WO2009023160A2 - Novel inhibitors of bacterial sortase enzymes and methods of using the same - Google Patents

Novel inhibitors of bacterial sortase enzymes and methods of using the same Download PDF

Info

Publication number
WO2009023160A2
WO2009023160A2 PCT/US2008/009577 US2008009577W WO2009023160A2 WO 2009023160 A2 WO2009023160 A2 WO 2009023160A2 US 2008009577 W US2008009577 W US 2008009577W WO 2009023160 A2 WO2009023160 A2 WO 2009023160A2
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
group
alkenyl
pharmaceutically acceptable
Prior art date
Application number
PCT/US2008/009577
Other languages
French (fr)
Other versions
WO2009023160A3 (en
WO2009023160A8 (en
Inventor
Sadanandan E. Velu
Sthanam V.L. Narayana
Aaron L. Lucias
Original Assignee
The Uab Research Foundation
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 Uab Research Foundation filed Critical The Uab Research Foundation
Publication of WO2009023160A2 publication Critical patent/WO2009023160A2/en
Publication of WO2009023160A3 publication Critical patent/WO2009023160A3/en
Publication of WO2009023160A8 publication Critical patent/WO2009023160A8/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/04Nitro compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/443Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5355Non-condensed oxazines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present disclosure relates generally to inhibitors of bacterial virulence and infectivity.
  • the present disclosure relates to compounds having the formula I (including compounds having the formula Ia, Ib and Ic) through XII and the identification of such compounds as inhibitors of bacterial sortase enzymes.
  • the present disclosure also relates to methods of using the compounds disclose herein, pharmaceutical compositions comprising the compounds disclosed herein and methods for producing the compounds disclosed herein.
  • Gram-positive bacteria capable of causing serious or fatal infections in humans include, but are not limited to, Staphylococcus, Streptococcus, Enterococcus, Bacillus, and Listeria. Infections caused by these pathogens are particularly severe and difficult to treat in immunologically compromised patients and patients with pre-existing diseases and/or conditions. These include patients suffering from infection with the Human Immunodeficiency Virus (HIV), the virus that causes AIDS, as well as patients given immune-suppressive agents for treatment of cancer or autoimmune diseases. Gram-negative bacteria can also give rise to serious or fatal infections in humans.
  • HIV Human Immunodeficiency Virus
  • a unique characteristic of bacterial pathogens including Gram-positive and Gram- negative bacteria, is their surface display of proteins anchored to the cell wall. In fact, many of these molecules are known to be involved in increasing bacterial virulence which mediates the ability of bacterial cells to infect cells in a susceptible host. Thus, a possible disruption in this anchoring process of these surface proteins may prove to be an effective treatment against bacteria infection and a mechanism to decrease bacterial virulence.
  • Staphylococci are responsible for more than a million hospital-acquired bacterial infections every year. In addition, they are to blame for a number of minor infections ranging from pustules to boils in regular day- to-day life, and for a significant number of serious infections like endocarditis, osteomyelitis, and septic arthritis (Petti, et al., Cardiol Clin 2003, 21, (2), 219-33, vii; Lobati, et al., Infection 2001, 29, (6), 333-6; Ish-Horowicz, et al., Pediatr Infect Dis J 1992, 11, (2), 82-7).
  • Streptococcus pneumoniae is responsible for a major portion of 3 million deaths worldwide of children from pneumonia and meningitis and for a large number of deaths from pneumonia that occurs in elderly individuals. They are also responsible for a major percentage of ear infections, sinusitis, and additional severe invasive infections in asplenic persons and those with sickle cell disease. Compounding their importance is the recent steep increase in multi-drug resistance found in bacteria and the potential of 'drug resistant bacteria' to be used in biowarfare and bioterrorism (Pappas, et al., Infect Dis Clin North Am 2006, 20, (2), 395-421, x; Clarke, et al., Br J Biomed Sci 2005, 62, (1), 40-6). Hospital strains of S.
  • aureus are usually resistant to a variety of different antibiotics. A few strains are resistant to all clinically useful antibiotics except vancomycin, and vancomycin-resistant strains are increasingly reported (Mohr, et al., Clin Infect Dis 2007, 44, (12), 1536-42; Webster, et al., Diagn Microbiol Infect Dis 2007, 57, (2), 177-81; Weigel, et al., Antimicrob Agents Chemother 2007, 51, (1), 231-8). Methicillin resistant S. aureus (MRSA) is widespread and most methicillin-resistant bacterial strains are also resistant against multiple antibiotics, necessitating the need for identifying new therapeutic agents (Baggett, et al.
  • MRSA Methicillin resistant S. aureus
  • Bacterial infections are very complex and involve the action of a large sophisticated arsenal of virulence factors, many of which are surface-bound or secreted. Surface proteins often fall into one of four functional categories: microbial adhesion to host tissues, protection from host defense mechanisms, acquisition of nutrients for bacterial growth, and secretion of toxins and invasins. These virulence factors carry out important roles in the infection process.
  • the possibility of human interventions interrupting this process through interference with virulence is a possibility, but not yet a reality.
  • Gram-positive bacteria such as, but not limited to, Staphylococci
  • Bacterial sortases are cysteine transpeptidases that participate in secretion and anchoring of many cell wall proteins by a mechanism highly conserved in almost the entire class of Gram-positive bacteria. Sortases and sortase homologs have also been identified in certain Gram-negative bacteria and appear to use the same mechanism. Sortases, because of their control over the cellular location of multiple virulence factors, is an attractive potential target for interrupting bacterial virulence and thereby reducing the infectivity of bacteria towards a host.
  • Proteins that are covalently attached to the cell wall share conserved regions known as the "sorting signal” or cell wall anchors (Fischetti, V. et al., MoI Microbiol 1990, 4, (9), 1603-5; 15; Schneewind, et al., Cell 1992, 70, (2), 267-81).
  • Sortases from Gram- positive bacteria are currently grouped into four, sometimes five, families designated Sortase (Srt) A, SrtB, SrtC (Family 3), and SrtD (Families 4 and 5), with each family recognizing a slightly different sorting signal.
  • the sortases found in gram-negative bacteria have been grouped into another separate family (Family 6).
  • the sorting signal for SrtA includes a conserved amino acid motif, usually LPXaaTG (SEQ ID NO: 1), where Xaa can.
  • sorting signal for SrtB includes the conserved amino acid motif NPQTXaaN (SEQ ID NO: 2), where Xaa can be any one of the naturally occurring L- amino acids. Additional sorting signals are also known to be present in Gram-positive bacteria.
  • precursor proteins are directed into a secretory pathway by their N-terminal signal peptides (Schneewind, et al., Cell 1992, 70, (2), 267-81; Navarre, et al., MoI Microbiol 1994, 14, (1), 115-21; Navarre, et al.,.
  • the enzyme sortase acts at this point to cleave the protein at a point in the sorting signal (for example, SrtA cleaves between the threonine (T) and the glycine (G) of SEQ ID NO: 1) (Navarre, et al., MoI Microbiol 1994, 14, (1), 115-21).
  • the end residue of the sorting signal is then covalently linked to a cell wall component of the bacteria.
  • the bacterial cell wall target can vary depending on the bacterial species and the sortase involved.
  • sortase is an enzyme that recognizes a specialized cell wall "sorting signals" in proteins destined for surface attachment and covalently links them to the bacterial cell wall (Navarre, et al., Microbiol MoI Biol Rev 1999, 63, (1), 174-229). Sortase-defective strains of various pathogens were shown to be faulty in the display of surface proteins and are less virulent (Jonsson, et al., Microbes Infect 2003, 5, (9), 775-80; Mazmanian, et al., Proc N ⁇ tl Ac ⁇ d Sci US A 2000, 97, (10), 5510-5).
