain embodiments, the at least one additional antibacterial agent is coadministered with the compound to the subject. In other embodiments, the at least one additional antibacterial agent and the compound are coformulated. In yet other embodiments, upon administering to the subject the compound and the at least one additional antibacterial agent, the subject is administered the at least one additional antibacterial agent at a lower dose or frequency as compared to the administering of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof. In yet other embodiments, upon administering to the subject the compound and the at least one additional antibacterial agent, the subject develops a lower or slower rate of resistance to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof. In yet other embodiments, upon administering to the subject the compound and the at least one additional antibacterial agent, the subject develops lower toxicity due to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
A synergistic effect may be calculated, for example, using suitable methods such as, for example, the Sigmoid-Emax equation (Holford & Scheiner, 19981, Clin.
Pharmacokinet. 6: 429-453), the equation of Loewe additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol. 114: 313-326) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul. 22: 27-55). Each equation referred to above may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. The corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively. Administration/Dosage/Formulations
The regimen of administration may affect what constitutes an effective amount. The therapeutic formulations may be administered to the patient either prior to or after the onset of a condition considered herein. Further, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be
continuously infused, or may be a bolus injection. Further, the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
Administration of the compositions of the present invention to a patient, preferably a mammal, more preferably a human, may be carried out using known procedures, at dosages and for periods of time effective to treat a condition considered herein in the patient. An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the state of the disease or disorder in the patient; the age, sex, and weight of the patient; and the ability of the therapeutic compound to treat a condition considered herein in the patient. Dosage regimens 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. A non-limiting example of an effective dose range for a therapeutic compound of the invention is from about 1 and 5,000 mg kg of body weight/per day. One of ordinary skill in the art would be able to study the relevant factors and make the
determination regarding the effective amount of the therapeutic compound without undue experimentation.
Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
In particular, the selected dosage level will depend upon a variety of factors including the activity of the particular compound employed, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds or materials used in combination with the compound, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well, known in the medical arts.
A medical doctor, e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
In particular embodiments, it is especially advantageous to formulate the compound in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the patients to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle. The dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of
compounding/formulating such a therapeutic compound for the treatment of a condition considered herein in a patient.
In certain embodiments, the compositions of the invention are formulated using one or more pharmaceutically acceptable excipients or carriers. In certain
embodiments, the pharmaceutical compositions of the invention comprise a therapeutically effective amount of a compound of the invention and a pharmaceutically acceptable carrier.
The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity may be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition. Prolonged absorption of the injectable compositions may be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
In certain embodiments, the compositions of the invention are administered to the patient in dosages that range from one to five times per day or more. In other
embodiments, the compositions of the invention are administered to the patient in range of dosages that include, but are not limited to, once every day, every two, days, every three days to once a week, and once every two weeks. It will be readily apparent to one skilled in the art that the frequency of administration of the various combination compositions of the invention will vary from individual to individual depending on many factors including, but not limited to, age, disease or disorder to be treated, gender, overall health, and other factors. Thus, the invention should not be construed to be limited to any particular dosage regime and the precise dosage and composition to be administered to any patient will be determined by the attending physical taking all other factors about the patient into account.
Compounds of the invention for administration may be in the range of from about 1 μg to about 10,000 mg, about 20 μg to about 9,500 mg, about 40 μg to about 9,000 mg, about 75 μg to about 8,500 mg, about 150 μg to about 7,500 mg, about 200 μg to about 7,000 mg, about 3050 μg to about 6,000 mg, about 500 μg to about 5,000 mg, about 750 μg to about 4,000 mg, about 1 mg to about 3,000 mg, about 10 mg to about 2,500 mg, about 20 mg to about 2,000 mg, about 25 mg to about 1,500 mg, about 30 mg to about 1,000 mg, about 40 mg to about 900 mg, about 50 mg to about 800 mg, about 60 mg to about 750 mg, about 70 mg to about 600 mg, about 80 mg to about 500 mg, and any and all whole or partial increments therebetween.
In some embodiments, the dose of a compound of the invention is from about 1 mg and about 2,500 mg. In some embodiments, a dose of a compound of the invention used in compositions described herein is less than about 10,000 mg, or less than about 8,000 mg, or less than about 6,000 mg, or less than about 5,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1,000 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg. Similarly, in some embodiments, a dose of a second compound (i.e., a drug used for treating Parkinson's Disease ) as described herein is less than about 1,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof.
In certain embodiments, the present invention is directed to a packaged pharmaceutical composition comprising a container holding a therapeutically effective amount of a compound of the invention, alone or in combination with a second
pharmaceutical agent; and instructions for using the compound to treat, prevent, or reduce one or more symptoms of a condition considered herein in a patient.
Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for oral, parenteral, nasal, intravenous, subcutaneous, enteral, or any other suitable mode of administration, known to the art. The pharmaceutical preparations may be sterilized and if desired mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or aromatic substances and the like. They may also be combined where desired with other active agents, e.g., other analgesic agents.
Routes of administration of any of the compositions of the invention include nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous. The compounds for use in the invention may be formulated for administration by any suitable route, such as for oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and
perivaginally), (intranasal and (trans )rectal), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
Suitable compositions and dosage forms include, for example, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for intravesical administration and the like. It should be understood that the formulations and compositions that would be useful in the present invention are not limited to the particular formulations and compositions that are described herein.
Oral Administration
For oral application, particularly suitable are tablets, dragees, liquids, drops, suppositories, or capsules, caplets and gelcaps. The compositions intended for oral use may be prepared according to any method known in the art and such compositions may contain one or more agents selected from the group consisting of inert, non-toxic pharmaceutically excipients that are suitable for the manufacture of tablets. Such excipients include, for example an inert diluent such as lactose; granulating and disintegrating agents such as cornstarch; binding agents such as starch; and lubricating agents such as magnesium stearate. The tablets may be uncoated or they may be coated by known techniques for elegance or to delay the release of the active ingredients. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert diluent.
For oral administration, the compounds of the invention may be in the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropylmethylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate). If desired, the tablets may be coated using suitable methods and coating materials such as OPADRY™ film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRY™ OY Type, OYC Type, Organic Enteric OY-P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRY™ White, 32K18400). Liquid preparation for oral administration may be in the form of solutions, syrups or suspensions. The liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxy benzoates or sorbic acid).
Granulating techniques are well known in the pharmaceutical art for modifying starting powders or other particulate materials of an active ingredient. The powders are typically mixed with a binder material into larger permanent free-flowing agglomerates or granules referred to as a "granulation." For example, solvent-using "wet" granulation processes are generally characterized in that the powders are combined with a binder material and moistened with water or an organic solvent under conditions resulting in the formation of a wet granulated mass from which the solvent must then be evaporated. Melt granulation generally consists in the use of materials that are solid or semi-solid at room temperature (i.e. having a relatively low softening or melting point range) to promote granulation of powdered or other materials, essentially in the absence of added water or other liquid solvents. The low melting solids, when heated to a temperature in the melting point range, liquefy to act as a binder or granulating medium. The liquefied solid spreads itself over the surface of powdered materials with which it is contacted, and on cooling, forms a solid granulated mass in which the initial materials are bound together. The resulting melt granulation may then be provided to a tablet press or be encapsulated for preparing the oral dosage form. Melt granulation improves the dissolution rate and bioavailability of an active (i.e. drug) by forming a solid dispersion or solid solution.
U.S. Patent No. 5, 169,645 discloses directly compressible wax-containing granules having improved flow properties. The granules are obtained when waxes are admixed in the melt with certain flow improving additives, followed by cooling and granulation of the admixture. In certain embodiments, only the wax itself melts in the melt combination of the wax(es) and additives(s), and in other cases both the wax(es) and the additives(s) will melt.
The present invention also includes a multi-layer tablet comprising a layer providing for the delayed release of one or more compounds of the invention, and a further layer providing for the immediate release of a medication for treatment of Parkinson's Disease. Using a wax/pH-sensitive polymer mix, a gastric insoluble composition may be obtained in which the active ingredient is entrapped, ensuring its delayed release.
Parenteral Administration
For parenteral administration, the compounds of the invention may be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose and/or continuous infusion.
Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing and/or dispersing agents may be used. Additional Administration Forms
Additional dosage forms of this invention include dosage forms as described in U.S. Patents Nos. 6,340,475; 6,488,962; 6,451,808; 5,972,389; 5,582,837; and 5,007,790. Additional dosage forms of this invention also include dosage forms as described in U.S. Patent Applications Nos. 20030147952; 20030104062; 20030104053; 20030044466;
20030039688; and 20020051820. Additional dosage forms of this invention also include dosage forms as described in PCT Applications Nos. WO 03/35041 ; WO 03/35040; WO 03/35029; WO 03/35177; WO 03/35039; WO 02/96404; WO 02/32416; WO 01/97783; WO 01/56544; WO 01/32217; WO 98/55107; WO 98/11879; WO 97/47285; WO 93/18755; and WO 90/11757.
Controlled Release Formulations and Drug Delivery Systems
In certain embodiments, the formulations of the present invention may be, but are not limited to, short-term, rapid-offset, as well as controlled, for example, sustained release, delayed release and pulsatile release formulations.
The term sustained release is used in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that may, although not necessarily, result in substantially constant blood levels of a drug over an extended time period. The period of time may be as long as a month or more and should be a release which is longer that the same amount of agent administered in bolus form.
For sustained release, the compounds may be formulated with a suitable polymer or hydrophobic material which provides sustained release properties to the compounds. As such, the compounds for use the method of the invention may be administered in the form of microparticles, for example, by injection or in the form of wafers or discs by implantation.
In one embodiment of the invention, the compounds of the invention are administered to a patient, alone or in combination with another pharmaceutical agent, using a sustained release formulation.
The term delayed release is used herein in its conventional sense to refer to a drug formulation that provides for an initial release of the drug after some delay following drug administration and that mat, although not necessarily, includes a delay of from about 10 minutes up to about 12 hours.
The term pulsatile release is used herein in its conventional sense to refer to a drug formulation that provides release of the drug in such a way as to produce pulsed plasma profiles of the drug after drug administration.
The term immediate release is used in its conventional sense to refer to a drug formulation that provides for release of the drug immediately after drug administration.
As used herein, short-term refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes and any or all whole or partial increments thereof after drug administration after drug administration. As used herein, rapid-offset refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes, and any and all whole or partial increments thereof after drug administration.
Dosing
The therapeutically effective amount or dose of a compound of the present invention will depend on the age, sex and weight of the patient, the current medical condition of the patient and the progression of a condition considered herein in the patient being treated. The skilled artisan will be able to determine appropriate dosages depending on these and other factors.
A suitable dose of a compound of the present invention may be in the range of from about 0.01 mg to about 5,000 mg per day, such as from about 0.1 mg to about 1,000 mg, for example, from about 1 mg to about 500 mg, such as about 5 mg to about 250 mg per day. The dose may be administered in a single dosage or in multiple dosages, for example from 1 to 4 or more times per day. When multiple dosages are used, the amount of each dosage may be the same or different. For example, a dose of 1 mg per day may be administered as two 0.5 mg doses, with about a 12-hour interval between doses.
It is understood that the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days. For example, with every other day administration, a 5 mg per day dose may be initiated on Monday with a first subsequent 5 mg per day dose administered on Wednesday, a second subsequent 5 mg per day dose administered on Friday, and so on.
In the case wherein the patient's status does improve, upon the doctor's discretion the administration of the inhibitor of the invention is optionally given
continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a "drug holiday"). The length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday includes from 10%- 100%, including, by way of example only, 10%, 15%,20%,25%,30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.
Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of