  • SrtA and SrtB Two sortases from Staphylococcus aureus, SrtA and SrtB have been cloned, sequenced, expressed and purified (Mazmanian, et al., Science 1999, 285, (5428), 760-3; Mazmanian, et al., Proc Natl Acad Sci U S A 2002, 99, (4), 2293-8).
  • S. aureus SrtA is the best-characterized enzyme from the family of bacterial sortases.
  • SrtA is a 206-amino acid protein with a cysteine residue at position 184, which was shown to be critical for its function (Ton-That, et al, Proc Natl Acad Sci USA 1999, 96, (22), 12424-9).
  • SrtB exhibiting 23% primary sequence homology with SrtA, is the second identified S. aureus sortase.
  • SrtA is responsible for anchoring all LPXTG- containing surface proteins including CIfA, CIfB, FnbpA, FnbpB, SdrC, and Cna.
  • the amino acid sequence of SrtA (SEQ ID NO: 3) and SrtB (SEQ ID NO: 4) are shown in FIGS. IB and 1C, respectively. Despite an overall homology of 20-30% among different bacterial sortases, the homology in the active site regions is significantly higher.
  • All sortases share various degrees of homology to the archetypal sortase, SrtA of S. aureus; however, all known SrtA homologs (all sortases) contain structurally conserved active- site residues of histidine, arginine, and cysteine (SrtA residues Hisl20, Argl97, and Cysl84) (Marraffmi, et al., Microbiol MoI Biol Rev 2006, 70, (1), 192-221). This absolute conservation includes the SrtB family, which has the most divergent consensus sorting signal of all the sortase families (Maresso, et al., J Bacteriol 2006, 188, (23), 8145-52).
  • aureus greatly impaired transpeptidation activity suggests that compounds interacting with or blocking access to the Argl97 residue (or appropriate arginine residue in the SrtA homologs) would inhibit the enzymatic activity of any SrtA homolog.
  • Sortases are virtually universal among Gram-positive bacteria. However, some examples are also found in limited number of archaeal and Gram-negative bacteria (Pallen, et al., Trends Microbiol 2001, 9, (3), 97-102).
  • sortases Due to their universal presence in Gram-positive bacteria and other bacterial species, sortases are attractive pharmacotherapeutic targets (Cossart, et al., Proc Natl Acad Sci U S A 2000, 97, (10), 5013-5).
  • the sorting signal of proteins (SEQ ID NO: 1) recognized by the main sortase is nearly identical among Gram-positive bacteria (Pallen, et al., Trends Microbiol 2001, 9, (3), 97-102; Comfort, et al., Infect Immun 2004, 72, (5), 2710-22).
  • virulence factors such as, but not limited to, surface proteins
  • altering the targeting and/or distribution of cell surface proteins on the cell wall offers a novel strategy against bacterial infections.
  • sortase inhibitors have the potential to be powerful therapeutic agents when used in conjunction with existing traditional antibiotics as a combination therapy since these function by a complementary mechanism.
  • the present disclosure described the identification of several new low micromolar sortase inhibitors identified through structure based inhibitor design studies.
  • the present disclosure also described pharmaceutical compositions containing these inhibitors methods of using the identified inhibitors and pharmaceutical compositions.
  • FIG. IA illustrates the reaction catalyzed by the S. aureus Sortase A.
  • FIG. IB shows the amino acid sequence of the Sortase A enzyme from S. aureus.
  • FIG. 1C shows the amino acid sequence of the Sortase B enzyme from S. aureus.
  • FIG. 2 shows the structure of compound 1.
  • FIG. 3 shows a FlexX model of compound 1 in the SrtA ⁇ sg active site.
  • FIG. 4 shows the interactions of compound 1 with amino acid residues in the SrtA ⁇ 59 active site.
  • FIG. 5 shows an exemplary synthesis of compounds 1, 13, 12a and 12b.
  • FIG. 6 shows an exemplary synthesis of compounds 14, 15, 16 and 17.
  • FIG. 7 shows an exemplary synthesis of compounds 18a, 18b, 19a and 19b.
  • FIG. 8 shows an exemplary synthesis of compound 20.
  • FIG. 9 shows an exemplary synthesis of compounds 21-24.
  • FIG. 10 shows an exemplary synthesis of compounds 25-28.
  • FIG. 11 shows an exemplary synthesis of compounds 29-30.
  • FIG. 12 shows an exemplary synthesis of compounds 31-34.
  • FIG. 13 shows an exemplary synthesis of compounds 43-46.
  • FIG. 14 shows an exemplary synthesis of compounds 51-56.
  • FIG. 15 shows an exemplary synthesis of compound 57.
  • alkyl includes straight hydrocarbon groups comprising from one to twenty carbon atoms.
  • straight chain alkyl groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • the phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -CH(CHj) 2 , -CH(CH 3 )(CH 2 CH 3 ), -CH(CH 2 CH 3 ) 2 , -C(CH 3 ) 3 , - C(CH 2 CH 3 ) 3 , -CH 2 CH(CH 3 ) 2) -CH 2 CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH(CH 2 CH 3 );, -CH 2 C(CH 3 ) 3 , - CH 2 C(CH 2 CH 3 );,-, -CH(CH 3 )CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH 2 CH(CH 3 ) 2 , CH 2 CH 2 CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH 2 CH(CH 2 CH 3 ) Z , -CH 2 CH 2 C(CH 3 ) 3 ,
  • the phrase also includes cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • the phrase also includes polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • alkenyl whether used alone or as part of a substituent group, includes an alkyl group having at least one double bond between any two adjacent carbon atoms.
  • alkynyl whether used alone or as part of a substituent group, includes an alkyl group having at least one triple bond between any two adjacent carbon atoms.
  • unsubstituted alkyl refers to alkyl, alkenyl and alkynyl groups that do not contain heteroatoms.
  • substituted alkyl refers to alkyl, alkenyl and alkynyl groups as defined above in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen or non-carbon atoms such as, but not limited to, a halogen atom, such as F, Cl, Br, and I; and oxygen atom in groups such as carbonyl, carboxyl, hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamine
  • alkyl groups include those in which one or more bonds to a carbon or hydrogen atom is replaced by a bond to an oxygen atom such that the substituted alkyl group contains a hydroxyl, alkoxy, aryloxy group, or heterocyclyloxy group.
  • Still other alkyl groups include alkyl groups that have an amine, alkylamine, dialkylamine, arylamine, (alkyl)(aryl)amine, diarylamine, heterocyclylamine, (alkyl)(heterocyclyl)amine, (aryl)(heterocyclyl)amine, or diheterocyclylamine group.
  • unsubstituted aryl refers to monocyclic or bicyclic aromatic hydrocarbon groups having 6 to 12 carbon atoms in the ring portion, such as, but not limited to, phenyl, naphthyl, anthracenyl, biphenyl and diphenyl groups, that do not contain heteroatoms.
  • unsubstituted aryl includes groups containing condensed rings such as naphthalene, it does not include aryl groups that have other groups such as alkyl or halogen groups bonded to one of the ring members, as aryl groups such as tolyl are considered herein to be substituted aryl groups as described below.
  • Unsubstituted aryl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
  • substituted aryl group has the same meaning with respect to unsubstituted aryl groups that substituted alkyl groups had with respect to unsubstituted alkyl groups.
  • a substituted aryl group also includes aryl groups in which one of the aromatic carbons is bonded to one of the non-carbon or non-hydrogen atoms described above and also includes aryl groups in which one or more aromatic carbons of the aryl group is bonded to a substituted and/or unsubstituted alkyl, alkenyl, or alkynyl group as defined herein.