administration, or both, is reduced, as a function of the viral load, to a level at which the improved disease is retained. In certain embodiments, patients require intermittent treatment on a long-term basis upon any recurrence of symptoms and/or infection.
The compounds for use in the method of the invention may be formulated in unit dosage form. The term "unit dosage form" refers to physically discrete units suitable as unitary dosage for patients undergoing treatment, with each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier. The unit dosage form may be for a single daily dose or one of multiple daily doses (e.g., about 1 to 4 or more times per day). When multiple daily doses are used, the unit dosage form may be the same or different for each dose.
Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD50 and ED50. Capsid assembly inhibitors exhibiting high therapeutic indices are a specific example. The data obtained from cell culture assays and animal studies is optionally used in formulating a range of dosage for use in human. The dosage of such capsid assembly inhibitors lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures, embodiments, claims, and examples described herein. Such equivalents were considered to be within the scope of this invention and covered by the claims appended hereto. For example, it should be understood, that modifications in reaction conditions, including but not limited to reaction times, reaction size/volume, and experimental reagents, such as solvents, catalysts, pressures, atmospheric conditions, e.g., nitrogen atmosphere, and reducing/oxidizing agents, with art- recognized alternatives and using no more than routine experimentation, are within the scope of the present application.
It is to be understood that wherever values and ranges are provided herein, all values and ranges encompassed by these values and ranges, are meant to be encompassed within the scope of the present invention. Moreover, all values that fall within these ranges, as well as the upper or lower limits of a range of values, are also contemplated by the present application.
The following examples further illustrate aspects of the present invention. However, they are in no way a limitation of the teachings or disclosure of the present invention as set forth herein.
EXAMPLES
The invention is now described with reference to the following Examples. These Examples are provided for the purpose of illustration only, and the invention is not limited to these Examples, but rather encompasses all variations that are evident as a result of the teachings provided herein.
Materials & Methods:
Unless otherwise noted, all starting materials and resins were obtained from commercial suppliers and used without purification.
Analytical Methods:
XH NMR were recorded on Varian Oxford 300 MHz, in OMSO-d6 unless otherwise noted. Chemical shifts (δ) are expressed in ppm downfield from tetramethylsilane (TMS).
Method A: LC/MS data were determined with a Waters Alliance 2695 HPLC/MS (Waters Symmetry CI 8 column, 4.6 x 75 mm, 3.5 μιη) with a 2996 diode array detector from 210-400 nm; the solvent system was 5-95% acetonitrile in water (with 0.1% TFA) over nine minutes using a linear gradient, and retention times were in minutes. Mass spectrometry was performed on a Waters ZQ using electrospray in positive mode.
Method B: Preparative reversed phase HPLC was performed on a Waters Sunfire column (19 x 150 mm, CI 8, 10 μιη) with a 20 min adjustable mobile phase gradient of acetonitrile/water with 0.1% TFA as buffer using 214 and 254 nm as detection
wavelengths. Injection and fraction collection were performed with a Gilson 215 liquid handling apparatus using UniPoint software.
Method C: LC/MS data were determined on a Shimadzu LC 20AD instrument with a Phenomenex Luna column (CI 8, 3.0 x 50 mm column, 3 μιη). Mobile phase consisted of water and acetonitrile with 0.1% formic acid buffer. Gradient was 10-90% acetonitrile over three minutes and held at 90% acetonitrile for two minutes. Detection was performed on diode array detector from 210-400 nm and retention times are in minutes. Mass specs were determined on an Applied Biosystems MDS Sciex API 2000 instrument using electrospray ionization positive or negative mode.
Antibacterial mechanism of action:
Bacteria (107-109 colony forming units, CFU) are spread onto the surface of agar plates containing 2x, 4x or lOx MIC of each organism/compound combination. Plates are incubated for 48 hr at the appropriate temperature. Colonies that appear are purified on agar containing the appropriate concentration of compound, and resistance is confirmed by MIC determination. Total DNA is prepared from colonies resistant to the different compounds and their parent strains and the A baumannii NDH-1 operon consisting of 14 structural genes (nuoA-N) is amplified by PCR. DNA is sequenced, and DNA sequences of parent and mutant genes are compared to identify any changes.
To confirm whether the identified mutation(s) in the NDH- 1 genes confer resistance, the mutant gene is PCR amplified and cloned into an appropriate vector and expressed in the wild type strain. Alternatively, a wild type gene is expressed in isolated mutants to determine whether the expression of wild type protein is able to reverse resistance to the compound tested. These tests confirm if NDH-1 is the target for the compound. If a compound does not appear to target NDH-1, the genome is sequenced in an attempt to identify the target protein whose mutation conferred resistance to the organism.
MIC determination:
The MIC is defined as the lowest concentration of compound that inhibits visible growth of the organism. Determinations are performed according to the Clinical and Laboratory Standards Institute recommended procedures for aerobic bacteria (Performanc e Standards for Antimicrobial Susceptibility Testing; 18th Informational Supplement CSLI document M100-S18, Wayne, PA: Clinical and Laboratory Standards Institute, 2008). A comparator antibiotics is used. A positive growth control well containing medium and the test isolate is included on each plate.
MIC90 determinations against drug-resistant and drug-susceptible Ab clinical isolates:
The Walter Reed Army Institute of Research (WRAIR) has developed a diversity set of -120 clinical isolates of A. baumannii that harbors various strains isolated from different infection sites, with different antibiotic resistance profiles and with varied pulsed- field gel electrophoresis (PFGE) patterns that includes representatives of each of the three International Clonal Complex (ICC) groups as well as 34 minor clades as defined by PFGE (Zurawski, et ah, 2012, J. Bacteriol. 194:819-20). Many of the isolates are considered multidrug-resistant (MDR - resistant to more than one conventional antibiotic). Some can even be considered extremely drug-resistant (XDR), which means resistant to both carbapenems (CRE) and the other antibiotic of last resort, colistin. A subset of 33 isolates of this larger diversity set has been previously described and characterized in various studies that showed that there were distinct and observable differences in motility, biofilm, antibiotic resistance genes, and virulence, and these differences could be matched by the phylogeny and genotyping differences found in PFGE, optical mapping, and sequencing (Clinical and Laboratory Standards Institute. 2008. Performance standards for antimicrobial susceptibility testing; 18th informational supplement, vol. 28, no 1. Document no. M100-S18. Clinical and Laboratory Standards Institute, Wayne, PA).
The WRAIR tests the BDI derivatives that were found to inhibit baumannii oxidative phosphorylation. Specifically, minimum inhibitory concentration (MIC) are determined according to CLSI standards (Yu, et ah, 1996, J. Antimicrob. Chemother. 37:233- 42) as well as using a spiral plating method, which can calculate exact MIC values (White, et ah, 1996, Antimicrob Agents Chemother. 40: 1914-8). Subsequently, a time-kill assay (Thompson, et ah, 2012, Antimicrob. Agents Chemother. 56:5419-2) is performed to assess whether or not the compounds are bacteriostatic or bactericidal (Eveillard, et ah, 2010, J. Infect. 60: 154-161) and confirm the MIC results over a time course. MIC results are interpreted visually. Each well is assessed for growth or no-growth. The interface between growth and no-growth wells represents the range in which the MIC exists. Subsequent rounds of MIC determination tests are performed to allow for a narrower MIC range to be determined.
Activity in the mammalian submitochondrial particle (SMP) OxPhos assay:
Bovine heart mitochondria is prepared (Smith, 1967, Methods Enzymol. 10:81-88), and SMPs are prepared (Matsuno-Yagi & Hatefi, 1985, J. Biol. Chem. 260: 14424- 14427) and IC50 values for ATP synthesis are determined in the same way as bacterial membranes.
In vivo efficacy in relevant animal models of lung and wound-based A. baumannii infection: The efficacy experiments are carried out as previously described (Taitt, et ah, 2014, Antimicrob. Agents Chemother. 58:767-81). In order to promote d, baumannii infection, mice are rendered neutropenic via intraperitoneal (IP) administration of 150 mg/kg and 100 mg/kg cyclophosphamide in sterile saline on Day -4 and Day -1 prior to inoculation (Day 0), respectively. Both models require the use of AB5075, which is more virulent than other representative isolates and genetically tractable, which allows for larger therapeutic windows and subsequent studies. Both models require AB5075 grown overnight in LB broth and then sub-cultured to mid-exponential phase. The inoculating culture is then washed and re-suspended in PBS with OD6oo values corresponding to 2.0 x 108 CFU/mL.
The further distinctions between the models follow.
Murine pulmonary model of A. baumannii infection:
For the murine pulmonary infection model, mice are anesthetized with oxygenated isofluorane immediately prior to intranasal inoculation with 25 \L of AB5075 corresponding to 5.0 x 106 CFU. Subsequently, mice are injected IP with the selected derivatives diluted in sterile saline, starting at 4 hours post-infection. Four hours postinfection is chosen because by this time point AB5075 has grown 1-1.5 log CFU. Other time points and dosing schedule are determined based on pilot efficacy experiments. In certain embodiments, the schedule is no more than 2-3 times per day where the effective dose is no more than 25 mg/kg in no more than 200 \L volumes injected IP. Typically, a high dose (25 mg/kg) and a low dose (10 mg/kg) are tested in this model. Once the dosing schedule is determined, efficacy is assessed with the appropriate animal numbers (n = 10/group) to ensure the appropriate statistical power. Safety is also assessed in these experiments as well where no bacteria are inoculated as a control). The dosing regimen such as timing and amount are determined based upon the PK properties of the test compounds. A subset of mice is used for CFU/g wound tissue calculations, histopathology, and PK/PD evaluation. Formulation and vehicle are selected upon the physical properties of the test compounds.
Animal morbidity is scored twice daily for six days using a system evaluating mobility, coat condition, and conjunctivitis (Yano, et al, 2006, J. Biol. Chem. 281 : 11456- 11463). As mice become exceedingly moribund based on clinical score, they are humanely euthanized. To assess CFU burden in the lungs, mice are humanely euthanized according to protocol on Days 2 and 3 post- infection via an injection of ketamine (100 mg/kg) and xylazine (10 mg/kg). To quantify the pulmonary CFU burden, lungs are homogenized in 1 mL PBS and serial dilutions are plated using the Autoplate® Spiral Plating System
(Advanced Instruments, Norwood, MA) onto LB agar supplemented with 50 μg/mL carbenicillin. Bacterial load is reported as CFU per gram of lung tissue.
Murine wound model of A. baumannii infection:
The wound model of infection is carried out as previously described
(Thompson, et al, 2014, Antimicrob. Agents Chemother. 58: 1332-42). The protocol and mice species used for these studies are the same as the above lung model. Briefly, mice are pretreated with cyclophosphamide as described above, and after anesthesia, the dorsum of the mouse is shaved with an electric razor. The surgery area is sterilized with iodine and 70% alcohol. A full-thickness, excisional dermal wound is made on the back of each mouse with a 6 mm sterile biopsy punch, and 30 iL of AB5075 is inoculated in the wound with a 5.0 x 104 CFU dose (Fig. 10). Other groups of mice are wounded, but not inoculated with bacteria, and these mice serve as a negative control for efficacy and an evaluation of safety in this model of infection. Another negative control may include bacterial inoculum but no treatment.
The animals are then re-caged, and analgesics are subcutaneous ly given with buprenorphine at 0.05 mg/kg twice per day for 1-2 days (depending on the clinical condition). After 4 hr post- inoculum, mice are treated by IP dose of BDI derivative with a dosing schedule as determined by the murine pulmonary model experiments. A subset of mice is used for CFU/g wound tissue calculations, histopathology, and PK/PD evaluation.
For bacteria enumeration to calculate efficacy, the mice are monitored using an in vivo imaging system (IVIS Lumina XR, Caliper Life Sciences) every day during the first week, and every other day after the first week. For IVIS monitoring, mice are anesthetized by using isoflurane gas at 2~5% for induction, then 2% for maintenance anesthesia inside IVIS equipment. The bacterial load is also enumerated by counting the bacteria from the grounded wound tissues of the excisional dermal area (central cores from punch biopsy made with 4 mm sterile biopsy punch) or from other tissues if needed (spleens, lungs, livers, kidneys or draining lymphoid nodes, and so on) of euthanized mice at 3 and 5 days; 1, 2 and 3 weeks post-infection, comparing the efficiency with both IVIS and classical CFU enumeration. All tissue is weighed and suspended in 1 mL of PBS and individually homogenized in tissue grinders or mesh bags. Serial 10-fold saline dilutions of homogenates are cultured on LB plate for bacterial enumeration. The number of bacterial colonies are accounted and expressed as CFU per gram tissue.
Physiological observations, including body weight, mobility, skin ruffling, and temperature along with the general condition of the animals are conducted and recorded twice each week. For the immune response, blood is collected from each sacrificed mouse, and RT-PCR is performed to measure transcript of cytokines and chemokines. In order to do this, red blood cells are pelleted via centrifugation and plasma/serum is analyzed via PCR RT2 Profiler™ PCR Array (Qiagen Inc.) according to manufacturer's instructions. This PCR array was designed specifically for mice. Separately and to confirm the presence or absence of certain cytokines, ELISA-based assays are performed. Specifically, cytokines and chemokines associated with the innate immune response are monitored such as: IL-Ιβ, IL-6, IL-8, and TNFa, as these are indicators of sepsis and an inflammatory response. Lastly, some animals are sacrificed at different time points and skin biopsies are fixed for histopathology analysis. This analysis includes the characterization of the cell types at the wound site, inflammatory response, and immunohistochemistry staining to evaluate the presence of predatory bacteria in the wound. This may include hematoxylin and eosin as well as Brown and Brenn, and Brown and Hopps, methods for bacterial staining. One section per block is evaluated via light microscopy by a Veterinary Pathologist blinded as to treatment group. The following elements of wound healing are evaluated: Epithelial Coverage: the total linear distance of each wound and the linear distance covered by migrating epithelial cells aree measured visually and photographed, and the re-epithelialization percentage are calculated (RE% = distance covered by epithelial cells / total linear distance); Epithelial Maturation: the epithelial maturation is noted and scored based on the number of cell layers present (stratum basale, stratum spinosum, stratum granulosum, stratum corneum);
Granulation tissue: the amount of granulation tissue is scored; White cell infiltrate: the amount of subepithelial mixed leukocytic infiltrates is scored as an indicator of the degree of inflammation; Presence of Bacteria: Presence or absence of microscopically detectable bacteria is recorded. The score applied is: 1= absent, 2= mild, 3=moderate, 4= marked, 5=severe.
Wound closure is measured over time via a Silhouette (Aranz Medical Ltd., New Zealand). At each time point a picture is taken, and wound area is determined by this instrument to provide a time to close metric of efficacy, which is really an evaluation of both the antimicrobial activity as well as wound healing. Gross pathology, impact on biofilm (scanning electron microscopy of the wound bed and dressing), and histology of the wound are evaluated (Thompson, et al, 2014, Antimicrob. Agents Chemother. 58: 1332-42).