  • a fused ring system e.g. dihydronaphthyl or tetrahydronaphthyl.
  • substituted aryl includes, but is not limited to tolyl, and hydroxyphenyl among others.
  • Substituted aryl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
  • unsubstituted aralkyl refers to unsubstituted alkyl, alkenyl or alkyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkyl, alkenyl or alkyl group is replaced with a bond to an unsubstituted or substituted aryl group as defined above.
  • methyl (CH 3 ) is an unsubstituted alkyl group.
  • a hydrogen atom of the methyl group is replaced by a bond to a phenyl group, such as if the carbon of the methyl were bonded to a carbon of benzene, then the compound is an unsubstituted aralkyl group (i.e., a benzyl group).
  • Unsubstituted aralkyl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
  • substituted aralkyl has the same meaning with respect to unsubstituted aralkyl groups that substituted aryl groups had with respect to unsubstituted aryl groups.
  • a substituted aralkyl group also includes groups in which a carbon or hydrogen bond of the alkyl, alkenyl or alkyl part of the group is replaced by a bond to a non-carbon or a non-hydrogen atom.
  • unsubstituted heterocyclyl refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S.
  • unsubstituted heterocyclyl includes condensed heterocyclic rings such as benzimidazolyl, it does not include heterocyclyl groups that have other groups such as alkyl or halogen groups bonded to one of the ring members, as compounds such as 2-methylbenzimidazolyl are "substituted heterocyclyl" groups as defined below.
  • heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl; saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazoly
  • unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g.
  • 1,3-benzodioxoyl, etc. unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1 ,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing a sulfur atom and 1 to 2 oxygen atoms such as benzoxathiinyl.
  • unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl
  • saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1 ,4-oxathiane
  • Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones).
  • heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1 -dioxide.
  • Preferred heterocyclyl groups contain 5 or 6 ring members.
  • substituted heterocyclyl refers to an unsubstituted heterocyclyl group as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples, include, but are not limited to, 2-methylbenzimidazolyl, 5-methylbenzimidazolyl, 5- chlorobenzthiazolyl, 1 -methyl piperazinyl, and 2-chloropyridyl among others.
  • unsubstituted heterocyclylalkyl refers to unsubstituted alkyl, alkenyl or alkynyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkyl, alkenyl or alkynyl group is replaced with a bond to a substituted or unsubstituted heterocyclyl group as defined above.
  • methyl (-CH 3 ) is an unsubstituted alkyl group.
  • a hydrogen atom of the methyl group is replaced by a bond to a substituted or unsubstituted heterocyclyl group, such as if the carbon of the methyl were bonded to carbon 2 of pyridine (one of the carbons bonded to the N of the pyridine) or carbons 3 or 4 of the pyridine, then the compound is an unsubstituted heterocyclylalkyl group.
  • substituted heterocyclylalkyl refers to substituted alkyl, alkenyl or alkynyl groups as defined above in which a hydrogen or carbon bond of the substituted alkyl, alkenyl or alkynyl group is replaced with a bond to a substituted or unsubstituted heterocyclyl group as defined above.
  • a substituted heterocyclylalkyl group also includes groups in which a non-hydrogen atom is bonded to a heteroatom in the heterocyclyl group of the heterocyclylalkyl group such as, but not limited to, a nitrogen atom in the piperidine ring of a piperidinylalkyl group.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • pharmaceutically acceptable salts is v meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, oxalic, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galacrunoric acids and the like (see, for example, Berge, S. M., et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • prodrug is meant to include functional derivatives of the compounds disclosed which are readily convertible in vivo into the required compound.
  • administering shall encompass the treatment of the various disease states/conditions described with the compound specifically disclosed or with a prodrug which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs”, ed. H. Bundgaard, Elsevier, 1985.
  • suppression refers to administering a compound prior to the onset of clinical symptoms of a disease state/condition so as to prevent any symptom, aspect or characteristic of the disease state/condition. Such preventing and suppressing need not be absolute to be useful.
  • treat refers to administering a compound after the onset of clinical symptoms of a disease state/condition so as to reduce or eliminate any symptom, aspect or characteristic of the disease state/condition. Such treating need not be absolute to be useful.
  • in need of treatment refers to a judgment made by a caregiver that a patient requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, and may include the knowledge that the patient is ill as the result of a disease state/condition that is treatable by a compound or pharmaceutical composition of the disclosure.
  • in need of prevention refers to a judgment made by a caregiver that a patient requires or will benefit from prevention. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, and may include the knowledge that the patient may become ill as the result of a disease state/condition that is treatable by a compound or pharmaceutical composition of the disclosure.
  • mice or “patient” as used herein refers to any animal, including mammals, such as, but not limited to, mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, or humans.
  • mammals such as, but not limited to, mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, or humans.
  • the term may specify male or female or both, or exclude male or female; and
  • a therapeutically effective amount in reference to the treating, preventing or suppressing of a disease state/condition, refers to an amount of a compound either alone or as contained in a pharmaceutical composition that is capable of having any detectable, positive effect on any symptom, aspect, or characteristics of the disease state/condition.
  • a therapeutically effective amount is a tumor growth inhibiting amount. Such effect need not be absolute to be beneficial.
  • Summary of the Disclosure provides compounds of the general formula I (including compounds having the formula Ia, Ib and Ic) through XII, specifically compounds 1-8, 12-34 and 43-67. In one embodiment, these compounds are useful as inhibitors of bacterial sortase enzymes.
  • these compounds are useful as inhibitors of bacterial SrtA, SrtB, SrtC and/or SrtD, or homologs thereof.
  • the compounds are useful as inhibitors of bacterial SrtA.
  • the compounds of the present disclosure have utility in treating and/or preventing a variety of disease states and/or conditions involving bacterial infection and virulence, wherein said bacteria causing or contributing to the bacteria infection and virulence express a sortase enzyme.
  • the present disclosure provides a method of treating a disease state and/or condition in a subject which is caused by, at least in part, bacterial infection and virulence, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
  • the present disclosure provides a method of preventing or suppressing a disease state and/or condition in a subject which is caused by, at least in part, bacterial infection and virulence, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
  • the present disclosure provides for a method of reducing or eliminating bacterial virulence in a subject, the method comprising the administration to said subject a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
  • the present disclosure provides for methods of treating bacterial infection in a subject, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
  • the present disclosure provides for methods of preventing or suppressing bacterial infection in a subject, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
  • This present disclosure provides compounds of the general formula I (including compounds having the formula Ia, Ib and Ic) through XII and pharmaceutical compositions comprising the same or pharmaceutically acceptable salts thereof, or esters thereof, or prodrugs thereof and tautomers and polymorphic variants of any of the foregoing.
  • the compounds disclosed are useful as inhibitors of bacterial sortase enzymes.
  • Ring X is optional and when present is selected from substituted or unsubstituted aryl or substituted or unsubstituted heterocycle;
  • B is selected from S or O or NRi, where Ri is H or a substituted or unsubstituted alkyl; Li and L 2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and
  • L in Li and L 2 is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
  • the linking group I is a C 2 to C 5 substituted or unsubstituted alkyl, C 2 to C 5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C 2 to C 5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C 2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
  • ring Z is a substituted or unsubstituted 5 membered ring, including but not limited to, heterocylic compounds containing oxygen (such as furan), sulfur
  • A is selected from the group consisting of: S, CH 2 , O and N;
  • B is selected from the group consisting of: S, O and NRj, where R 1 is H or a substituted or unsubstituted alkyl;
  • E is selected from the group consisting of: CH 2 , O and N;
  • Li and L 2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and L in Li and L 2 is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
  • the linking group I is a C 2 to C 5 substituted or unsubstituted alkyl, C 2 to C 5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C 2 to C 5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C 2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
  • R 2 through R 5 are each independently selected from the group consisting of: H, OH, CORn, COORi 4 , substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C 7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl.