Toxicity in mammalian cell lines:
In order to assess cytotoxicity, activity against actively dividing cells of the mammalian cell lines NIH3T3, HepG2 and A549 is analyzed, tested at half log dilutions. Standard antibiotics are used as comparators. Cytochrome P450 (CYP) inhibition:
Inhibition of CYP metabolite formation is assessed for the isoforms CYP1A2, CYP2D6, and CYP3A4 in human and mouse liver microsomes using isoform-specific probe substrates. Appropriate positive control inhibitors are used.
DMPK studies:
A total of 6 male mice, aged approximately 6-8 weeks, aree used for each test compound assessed. The mice are divided into 2 dose groups (n=3 per group): intravenous and oral. Dosing of the test compound occurs once via the corresponding dose route for that group. Animals are fasted overnight prior to dosing and until 4 hours post-dose. Water is provided ad libitum. All animals are observed for clinical signs. Animals in the intravenous dose group are bled via jugular vein cannula at the following time points: pre-dose, 5, 15, and 30 minutes, and 1, 3, and 6 hours. Animals in the oral gavage dose group are bled via jugular vein cannula at the following time points: pre-dose, 30 minutes, and 1, 3, 6, 12, and 24 hours. Upon collection of the samples, the blood is processed to plasma for analysis of the test compound via LC-MS/MS.
Example 1: Screening Studies
Isolated A. baumannii (Ab) membranes were screened against a small- molecule library of about 10,000 compounds. Sub-mitochondrial particles (SMP), prepared from bovine heart, were used to estimate selectivity relative to mammalian OxPhos as a secondary assay.
The primary screen identified several drug-like scaffolds such as compound 1, which inhibited A. baumannii ATP synthesis. Compound 1 belongs to the 1H- benzo[d]imidazol-2(3H)-imine compound class, and was found to have IC50 of 1.9 μΜ for the inhibition of ATP synthesis, MIC of 17 μΜ against .4. baumannii, and good selectivity for A baumannii OxPhos over mammalian (SMP) OxPhos. A correlation between inhibition of ATP synthesis and MIC was observed, but no correlation was found between inhibition of SMP ATP synthesis and cytotoxicity.
In certain embodiments, the compounds of the invention do not inhibit mammalian NDH-1 up to about 100 μΜ. Example 2: Biochemical Investigations
The isolated membranes used for screening contain a fully functional OxPhos system and are amenable to biochemical investigation of the mechanism of action of compounds. Using this system, NDH-1 (the type 1 NADH-quinone oxidoreductase) was identified as a target of compound 1.
Accordingly, the mechanism of action of compound 5 was investigated in these membranes. Fig. 3A illustrates the result of monitoring O2 consumption by A.
baumannii membranes that were exposed to compound 5. Trace (1) (labelled "NADH") shows initial O2 consumption with NADH (ImM) as the substrate, which is abruptly terminated upon addition of compound 5 to give a concentration of 50 μΜ. Addition of ascorbate/TMPD (10 mM/0.1 mM in the assay) returned O2 consumption. Without wishing to be limited by any theory, these results suggest that compound 5 inhibits NDH-1
(preventing transfer of electrons to the quinone (Q) pool), and ascorbate/TMPD directly supplies electrons to the terminal oxidases, cytochrome bo3 oxidase and cytochrome bd oxidase, thereby circumventing the inhibited NDH-1. This is illustrated in Fig. 3B, which depicts the sequence of proteins that shuttle electrons through the OxPhos system of A.
baumannii to create a membrane potential to facilitate ATP synthase function. Similarly, trace (2) (labelled "Succinate") in Fig. 3A shows initial O2 consumption with succinate (50 mM) as the substrate, and no inhibition of O2 consumption upon addition of compound 5 to a 50 μΜ concentration or upon a second addition to give a 100 μΜ concentration. As can be seen from Fig. 3B, the succinate pathway, via succinate dehydrogenase (SDH), is not affected by inhibition of NDH-1. These characteristics mirror those of compound 1. In support of these findings, the IC50 values obtained by monitoring ATP synthesis or by direct monitoring of NADH consumption at 340 nm match closely for both compound 1 and compound 5.
Example 3: SAR Studies
Figure imgf000088_0001
Good activity was observed with o-chlorophenyl and biphenyl groups replacing the dichlorophenoxy group in 1. Removing the 2-(diethylamino)ethyl side chain from compound 1 (to yield compound 4) retained activity but lost selectivity (SMP IC50): the cell based MIC potency for A. baumannii (ATCC 19606, strain 2208) was decreased from 17 μΜ to 50 μΜ by removing the diethylamino functionality.
Removal of the chiral center by replacing the hydroxyl with a hydrogen in 5 (R=H) lowered cell based MIC to 13 μΜ, and binding IC50 improving to 0.6 μΜ from 1.9 μΜ. Selectivity was similar to lead compound 1. Shorter hydrophobic side chains
(compounds 6 and 7) were ten-fold less potent than compound 5.
B36 (R=C1) improved MIC to 6.25 μΜ from 13 μΜ, and B38 (R=4-FPh) had an improved MIC of 1.6 μΜ. Without wishing to be limited by any theory, addition of the 4- F group to the phenyl ring blocks certain metabolism there. Compound B41 (R=4-FPh) improved MIC to 0.8 μΜ from 1.6 μΜ by modifying the substitution on the amine- containing sidechain from diethylamino to pyrrolidino (fusion of ethyl groups to form a ring). Replacement of the 2,4-dichlorophenoxy of 5 with 4-CF3-biphenyl as in B43 yielded an improvement of MIC from 13.0 μΜ to 3.1 μΜ.
The SAR aspects of compound 5 were explored further. Diverse side chain replacements were made to the 2-(diethylamino)ethyl group, including compounds with ethyl (compound 13), butyl (compound 14), phenethyl (compound 15), di(alkyl)aminoethyl
(compounds 16-17), heterocyclic (compounds 18-27), amide (compound 28), and diether (compound 29) groups. Results are summarized in Figs. 6A-6C. Of the analogs prepared and tested, group, most showed low micromolar activity in the A. baumannii ATP synthesis screen, and some showed antibacterial activity at or below 50 μΜ. Congeners 17 and 18 (Figs. 6A-6B, N-propyl and cyclopentyl in place of ethyl) were as active as compound 5, although somewhat less selective.
Increasing hydrophobicity (LogD) by incorporation of additional substitution on the phenyl ring of the benzimidazole improved potency. For example, B36 improved MIC to 6.25 μΜ from 13 μΜ, and B38 had an improved MIC of 1.6 μΜ. Compound B41 improved MIC to 0.8 μΜ from 1.6 μΜ by modifying the substitution on amine-containing side chain from diethylamino to pyrrolidino.
Without wishing to be limited by any theory, increased MIC potency upon substitution on the benzimidazole ring may be related to increased hydrophobicity (increased LogD) and thus increased cell penetration. Without wishing to be limited by any theory, the 4-F group added to the phenyl ring blocks possible metabolism at that site.
Table 1.
Figure imgf000089_0001
The cytotoxicity evaluation is illustrated in Table 2 as an average between two cell lines (3T3/HepG2, EC50S), with the ratios given relative to MIC. The two cell lines were mouse 3T3 fibroblasts (ATCC, CRL-1658) and human transformed liver HepG2 cells (ATCC HB-8065), using an MTS viability assay (CellTiter 96® AQueous Non-Radioactive Cell Proliferation Assay) from Promega. Cytotoxicity selectivity was improved by incorporation of the 4-FPh to the benzimidazole, and improved by changing diethylamino to pyrrolidino (viz. B41).
Table 2.
Figure imgf000089_0002
Biological data for selected compounds of the invention are illustrated in
Table 3.
Table 3.
Figure imgf000090_0001
23 2925 18.3 49.7 2.7 100.0 323 89
24 2926 3.6 3.1 0.9 100.0 63 99
19 2927 3.6 22.4 6.2 50.0 83 37
25 2928 1.0 0.8 0.8 100.0 >1305 400
26 2929 0.9 0.9 1.0 50.0 35 22
28 2930 6.0 1 1.4 1.9 100.0 143 68
27 2931 1.9 6.1 3.2 100.0 168 94
29 2932 5.6 6.0 1.1 100.0 151 62
B13 2933 2.9 10.1 3.4 50.0 36 35
6 2934 6.9 18.3 2.7 100.0 187 59
B76 2935 4.6 1.1 0.2 100.0 84 76
B14 2936 1.6 1.6 1.0 50.0 38 42
B15 5.1 9.4 1.9 25.0 97 33
7 2937 5.1 9.4 1.9 25.0 97 33
B77 2939 21.4 12.0 0.6 100.0 813 345
B78 2941 36.7 156.1 4.2 100.0 114 137
14 2981 3.8 1.8 0.5 50.0 72 28
B79 2985 2.1 1.8 0.8 50.0 28 25
B80 2986 1.4 1.2 0.8 >100.0
B17 2987 3.5 2.2 0.6 50.0 65 26
B18 2988 2.2 1.0 0.5 50.0 51 39
B81 2989 2.7 1.2 0.5 50.0
B82 3002 >40.0 >100.0 >100.0 >1872 1096
B60 3003 10.0 13.2 1.3 100.0 151 55
B83 3004 38.4 42.0 1.1 >100.0 1385 828
B84 3005 31.0 29.0 0.9 >100.0 903 758
B56 3006 0.8 0.5 0.7 >100.0 173 152
B57 3007 2.7 5.7 2.1 >100.0 121 91
B58 3008 2.1 18.0 8.6 100.0 212 127
B62 3009 7.8 9.6 1.2 50.0 162 51
B85 3040 13.9 36.6 2.6 >100.0 218 1 18 B86 3041 4.4 6.8 1.5 >100.0 108 53
13 3042 6.4 8.8 1.4 50.0 96 24
B87 3128 75.6 58.8 0.8 >100.0 >2047 1140
16 3129 2.8 2.9 1.0 25.0 49 29
B88 3130 40.0 100.0 2.5 100.0 3130 3130
B89 3137 40.0 100.0 2.5 100.0 2181 2181
17 3145 1.7 2.5 1.4 13.0 37 19
18 3146 2.1 4.9 2.3 13.0 67 12
B23 3147 2.7 13.5 5.1 13.0 38 15
33 16.7 21.7 1.3 50.0 139 38
B90 3149 16.7 21.7 1.3 50.0 139 38
B25 3150 3.5 19.3 5.5 25.0 87 26
B26 3176 7.5 8.9 1.2 25.0 52 26
B27 3191 10.8 1 1.7 1.1 25.0 95 19
B91 3192 23.8 31.6 1.3 >100.0 287 40
B28 3193 15.3 28.2 1.8 50.0 47 25
B29 3212 6.0 18.3 3.0 25.0 11 1 35
B92 3284 22.7 34.8 1.5 >100.0 389 207
B30 3285 2.5 13.3 5.4 38.0 102 29
B93 3286 40.0 100.0 2.5 >100.0 2563 1012
B94 3287 13.3 81.4 6.1 >100.0 980 546
B31 3288 3.9 26.7 6.9 75.0 425 177
B32 3289 2.5 1.8 0.7 2.4 77 31
B33 3290 2.2 2.2 1.0 1.6 50 23
B34 3291 3.4 2.2 0.7 4.7 106 48
B95 3308 23.2 31.3 1.4 >100.0 >41.0 559 152
B35 3309 7.5 4.0 0.5 25.0 10 148 50
B96 3316 >40.0 >100.0 >100.0 >27 >2021 >2021
B36 3317 1.4 6.8 4.9 6.25 3 58 24
B97 3318 >40.0 22.5 >100.0 >31 670 517
B37 3319 5.8 7.0 1.2 1.6 1 33 29 B38 3329 0.7 3.8 5.1 3.1 1.6 61 126
B39 3330 1.2 5.1 4.3 3.1 1.8 65 86
B98 3341 5.9 3.0 0.5 >100.0 >37.0 57 76
B40 3383 4.5 8.2 1.8 3.1 1.7
B41 3385 1.3 1.0 0.8 0.8 0.4 78 35
B99 3387 11.9 12.3 1.0 >100.0 >43.0 90 93
B100 3388 8.5 2.1 0.2 >100.0 >59.0 45 39
B101 3389 3.8 9.0 2.3 >100.0 >39.0
B42 3402 76.7 20.0 0.3 >50 >16
B43 3403 1.7 1.5 0.9 3.1 1.5
B44 3404 4.1 3.9 1.0 12.5 7 57 57
B45 3436 3.3 3.1 0.9 12.5 7.0
B46 3437 1.3 1.1 0.8 6.25 3.2
B47 3438 12.6 19.3 1.5 50 21.8 193 105
B48 3441 8.1 1 1.5 1.4 >50
B49 3442 27.0 6.6 0.2 >50
B50 3443 1.1 3.8 3.6 3.1 1.6 94 26
B51 3444 2.1 1.5 0.8 3.1 1.8 43 31
B52 3445 >40.0 >100.0 >50 3468 2281
B53 3460 1.6 17.3 10.9 12.5 6.1 123 31
B54 3461 1.6 16.4 10.1 25 12.3 115 40
Al Gl 3.4 10.9 3.2 50.0
A2 G2 4.3 6.2 1.4 50.0
G3 or
A3/1 RMH- 1.9 15.4 8.1 17.0 84 40 0066645
RMH-
B102 3.6 5.9 1.6 100.0
0040272
RMH-
B103 2.9 9.0 3.1 50.0
0048597
RMH-
B104 37.5 37.5 1.0 100.0
0066629
RMH-
B105 17.4 28.7 1.6 100.0
0066630
RMH-
B106 22.6 37.5 1.7 100.0
0066631
RMH-
B107 0066632 37.5 37.5 1.0 100.0 RMH-
B108 37.5 37.5 1.0 100.0 0066633
RMH-
B109 0066634 13.6 37.5 2.8 100.0
RMH-
B110 7.4 24.5 3.3 100.0 0066635
RMH-
Bill 21.1 37.5 1.8 100.0 0066636
RMH-
B112 3.5 6.2 1.8 50.0 0066637
RMH-
B113 4.2 17.7 4.3 100.0 0066638
RMH-
B114 37.5 37.5 1.0 100.0 0066639
RMH-
B115 23.4 3.4
0066640 7.0 100.0
RMH-
B116 0066641 37.5 37.5 1.0 100.0
RMH-
B117 0066642 37.5 37.5 1.0 100.0
RMH-
B118 37.5 37.5 1.0 100.0 0066643
RMH-
B119 37.5 37.5 1.0 100.0 0066644
RMH-
B120 1.5 15.5 10.3 20.0 0066645
RMH-
B121 19.4 37.5 1.9 100.0 0066646
RMH-
B122 37.5 37.5 1.0 100.0 0066647
RMH-
B123 11.5 37.5 3.3 100.0 0066648
RMH-
B124 36.6 37.5 1.0 100.0 0066649
RMH-
B125 0066650 37.5 37.5 1.0 100.0
RMH-
B126 5.2 7.9 1.5 100.0 0066651
RMH-
B127 32.4 1.2
0066652 37.5 100.0
RMH-
B128 37.5 37.5 1.0 100.0 0066653
RMH-
B129 13.8 37.5 2.7 100.0 0066654
Figure imgf000094_0001
B39 3.13 1.56 ND >50 6.25 3.13 3.13
B41 3.13 3.13 ND 50 12.5 3.13 3.13
50 6.25 6.25 ND 50 50 3.13 6.25
Ciprofloxacin <0.024 0.39 ND 0.2 0.024 0.39 50
EC = Escherichia coli; SA = Staphylococcus aureus; EF = Enterococcus faecelis; PA = Pseudomonas aeruginosa; KP = Klebsiella pneumoniae; AB(BAA-1605) = drug-resistant strain of A. baumannii; ND = not determined Example 4: Further Characterization of Compound 5
Compound 5 was further evaluated in a panel of 23 clinical isolates of A. baumannii including multi-drug resistant (MDR) strains from various regions around the US, Europe and Latin America. The results are summarized in Figs. 5A-5B. The MIC90 for compound 5 was 8 μg/mL (13 μΜ) and the range was tight (4-16 μg/mL). The compound performed better against the drug-resistant strains than commercial antibiotics ceftazidime and imipenem, and was generally only a few fold less potent than standard of care colistin (MIC90 = 2 μg/mL), which displays dose-limiting toxicities.
Similar results were obtained with compound 4 in a second screen of the same panel of clinical isolates; MIC90 of 8 μg/mL and MIC range of 8-16 μg/mL. Susceptibility to either compound was comparable between drug-sensitive and drug-resistant strains, indicating that the resistance phenotypes were not effective against the OxPhos inhibitors.
Compound 5 showed low metabolic stability in mouse and human liver microsomes (t 2 of 13 min and 9.2 min, respectively), which translates to the potential for high hepatic clearance rates in vivo. The major metabolite generated in mouse liver microsomes was N-dealkylation of the diethyl amine, and minor amounts of hydroxy lated products were also observed. Because of this finding, other compounds not possessing the dialkylamino side chain were evaluated in mouse liver microsome stability assays including compounds 15, 25 and 26 (Figs. 6A-6B). Each of these three compounds showed considerably greater stability than 5 (48.2 min, 25.2 min and >90 min, respectively). These results indicate that metabolic stability can be improved through structural modifications, and that decreasing the dealkylation metabolic pathway does not result in some other pathway becoming overly prevalent.
Metabolic stability in liver microsomes was indeed improved by changing the amine group and adding substitution onto the phenyl portion of the benzimidazole ring. Compound 18 ( =H) has a 2.8 min half-life in rat liver microsomal stability compared to 1.7 min for 5 (change of amine substituent). Compound B41 with the 4-FPh group on the middle ring has a metabolic half-life of 12.2 min, improved from 2.8 min for 18. The mouse liver microsomal stability for 25 is t 2 = 24.2 mins.
Example 5: Chemical Synthesis
Benzimidazoles 2, 4-7, 13-29, B55 and B59-B62 were prepared using the synthetic schemes shown in Fig. 4. Compounds 2, 4-7 and 13-29 were prepared by sequential alkylation of 2-aminobenzimidazole (compound A) under relatively mild conditions to generate compound B, followed by a second alkylation under more rigorous conditions. 2-Aminobenzimidazoles B55 and B59-B62 were prepared by sequential reaction of 2-chlorobenzimidazole (compound C) to afford 2-aminobenzimidazoles D followed by alkylation on an internal nitrogen. Aminoalcohol B59 was prepared by reaction of 2- aminomethyl benzimidazole E with epoxide F derived upon reaction of epichlorohydrin and 2,4-dichlorophenol.
The method of preparation of 2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylethanamine (compound 5) is illustrated herein. A mixture of 2-aminobenzimidazole (350 mg, 2.63 mmol), 2-bromo-N,N- diethylethanamine (520 mg, 2.89 mmol), and cesium carbonate (1,880 mg, 5.77 mmol) in DMF (19 ml) was heated at 50 °C for three hours. An additional charge of 2-bromo-N,N- diethylethanamine (260 mg, 1.45 mmol) and cesium carbonate (940 mg, 2.88 mmol) was made and the reaction was heated for an additional fourteen hours. The reaction was concentrated in vacuo and partitioned between 3 N NaOH and ethyl acetate. The aqueous phase was extracted with two more portions of ethyl acetate. The combined ethyl acetate extracts were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the crude product. The crude was purified by silica chromatography using a gradient of 0-100% (90:9: 1 dichloromethane / methanol / ammonium hydroxide) to dichloromethane to generate N,N-diethyl-2-(2-imino-2,3-dihydro-lHbenzo[d]imidazol-yl)ethanamine (240 mg, 1.03 mmol, 39% yield).
A mixture of N,N-diethyl-2-(2-imino-2,3-dihydro-lH-benzo[d]imidazol- yl)ethanamine (50 mg, 0.215 mmol) and l-(3-bromopropoxy)-2,4-dichlorobenzene (73 mg, 0.258 mmol) in toluene (1 ml) was heated at 1 10 °C for 15 hours. Upon cooling, the precipitate was isolated to afford the title compound as the bis-HBr salt (73 mg, 0.122 mmol, 57% yield). HRMS (ESI+): calculated for C22H29C12 40 m/z [M+H] : 435.1713, observed 435.1624. 'H NMR (300MHZ, CD3OD) δ = 7.50 ( td, J=1.7, 7.6 Hz, 2H), 7.41-7.20 (m, 4H), 6.95 (d, J=8.8 Hz, 1H), 4.42 (t, J=6.6 Hz, 2H), 4.27-4.19 (m, 2H), 4.13 (t, J=5.3 Hz, 2H), 2.90-2.82 (m, 2H), 2.60 (q, J=7.3 Hz, 4H), 2.36 (td, J=6.1, 11.6 Hz, 2H), 1.01-0.91 (m, 6H).
Example 6: l-(2,4-Dichloro-phenoxy)-3-[2-(3-morpholin-4-yl-propylamino)- benzoimidazol-l-yl]-pro
Step 1. (lH-Benzoimida