  • R 6 through R 8 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl.
  • Ri 3 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi 5 Ri 6 ;
  • Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl.
  • Ri 5 and Ri 6 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl.
  • R 2 through R 5 are each independently selected from the group consisting of: H, COR J3 , COOR H and substituted or unsubstituted alkyl. In a further specific embodiment, R 2 through R 5 are each independently selected from the group consisting of: COOCH 3 , COOH, CHO, CH 2 OH and CONH 2 .
  • R 2 is selected from the group consisting of: H, COR1 3 ,
  • A is S
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 1.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • Lj and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 12a.
  • A is O
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • L 1 and L 2 are not present (i.e., 1 and 2 each equal 0)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 12b.
  • A is O
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 13.
  • A is S
  • B is NH
  • E is O
  • R 2 is CH 2 OH
  • R 3 through R 8 are H
  • L 1 and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 18a.
  • A is O
  • B is NH
  • E is O
  • R 2 is CH 2 OH
  • R 3 through R 8 are H
  • L 1 and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 18b.
  • A is S
  • B is NH
  • E is O
  • R 2 is CHO
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal 0)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 19a.
  • A is O
  • B is NH
  • E is O
  • R 2 is CHO
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 19b.
  • A is S
  • B is NH
  • E is O
  • R 2 is CONH 2
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 20.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkynyl and the compound has the structure of compound 21.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal 0)
  • the linking group I is a C 2 alkynyl and the compound has the structure of compound 22.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • L 1 and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the cis configuration and the compound has the structure of compound 23.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 8 are H
  • L 1 and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the cis configuration and the compound has the structure of compound 24.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 8 are H
  • Lj and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 29.
  • A is S
  • B is NH
  • E is CH 2
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal 0)
  • the linking group I is a C 3 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 31.
  • A is O
  • B is NH
  • E is CH 2
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • Lj and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 32.
  • A is S
  • B is NH
  • E is CH 2
  • R 2 is COOH
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 33.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOCH 2 CH 3
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 43.
  • A is O
  • B is NH
  • E is O
  • R 2 is COOCH 2 -C 6 Hi 2
  • R 3 through R 8 are H
  • Lj and L 2 are not present (i.e., 1 and 2 each equal 0)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 44.
  • A is O
  • B is NH
  • E is O
  • R 2 is COOCH 2 CH 2 CH 3
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 45.
  • A is O
  • B is NH
  • E is O
  • R 2 is COOCH 2 CH 2 CH 2 CH 3
  • R 3 through R 8 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 46.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • R 3 through R 8 are H
  • Li is not present (i.e., 1 is equal to O) and L 2 is CH 3
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 47.
  • A is S
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 8 are H
  • Li is not present (i.e., 1 is equal to O) and L 2 is CH 3
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 48.
  • A is selected from the group consisting of: S, CH 2 , O and N;
  • B is selected from the group consisting of: S, O and NRi, where Ri is H or a substituted or unsubstituted alkyl;
  • E is a heteroatom selected from the group consisting of: CH 2 , O and N;
  • F is a single, optional N heteroatom; when present, F may be in a para, meta or ortho position with respect to the point of attachment;
  • Li and L 2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and
  • L in Li and L 2 is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
  • the linking group I is a C 2 to C 5 substituted or unsubstituted alkyl, C 2 to C 5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C 2 to C 5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C 2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
  • R 2 through R 5 are each independently selected from the group consisting of: H, OH, CORn, COOR 15 , substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C 7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
  • R 9 through R) 3 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl; provided that when F is present at a given position (i.e., para, meta or ortho position), one of the R 9 through Ri 3 substituent groups will be absent;
  • R 14 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR 16 R 17 ;
  • R 15 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl;
  • Rj 6 and Ri 7 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl.
  • R 2 through R 5 are each independently selected from the group consisting of: H, CORi 4 , COOR 15 and substituted or unsubstituted alkyl.
  • R 2 through R 8 are each independently selected from the group consisting of:
  • R 2 is selected from the group consisting of: H, CORi 4 ,
  • R 2 is selected from the group consisting of: COOCH 3 , COOH, CHO, CH 2 OH and CONH 2 , while R 3 through R 5 and
  • Ri 2 and Ri 3 are H, Rn is absent, Lj and L 2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 51.
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • R 3 through R 5 and R 9 through R 13 are H
  • Li and L 2 are not present (i.e., 1 and 2 each equal 0)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 52.
  • F is N and is in the meta position
  • B is NH
  • E is O
  • R 2 is COOCH 3
  • Rn, Ri 2 and Rj 3 are H
  • R 10 is absent
  • L 1 and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 55.
  • F is N and is in the meta position
  • B is NH
  • E is O
  • R 2 is COOH
  • R 3 through R 5 and R 9 Rn
  • Ri 2 and Rj 3 are H
  • Ri 0 is absent
  • Li and L 2 are not present (i.e., 1 and 2 each equal O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 56.
  • A is selected from the group consisting of: S, CH 2 , O and N;
  • B is selected from the group consisting of: S, O and NRj, where R 1 is H or a substituted or unsubstituted alkyl;
  • L 1 is an optional linking groups, where 1 is selected from 0 or 1 and L in L 1 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
  • the linking group I is a C 2 to C 5 substituted or unsubstituted alkyl, C 2 to C 5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C 2 to C 5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C 2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration; R 2 through R 6 are each independently selected from the group consisting of: H, OH, CORi 0 ,
  • R 7 through R 9 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl;
  • Ri 0 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR] 2 R] 3 ;
  • Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl;
  • Ri 2 and R] 3 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl; and Ri 4 and R 15 are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl and substituted or unsubstituted aralkyl.
  • R 2 through R 6 are each independently selected from the group consisting of: H, OH, CORio, COORn, NR J4 R I 5 and substituted or unsubstituted alkyl.
  • R 2 through R 6 are each independently selected from the group consisting of: NHCH 2 -Phe, NH-Phe, N(CH 2 -Phe) 2 , N(CH 2 CH 2 OH) 2 , COOCH 3 , COOH, CHO, CH 2 OH and CONH 2 .
  • A is S
  • B is NH
  • R 2 is NH-CH 2 -Ph
  • R 3 is COOCH 3
  • R 4 through R 9 are H
  • Li is not present (i.e., 1 equals O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 58.
  • A is S
  • B is NH
  • R 2 is NH-Ph
  • R 3 is COOCH 3
  • R 4 through R 9 are H
  • Li is not present (i.e., 1 equals O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 60.
  • A is S
  • B is NH
  • R 2 is N-(CH 2 -Ph) 2
  • R 3 is COOCH 3
  • R 4 through R 9 are H
  • L 1 is not present (i.e., 1 equals 0)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 61.
  • A is S
  • B is NH
  • R 2 is N-(CH 2 -Ph) 2
  • R 3 is COOH
  • R 4 through R 9 are H
  • Li is not present (i.e., 1 equals O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 62.
  • A is S
  • B is NH
  • R 2 is N-(CH 2 CH 2 OH) 2
  • R 3 is COOH
  • R 4 through R 9 are H
  • L is not present (i.e., 1 equals O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 63.
  • A is S
  • B is NH
  • R 2 is N-(CH 2 -Phe) 2
  • R 3 through R 9 are H
  • Lj is not present (i.e., 1 equals O)
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 67.