Figure imgf000097_0001
A mixture of 2-chloro-lH-benzo[d]imidazole (580 mg, 3.82 mmol), 3-morpholino propan-1 -amine (1.10 g, 7.63 mmol), N-ethyl-N-isopropylpropan-2-amine (980 mg, 7.63 mmol) in dry NMP (2 mL) was heated at 165 °C in a sealed pressure tube. After five hours, the reaction was cooled and diluted with ethyl acetate (20 mL). The organic phase was washed with a mixture of 1 : 1 water/ IN NaOH (20 mL) and then water (2 x 10 mL), dried with solid K2CO3 and concentrated in vacuo. The resulting solid was triturated with ether (10 mL) and the solids collected by vacuum filtration to obtain the title compound as an off-white solid (140 mg, 14 % yield). LCMS Method A: (M+H)+ = 260.46.
Step 2. l-(2,4-Dichlorophenoxy)-3-(2-(3-morpholinopropylamino)-lH-benzo[d]imidazol-l- yl)propan-2-ol
Figure imgf000097_0002
Cesium carbonate (225 mg, 0.69 mmol) was added to a mixture of (1H- Benzoimidazol-2-yl)-(3-morpholin-4-yl-propyl)-amine (60 mg, 0.23 mmol, Example 6, Step 1), 2-((2,4-dichlorophenoxy)methyl)oxirane (59 mg, 0.27 mmol, Enamine) in dry NMP (500 μΚ). The reaction was heated to 100 °C for one hour then cooled to room temperature. The reaction was diluted with ethyl acetate (20 mL) and washed with water (3 x 10 mL), dried with solid K2CO3 and concentrated in vacuo. The crude material was purified by prep HPLC (Method B) to afford the title compound as the bis TFA salt (80 mg, 54% yield). LCMS Method A: (M+H)+ = 479.06, 481.03.
The following compounds were prepared using similar procedures to those used to prepare FC2861 :
Table 4.
Figure imgf000098_0002
Example 7: (lH-Benzoimidazol-2-ylmethyl)-[3-(2,4-dichloro-phenoxy)-propyl]-amine (FC3008, or Compound B58)
Figure imgf000098_0001
A mixture of (lH-benzo[d]imidazol-2-yl)methanamine dihydrochloride (230 mg, 1.06 mmol), potassium carbonate (728 mg, 5.28 mmol) and l-(3-bromopropoxy)-2,4- dichlorobenzene (299 mg, 1.06 mol) in acetonitrile (5 mL) was heated at 50 °C for two hours and then 63 °C overnight. The reaction was further heated at 70 °C for fourteen hours then allowed to cool. The reaction yielded a mixture of mono, di and trialkylated products. The solvent was removed in vacuo and the residue was partitioned between ethyl acetate (50 mL) and water (25 mL). The layers were separated and the organic phase was washed with brine, dried over K2CO3, filtered and concentrated in vacuo. To the material was added TFA (600 μΚ) and the mixture was again concentrated in vacuo. The concentrate was triturated with 60% methanol/water (8 mL) and filtered. The filtrate was by prep HPLC (Method B) to afford the bis TFA salt of the title compound as a white solid (73 mg, 12% yield). LCMS Method A: (M+H)+ = 349.94, 351.97.
Example 8: 2-{3-[3-(2,4-Dichloro-phenoxy)-propyl]-2-imino-2,3-dihydro- benzoimidazol-l-yl} or Compound 25)
Figure imgf000099_0001
Step 1. l-[3-(2,4-Dichloro-phenoxy)-propyl]-lH-benzoimidazol-2-ylamine
Figure imgf000099_0002
aminobenzimidazole (1.00 g, 7.51mmol, AK Scientific) in dry DMF (38 mL). The mixture was stirred for 20 minutes before adding neat 2-(3-bromopropoxy)-2,4-dichlorobenzene (2.13 g, 7.51 mmol, Acros). The mixture was heated overnight at 50 °C. The reaction was cooled and partitioned between water and warm ethyl acetate. The layers were separated and the product was further extracted with warm ethyl acetate. The combined organic extracts were washed with brine, filtered and concentrated in vacuo. The resulting slurry was triturated with dichloromethane and the solids were collected by vacuum filtration. The title compound was obtained as an off-white powder (2.25 g, 89% yield). LCMS Method A: (M+H)+ = 335.94, 337.91.
Step 2. 2-{i-[i-(2,4-Dichloro-phenoxy)-propyl]-2 mino-2,i-dihydro-benzoimidazol-l-yl}- l-(4-phenyl-pip
Figure imgf000099_0003
A mixture of l-[3-(2,4-dichloro-phenoxy)-propyl]-lH-benzoimidazol-2- ylamine (100 mg, 0.297 mmol, Example 8, Step 1), 2-Chloro-l-(4-phenylpiperazino) ethanes - 1-one (142 mg, 0.595 mmol, Maybridge), cesium carbonate (194 mg, 0.595 mmol, and sodium bromide (31 mg, 0.297 mmol) in DMF (2 mL) was heated overnight at 120 °C. The reaction was cooled, filtered and purified directly by prep HPLC (Method B) to obtain the bis TFA salt of the title compound as a yellow glass (41 mg, 18% yield). XH NMR (300MHz, DMSO-d6) δ = 8.85 (s, 1H), 7.60-7.52 (m, 3H), 7.39-7.28 (m, 3H), 7.28-7.23 (m, 2H), 7.23- 7.14 (m, 1H), 7.13-7.08 (m, 1H), 7.03-6.99 (m, 1H), 6.88-6.79 (m, 1H), 4.46 (s, 2H), 4.41- 4.36 (m, 1H), 4.33-4.27 (m, 1H), 4.20-4.05 (m, 3H), 3.74-3.58 (m, 2H), 3.57 (s, 2H), 3.39- 3.30 (m, 1H), 3.23-3.13 (m, 1H), 2.36-2.17 (m, 3H). LCMS Method A: (M+H)+ = 538.08, 540.12.
The following compounds were prepared using similar procedures to those used to prepare FC2928:
Table 5.
Figure imgf000100_0003
Example 9: l-(2-Diethylamino-ethyl)-2-imino-3-[2-(4'-trifluoromethyl-biphi ethyl]-2,3-dihydro-l )
Figure imgf000100_0001
Step 1. l-[2-(4-Bromo^henyl)-ethyl]-3-(2-diethylamino-ethyl)-2-imino-2,3-dihydro-lH- benzoimidazole
Figure imgf000100_0002
In a similar manner to Example 11, Step 5, the title compound was prepared (290 mg, 73% yield) by using l-(2-diethylamino-ethyl)-lH4jenzoimidazol-2-ylamine (Example 12, Step 1) as the starting material with 4-bromophenethyl bromide (Acros).
LCMS Method A: (M+H)+=415.11,417.13.
Step 2. l-(2-Diethylamino-ethyl)-2 mino-3-[2-(4'-trifluoromethyl-biphenyl-4-yl)^
2,3-dihydro-lH-benzoimidazole
Figure imgf000101_0001
In a similar manner to as Example 1 1, Step 4, l-[2-(4-Bromo-phenyl)-ethyl]- 3-(2-diethylamino-ethyl)-2-imino-2,3-dihydro-lH-benzoimidazole (Example 9, Step 1) was used as the starting material to afford the title compound as the bis-TFA salt (3 mg, 2% yield). LCMS Method A: (M+H)+=481.21.
The following compounds were prepared using similar procedures to those used to prepare FC3403:
Table 6.
Figure imgf000101_0003
Example 10: 5-Chloro-l-[3-(2,4-dichloro-phenoxy)-propyl]-3-(2-diethylamino-ethyl)-2- imino-2,3-dihydro-lH-b 36):
Figure imgf000101_0002
FC3317 was prepared by two procedures A and B below. Using procedure A. regiochemistry of the final product is unambiguous, as the procedure starts from starting material 4-chloro-2-fluoro-l -nitrobenzene. The same final product was obtained using both procedures A and B, indicating that chlorination step 2 of procedure B yields the chlorine atom in the position described. Procedure A
Step 1. N'-(5-Chloro-2-nitro-phenyl)-N,N-diethyl-ethane-l,2-diamine
Figure imgf000102_0001
In a similar fashion as Example 13, Step 2, 4-chloro-2-fluoro-l -nitrobenzene was used to prepare the title compound (6.40 g, 98% yield). LCMS Method A: (M+H)+ = 272.15.
Step 2. 4-Chloro-N2-(2-diethylam mine
Figure imgf000102_0002
In a similar fashion as Example 13, Step 3, N'-(5-chloro-2-nitro-phenyl)-N,N- diethyl-ethane- 1 ,2-diamine was used as a starting material to prepare the titled compound (1.20 g, 21% yield). LCMS Method A: (M+H)+ = 242.12.
Step 3. 6-Chloro-l-(2-diethylamin idazol-2-ylamine
Figure imgf000102_0003
In a similar fashion as Example 13, Step 4, 4-chloro-N2-(2-diethylamino- ethyl)-benzene-l,2-diamine was used to prepare the titled compound (600 mg, 83% yield). LCMS Method A: (M+H)+ = 267.09.
Step 4. 5-Chloro-l-[3-(2,4-dichloro-phenoxy)-propyl]-3-(2-diethylamino-ethyl)-2-imino- 2,3-dihydro-lH-benzoimidazole
Figure imgf000102_0004
In a similar fashion as Example 13, Step 5, 6-Chloro- 1 -(2-diethylamino- ethyl)-lH-benzoimidazol-2-ylamine was used as a starting material to prepare the bis-TFA salt of the titled compound (46 mg, 18 % yield). 1H NMR (300MHz, DMSO-d6) 5 = 9.17 (br. s., 1H), 7.87-7.79 (m, 1H), 7.65-7.50 (m, 2H), 7.42-7.29 (m, 2H), 7.13-7.04 (m, 1H), 4.57-
4.42 (m, 2H), 4.38-4.26 (m, 2H), 4.18-4.07 (m, 2H), 3.48-3.09 (m, 6H), 2.30-2.12 (m, 2H),
1.20 (t, J=7.0 Hz, 6H). LCMS Method A: (M+H)+ = 469.10, 471.14.
Procedure B;
Step 1. l-(2-Diethylamino-ethyl)-lH-benzoimidazol-2-ylamine
Figure imgf000103_0001
A mixture of 2-amino enzimidazole (1.94 g, 14.5 mmol), 2-bromo-N,N- diethylethanamine hydrobromide* (4.51 g, 17.4 mmol, Acros), cesium carbonate (23.6 g, 72.5 mmol) and sodium bromide (2.98 g, 29 mmol) in DMF (20 ml) was heated at 50 C for 16 hrs. The reaction was diluted with ethyl acetate (60 ml ) and washed with water (3 x 40 mL), dried with sodium sulfate, filtered, and concentrated in vacuo to afford the crude product. The crude product was purified by flash chromatography (80 g silica, 0-100% A/B where A = 90:9: 1 DCM/MeOH/NH4OH and B = DCM) to obtain the title compound (1.04 g, 31% yield). LCMS Method A: (M+H)+ = 233.17.
*For synthesizing diisopropyl analogs, 2-bromo-N,N-diisopropylethanamine hydrochloride was used: To a solution of 2-(diisopropylamino)ethanol (5.68 g, 39.1 mmol) in 1,2-dichloroethane (25 mL) was added thionyl chloride ( 9.69 g, 97.9 mmol) dropwise over 20 minutes. After aging 15 minutes, the reaction was warmed to 70 °C overnight under nitrogen. The reaction was concentrated under vacuum to yield a solid which was triturated with Et20. The solids were collected by vacuum filtration to afford 2-bromo-N,N- diisopropylethanamine HC1 (7.52 g, 82% yield).
Step 2. 6-Chloro-l-(2-diethylamin idazol-2-ylamine
Figure imgf000103_0002
l-(2-Diethylamino-ethyl)-lH-benzoimidazol-2-ylamine (325 mg, 1.40 mmol, Example 10, Step 1) was dissolved in thionyl chloride (7 mL) in a sealable tube. The tube was capped and heated overnight at 90 °C. The thionyl chloride was removed in vacuo and the residue was partitioned between aqueous saturated NaHC03 and EtOAc. The layers were separated and the product extracted with EtOAc twice more. The combined organic extracts were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to obtain the title compound as an orange solid (373 mg, quantitative yield). LCMS Method A: (M+H)+ = 267.16. 'H NMR (300MHZ, DMSO) δ = 9.79 (bs, 1H), 8.82 (bs, 2H), 7.76 (m, 1H), 7.34 (m, 2H), 4.45 (m, 2H), 3.41 (m, 2H), 3.34-3.10 (m, 4H), 1.19 (m, 6H).
Step 3. 5-Chloro-l-[3-(2,4-dichloro^henoxy)^ropyl]-3-(2-diethylamino-ethyl)-2-imino- 2,3-dihydro-lH-benzoimidazole
Figure imgf000104_0001
In a similar manner as Example 11, Step 5, 6-Chloro-l-(2-diethylamino- ethyl)-lH-benzoimidazol-2-ylamine (50 mg, 0.19 mmol, Example 10, Step 2) was used as the starting material to prepare the title compound as the bis TFA salt (46 mg, 34% yield). XH NMR (300MHz, DMSO-d6) δ = 10.03 (br. s., 1H), 9.21 (br. s., 2H), 7.90-7.79 (m, 1H), 7.65- 7.47 (m, 2H), 7.46-7.29 (m, 2H), 7.13-7.04 (m, 1H), 4.58-4.48 (m, 2H), 4.38-4.26 (m, 2H), 4.21-4.09 (m, 2H), 3.46-3.36 (m, 2H), 3.36-3.15 (m, 4H), 2.27-2.12 (m, 2H), 1.27-1.15 (m, 6H). LCMS Method A: (M+H)+ = 469.10, 471.08.
Example 11 : l-[3-(2,4-Dichloro-phenoxy)-propyl]-3-(3-diethylamino-propyl)-2-imino-5- 32)
Figure imgf000104_0002
A mixture of 2-aminobenzimidazole (2.00 g, 15.0 mmol), 2-chloro-l-propanol (3.76 mL, 45.0 mmol, Alfa Aesar), and cesium carbonate (14.7 g, 45.0 mmol) in dry DMA (75 mL) was heated at 50 °C for 2 hours. The reaction was cooled and filtered. The filtrate was concentrated in vacuo to obtain an orange slurry. The slurry was diluted with DCM and allowed to stir at room temperature overnight. The white precipitate was collected by vacuum filtration, washing with DCM, to obtain the title compound as a white powder (1.35 g, 47% yield). LCMS Method A: (M+H)+ = 192.15.
Step 2. 6-Chloro-l-(3-chloro-propyl)-lH-benzoimidazol-2-ylamine and 5,6-Dichloro-l-(3- chloro-propyl)-lH-benzoimidazol-2-ylamine
Figure imgf000105_0001
3-(2-Amino-benzoimidazol-l-yl)-propan-l-ol (1.35 g, 5.44 mmol, Example 11, Step 1) in thionyl chloride (5.2 mL) was heated overnight at 90 °C in a sealed tube. The reaction was cooled, and the thionyl chloride was removed in vacuo. The residue was diluted with DCM and concentrated in vacuo again to obtain the mixture of the two products as a yellow foam (2.07g, > quantitative yield), which was taken on without further purification. LCMS Method A: (M+H)+ = 244.09, 246.06 and 278.02, 279.99.
Step 3. 6-Chloro-l-(3-diethylamino-propyl)-lH-benzoimidazol-2-ylamine and 5,6- Dichloro-l-(3
Figure imgf000105_0002
A mixture of 6-chloro-l-(3-chloro-propyl)-lH-benzoimidazol-2-ylamine and 5,6-dichloro-l-(3-chloro-propyl)-lH-benzoimidazol-2-ylamine (2.07 g, exact mmol unknown, Example 1 1, Step 2) was heated in a sealed tube at 50 °C in diethylamine (13 mL) overnight. More diethylamine (6 mL) was added and the reaction heated at 90 °C for another 3 hours. The solvent was removed in vacuo and the residue was purified by flash chromatography (80 g silica, 0-100% A/B where A = 90:9: 1 DCM/MeOH/NH4OH and B = DCM). A mixture of the title compounds was obtained as an orange-brown solid (1.79 g, exact mmol unknown). LCMS Method A: (M+H)+ = 281.07 and 315.02, 316.99.
Step 4. l-(3-Diethylamino-propyl)-6-phenyl-lH-benzoimidazol-2-ylamine, 5-Chloro-l-(3- diethylamino-propyl)-6-phenyl-lH-benzoimidazol-2-ylamine, l-(3-Diethylamino-propyl)-
Figure imgf000106_0001
The product mixture from Example 1 1, Step 3, of 6-Chloro-l-(3- diethylamino-propyl)-lH-benzoimidazol-2-ylamine and 5,6-Dichloro-l-(3-diethylamino- propyl)- lH-benzoimidazol-2-ylamine (200 mg, exact mmol unknown), phenyl boronic acid (130 mg, 1.07 mmol), and 2M aqueous potassium carbonate (1.07 mL, 2.14 mmol) in dioxane (3.5 mL) in a 20 mL scintillation vial was flushed with nitrogen gas for 5 minutes before adding X-Phos (85 mg, 0.18 mmol) and palladium acetate (16 mg, 0.07 mmol). The vial was capped and heated at 80 °C for two hours. The reaction was poured into water and the product was extracted with EtOAc three times. The combined organic extracts were washed with brine, dried over a2S04, filtered and concentrated in vacuo to a brown foam. The crude material was purified by prep HPLC (Method B). The three products as bis-TFA salts eluted as three separate peaks in the same order as in the title: l-(3-diethylamino- propyl)-6-phenyl-lH-benzoimidazol-2-ylamine (52 mg, LCMS Method C: (M+H)+ = 322.9) and 5 -chloro- 1 -(3 -diethylamino-propyl)-6-phenyl- 1 H-benzoimidazol-2-ylamine (47 mg, LCMS Method C: (M+H)+ = 356.9) and l-(3-diethylamino-propyl)-5,6-diphenyl-lH- benzoimidazol-2-ylamine (31 mg, LCMS Method C: (M+H)+ = 398.9).
Step 5. l-[3-(2,4-Dichloro^henoxy)^ropyl]-3-(3-diethylamino-propyl)-2-imino-5-phenyl- 2,3-dihydro-lH-benzoimidazole
Figure imgf000106_0002
A solution of the freebase of l-(3-diethylamino-propyl)-6-phenyl benzoimidazol-2-ylamine (28 mg, 0.087 mmol, Example 11, Step 4) and 2-(3- bromopropoxy)-2,4-dichlorobenzene (30 mg, 01 mmol, Acros) in toluene (500 μί) in a sealed one dram vial was heated overnight at 105 °C. The reaction was purified directly by prep HPLC (Method B) to obtain the bis TFA salt of the title compound as a yellow powder after freeze drying from dioxane overni ght (43 mg, 66% yield). XH NMR (300MHz, DMSO- d6) δ = 9.47 (br. s., 1H), 8.97 (br. s., 2H), 7.94-7.90 (m, 1H), 7.73-7.67 (m, 2H), 7.66-7.60 (m, 1H), 7.60-7.55 (m, 1H), 7.55-7.52 (m, 1H), 7.52-7.45 (m, 2H), 7.42-7.37 (m, 1H), 7.37- 7.32 (m, 1H), 7.13-7.07 (m, 1H), 4.43-4.32 (m, 2H), 4.30-4.21 (m, 2H), 4.21-4.13 (m, 2H), 3.24-3.04 (m, 6H), 2.33-2.17 (m, 2H), 2.15-1.95 (m, 2H), 1.24-1.12 (m, 6H). LCMS Method A: (M+H)+ = 525.15, 527.19.
The following compounds were prepared using similar procedures to those used to prepare FC3289:
Figure imgf000107_0002
Example 12: l-[3-(2,4-Dichloro-phenoxy)-propyl]-3-(2-diethylamino-ethyl)-2-imino-5- phenyl-2,3-dihydro-lH-b B37)
Figure imgf000107_0001
Step 1. l-(2-Diethylamino-ethyl)-6-phenyl-lH-benzoimidazol-2-ylamine
Figure imgf000108_0001
6-Chloro- 1 -(2-diethylamino-ethyl)- lH-benzoimidazol-2-ylamine (75mg, 0.28 mmol, Example 10, Method A, Step 2 or Method B, Step 3), phenyl boronic acid (52 mg, 0.42 mmol), and 2M aqueous potassium carbonate (423 μί, 0.85 mmol) in dioxane (1.5 mL) in a 20 mL scintillation vial was flushed with nitrogen gas for 5 minutes before adding X- Phos (34 mg, 0.071 mmol) and palladium acetate (6.3 mg, 0.028 mmol). The vial was capped and heated at 80 °C overnight. The reaction was poured into water and the product was extracted with EtOAc three times. The combined organic extracts were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to an orange foam. The crude product was purified by flash chromatography (4 g silica, 0-100% A/B where A = 90:9: 1 DCM/MeOH/NH4OH and B = DCM). The title compound was obtained as an orange foam (56 mg, 64%). LCMS Method A: (M+H)+ = 309.18.
Step 2. l-[3-(2,4-Dichloro^henoxy)^ropyl]-3-(2-diethylamino-ethyl)-2-imino-5-phenyl- 2,3-dihydro-lH-benzoimidazole
Figure imgf000108_0002
In a similar manner as Example 11, Step 5, l-(2-Diethylamino-ethyl)-6- phenyl-lH-benzoimidazol-2-ylamine (56 mg, 0.18 mmol, Example 12, Stepl) was used as the starting material to prepare the title compound as the bis-TFA salt (46 mg, 34% yield). ¾ NMR (300MHz, DMSO-d6) δ = 9.89 (br. s., 1H), 9.06 (s, 2H), 7.92-7.89 (m, 1H), 7.74- 7.68 (m, 2H), 7.67-7.58 (m, 2H), 7.56-7.53 (m, 1H), 7.52-7.45 (m, 2H), 7.43-7.32 (m, 2H), 7.13-7.07 (m, 1H), 4.64-4.58 (m, 2H), 4.40-4.33 (m, 2H), 4.21-4.15 (m, 2H), 3.51-3.39 (m, 2H), 3.38-3.17 (m, 4H), 2.30-2.18 (m, 2H), 1.25-1.17 (m, 6H). LCMS Method A: (M+H)+ = 511.21, 513.25.
The following compounds were prepared using similar procedures to those used to prepare FC3319:
Figure imgf000109_0001
Example 13: l-[3-(2,4-Dichloro-phenoxy)-propyl]-5-(4-fluoro-phenyl)-3-(2-pyrrolidin- l-yl-ethyl)-l,3-dihydro-b or Compound B41)
Figure imgf000110_0001
Step 1. 3,4'-Difluoro-4-nitro-biphenyl
Figure imgf000110_0002
To a mixture of 4-bromo-2-fluoro-l -nitrobenzene (9.30 g, 42.3 mmol) , 4- fluorophenylboronic acid (7.69 g, 55.0 mmol), palladium acetate (238 mg, 1.06 mmol), S- Phos (1.20 g, 2.96 mmol) in dioxane (140 mL) was added 2.0 M aqueous potassium carbonate (74 mL, 148 mmol). The biphasic system was stirred and purged with nitrogen gas for ten minutes. The flask was added to an oil bath preheated to 50 °C, and the reaction was heated for seven hours at 70 °C. The reaction was cooled and concentrated under vacuum. The resulting solid was partitioned between ethyl acetate (80 mL) and water (40 mL). The layers were separated and the ethyl acetate phase was dried over Na2S04, filtered and concentrated in vacuo to afford the title compound as a crude black solid (12.5 g, » theoretical yield), which was taken on without further purification. Assumed theoretical yield for next reaction.
Step 2. 4 '-Fluoro-4-nitro-N-(2-(pyrrolidin-l-yl)ethyl)biphenyl-3-amine
Figure imgf000110_0003
To a mixture of 3,4'-difluoro-4-nitro-biphenyl 9.9 g , 42 mol, Example 13, Step 1) in dry ACN (105 mL) was added 2-(pyrrolidin-l-yl)ethanamine ( 5.4 mL, 43 mmol, Aldrich) dropwise over 10 minutes. After stirring five minutes, DIEA (8.7 mL, 53 mmol) was added over 15 minutes, and then the reaction was stirred overnight at room temp. The reaction was concentrated in vacuo and the residue was diluted with water (100 mL), brine (25 niL) and warm EtOAc (200 mL). The layers were separated and product was extracted from the aqueous layer with warm ethyl acetate (2 x 50 mL). The combined organic extracts were washed with water, brine, dried over solid K2CO3, filtered and concentrated in vacuo to afford the title compound (13.9 g, quantitative yield) which was taken on without further purification. LCMS Method A: (M+H)+ = 330.17
Step 3. 4 '-Fluoro-N3-(2-pyrrolidin-l-yl-ethyl)-biphenyl-3,4-diamine
Figure imgf000111_0001
To a solution of 4'-fluoro-4-nitro-N-(2-(pyrrolidin-l-yl)ethyl)bipheny 1-3 -amine (13.9 g, 42.2 mmol, Example 13, Step 2) in a 3 :4 THF/MeOH mixture (341 mL) was added solid ammonium chloride (27.4 g, 507 mmol) and a catalytic amount of acetic acid (12.2 mL).
Zinc dust (19.2 g, 296 mmol) was added in three portions 1.5 hours apart, while the reaction was at 45 °C. The reaction was heated at 45 °C overnight then cooled, filtered through a thin pack of Celite, while rinsing with methanol, and concentrated in vacuo. The residue was partitioned between ethyl acetate (222 mL) and IN HC1 (139 mL), and the mixture was stirred vigorously for one hour. The layers were separated and the aqueous phase was made basic with -30% aq NaOH (30 mL). The solid white zinc oxide that precipitated out of the solution was filtered off. The product was extracted using ethyl acetate (3 x 100 mL). The combined EtOAc extracts were dried over sodium sulfate and concentrated in vacuo to afford title compound as a dark brown oil (6.6 g, 52 % yield), which was taken on without further purification. LCMS Method A: (M+H)+ =300.22
Step 4. 6-(4-Fluoro-phenyl)-l-(2-pyrrolidin-l-yl-ethyl)-l,3-dihydro-benzoimidazol-2- ylideneamine
Figure imgf000111_0002
To a cloudy solution of 4'-fluoro-N3-(2-pyrrolidin-l-yl-ethyl)-biphenyl-3,4- diamine (1.0 g ,3.3 mmol, Example 13, Step 3) dissolved in MeOH (30 mL) and water ( 30mL) was added solid cyanogen bromide (1.06 g, 10 mmol). The mixture was heated at 50 °C for two hours to give a 1 : 1 mixture of the title compound and the over-cyanated product, 6-(4-fluoro-phenyl)- 1 -(2-pyrrolidin- 1 -yl-ethyl)- 1 H-benzoimidazol-2-yl-cyanamide. The reaction was cooled and the methanol was mostly removed in vacuo. Impurities were extracted from the acidic aqueous layer with DCM (3 x 30 mL) and placed aside. The pH of the aqueous layer was adjusted to around a pH 10 using IN NaOH. The mixture of products was extracted with DCM (3 x 30 mL). The combined organic extracts were dried over Na2S04, filtered and concentrated in vacuo. The crude product was purified by flash chromatography (40 g silica, 0-100% A/B where A = 90:9: 1 DCM/MeOH/NH4OH and B = DCM). The title compound was obtained as a pale tan solid (310 mg, 29% yield). LCMS Method A: (M+H)+ =325.14.
Step 5. l-[3-(2,4-Dichloro-phenoxy)-propyl]-5-(4-fluoro-phenyl)-3-(2-pyrrolidin-l-yl-
Figure imgf000112_0001
In a similar fashion as Example 1 1, Step 5, 6-(4-fluoro-phenyl)-l-(2- pyrrolidin-1 -yl-ethyl)- 1, 3 -dihydro-benzoimidazol-2-ylideneamine (Example 13, Step 4) was used as a starting material to afford the bis TFA salt of the title compound (47 % yield). lH NMR (300MHz, methanol-d4) δ = 7 '.79-7 '.72 (m, 1H), 7.71-7.61 (m, 2H), 7.60-7.45 (m, 2H), 7.42-7.37 (m, 1H), 7.27-7.14 (m, 3H), 7.02-6.92 (m, 1H), 4.49-4.35 (m, 4H), 4.23-4.14 (m, 2H), 3.17-2.99 (m, 2H), 2.94-2.60 (m, 4H), 2.45-2.32 (m, 2H), 1.95-1.79 (m, 4H). LCMS Method A: (M+H)+ =527.26, 529.23.
Example 14: 2-(3-(3-(2,4-Dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol-l-yl)-N,N-diet
Figure imgf000112_0002
- I l l - A mixture of 2-aminobenzimidazole (350 mg, 2.63 mmol), 2-bromo-N,N- diethylethanamine (520 mg, 2.89 mmol), and cesium carbonate (1880 mg, 5.77 mmol) in DMF (19 ml) was heated at 50 C for 3 hrs. Additional 2-bromo-N,N-diethylethanamine (260 mg, 1.45 mmol) and cesium carbonate (940 mg, 2.88 mmol) was added and the reaction was heated for an additional 14 hrs. The reaction was concentrated in vacuo and partitioned between 3 Ν NaOH and ethyl acetate. The aqueous phase was extracted with 2 more portions of ethyl acetate. The combined ethyl acetate extracts were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the crude product, which was purified by silica chromatography using a gradient of 0-100% (90:9: 1 DCM/methanol/ammonium hydroxide) to DCM to obtain N,N-diethyl-2-(2-imino-2,3-dihydro-lH-benzo[i/]imidazol-yl)ethanamine (240 mg, 1.03 mmol, 39% yield). A mixture of N,N-diethyl-2-(2-imino-2,3-dihydro-lH- benzo[i/]imidazol-yl)ethanamine (50 mg, 0.215 mmol) and l-(3-bromopropoxy)-2,4- dichlorobenzene (73 mg, 0.258 mmol) in toluene (1 mL) was heated at 110 C for 15 hrs. Upon cooling, the precipitate was isolated to afford the title compound as the HBr salt (73 mg, 0.142 mmol, 66%). HRMS (ESI+): calculated for C22H29C12 40 m/z [M+H]+:
435.1713, observed 435.1624; XH NMR (300MHz, CD3OD) δ 7.50 (td, J=1.7, 7.6 Hz, 2H), 7.41 - 7.20 (m, 4H), 6.95 (d, J=8.8 Hz, 1H), 4.42 (t, J=6.6 Hz, 2H), 4.27 - 4.19 (m, 2H), 4.13 (t, J=5.3 Hz, 2H), 2.90 - 2.82 (m, 2H), 2.60 (q, J=7.3 Hz, 4H), 2.36 (td, J=6.1, 11.6 Hz, 2H), 1.01 - 0.91 (m, 6H).
Example 15:
Interest in polymyxins has increased as they have become the last line of defense against multidrug resistant (MDR) Gram negative bacteria such MDR baumannii. Polymyxin E (colistin) is used clinically but suffers from dose-limiting toxicity. Polymyxin B and colistin (polymyxin E) are lipopeptide antibiotics that are very similar, differing by one amino acid (D-Phe changed to D-Leu). In the present synergy studies, polymyxin B and E behave in the same way, and polymyxin B/colistin herein is used to indicate either polymyxin B or colistin.
Polymyxin B/colistin is believed to target bacterial membrane lipopolysaccharides and phospholipids, and to disrupt their integrity by increasing permeability and disturbing membrane surface charges. Polymyxin B/colistin also affects numerous membrane-associated cellular functions such as OxPhos and elevates drug penetration into bacterial cells. In certain embodiments, combination of the compounds of the present invention with polymyxin B/colistin allows for lower toxicity by using reduced levels of both compounds and/or reduces the frequency of resistance encountered with monotherapy.
As demonstrated herein, synergy was observed between compounds of the invention and polymyxin B/colistin. Activity of the combination of polymyxin B and colistin with the BDIs was determined via a checkerboard analysis of MIC values as illustrated in Figs. 1 lA-1 IB. The Fractional Inhibitory Concentration Index (FICI) was calculated using the formula
FICI = FICx + FICy,
wherein FICx = X/MICx, FICy = Y/MICy, X is the concentration of compound x, MICx is the MIC of compound x alone, Y is the concentration of compound y, and MICy is the MIC for compound y alone. The FICI was displayed as an isobologram (Fig. 12B).
Colistin, while fairly toxic, is prescribed clinically as the last line of defense against MDR- baumannii. The presently described synergy between compounds of the invention and polymyxin B/colistin that combination therapy reduces the dose of colistin required for therapeutic effect. The isobolograms for B41, 25 and B45 and polymyxin B against .4. baumannii are shown in Figs. 12A-12B. Synergy with polymyxin B was observed for both 25 and B45, and more modestly for B41. Compound 25 demonstrated a 1 μΜ ICso when tested for its ability to inhibit ATP synthesis in A. baumannii membranes (Table 9). However, the potency in isolated membranes did not translate into potent antibacterial activity with an MIC of 400 μΜ against A. baumannii. This compound showed 250 to 500 fold lower MIC values with polymyxin B concentrations of 0. 0625 and 0.125 μg/mL respectively (4 and 2X lower respectively than the polymyxin B MIC for A. baumannii alone). The compound was not cytotoxic, with IC50S of >1,300 μΜ and 400 μΜ against HepG2 and 3T3 cells, respectively (Table 9). In mouse liver microsomes, 25 demonstrated a half-life of 25.2 min.
FICI values for B41, 25, B45, 15 and 26 with A baumannii are shown in Table 10. The synergy of 25 with polymyxin B was also examined against Klebsiella pneumonia (ATCC 4352) as shown in Fig. 12C and Table 11. Based on this analysis, there is at least comparable synergy in the action of 25 and polymyxin B on K. pneumonia, to that observed with A baumannii. Related analogs B55-B58 also showed significant synergy with colistin (Table 12), but they have poor MICs as a single agent against A. baumannii (MICs > 100 μΜ). However, in the presence of sub-MIC concentrations of colistin, their MIC values dropped substantially, similar to the effect seen for 25, B45, 15 and 26. The checkerboard analysis of B56 with colistin against A. baumannii is shown in Fig. 13. As for their parent compounds, they are inhibitors of NDH- 1 as determined by the oxygen consumption assay and other biochemical assays discussed below. Addition of polymyxin B had minimal effect on cytotoxicity of 25 (139 μΜ IC50) in HepG2 (Fig. 14A) and NIH3T3 cells. Table 9. Exemplary compounds of the invention that show synergistic effects with polymyxin B.
Figure imgf000115_0001
Figure imgf000115_0003
a All compounds are bis-TFA salts. Ab ATP synthesis IC50. c AB minimum inhibitory concentration. d nd, not determined Table 10. FICI values for compounds synergy with polymyxin B against A baumannii (ATCC 19606). The MIC with polymyxin B alone was 0.25 μg/ml.
Figure imgf000116_0001
Table 11. FICI values for compound 25 synergy with polymyxin B against K. pneumonia. The MIC with polymyxin B alone was 0.125 μg/ml.
Figure imgf000116_0002
Table 12. Exemplary compounds of the invention that show synergistic effects with colistin.
Figure imgf000116_0003
a All compounds are bis-TFA salts. Ab ATP synthesis IC50. c AB minimum inhibitory concentration.
Table 13 illustrates MIC values for compounds of the invention against colistin resistant ^/? strains. MIC values of these strains for colistin were >128 μg/ml. Their growth are much slower than wild type, suggesting impaired fitness.
Table 13.
Figure imgf000116_0004
colistin) μ^ιηΐ #1 MIC #2 MIC colistin) μΜ (no μΜ (no colistin) colistin)
2928 25 + 400 <1.56 >100 >100
2861 B55 + 50 <1.56 25 12.5
2837 5 + 12.5 1.56 6.25 6.25
3385 B41 - 0.8 0.67 0.2 0.2
Synergistic inhibitory effects can be observed when two or more inhibitors interact at separate sites of an enzyme or a pathway and produce an effect greater than the sum of the individual effects. Without wishing to be limited by any theory, the strong synergy of polymyxin B/colistin and compounds of the invention may be due to the fact that both polymyxin B, colistin and BDIs target bacterial membranes. Polymyxin B and colistin are believed to increase membrane permeability thereby permitting higher influx of compounds into bacterial cells. Furthermore, permeablized membranes may lose the ability to maintain the chemical potential across the membrane that is generated by OxPhos. ATP synthesis activity depends on this potential and thus may be significantly compromised in the presence of low concentrations of polymyxin B or colistin. Furthermore, drugs are often removed from cells by a wide array of efflux pumps (1) ABC transporters, (2) major facilitator superfamily (MFS) proteins, and (3) small multidrug resistance (SMR) family. ABC transporters utilize ATP whereas the MFS and SMR families utilize H+/Na+ motive forces. All efflux pumps depend on the OxPhos activity (Fig. 15), thus compounds of the invention, even in low concentrations, render bacteria more susceptible to treatment with polymyxin B and/or colistin.
In order to better understand the underlying mechanism of synergy observed for compounds of the invention, oxygen consumption of live A. baumannii cells was monitored in the presence of selected compounds. Compound 5, which does not show synergistic activity with colistin, instantaneously inhibited bacterial respiration (Figs. 16A- 16B), which was rescued by the addition of ascorbate/TMPD, direct electron donors to the terminal oxidase, further confirming that compounds of the invention inhibit NDH-1. By contrast, B55, with significant synergistic effects with colistin, inhibited A. baumannii respiration only when colistin was added (Figs. 16A-16B). The associated oxygen consumption gradually slowed down and eventually stopped regardless of the order of addition of B55 or colistin. Furthermore, B55 exerts bactericidal effects only with colistin. Without wishing to be limited by any theory, colistin may enhance drug delivery to the target site and inhibition of respiration decreases PMF and intracellular ATP, thus attenuating efflux pump activities.
Combination doses and ratios of compounds of the invention and colistin for in vivo testing may be determined using a rapid, low-cost, invertebrate wax worm model of infection, followed by evaluation in the mouse A. baumannii wound model. The Galleria mellonella (wax worm) infection model can be used to assess the safety and efficacy of novel therapeutics against bacterial pathogens including ^, baumannii. Moreover, evaluation in G. mellonella can be used to evaluate combination therapy with colistin, specifically against A. baumannii and is useful as a low cost prescreen prior to going into rodent models. The Galleria mellonella model is relatively easy to implement and does not require animal protocol approvals. Twenty worms per group can be used to ensure appropriate statistical power. Three doses of a compound of the invention and colistin each are selected based on the in vitro potency of the compound and the reported efficacy of colistin, along with groups tested with the compound and colistin alone, for 1 1 treatment groups.
Multiple Ab bacterial strains are grown overnight in LB in an orbital shaker (37°C, 200 rpm), and overnight cultures are then diluted 100-fold into fresh medium and grown for 3 hours. Cells are collected by centrifugation, washed once in PBS, and resuspended in PBS to a final OD600 of 1.0. The number of bacterial cells in each injected sample are enumerated by plating 10-fold serial dilutions on LB agar plates and counting CFU after overnight incubation. G. mellonella larvae are infected with an approximate dose of 1.0 x 106 CFU, which is lethal for each GNB isolate. After 4 hours of infection (which allows for one loglO of in vivo growth), 5 of purified product ranging from 10 - 500 μg/mL are injected into the last left proleg of the larvae using a 10 μΐ^ glass syringe fitted with a 30G needle. Each experiment includes control groups of non- injected larvae or larvae injected with 5 sterile PBS. Another control of uninfected larvae is used to assess the toxicity of the purified compounds. Injected larvae are incubated at 37°C, assessing death at 24 hour intervals over six days. Larvae are considered dead if they turn black and do not respond to physical stimuli. Experiments are repeated three times using 20 larvae per experimental group for appropriate statistical analysis. Combinations prequalified from the G. mellonella screen are evaluated in the mouse wound model of A. baumannii infection.
The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety. While the invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.