  • A is selected from the group consisting of: S, CH 2 , O and NRi 8 , where Ri 8 is H or a substituted or unsubstituted alkyl
  • the linking group I is a C 2 to C 5 substituted or unsubstituted alkyl, C 2 to C 5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C 2 to C 5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C 2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
  • R 1 through R 9 are each independently selected from the group consisting of: H, OH, NRi 6 R 17 ,
  • CORi 2 COOR 13 , substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, C 1 to C 7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
  • R 10 and Rn are each independently selected from the group consisting of: H substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
  • R 12 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR 14 Rj 5 ;
  • Ri 3 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; Ri 4 and Rn are each independently selected from the group consisting of: H, O and substituted or unsubstituted alkyl.
  • Ri through R 9 are each independently selected from the group consisting of: H, NRi 6 Ri 7 , COR 12 , COORi 3 and substituted or unsubstituted alkyl.
  • Ri is NO 2 or NH 2 .
  • Ri is selected from the group consisting of: H, NRi 6 Ri 7 ,
  • Rn are CH 3 , A is O and I is a C 2 alkenyl.
  • Ri is NO 2 or NH 2 , while R 2 through R 9 are at H, Ri 0 through Ri 1 are CH 3 , A is O and I is a C 2 alkenyl.
  • A is O
  • Ri is NO 2
  • R 2 through R 9 are H
  • the linking group I is a C 2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 57.
  • FlexX is an ideal tool for screening large libraries of ligands to find leads for drug design.
  • FlexX has the feature of allowing conformational flexibility of the ligand in complementing the active site, when active site is used as a constraint, while docking.
  • FlexX allows complete specification of the receptor active site, including oxidation states, metal ions, side chain protonation states and automatic ligand positioning, while docking. FlexX allows the ability to dock and score small combinatorial or related libraries of compounds to identify lead structural scaffolds.
  • RDF receptor description file
  • a RDF for SrtA ⁇ 59 was created using the receptor PDB file, the "active site file” which is constituted of all residues within 6.5 A distance from the center of the active site and a "pocket file” which is composed of a few hand-picked residues that are directly interacting with the substrate peptide.
  • 2D compound data sets from commercial sources were transformed into three dimensional molecular structures using COMPARE program. All compounds were generated in the protonated state that can be assumed under physiological conditions. The following limits were applied for the virtual screening: molecular weight >150 ⁇ 500, ClogP ⁇ 5. In addition, compounds bearing metal ions were omitted from the data set.
  • Inhibitor 1 has a three-ring structure. The left end is a thiophene ring (ring A), the middle ring is phenyl ring (ring B) and the right end is a morpholine ring (ring C). Ring A and B are connected by an acrylamido linkage in which the double bond has trans stereochemistry. Middle ring B bears a carboxylic acid group at a position meta to the point of attachment of acrylamido linkage. Ring C is connected to the middle ring B at the ortho position to COOH group through the morpholine ring N atom.
  • FlexX docking model of inhibitor 1 revealed several interactions of the inhibitor within the active site ( Figure 4). Morpholine ring oxygen atom has a hydrogen bonding interaction with amide backbone NH of Trpl94.
  • the cysteine residue (Cysl84) is at 2.94A distance from the morpholine ring carbon atom next to the oxygen. If the morpholine ring is appropriately substituted with a hydrogen bond acceptor or donor the modification may result in additional hydrogen bonding interactions.
  • the carboxylic acid on the middle phenyl ring has a direct salt bridge interaction with the guanidine side chain of Argl97.
  • the amide NH group of the linker has a hydrogen bonding interaction with the carboxylic acid side chain of GIu 105 and the amide carbonyl oxygen of the linker has a hydrogen bonding interaction with the OH side chain of Serl l ⁇ .
  • the thiophene ring (ring A) of inhibitor 1 is in close proximity to Glnl72 (3.2A) and Asnl 14 (3.1 A). Appropriate substitution to the thiophene ring may result in additional hydrogen bonding interactions with Glnl72 and Asnl 14.
  • FlexX docking model of inhibitor 57 also revealed several interactions of the inhibitor within the active site.
  • the carbonyl group present in the molecule showed an interaction with an arginine residue (Arg 197) that is conserved in the active site of sortase family enzymes.
  • the nitro group present in the molecule showed critical interactions with several residues in the active site (Asp 170, GIu 171, GIn 172 and GIn 178). Furthermore, the nitrogen of dimethylamino group was found to be in close proximity to Serl 16 and Glul05.
  • SAR structure-activity relationship
  • Derivatives of compound 1 were synthesized in order to derive preliminary structure activity relationship data.
  • the activities of the derivatives against the activity of the SrtA ⁇ 5 9 protein were determined using the FRET assay as described. Structures and IC 50 values of the newly synthesized derivative compounds are given in Table 2.
  • Compound 13 is a furan analog of compound 1. Design of this structure was inspired by the comparison of activities of thiophene and furan compounds 3 and 6 (Table 1). These two compounds have the same structure except that compound 3 contained a furan ring and compound 6 contained a thiophene ring. Furan compound 3 is more than two fold more active than the thiophene compound, 6. Therefore, a furan substitution of the thiophene ring of compound 1 may increase its activity.
  • Compound 13 was synthesized by a similar procedure as described for the synthesis of compound 1 (Scheme 1, FIG. 5).
  • Compounds 14-17 were synthesized to evaluate the importance of the double bond present in compounds 1 and 13 for their inhibitory activity.
  • Compounds 14 and 16 are the saturated analogs of the carboxylic acid derivatives 1 and 13, respectively.
  • Compounds 15 and 17 are the saturated analogs of the methyl ester derivatives 12a and 12b, respectively.
  • the SrtA ⁇ 59 protein assay showed that all four saturated compounds 14-17 were inactive up to a concentration of 600 ⁇ M, indicating that the double bond is necessary for the activity of the disclosed compounds.
  • Synthesis of compounds 14 - 17 is outlined in Scheme 2 (FIG. 6).
  • Derivative included the following substitutions: CH 2 OH (18b) and CHO (19b). These compounds showed improved inhibition as compared to parent furan compound 13. Compound 18b showed an ICs 0 value of 111 ⁇ M and compound 19b showed an IC 50 value of 107 ⁇ M.
  • Synthesis of compounds 18a, 18b, 19a and 19b is outlined in Scheme 3 (FIG. 7). Compounds 12a and 12b were reduced using DIBAL in a mixture of anhydrous dichloromethane and THF to afford the alcohol derivatives 18a and 18b. Oxidation of alcohols using PCC in anhydrous THF afforded the aldehydes 19a and 19b.
  • Synthesis of amide compound 20 is outlined in Scheme 4 (FIG. 8). Compound 20 was prepared from compound 1 by treatment with SOCl 2 followed by ammonia.
  • the acid 35 was coupled with the amino compound 36 in the presence of EDAC and DMAP in 1,2- dichloroethane afforded the alkyne ester 21.
  • the ester group present in compound 21 was hydrolyzed using NaOH in MeOH to afford the acetylenic acid 22.
  • the partial hydrogenation of the acetylenic ester 21 using Lindlar Pd catalyst did not work as expected possibly due to the catalytic poisoning effect of the thiophene ring already present in the molecule.
  • reduction worked smoothly to a cis double bonded product 23 when it was hydrogenated with H 2 in the presence of Pd/C in ethyl acetate.
  • the ester group present in compound 23 was hydrolyzed using NaOH in MeOH to afford the cis acid 24.
  • Synthesis is of compound 29-30 is outlined in Scheme 7 shown in FIG. 11. These compounds were designed to evaluate the nature of the linking group I on the activity of the compounds.