Claims

CLAIMS What is claimed is:
1. At least one compound, or a salt, solvate or N-oxide thereof, selected from the group consisting of:
Figure imgf000120_0001
wherein in (I)-(III):
ring A is a fused aryl or heteroaryl ring selected from the group consisting
Figure imgf000120_0002
each occurrence of R1 is independently a bond or -CH2-;
R2 is selected from the group consisting of -(CH2)i-3-0-(CH2)o-2-,
Figure imgf000120_0003
and -(CH2)!_6, wherein each of the CH2 groups in R2 is independently optionally substituted with a substituent selected from the group consisting of C1-C6 alkyl, C1-C6 fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7;
R4 is -(CH2)o-4;
each occurrence of R6 is independently CH, CR10 or N, wherein 0-2 occurrences of R6 in ring A are N;
each occurrence of R7 is independently H, C1-C6 alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted; each occurrence of R8 is independently C1-C6 alkyl, heterocyclyl, aryl or heteroaryl, wherein the alkyl, heterocyclyl, aryl or heteroaryl group is optionally substituted;
each occurrence of R9 is independently O, S or R7;
each occurrence of R10 is independently selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7; wherein in (I):
X1 and X2 are independently selected from the group consisting of H and optionally substituted Ci-C6 alkyl, X1 and X2 combine to form =NH, =N(Ci-C6 alkyl), =CH-CN, =N- CN, or =CH-N02, or X1 is absent and X2 is -N=CH-NMe2; wherein in (I) and (II):
each occurrence of R3 is independently -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci- Ce alkyl), pyrrolidin- 1 -yl, pyperidin-l -yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7; wherein in (I) and (III):
R5 is selected from the group consisting of H, prop-2-en- l-yl, C1-C6 alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2, -(C=O)0-i-(OCH2CH2)1_
3-OR
Figure imgf000121_0001
, wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1 ; wherein in (II):
RR55 iiss sseelected from the group consisting of H, prop-2-en- 1 -yl, Ci-Ce alkyl, -(C=0)o-i- aryl, -(C=O)0-i -heteroaryl, -(C=O)0-i -heterocyclyl, -(C=O)0-i-N(R7)2,
-(C=0)o-i-(OCH2CH2)1_3-OR7,
Figure imgf000121_0002
; wherein the alkyl, aryl, heteroaryl or heterocyclic group is optionally substituted; m is 2, 3, 4 or 5; and n is 0 or 1 ; or R4 is a bond and R5 combines with the N atom to which it is bound to form morpholyn- 1 -yl, piperidin- 1-yl, 4-cyano-piperidin- 1 -yl and piperazin- 1 -yl, wherein N4 of the piperazinyl is optionally substituted with Ci-Ce alkyl; wherein in (III):
each occurrence of R3 is independently H, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(C1-C6 alkyl), pyrroli din- 1 -yl, pyperidin- l-yl, aryl or heteroaryl, wherein the aryl or heteroaryl group is optionally substituted with at least one substituent selected from the group consisting of optionally substituted Ci-Ce alkyl, Ci-Ce fluoroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8, NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7; and any mixtures thereof, with the proviso that the compound is not:
1 -(2,4-dichlorophenoxy)-3 -(2-imino-3-propyl-2,3-dihydro- 1 H-benzo[d]imidazol- 1 -yl) propan-2-ol (Al);
1 -(3-allyl-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-2-ol (A2);
l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro- lH- benzo[d] imidazol- l-yl)propan-2-ol (1);
1 -(3 -benzyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-2-ol (A4); or
1 -(2,4-dichlorophenoxy)-3 -(2-imino-3 -(2-(piperidin- 1 -yl)ethyl)-2, 3 -dihydro- 1 H- benzo[d] imidazol- l-yl)propan-2-ol (A5).
2. The compound of claim 1 , wherein the at least one compound is selected from the group consisting of:
Figure imgf000122_0001
(Ilia).
3. The compound of claim 1, in (I), when X1 and X2 combine to form =NH and ring A is a six-membered ring, then at least one occurrence of R6 is CR10 or .
4. The compound of claim 1 , wherein (I) is not a compound of formula
(lb):
Figure imgf000123_0001
(lb), wherein in (lb):
RA is Ci-C6 alkyl, C2-C7 alkenyl, -CH2CH2NEt2 or -CH2CH2-(N1-morpholinyl), or benzyl optionaly substituted with at least one halogen or C1-C3 alkyl;
RB is phenyl optionally substituted with at least one halogen; aryl; aroxy; naphthyl; phenoxymethyl optionally substituted with at least one halogen, C1-C3 alkyl and/or C1-C3 alkoxy; phenyl isoxazole optionally substituted with at least one halogen; or 4-methyl-3- phenyl- 1 ,2,4-oxadiazole-5 (4H)-one.
5. The compound of claim 1 , wherein ring A is
Figure imgf000123_0002
, wherein R6 is
10
CH or CR
6. The compound of claim 1 , wherein at least one occurrence of R6 in ring A is CR .
7. The compound of claim 1 , wherein ring A is
Figure imgf000123_0003
or
Figure imgf000123_0004
8. The compound of claim 7, wherein R10 is F, CI, Br, I or C(=0)OR7.
9. The compound of claim I, wherein ring A is selected from the group consisting of
Figure imgf000124_0001
The compound of claim 1 , wherein ring A is selected from the group consisting of:
Figure imgf000124_0002
wherein Hal is halogen.
11. The compound of claim 9, wherein R is optionally substituted phenyl or optionally substituted pyridyl.
12. The compound of claim 1 1, wherein R is 4-fluorophenyl, p-tolyl or phenyl.
13. The compound of claim 10, wherein R9 is O or NR7.
14. The compound of claim 13, wherein R7 is methyl or 4-fluorophenyl
The compound of claim 1 , wherein both occurrences of R1 are a bond.
The compound of claim 1, wherein R is selected from the group consisting of -(CH2)1-3-0-, -(CH^s-NR7-, -(CH2)1-3-S-, and -(CH2)i^-.
17. The compound of claim 1 , wherein at least one of the CH2 groups in R2 is independently substituted with OR7.
18. The compound of claim 1 , wherein R2 is -CH2CH2-, -CH2CH2CH2-, - CH2CH20-, -CH2CH2CH20-, or -CH2CH(OH)CH20-.
19. The compound of claim 1 , wherein R3 or R3 is phenyl, pyridyl, pyrimidyl, 1 ,2-biphenyl, 1,3-biphenyl, 1 ,4-biphenyl, or naphthyl, wherein the aryl or heteroaryl group is optionally substituted with at least one selected from the group consisting of Ci-C6 alkyl, Ci-C6 fluoroalkyl, F, CI, Br, I, CN, OR7, SR7, S(=0)R8, S(=0)2R8,
NHS(=0)2R8, C(=0)R7, OC(=0)R8, C(=0)OR7, OC(=0)OR8, N(R7)2, C(=0)N(R7)2, OC(=0)N(R7)2, NHC(=0)NHR7, NHC(=0)R7, and NHC(=0)OR7.
20. The compound of claim 1 , wherein R3 or R3 is phenyl, 2- methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-chlorophenyl, 2-methoxyphenyl, 3- methoxyphenyl, 4-methoxyphenyl, 3-chlorophenyl, 4-chlorophenyl, 2-bromophenyl, 3- bromophenyl, 4-bromophenyl, 2-trifluoromethylphenyl, 3 -trifluoromethylphenyl, 4- trifluoromethylphenyl, 2,4-difluorophenyl, 2,4-dichlorophenyl, 1,2-biphenyl, 1,3-biphenyl, 1 ,4-biphenyl, 4-trifluoromethylmethyl- 1 ,4-biphenyl, 4'-fluoro-2-chloro- l,4-biphenyl, 2- trifluoromethyl-pyridin-5-yl, and 2-furyl.
21. The compound of claim 1 , wherein R5 or R5 is selected from the group consisting of H, Ci-Ce alkyl, aryl, N(R7)2, pyrrolidin- l-yl, piperidin-l -yl, N^H-piperidin^- yl, 1-(Ci-C6 alkyl)-piperidin-4-yl, N1-(tBoc)-piperidin-4-yl, morpholin-l -yl, imidazol-l -yl, imidazol-2-yl, pyrrolidin-2-one- l-yl, pyridyl, and
Figure imgf000125_0001
22. The compound of claim 1 , wherein R5 or R5 is N(R7)2, wherein each occurrence of R7 is independently H, methyl, ethyl, w-propyl or isopropyl.
23. The compound of claim 1 , wherein R5 or R5 is selected from the group consisting of pyrrolidin- l-yl, piperidin- l-yl, N1-H-piperidin-4-yl, 1-(Ci-C6 alkyl)-piperidin- 4-yl, N1-(tBoc)-piperidin-4-yl, morpholin-l -yl, imidazol- l -yl, imidazol-2-yl, pyrrolidin-2- one-l-yl, pyridyl, 3,5-dimethylisoxazol-4-yl, 6-trifluoromethyl-pyridin-3-yl, -(C=0)o-i- (OCH2CH2)1_3-0(Ci-C6 alkyl), and -(C=O)0-i-( 4-phenyl-piperazin-l-yl).
24. The compound of claim I, wherein each occurrence of R6 is CR7.
25. The compound of claim 24, wherein each occurrence of R6 is CH.
26. The compound of claim I, wherein the compound of formula (I) is at least one selected from the group consisting of:
1- (3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-amine (4);
2- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (5);
2-(3-(2-(2,4-dichlorophenoxy)ethyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (6);
2-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (7);
l-(3-(2,4-dichlorophenoxy)propyl)-3-ethyl-l,3-dihydro-2H-benzo[d]imidazol-2-imine (13); l-butyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (14); l-(3-(2,4-dichlorophenoxy)propyl)-3-(3-phenylpropyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (15);
N-(2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl) ethyl)-N-propylpropan- 1 -amine (16);
N-(2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 - yl)ethyl)-N-isopropylpropan-2-amine (17);
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -(2-(pyrrolidin- 1 -yl)ethyl)- 1 ,3 -dihydro-2H- benzo[d]imidazol-2-imine (18);
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -(2-(piperidin- 1 -yl)propyl)- 1 ,3 -dihydro-2H- benzo[d]imidazol-2-imine (19);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-morpholinoethyl)-l,3-dihydro-2H-benzo[d]imidazol-
2-imine (20);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyridin-2-yl)ethyl)-l,3-dihydro-2H-benzo[d]
imidazol-2-imine (21);
tert-butyl 4-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]
imidazol- 1 -yl)ethyl)piperidine- 1 -carboxylate (22); 1 -(2-( 1 H-imidazol- 1 -yl)ethyl)-3 -(3 -(2,4-dichlorophenoxy)propyl)- 1 ,3 -dihydro-2H- benzo[d]imidazol-2-imine (23);
1- (2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethyl)pyrrolidin-2-one (24);
2- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-l-(4- phenylpiperazin- 1 -yl)ethan- 1 -one (25);
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -((6-(trifluoromethyl)pyridin-3 -yl)methyl)- 1 ,3 -dihydro- 2H-benzo[d]imidazol-2-imine (26);
1- (3-(2,4-dichlorophenoxy)propyl)-3-((3,5-dimethylisoxazol-4-yl)methyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (27);
2- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylacetamide (28);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(2-ethoxyethoxy)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (29);
3 -(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2, 3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-N,N- diethylpropan-1 -amine (33);
3- (5-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol-
1 -yl)-N,N-diethylpropan- 1 -amine (34);
l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B4);
(R)-l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B5);
((S)-l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B6);
l-(2-(2,4-dichlorophenoxy)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B13);
1- (3-(2,4-dichlorophenyl)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B14);
2- (3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (B15);
l-(cyclopropylmethyl)-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B17);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (B18);
3- (6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N,N-diethylpropan- 1 -amine (B23); 3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- dipropylpropan-1 -amine (B25);
N-benzyl-2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N-ethylethan- 1 -amine (B26);
2- (3-(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2, 3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N- ethylethan-1 -amine (B27);
N-benzyl-2-(3-(3-(2-chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N-ethylethan-l -amine (B28);
N,N-diethyl-2-(2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B29);
3 -(2-imino-3 -(3 -(2-(trifluoromethyl)phenoxy)propyl)-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 - yl)-N,N-dipropylpropan-l -amine (B30);
N,N-diethyl-3-(2-imino-5,6-diphenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-l -amine
(B31);
3- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N,N-diethylpropan- 1 -amine (B32);
3 -(5 -chloro-3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3 -dihydro- 1 H- benzo[d] imidazol- l-yl)-N,N-diethylpropan-l -amine (B33);
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-5,6-diphenyl-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)-N,N-diethylpropan- 1 -amine (B34);
2-(3 -(2-([ 1 , 1 '-biphenyl] -4-yl)ethyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N- diethylethan- 1 -amine (B35);
2-(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N,N-diethylethan- 1 -amine (B36);
2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N,N-diethylethan- 1 -amine (B37);
2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B38);
2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-6-(4-trifluoromethylphenyl)-2-imino-2,3 -dihydro- 1H- benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B39);
1 -(3 -(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3 -(2-(piperidin- 1 -yl)ethyl)- 1,3- dihydro-2H-benzo[d]imidazol-2-imine (B40);
l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(pyrrolidin-l-yl)ethyl)-l,3- dihydro-2H-benzo[d]imidazol-2-imine (B41); 6-(4-fluorophenyl)- l-(2-(pyrrolidin- 1 -yl)ethyl)- lH-benzo[d]imidazol-2-amine (B42);
N,N-diethyl-2-(2-imino-3-(2-(4'-(rt^
lH-benzo[d]imidazol-l-yl)ethan-l-amine (B43);
N-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)ethyl)-N-isopropylpropan-2-amine (B44);
N-(2-(6-(4-fluorophenyl)-2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo[d]imidazol-l-yl)ethyl)-N-isopropylpropan-2-amine (B45);
N,N-diethyl-2-(2-imino-3-(3-((4'-(trifluoromethyl)-[l, l'-biphenyl]-4-yl)oxy)propyl)-2,3- dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B46);
2- (l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-l,2-dihydro-3H-imidazo[4,5-b]pyridin-3-yl)-
N,N-diethylethan-l -amine (B47);
l-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-lH-benzo[d]imidazol-2-amine (B48); 1 -(3 -((3 -chloro-4'-fluoro-[ 1 , 1 '-biphenyl]-4-yl)oxy)propyl)-6-(4-fluorophenyl)- 1H- benzo[d]imidazol-2-amine (B49);
3 -(3 -(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)- 1 -(2-(pyrrolidin- 1 -yl)ethyl)- 1,3- dihydro-2H-benzo[d]imidazol-2-imine (B50);
3- (3-((3-chloro-4'-fluoro-[l, -biphenyl]-4-yl)oxy)propyl)-5-(4-fluorophenyl)-l-(2-
(pyrrolidin- 1 -y l)ethy 1)- 1 , 3 -dihy dro-2H-benzo [d] imidazol-2 -imine (B51 ) ;
methyl 2-amino-l-(2-(pyrrolidin-l-yl)ethyl)-lH-benzo[d]imidazole-6-carboxylate (B52); methyl 3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 1 -(2-(pyrrolidin- 1 -yl)ethyl)-2,3-dihydro-
1 H-benzo [d] imidazole-5 -carboxylate (B53);
methyl l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-3-(2-(pyrrolidin-l-yl)ethyl)-2,3-dihydro-
1 H-benzo [d] imidazole-5 -carboxylate (B54);
1- (3-((2,4-dichlorophenyl)thio)propyl)-lH-benzo[d]imidazol-2-amine (B76);
2- (2-amino- 1 H-benzo [d] imidazol- 1 -yl)-N-(2,4-dichlorophenyl)acetamide (B77);
N-(2,4-dichlorophenyl)-2-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)acetamide (B78);
(E)-N, l-dibutyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (B79);
(E)-l-(3-(2,4-dichlorophenoxy)propyl)-N,3-bis(4-methylpentyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B81);
(E)-N'-( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo [d] imidazol-2 -yl)-N,N- dimethylformimidamide (B80);
l-(2-(dipropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B87); l-(2-(diisopropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B88);
1- (2-(pyrrolidin-l-yl)ethyl)-lH-benzo[d]imidazol-2-amine (B89);
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylpropan- 1 -amine (B90);
2- (3 -(3 -(2 -chlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N- ethylethan-1 -amine (B91);
2-(3-(3-(2,4-difluorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (B92);
1 -(3 -(diethylamino)propyl)-6-phenyl- 1 H-benzo [d] imidazol-2-amine (B93);
5- chloro-l-(3-(diethylamino)propyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B94);
2-(3-(4-bromophenethyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylethan-
1 -amine (B95);
l-(2-(diethylamino)ethyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B97);
6- chloro- 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo [d] imidazol-2-amine (B98);
l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-lH-benzo[d]imidazol-2-amine (B99); l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-((6-(trifluoromethyl)pyridin-3- yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B100);
5 -(4-fluorophenyl)-l-((6-(trifluoromethyl)pyridin-3-yl)methyl)-l H-benzo [d]imidazol-2- amine (B101);
1 -([ 1 , 1 '-biphenyl] -4-yl)-2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -ol (B102);
1- (3-(2,4-dichlorophenoxy)propyl)-3-propyl-l,3-dihydro-2H-benzo[d]imidazol-2-imine
(B103);
2- (3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(p-tolyl)ethan- 1 -ol (B104); 2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(4-bromophenyl)ethan- 1 -ol
(B105);
2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(3 -bromophenyl)ethan- 1 -ol (B106);
2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(furan-2-yl)ethan- 1 -ol (B107); 2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(4-methoxyphenyl)ethan- 1 -ol (B108);
l-(4-chlorophenoxy)-3-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B109);
1 -(3 -propyl-2-imino-2,3 -dihydro- lH-benzo[d]imidazol- 1 -yl)-3 -(4-chlorophenoxy)propan-2- ol (B110);
l-(4-chlorophenoxy)-3-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2- ol (Bill);
1 -(3 -allyl-2-imino-2,3-dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy)propan- 2-ol (B112);
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol (B113);
l-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B114); 1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(4-chlorophenoxy)propan-2-ol (B115);
l-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B116); l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B117); l-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2-methoxyphenoxy)propan- 2-ol (B118);
l-(2-chlorophenoxy)-3-(3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol
(B119);
l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B120);
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B121); l-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B122); l-(4-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B123);
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(p-tolyloxy)propan-2-ol
(B124);
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2-methoxyphenoxy)propan-2- ol (B125);
l-(2-chlorophenoxy)-3-(2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B126); l-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-methoxyphenoxy)propan-2- ol (B127);
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(m-tolyloxy)propan-2-ol (B128); and
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(o-tolyloxy)propan-2-ol (B129).
27. The compound of claim 1, wherein the compound of formula (II) is at least one selected from the group consisting of:
1 -(2,4-dichlorophenoxy)-3 -(2 -((3 -morpholinopropyl)amino)- 1 H-benzo [d] imidazol- 1 - yl)propan-2-ol (B55);
l-(2,4-dichlorophenoxy)-3-(2-(phenethylamino)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B56);
1 -(2-(( 1 -benzylpiperidin-4-yl)amino)- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4- dichlorophenoxy)propan-2 -ol (B57) ;
l-(2,4-dichlorophenoxy)-3-(2-((2-(diethylamino)ethyl)amino)-lH-benzo[d]imidazol-l- yl)propan-2-ol (B60);
1 -(2,4-dichlorophenoxy)-3-(2-((3-(diethylamino)propyl)amino)- 1 H-benzo [d] imidazol- 1 - yl)propan-2-ol (B61);
1 -( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo[d]imidazol-2-yl)-N3 ,N3 -diethylpropane- 1,3 -diamine (B62);
l-(2,4-dichlorophenoxy)-3-(2-(4-ethylpiperazin-l-yl)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B66);
l-(2,4-dichlorophenoxy)-3-(2-((5-(diethylamino)pentan-2-yl)amino)-lH-benzo[d]imidazol-l- yl)propan-2-ol (B73); and
1 -( 1 -(3 -(2,4-dichlorophenoxy)-2-hydroxypropyl)- 1 H-benzo [d] imidazol-2-yl)piperidine-4- carbonitrile (B74).
28. The compound of claim 1, wherein the compound of formula (III) is at least one selected from the group consisting of:
N-((lH-benzo[d]imidazol-2-yl)methyl)-3-(2,4-dichlorophenoxy)propan-l-amine (B58); l-(((lH-benzo[d]imidazol-2-yl)methyl)amino)-3-(2,4-dichlorophenoxy)propan-2-ol (B59); 1 -(2,4-dichlorophenoxy)-3 -((1 -(2-(diethylamino)ethyl)- 1 H-benzo [d] imidazol-2- yl)oxy)propan-2-ol (B64);
N-pentyl- 1 H-benzo [d] imidazol-2-amine (B83);
N-phenethyl- 1 H-benzo [d] imidazol-2-amine (B84);
1 -(( 1 H-benzo[d]imidazol-2-yl)methyl)-N 1 -(3 -(2,4-dichlorophenoxy)propyl)-N2,N2- diethylethane-l,2-diamine (B85); and
1 -(3 -(2,4-dichlorophenoxy)propyl)-N 1 -(( 1 -(2-(diethylamino)ethyl)- 1 H-benzo[d] imidazol-
2-yl)methyl)-N2,N2-diethylethane-l,2-diamine (B86).
29. A pharmaceutical composition comprising at least one compound of claim 1 and at least one pharmaceutically acceptable carrier.