  • Compounds 29-30 contain a C3 alkenyl as the linking group as compared to a C 2 alkenyl linking group in the previous compounds. Generally, the compounds containing a C 3 alkenyl as the linking group I showed less activity than compounds containing a C 2 alkenyl as the linking group I.
  • Compounds 29 and 30 showed IC 50 values of about 250 ⁇ M and greater (Table 2). Reductive amination of the known aldehyde 39 with the amine 36 in the presence of NaCNBH 3 and ZnCl 2 in methanol afforded the ester compound 29. Basic hydrolysis of compound 29 using NaOH in MeOH afforded the final acid product 30.
  • Synthesis of compounds 51-56 is outlined in Scheme 10 shown in FIG. 14. These compounds were designed to evaluate substitution of the compounds at the position of the 5- membered (left most) ring on activity of the compounds.
  • Compounds 51-56 contain benzyl and piperidine in place of thiophene and furan groups generally present on the parent compounds. Generally, the compounds 51-56 showed similar IC 50 values to previous compounds.
  • Compounds 51-56 showed IC 50 values of 208 ⁇ M, 245 ⁇ M, >300 ⁇ M, n.d.,204 ⁇ M and 242 ⁇ M, respectively (Table 2).
  • Compounds 58, 60-63 and 67 are presented in Table 2. These compounds were generated as a result of the SAR studies discussed above. Compounds 58, 60-63 and 67 are represented by the general formula Ic and share structural similarity to the general formulas provided in structures Ia and Ib. Specifically, these compounds provide additional variation along the phenyl group to provide configurations similar to the morpholine and piperidine groups present in many of the previous compounds. Compounds 59, 64 and 66 also share similarity with the compounds discussed above in that they share a thiophene group linked to an N group via a C 2 -C 3 alkenyl linkage containing at least one double bond. Although compounds 58-64 and 66- 67 have not yet been tested against sortase enzymes in vitro, they can be expected to display inhibitory activity based on the similarity to the compounds disclosed herein. Pharmaceutical Compositions and Modes of Administration
  • the present disclosure provides compounds of the general formula (I), (Ia), (Ib), (Ic) and II-XII and pharmaceutical compositions comprising the same.
  • the present disclosure provides compounds 12-13, 18-20, 23-24, 31-32 and 43-46 and pharmaceutical compositions comprising the same.
  • these compounds are useful as inhibitors of bacterial sortase enzymes.
  • these compounds are useful as inhibitors of bacterial SrtA, SrtB, SrtC and/or SrtD or homologs thereof.
  • the compounds are useful as inhibitors of bacterial SrtA.
  • the compounds of the present disclosure have utility in treating and/or preventing a variety of disease states and/or conditions involving bacterial infection and virulence, wherein said bacteria causing or contributing to the bacteria infection and virulence express a sortase enzyme.
  • the present disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one compound of the present disclosure as described herein.
  • Such pharmaceutical compositions may be used in the manufacture of a medicament for treating and/or preventing a disease or condition in which it is desirable to inhibit involving bacterial infection and virulence.
  • Such pharmaceutical compositions and medicaments may also comprise a pharmaceutically acceptable carrier.
  • the compounds of the disclosure are useful in both free form and in the form of pharmaceutically acceptable salts.
  • the pharmaceutically acceptable carriers described herein including, but not limited to, vehicles, adjuvants, excipients, or diluents, are well-known to those who are skilled in the art.
  • the pharmaceutically acceptable carrier is chemically inert to the active compounds and has no detrimental side effects or toxicity under the conditions of use.
  • the pharmaceutically acceptable carriers can include polymers and polymer matrices.
  • the compounds described in the instant disclosure can be administered by any conventional method available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in combination with additional therapeutic agents.
  • the compounds described are administered in therapeutically effective amount.
  • the therapeutically effective amount of the compound and the dosage of the pharmaceutical composition administered will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration, the age, health and weight of the recipient; the severity and stage of the disease state or condition; the kind of concurrent treatment; the frequency of treatment; and the effect desired.
  • a daily dosage of active ingredient can be expected to be about 0.001 to 1000 milligrams (mg) per kilogram (kg) of body weight.
  • the total amount is between about 0.1 mg/kg and about 1000 mg/kg of body weight; in an alternate embodiment between about 1.0 mg/kg and about 100 mg/kg of body weight; in yet another alternate embodiment between 0.1 mg/kg and about 30 mg/kg of body weight.
  • the above described amounts may be administered as a series of smaller doses over a period of time if desired.
  • the dosage of active ingredient may be given other than daily if desired.
  • the total amount of the compound administered will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side effects that might accompany the administration of the compound and the desired physiological effect. It will be appreciated by one skilled in the art that various conditions or disease states, in particular chronic conditions or disease states, may require prolonged treatment involving multiple administrations.
  • Dosage forms of the pharmaceutical compositions described herein contain from about 0.1 mg to about 500 mg of active ingredient (i.e. the compounds disclosed) per unit.
  • the active ingredient will ordinarily be present in an amount of about 0.5-95% weight based on the total weight of the composition.
  • Multiple dosage forms may be administered as part of a single treatment.
  • the active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups and suspensions. It can also be administered parenterally, in sterile liquid dosage forms.
  • the active ingredient can also be administered intranasally (nose drops) or by inhalation via the pulmonary system, such as by nebulizers, propellant based metered dose inhalers or dry powders inhalation devices.
  • Other dosage forms are potentially possible such as administration transdermally, via a patch mechanism or an ointment.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as a pharmaceutically effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined pharmaceutically effective amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, propylene glycol, glycerin, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • diluents such as water and alcohols, for example, ethanol, benzyl alcohol, propylene glycol, glycerin, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch.
  • Tablet forms can include one or more of the following: lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acadia, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acadia, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti -oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the patient, and aqueous and non- aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the compound can be administered in a physiologically acceptable diluent in a pharmaceutically acceptable carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol such as poly(ethyleneglycol) 400, glycerol ketals, such as 2,2-dimethyl-l,3-dioxolane-4-methanol, ethers, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other
  • Oils which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyldialkylammonium halides, and alkylpyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl .beta.-aminopropionates, and 2-alkylimidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the parenteral formulations typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5% to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • HLB hydrophile-lipophile balance
  • compositions of the present invention are also well-known to those who are skilled in the art. The choice of excipient will be determined in part by the particular compound, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention. The following methods and excipients are merely exemplary and are in no way limiting.
  • the pharmaceutically acceptable excipients preferably do not interfere with the action of the active ingredients and do not cause adverse side-effects.
  • Suitable carriers and excipients include solvents such as water, alcohol, and propylene glycol, solid absorbants and diluents, surface active agents, suspending agent, tableting binders, lubricants, flavors, and coloring agents.
  • the compounds of the present disclosure can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, and nitrogen.
  • Such aerosol formulations may be administered by metered dose inhalers. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
  • the formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • sterile liquid excipient for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets.
  • the requirements for effective pharmaceutically acceptable carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J.B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, Eds., 238- 250 (1982) and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., 622-630 (1986).
  • Formulations suitable for topical administration include pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, as well as creams, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
  • an inert base such as gelatin and glycerin, or sucrose and acacia
  • formulations suitable for rectal administration may be presented as suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • teachings of the present disclosure provide for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in a method of treating a bacterial infection or a disease state and/or condition which is caused by, at least in part, bacterial infection and/or virulence.
  • disease states and conditions include, but are not limited to, hospital-acquired bacterial infections, endocarditis, osteomyelitis, septic arthritis pneumonia, toxic shock syndrome, skin infections, meningitis, anthrax, B.
  • cereus food poisoning tetanus, clostridial myonecrosis, pneumonia, toxic shock syndrome, skin infections, meningitis, ear infections, strep throat, rheumatic fever, scarlet fever, puerperal fever, listeriosis, perinatal septicemia, encephalitis, intrauterine infections sinusitis and severe invasive infections; however, other disease states and conditions may be treated as would be obvious to one of ordinary skill in the art.
  • the method of treatment comprises the steps of providing such composition or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection and/or virulence in a patient in need of such treatment.
  • One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure.
  • the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject and/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the disease state and/or condition which is caused by, at least in part, bacterial infection and/or virulence is thereby treated in the subject.
  • teachings of the present disclosure provide for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in a method of preventing or suppressing a bacterial infection or a disease state and/or condition which is caused by, at least in part, bacterial infection and/or virulence.
  • disease states and conditions include, but are not limited to, hospital- acquired bacterial infections, endocarditis, osteomyelitis, septic arthritis pneumonia, toxic shock syndrome, skin infections, meningitis, anthrax, B.
  • cereus food poisoning tetanus, clostridial myonecrosis, pneumonia, toxic shock syndrome, skin infections, meningitis, ear infections, strep throat, rheumatic fever, scarlet fever, puerperal fever, listeriosis, perinatal septicemia, encephalitis, intrauterine infections sinusitis and severe invasive infections; however, other disease states and conditions may be treated as would be obvious to one of ordinary skill in the art.
  • the method of preventing or suppressing comprises the steps of providing such compound or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection and/or virulence in a patient in need of such preventing or suppressing.
  • One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure.
  • the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject and/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the disease state and/or condition which is caused by, at least in part, bacterial infection and virulence is thereby prevented or suppressed in the subject.
  • the disease state or condition caused, at least in part, by a bacterial infection or virulence is endocarditis.
  • Endocarditis may be related infection of the endocardium and/or the heart valves by bacteria.
  • Staphylococcus aureus is a common cause of endocarditis.
  • the sortase enzyme such as, but not limited to SrtA
  • the ability of bacteria, such as, but not limited to, Staphylococcus aureus to bind to and infect the endocardium and/or heart valves may be reduced; furthermore, pre-existing bacterial infections may be eliminated.
  • the foregoing leads to a decrease in disease.
  • the present disclosure provides for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in methods of reducing or eliminating bacterial virulence in a subject in need of such reduction.
  • the method of reducing or eliminating comprises the steps of providing such compound or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to reduce or eliminate bacterial virulence in a patient in need of such reducing or eliminating.
  • One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure.
  • the present disclosure provides for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in methods of treating bacterial infection.
  • the method of treatment comprises the steps of providing such composition or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection in a patient in need of such treatment.
  • One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure.
  • the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject an/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the bacterial infection is thereby treated in the subject.
  • the present disclosure provides for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in methods of preventing or suppressing bacterial infection.
  • the method of preventing or suppressing comprises the steps of providing such compound or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection in a patient in need of such preventing or suppressing.
  • One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure.
  • the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject an/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the bacterial infection is thereby prevented or suppressed in the subject.
  • the bacteria may be any bacteria or other organism that contains and expresses a sortase enzyme, hi one embodiment, the sortase enzyme is SrtA, SrtB, SrtC, and/or SrtD, and/or homologues of any of the foregoing. In a specific embodiment, the sortase enzyme is SrtA. In another embodiment, the bacteria is Staphylococcus aureus.
  • the bacteria may one that is resistant to a commonly used antibiotic, such as methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococcus (VRE), vancomycin-intermediate Staphylococcus aureus (VISA) and vancomycin-resistant Staphylococcus aureus (VRSA).
  • MRSA methicillin-resistant Staphylococcus aureus
  • VRE vancomycin-resistant enterococcus
  • VISA vancomycin-intermediate Staphylococcus aureus
  • VRSA vancomycin-resistant Staphylococcus aureus
  • the bacteria may be Gram-positive bacteria, Gram-negative bacteria or combinations of the foregoing that expresses a sortase enzyme; in one embodiment, the bacteria is a Gram-positive bacteria that expresses the sortase enzyme.
  • Representative bacteria that have been documented to express a sortase enzyme include, but are not limited to, Acidothermus cellulolyticus, Actinomyces naeslundii, Actinomyces viscosus, Arthrobacter aurescens, Arthrobacter sp.
  • JS614 Oceanobacillus iheyensis, Pediococcus pentosaceus, Pseudoalteromonas atlantica, Pseudomonas mendocina, Rhodobacter sphaeroides, Rubrobacter xylanophilus, Saccharophagus degradans, Shewanella amazonensis, Shewanella baltica, Shewanella frigidimarina, Shewanella oneidensis, Shewanella sp. ANA-3, Shewanella sp. MR-4, Shewanella sp. MR-7, Shewanella sp.
  • Solibacter usitatus Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus haemolyticus, Staphylococcus saprophyticus, Streptococcus agalactiae, Streptococcus gordonii, Streptococcus mutans, Streptococcus oralis, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus sanguinis, Streptococcus suis, Streptococcus thermophilus, Streptomyces avermitilis, Streptomyces coelicolor, Symbiobacterium thermophilum, Thermobifida fusca and Tropheryma whipplei.
  • Sortase enzyme Representative other organisms that have been demonstrated to contain a sortase enzyme include, but are not limited to, Methanothermobacter thermautotrophicus. However, any bacteria or organism that expresses a sortase enzyme may be subject to the methods of the present disclosure.
  • the methods of use described herein may further comprise the administration of a therapeutically effective amount of one or more additional agents to further improve the efficacy of the methods described herein.
  • the additional agents may be administered at the same time or a different time than a compound of the present disclosure.
  • the additional agent has a mechanism of action that is distinct from the compounds of the present disclosure (i.e., the second compound is not an inhibitor of a sortase enzyme).
  • the identity and nature of the additional agent may be determined by the healthcare provider based on his/her knowledge of the subject's condition.
  • the additional agent is an antibiotic. Any antibiotic or antibiotic combinations may be used as the additional agent. As the compounds of the present disclosure operate in a manner that is different and distinct from the mechanisms of action of currently known antibiotics, the combination of the compounds of the present disclosure and antibiotics are expected to be effective in the methods described herein.
  • Representative antibiotics may be selected from the classes of aminoglycosides, ansamycins, carbacephem, carbapenems, cephalosporins, glycopeptides, macrolides, monobactams, penicillins, polypeptides, quinolones, sulfonamides and/or tetracyclines; other classes of antibiotics may also be used.
  • antibiotics include, but are not limited to, Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Streptomycin, Tobramycin, Geldanamycin, Herbimycin, Loracarbef, Ertapenem, Imipenem/Cilastatin, Meropenem, Cefadroxil, Cefazolin, Cephalexin, Cefaclor, Cefamandole, Cefoxitin, Cefprozil, Cefuroxime, Cefixime, Cefdinir, Cefditoren, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefepime, Teicoplanin, Vancomycin, Methicillin, Azithromycin, Clarithromycin, Dirithromycin, Erythromycin, Roxithromycin, Troleandomycin, Aztreonam, Amoxicillin, Ampicillin, Az
  • FRET fluorescenceresonance energy transfer
  • this assay is performed in a fluorometer under steady-state or multiple turnover conditions, where steady-state conditions are defined as the substrate being in large excess over the enzyme.
  • steady-state conditions are defined as the substrate being in large excess over the enzyme.
  • a constant enzyme concentration is titrated with increasing concentrations of the peptide substrate.
  • the EDANS fluorophore is excited at 350 nm and fluorescence from EDANS is observed at 495 nm over a period of time. To reduce the correction for the inner filter effect the reaction was monitored at an emission of 590 nm.