30. The composition of claim 29, further comprising at least one additional antibacterial agent.
31. The composition of claim 30, wherein the at least one compound and the at least one additional antibacterial agent are synergistic.
32. The composition of claim 31, wherein the Fractional Inhibitory Concentration Index (FICI) determined by the combination of the at least one compound and the at least one additional antibacterial agent is equal to or lower than about 0.50.
33. The composition of claim 30, wherein the at least one additional antibacterial agent comprises a lipoprotein antibiotic.
34. The composition of claim 33, wherein the at least one additional antibacterial agent comprises a polymyxin.
35. The composition of claim 30, wherein the at least one compound comprises at least one selected from the group consisting of 5, 25, and B55-B58.
36. A pharmaceutical composition comprising at least one additional antibacterial agent and at least one compound selected from the group consisting of l-(2,4- dichlorophenoxy)-3 -(2-imino-3 -propyl-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl) propan-2-ol; 1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan- 2-ol; l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol; l-(3-benzyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-2-ol; 1 -(2,4-dichlorophenoxy)-3 -(2-imino-3 -(2- (piperidin- 1 -yl)ethyl)-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)propan-2-ol.
37. The pharmaceutical composition of claim 36, wherein the at least one compound and the at least one additional antibacterial agent are synergistic.
38. The phatmaceutical composition of claim 37, wherein the FICI determined by the combination of the at least one compound and the at least one additional antibacterial agent is equal to or lower than about 0.50.
39. A method of treating or preventing a infection by a bacterium in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of: (a) at least one compound of claim 1 , or a salt, solvate, tautomer or N- oxide thereof, or (b) at least one compound, or a salt, solvate, tautomer or N-oxide thereof, selected from the group consisting of l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3- dihydro- lH-benzo[d]imidazol- 1 -yl) propan-2-ol; 1 -(3 -allyl-2-imino-2,3 -dihydro- 1H- benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan-2-ol; 1 -(2,4-dichlorophenoxy)-3 -(3 - (2-(diethylamino)ethyl)-2-imino-2,3 -dihydro- 1 H-benzo[d] imidazol- 1 -yl)propan-2-ol; 1 -(3 - benzyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2- ol; l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin-l-yl)ethyl)-2,3-dihydro-lH-benzo[d] imidazol- 1 -yl)propan-2-ol,
and any mixtures thereof,
whereby the infection by the bacterium in the subject is treated or prevented.
40. The method of claim 39, wherein, when X1 and X2 combine to form =NH and ring A is a six-membered ring, then at least one occurrence of R6 is N.
41. The method of claim 39, wherein the compound of formula (I) is not (2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl) propan- 2-ol (Al); l-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4- dichlorophenoxy) propan-2-ol (A2); l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2- imino-2,3-dihydro-lH-benzo[d] imidazol- l-yl)propan-2-ol (A3, also known as 1); l-(3- benzyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2,4-dichlorophenoxy) propan-2-ol (A4); 1 -(2,4-dichlorophenoxy)-3-(2-imino-3-(2-(piperidin- 1 -yl)ethyl)-2,3-dihydro- 1H- benzo[d] imidazol- l-yl)propan-2-ol (A5).
42. The method of claim 39, wherein (I) is not a compound of formula
(Ib):
Figure imgf000135_0001
(lb), wherein in (lb):
RA is Ci-C6 alkyl, C2-C7 alkenyl, -CH2CH2NEt2 or -CH2CH2-(N1-morpholinyl), or benzyl optionaly substituted with at least one halogen or C1-C3 alkyl;
RB is phenyl optionally substituted with at least one halogen; aryl; aroxy; naphthyl; phenoxymethyl optionally substituted with at least one halogen, C1-C3 alkyl and/or C1-C3 alkoxy; phenyl isoxazole optionally substituted with at least one halogen; or 4-methyl-3- phenyl- 1 ,2,4-oxadiazole-5 (4H)-one.
43. The method of claim 39, wherein the bacterium is Gram-negative.
44. The method of claim 39, wherein the bacterium is obligatory aerobic.
45. The method of claim 39, wherein the bacterium belongs to the Acinetobacter genus.
46. The method of claim 45, wherein the bacterium comprises A.
baumannii.
47. The method of claim 39, wherein the compound interferes with or inhibits the Oxidative Phosphorylation (OxPhos) pathway of the bacterium.
48. The method of claim 47, wherein the compound selectively inhibits the bacterial OxPhos over mammalian mitochondrial OxPhos.
49. The method of claim 39, wherein the compound is at least one selected from the group consisting of:
l-(2,4-dichlorophenoxy)-3-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)
propan-2-ol (Al);
1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy) propan- 2-ol (A2); l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH-benzo[d] imidazol-l-yl)propan-2-ol (1);
1 -(3 -benzyl-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy)
propan-2-ol (A4);
l-(2,4-dichlorophenoxy)-3-(2-imino-3-(2-( iperidin-l-yl)ethyl)-2,3-dihydro-lH-benzo[d] imidazol-l-yl)propan-2-ol (A5);
1- (3-(2,4-dichlorophenoxy)propyl)-lH-benzo[d]imidazol-2-amine (4);
2- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (5);
2-(3-(2-(2,4-dichlorophenoxy)ethyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (6);
2-(3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (7);
l-(3-(2,4-dichlorophenoxy)propyl)-3-ethyl-l,3-dihydro-2H-benzo[d]imidazol-2-imine (13); l-butyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (14); l-(3-(2,4-dichlorophenoxy)propyl)-3-(3-phenylpropyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (15);
N-(2-(3 -(3 -(2,4-dichlorophenoxy )propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl) ethyl)-N-propylpropan- 1 -amine (16);
N-(2-(3 -(3 -(2,4-dichlorophenoxy )propyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 - yl)ethyl)-N-isopropylpropan-2-amine (17);
1 -(3 -(2,4-dichlorophenoxy )propyl)-3-(2-(pyrrolidin- 1 -yl)ethyl)- 1 ,3 -dihydro-2H- benzo[d]imidazol-2-imine (18);
1 -(3 -(2,4-dichlorophenoxy )propy l)-3 -(2-(piperidin- 1 -yl)propyl)- 1 ,3 -dihydro-2H- benzo[d]imidazol-2-imine (19);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-morpholinoethyl)-l,3-dihydro-2H-benzo[d]imidazol-
2-imine (20);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(pyridin-2-yl)ethyl)-l,3-dihydro-2H-benzo[d]
imidazol-2-imine (21);
tert-butyl 4-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]
imidazol- 1 -yl)ethyl)piperidine- 1 -carboxylate (22);
1 -(2-( 1 H-imidazol- 1 -yl)ethyl)-3 -(3 -(2,4-dichlorophenoxy)propyl)- 1 ,3 -dihydro-2H- benzo[d]imidazol-2-imine (23);
l-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)ethyl)pyrrolidin-2-one (24);
2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-l-(4- phenylpiperazin- 1 -yl)ethan- 1 -one (25);
1 -(3 -(2,4-dichlorophenoxy)propyl)-3 -((6-(trifluoromethyl)pyridin-3 -yl)methyl)- 1 ,3 -dihydro-
2H-benzo[d]imidazol-2-imine (26);
1- (3-(2,4-dichlorophenoxy)propyl)-3-((3,5-dimethylisoxazol-4-yl)methyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (27);
2- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylacetamide (28);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(2-(2-ethoxyethoxy)ethyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (29);
3 -(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2, 3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-N,N- diethylpropan-1 -amine (33);
3 - (5 -chloro-3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d] imidazol-
1 -yl)-N,N-diethylpropan- 1 -amine (34);
l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B4);
(R)-l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B5);
((S)-l-(2,4-dichlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)propan-2-ol (B6);
l-(2-(2,4-dichlorophenoxy)ethyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B13);
1- (3-(2,4-dichlorophenyl)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B14);
2- (3-(3-(2,4-dichlorophenyl)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (B15);
l-(cyclopropylmethyl)-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B17);
l-(3-(2,4-dichlorophenoxy)propyl)-3-(4-methylpentyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (B18);
3 -(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 - yl)-N,N-diethylpropan- 1 -amine (B23);
3 -(3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2, 3 -dihydro- 1 H-benzo [d]imidazol- 1 -yl)-N,N- dipropylpropan-1 -amine (B25);
N-benzyl-2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol- 1 -yl)-N-ethylethan- 1 -amine (B26);
2- (3-(3 -(2,4-dichlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N- ethylethan-1 -amine (B27);
N-benzyl-2-(3-(3-(2-chlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N-ethylethan-l -amine (B28);
N,N-diethyl-2-(2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B29);
3 -(2-imino-3 -(3 -(2-(trifluoromethyl)phenoxy)propyl)-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 - yl)-N,N-dipropylpropan-l -amine (B30);
N,N-diethyl-3-(2-imino-5,6-diphenyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-l -amine
(B31);
3- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N,N-diethylpropan- 1 -amine (B32);
3 -(5 -chloro-3 -(3 -(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3 -dihydro- 1 H- benzo[d] imidazol- l-yl)-N,N-diethylpropan-l -amine (B33);
3-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-5,6-diphenyl-2,3-dihydro-lH- benzo [d] imidazol- 1 -yl)-N,N-diethylpropan- 1 -amine (B34);
2-(3 -(2-([ 1 , 1 '-biphenyl] -4-yl)ethyl)-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-N,N- diethylethan- 1 -amine (B35);
2-(6-chloro-3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l- yl)-N,N-diethylethan- 1 -amine (B36);
2-(3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-6-phenyl-2,3-dihydro-lH-benzo[d]imidazol-
1 -yl)-N,N-diethylethan- 1 -amine (B37);
2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B38);
2-(3 -(3 -(2,4-dichlorophenoxy)propyl)-6-(4-trifluoromethylphenyl)-2-imino-2,3 -dihydro- 1H- benzo [d] imidazol- 1 -yl)-N,N-diethylethan- 1 -amine (B39);
1 -(3 -(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3 -(2-(piperidin- 1 -yl)ethyl)- 1,3- dihydro-2H-benzo[d]imidazol-2-imine (B40);
l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-(2-(pyrrolidin-l-yl)ethyl)-l,3- dihydro-2H-benzo[d]imidazol-2-imine (B41);
6-(4-fluorophenyl)- l-(2-(pyrrolidin- 1 -yl)ethyl)- 1 H-benzo [d]imidazol-2-amine (B42);
N,N-diethyl-2-(2-imino-3-(2-(4'-(trifluoromethyl)-[l, l'-biphenyl]-4-yl)ethyl)-2,3-dihydro- lH-benzo[d]imidazol-l-yl)ethan-l-amine (B43); N-(2-(3-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-2-imino-2,3-dihydro-lH- benzo[d]imidazol-l-yl)ethyl)-N-isopropylpropan-2-amine (B44);
N-(2-(6-(4-fluorophenyl)-2-imino-3-(3-(2-(trifluoromethyl)phenoxy)propyl)-2,3-dihydro-lH- benzo[d]imidazol-l-yl)ethyl)-N-isopropylpropan-2-amine (B45);
N,N-diethyl-2-(2-imino-3-(3-((4'-(trifluoromethyl)-[l, l'-biphenyl]-4-yl)oxy)propyl)-2,3- dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -amine (B46);
2- (l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-l,2-dihydro-3H-imidazo[4,5-b]pyridin-3-yl)-
N,N-diethylethan-l -amine (B47);
l-(3-(2,4-dichlorophenoxy)propyl)-6-(4-fluorophenyl)-lH-benzo[d]imidazol-2-amine (B48); 1 -(3 -((3 -chloro-4'-fluoro-[ 1 , 1 '-biphenyl]-4-yl)oxy)propyl)-6-(4-fluorophenyl)- 1H- benzo[d]imidazol-2-amine (B49);
3 -(3 -(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)- 1 -(2-(pyrrolidin- 1 -yl)ethyl)- 1,3- dihydro-2H-benzo[d]imidazol-2-imine (B50);
3- (3-((3-chloro-4'-fluoro-[l, -biphenyl]-4-yl)oxy)propyl)-5-(4-fluorophenyl)-l-(2-
(pyrrolidin- 1 -y l)ethy 1)- 1 , 3 -dihy dro-2H-benzo [d] imidazol-2 -imine (B51 ) ;
methyl 2-amino-l-(2-(pyrrolidin-l-yl)ethyl)-lH-benzo[d]imidazole-6-carboxylate (B52); methyl 3-(3-(2,4-dichlorophenoxy)propyl)-2-imino- 1 -(2-(pyrrolidin- 1 -yl)ethyl)-2,3-dihydro-
1 H-benzo [d] imidazole-5 -carboxylate (B53);
methyl l-(3-(2,4-dichlorophenoxy)propyl)-2-imino-3-(2-(pyrrolidin-l-yl)ethyl)-2,3-dihydro-
1 H-benzo [d] imidazole-5 -carboxylate (B54);
1- (3-((2,4-dichlorophenyl)thio)propyl)-lH-benzo[d]imidazol-2-amine (B76);
2- (2-amino- 1 H-benzo [d] imidazol- 1 -yl)-N-(2,4-dichlorophenyl)acetamide (B77);
N-(2,4-dichlorophenyl)-2-(3-(2-(diethylamino)ethyl)-2-imino-2,3 -dihydro- 1H- benzo [d] imidazol- 1 -yl)acetamide (B78);
(E)-N, l-dibutyl-3-(3-(2,4-dichlorophenoxy)propyl)-l,3-dihydro-2H-benzo[d]imidazol-2- imine (B79);
(E)-l-(3-(2,4-dichlorophenoxy)propyl)-N,3-bis(4-methylpentyl)-l,3-dihydro-2H- benzo[d]imidazol-2-imine (B81);
(E)-N'-( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo [d] imidazol-2 -yl)-N,N- dimethylformimidamide (B80);
l-(2-(dipropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B87);
l-(2-(diisopropylamino)ethyl)-lH-benzo[d]imidazol-2-amine (B88);
l-(2-(pyrrolidin-l-yl)ethyl)-lH-benzo[d]imidazol-2-amine (B89);
3- (3-(3-(2,4-dichlorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylpropan- 1 -amine (B90);
2-(3 -(3 -(2 -chlorophenoxy)propyl)-2-imino-2,3 -dihydro- 1 H-benzo[d]imidazol- 1 -yl)-N- ethylethan-1 -amine (B91);
2-(3-(3-(2,4-difluorophenoxy)propyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N- diethylethan- 1 -amine (B92);
1 -(3 -(diethylamino)propyl)-6-phenyl- 1 H-benzo [d] imidazol-2-amine (B93);
5- chloro-l-(3-(diethylamino)propyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B94);
2-(3-(4-bromophenethyl)-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-N,N-diethylethan-
1 -amine (B95);
l-(2-(diethylamino)ethyl)-6-phenyl-lH-benzo[d]imidazol-2-amine (B97);
6- chloro- 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo [d] imidazol-2-amine (B98);
l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-lH-benzo[d]imidazol-2-amine (B99); l-(3-(2,4-dichlorophenoxy)propyl)-5-(4-fluorophenyl)-3-((6-(trifluoromethyl)pyridin-3- yl)methyl)-l,3-dihydro-2H-benzo[d]imidazol-2-imine (B100);
5 -(4-fluorophenyl)-l-((6-(trifluoromethyl)pyridin-3-yl)methyl)-l H-benzo [d]imidazol-2- amine (B101);
1 -([ 1 , 1 '-biphenyl] -4-yl)-2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)ethan- 1 -ol (B102);
1- (3-(2,4-dichlorophenoxy)propyl)-3-propyl-l,3-dihydro-2H-benzo[d]imidazol-2-imine
(B103);
2- (3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(p-tolyl)ethan- 1 -ol (B104); 2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(4-bromophenyl)ethan- 1 -ol
(B105);
2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(3 -bromophenyl)ethan- 1 -ol (B106);
2-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-l-(furan-2-yl)ethan-l-ol (B107); 2-(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)- 1 -(4-methoxyphenyl)ethan- 1 -ol (B108);
l-(4-chlorophenoxy)-3-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B109);
1 -(3 -propyl-2-imino-2,3 -dihydro- lH-benzo[d]imidazol- 1 -yl)-3 -(4-chlorophenoxy)propan-2- ol (B110);
l-(4-chlorophenoxy)-3-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2- ol (Bill); 1 -(3 -allyl-2-imino-2,3-dihydro- 1 H-benzo[d]imidazol- 1 -yl)-3 -(2,4-dichlorophenoxy)propan- 2-ol (B112);
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-chlorophenoxy)propan-2-ol (B113);
l-(3-ethyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B114); 1 -(3 -allyl-2-imino-2,3 -dihydro- 1 H-benzo [d] imidazol- 1 -yl)-3 -(4-chlorophenoxy)propan-2-ol (B115);
l-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B116); l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B117); l-(2-imino-3-propyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2-methoxyphenoxy)propan- 2-ol (B118);
l-(2-chlorophenoxy)-3-(3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol
(B119);
l-(2-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B120);
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B121); l-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-phenoxypropan-2-ol (B122); l-(4-chlorophenoxy)-3-(3-(2-(diethylamino)ethyl)-2-imino-2,3-dihydro-lH- benzo[d] imidazol- l-yl)propan-2-ol (B123);
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(p-tolyloxy)propan-2-ol
(B124);
l-(3-butyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(2-methoxyphenoxy)propan-2- ol (B125);
l-(2-chlorophenoxy)-3-(2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)propan-2-ol (B126); l-(3-allyl-2-imino-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(4-methoxyphenoxy)propan-2- ol (B127);
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(m-tolyloxy)propan-2-ol (B128);
l-(2-imino-3-methyl-2,3-dihydro-lH-benzo[d]imidazol-l-yl)-3-(o-tolyloxy)propan-2-ol (B129);
1 -(2,4-dichlorophenoxy)-3 -(2-((3 -morpholinopropyl)amino)- 1 H-benzo [d] imidazol- 1 - yl)propan-2-ol (B55);
l-(2,4-dichlorophenoxy)-3-(2-(phenethylamino)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B56); 1 -(2-(( 1 -benzylpiperidin-4-yl)amino)- 1 H-benzo [d] imidazol- 1 -yl)-3 -(2,4- dichlorophenoxy)propan-2-ol (B57);
l-(2,4-dichlorophenoxy)-3-(2-((2-(diethylamino)ethyl)amino)-lH-benzo[d]imidazol-l- yl)propan-2-ol (B60);
1 -(2,4-dichlorophenoxy)-3-(2-((3-(diethylamino)propyl)amino)- 1 H-benzo [d] imidazol- 1 - yl)propan-2-ol (B61);
1 -( 1 -(3 -(2,4-dichlorophenoxy)propyl)- 1 H-benzo[d]imidazol-2-yl)-N3 ,N3 -diethylpropane- 1,3 -diamine (B62);
l-(2,4-dichlorophenoxy)-3-(2-(4-ethylpiperazin-l-yl)-lH-benzo[d]imidazol-l-yl)propan-2-ol (B66);
l-(2,4-dichlorophenoxy)-3-(2-((5-(diethylamino)pentan-2-yl)amino)-lH-benzo[d]imidazol-l- yl)propan-2-ol (B73);
1 -( 1 -(3 -(2,4-dichlorophenoxy)-2-hydroxypropyl)- 1 H-benzo [d] imidazol-2-yl)piperidine-4- carbonitrile (B74);
N-((lH-benzo[d]imidazol-2-yl)methyl)-3-(2,4-dichlorophenoxy)propan-l-amine (B58); l-(((lH-benzo[d]imidazol-2-yl)methyl)amino)-3-(2,4-dichlorophenoxy)propan-2-ol (B59); 1 -(2,4-dichlorophenoxy)-3 -(( 1 -(2-(diethylamino)ethyl)- 1 H-benzo [d] imidazol-2- yl)oxy)propan-2-ol (B64);
N-pentyl- 1 H-benzo [d] imidazol-2-amine (B83);
N-phenethyl- 1 H-benzo [d] imidazol-2-amine (B84);
1 -(( 1 H-benzo[d]imidazol-2-yl)methyl)-N 1 -(3 -(2,4-dichlorophenoxy)propyl)-N2,N2- diethylethane-l,2-diamine (B85);
1 -(3-(2,4-dichlorophenoxy)propyl)-N 1 -(( 1 -(2-(diethylamino)ethyl)- lH-benzo[d]imidazol-
2-yl)methyl)-N2,N2-diethylethane-l,2-diamine (B86).
50. The method of claim 39, wherein the compound is administered to the subject by a route selected from the group consisting of nasal, inhalational, topical, oral, buccal, rectal, pleural, peritoneal, vaginal, intramuscular, subcutaneous, transdermal, epidural, intratracheal, otic, intraocular, intrathecal and intravenous.
51. The method of claim 39, wherein the subject is further administered at least one additional antibacterial agent.
52. The method of claim 51 , wherein the at least one additional antibacterial agent is co-administered with the compound to the subject.
53. The method of claim 52, wherein the at least one additional antibacterial agent and the compound are coformulated.
54. The method of claim 51, wherein the at least one compound and the at least one additional antibacterial agent are synergistic.
55. The method of claim 54, wherein the FICI determined by the combination of the at least one compound and the at least one additional antibacterial agent is equal to or lower than about 0.50.
56. The method of claim 51 , wherein the at least one additional antibacterial agent comprises a lipoprotein antibiotic.
57. The method of claim 56, wherein the at least one additional antibacterial agent comprises a polymyxin.
58. The method of claim 51, wherein the at least one compound comprises at least one selected from the group consisting of 5, 25, and B55-B58.
59. The method of claim 51, wherein the subject is administered the at least one additional antibacterial agent at a lower dose or frequency as compared to the administering of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
60. The method of claim 51 , wherein the subject develops a lower or slower rate of resistance to the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
61. The method of claim 51, wherein the subject develops lower toxicity from the at least one additional antibacterial agent as compared to the administration of the at least one additional antibacterial agent alone that is required to achieve similar results in treating or preventing the bacterial infection in a subject in need thereof.
62. The method of claim 39, wherein the subject is a mammal.
63. The method of claim 62, wherein the mammal is human.
PCT/US2015/060315 2014-11-12 2015-11-12 Novel anti-infective compounds and methods using same WO2016077541A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462078726P 2014-11-12 2014-11-12
US62/078,726 2014-11-12

Publications (1)

Publication Number Publication Date
WO2016077541A1 true WO2016077541A1 (en) 2016-05-19

Family

ID=55955030

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/060315 WO2016077541A1 (en) 2014-11-12 2015-11-12 Novel anti-infective compounds and methods using same

Country Status (1)

Country Link
WO (1) WO2016077541A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109503495A (en) * 2018-11-21 2019-03-22 徐州医科大学 A kind of 2- imines benzimidazoles compound and medical usage
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
CN114656509A (en) * 2022-04-08 2022-06-24 国科温州研究院(温州生物材料与工程研究所) N-heterocyclic carbene-gold palladium alloy nanoparticle and preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020061914A1 (en) * 1999-05-12 2002-05-23 Jensen Bo Skaaning Ion channel modulating agents
US20050187390A1 (en) * 2003-08-01 2005-08-25 Genelabs Technologies, Inc. Bicyclic heteroaryl derivatives
US20050228022A1 (en) * 2002-06-17 2005-10-13 Taktix Corporation Guanidine compounds as anesthetics and for treatment of nervous system disorders
US20080200529A1 (en) * 2005-01-11 2008-08-21 Neuro Search A/S Novel 2-Amino Benzimidazole Derivatives and Their Use As Modulators Of Small-Conductance Calcium-Activated Potassium Channels
US20090074839A1 (en) * 1997-01-22 2009-03-19 Marton Milankovits Pharmaceutical Compositions Primarily for the Treatment and Prevention of Genitourinary Infections and their Extragenital Complications

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090074839A1 (en) * 1997-01-22 2009-03-19 Marton Milankovits Pharmaceutical Compositions Primarily for the Treatment and Prevention of Genitourinary Infections and their Extragenital Complications
US20020061914A1 (en) * 1999-05-12 2002-05-23 Jensen Bo Skaaning Ion channel modulating agents
US20050228022A1 (en) * 2002-06-17 2005-10-13 Taktix Corporation Guanidine compounds as anesthetics and for treatment of nervous system disorders
US20050187390A1 (en) * 2003-08-01 2005-08-25 Genelabs Technologies, Inc. Bicyclic heteroaryl derivatives
US20080200529A1 (en) * 2005-01-11 2008-08-21 Neuro Search A/S Novel 2-Amino Benzimidazole Derivatives and Their Use As Modulators Of Small-Conductance Calcium-Activated Potassium Channels

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ZAVASCKI, AP ET AL.: "Polymyxin B for the treatment of multidrug-resistant pathogens: a critical review", JOURNAL OF ANTIMICROBIAL CHEMOTHERAPY, vol. 60, 2007, pages 1206 - 1215 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
CN109503495A (en) * 2018-11-21 2019-03-22 徐州医科大学 A kind of 2- imines benzimidazoles compound and medical usage
CN114656509A (en) * 2022-04-08 2022-06-24 国科温州研究院(温州生物材料与工程研究所) N-heterocyclic carbene-gold palladium alloy nanoparticle and preparation method and application thereof

Similar Documents

Publication Publication Date Title
EP2114903B1 (en) Bicyclic pyrimidinones and uses thereof
US8999991B2 (en) Inhibitors of the renal outer medullary potassium channel
WO2019042445A1 (en) Compound having bruton&#39;s tyrosine kinase (btk)-inhibition and degradation activity
AU2013358591B2 (en) Pyrimido [4,5-b]quinoline-4,5 (3H,10H)-diones as nonsense mutation suppressors
WO2009022730A1 (en) P2x4 receptor antagonist
JP2008519083A (en) Aminoquinazoline compounds
WO2016077541A1 (en) Novel anti-infective compounds and methods using same
AU2018326721B2 (en) Compounds and compositions for IRE1 inhibition
EP3307715A1 (en) Aromatic sulfonamide derivatives
JP2013532657A (en) Cyclic N, N&#39;-diarylthiourea and N, N&#39;-diarylurea-androgen receptor antagonists, anticancer agents, methods for their preparation and uses
US8426405B2 (en) Thiazopyrimidinones and uses thereof
WO2019052535A1 (en) Compound for inhibiting and degrading cdk
BR112020001327A2 (en) chemical compounds
JP2018138609A (en) Naphthyridinedione derivatives
US9879002B2 (en) PDE10 inhibitors and related compositions and methods
CN100526316C (en) Triazole miazines derivative
WO2004018485A1 (en) Azole derivatives as antifungal agents
US20220387401A1 (en) Estrogen-related receptor alpha modulators
CN109963853B (en) Compound with activity of degrading tyrosine protein kinase JAK3
WO2019042442A1 (en) Compound having tyrosine protein kinase jak1- or jak2-inhibittion and degradation activity
EP3572411A1 (en) Thiazole derivatives as metallo-beta-lactamase inhibitors
JP2023538255A (en) Urea derivatives as pyruvate kinase activating compounds
CN108976227B (en) Benzothiazine-4-ketone compound containing basic bridged ring segment and preparation method thereof
US20150320733A1 (en) Indazole compounds as aldosterone synthase inhibitors
AU2016304331A1 (en) Method of treating cancer with a combination of benzylideneguanidine derivatives and chemotherapeutic agent.

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: 15859524

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: 15859524

Country of ref document: EP

Kind code of ref document: A1