  • fluorescence is low due to a high FRET efficiency, but, as the peptide is cleaved the fluorescence increases linearly. The slope of this linear increase is taken to be the initial velocity, V 0 .
  • the initial velocity as a function of peptide concentration will exhibit Michaelis-Menten kinetics, i.e. a hyperbolic dependence on substrate concentration, and can be analyzed to yield the steady-state kinetic constants K n , and k cat .
  • the effect of inhibitors on V 0 was monitored at select enzyme and substrate concentration.
  • Solvent evaporations were carried out in vacuo with a rotary evaporator.
  • Analytical samples were prepared by drying the samples in vacuo (0.2 mmHg) in an Abderhalden drying apparatus over P 2 O 5 and ethyl acetate at reflux temperature.
  • Thin layer chromatography (TLC) was performed on silica gel plates with fluorescent indicator (Whatmann, silica gel, UV254, 25 ⁇ m plates). Spots were visualized by UV light (254 and 365 ran).
  • AU analytical samples were single spots on TLC in at least two different solvent systems. Purification by column and flash chromatography was carried out using 'BAKER' silica gel (40 ⁇ m) in the solvent systems indicated.
  • the amount (weight) of silica gel for column chromatography was in the range of 50- 100 times the amount (weight) of the crude compounds being separated. Melting points were determined on a Mel-Temp II melting point apparatus and are uncorrected. Proton nuclear magnetic resonance ( 1 H NMR) and carbon nuclear magnetic resonance ( 13 C NMR) spectra were recorded on a Brucker DPX-300 spectrometer using TMS as internal standard. The values of chemical shifts ( ⁇ ) are given in ppm and coupling constants (J) in Hz. The chemical shift values are reported as parts per million (ppm) relative to tetramethylsilane as internal standard. Mass spectra were recorded on a Micromass Platform LCC instrument.
  • reaction mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated NaHCO 3 solution (2 x 25 mL), water (1 x 25 mL), brine (1 x 25 mL) and dried over anhydrous Na 2 SO 4 .
  • the organic layer was separated and washed with saturated NaHCO 3 solution (2 x 15 mL), water (2 x 15 mL), brine (1 x 15 mL) and dried (Na 2 SO 4 ). The drying agent was filtered off and the solvent was removed under vacuum to obtain the crude product.
  • reaction mixture was cooled to 0 °C and was quenched by careful addition of methanol (3 mL) and IN. HCl (0.5 mL). The solvent was removed under reduced pressure and the residue obtained was dissolved in CHCl 3 (300 mL) and washed with water (2 x 75 mL), brine (1 x 150 mL) and dried (Na 2 SO 4 ).
  • IC 50 Values were determined by fluorescence resonance energy transfer (FRET) assay Measurements were carried out in triplicate and average value with standard deviation is reported.
  • Table 2 S. aureus SrtAa 5 9 inhibition of the synthesized compounds.
  • IC 50 Values were determined by fluorescence resonance energy transfer (FRET) assay. Measurements were carried out in triplicate and average value with standard deviation is reported.
  • FRET fluorescence resonance energy transfer

Abstract

The present disclosure describes novel small-molecule inhibitors of bacterial sortases. The identified inhibitors may be used in the methods of treatment and prevention described in the present disclosure.

Description

Novel Inhibitors of Bacterial Sortase Enzymes and Methods of Using the Same
Inventors Sadanandan E. VeIu, Sthanam V. L. Narayana and Aaron L. Lucius
The present application claims the benefit of U.S. provisional application serial no. 60/955,353, filed 8-11-2007.
FIELD OF THE DISCLOSURE The present disclosure relates generally to inhibitors of bacterial virulence and infectivity.
Specifically, the present disclosure relates to compounds having the formula I (including compounds having the formula Ia, Ib and Ic) through XII and the identification of such compounds as inhibitors of bacterial sortase enzymes. The present disclosure also relates to methods of using the compounds disclose herein, pharmaceutical compositions comprising the compounds disclosed herein and methods for producing the compounds disclosed herein.
BACKGROUND
Bacteria can cause serious or fatal infections in humans. Gram-positive bacteria capable of causing serious or fatal infections in humans include, but are not limited to, Staphylococcus, Streptococcus, Enterococcus, Bacillus, and Listeria. Infections caused by these pathogens are particularly severe and difficult to treat in immunologically compromised patients and patients with pre-existing diseases and/or conditions. These include patients suffering from infection with the Human Immunodeficiency Virus (HIV), the virus that causes AIDS, as well as patients given immune-suppressive agents for treatment of cancer or autoimmune diseases. Gram-negative bacteria can also give rise to serious or fatal infections in humans. A unique characteristic of bacterial pathogens, including Gram-positive and Gram- negative bacteria, is their surface display of proteins anchored to the cell wall. In fact, many of these molecules are known to be involved in increasing bacterial virulence which mediates the ability of bacterial cells to infect cells in a susceptible host. Thus, a possible disruption in this anchoring process of these surface proteins may prove to be an effective treatment against bacteria infection and a mechanism to decrease bacterial virulence.
Gram-positive bacteria are formidable human pathogens. For example, Staphylococci are responsible for more than a million hospital-acquired bacterial infections every year. In addition, they are to blame for a number of minor infections ranging from pustules to boils in regular day- to-day life, and for a significant number of serious infections like endocarditis, osteomyelitis, and septic arthritis (Petti, et al., Cardiol Clin 2003, 21, (2), 219-33, vii; Lobati, et al., Infection 2001, 29, (6), 333-6; Ish-Horowicz, et al., Pediatr Infect Dis J 1992, 11, (2), 82-7). Streptococcus pneumoniae is responsible for a major portion of 3 million deaths worldwide of children from pneumonia and meningitis and for a large number of deaths from pneumonia that occurs in elderly individuals. They are also responsible for a major percentage of ear infections, sinusitis, and additional severe invasive infections in asplenic persons and those with sickle cell disease. Compounding their importance is the recent steep increase in multi-drug resistance found in bacteria and the potential of 'drug resistant bacteria' to be used in biowarfare and bioterrorism (Pappas, et al., Infect Dis Clin North Am 2006, 20, (2), 395-421, x; Clarke, et al., Br J Biomed Sci 2005, 62, (1), 40-6). Hospital strains of S. aureus are usually resistant to a variety of different antibiotics. A few strains are resistant to all clinically useful antibiotics except vancomycin, and vancomycin-resistant strains are increasingly reported (Mohr, et al., Clin Infect Dis 2007, 44, (12), 1536-42; Webster, et al., Diagn Microbiol Infect Dis 2007, 57, (2), 177-81; Weigel, et al., Antimicrob Agents Chemother 2007, 51, (1), 231-8). Methicillin resistant S. aureus (MRSA) is widespread and most methicillin-resistant bacterial strains are also resistant against multiple antibiotics, necessitating the need for identifying new therapeutic agents (Baggett, et al. J Infect Dis 2004, 189, (9), 1565-73; Deresinski, S., Clin Infect Dis 2005, 40, (4), 562-73; Salerno, et al., Ann Thorac Surg 2007, 83, (5), 1888-91). Ideally, such agents would not promote the emergence of resistant strains or do so to a lesser degree than other currently used drugs.
New approaches for the prevention and treatment of bacterial infections and reduction of bacterial virulence require greater understanding of the molecular structure and mechanisms of the chosen intervention targets and of the pathogenic role played by the target in the infection process. Bacterial infections are very complex and involve the action of a large sophisticated arsenal of virulence factors, many of which are surface-bound or secreted. Surface proteins often fall into one of four functional categories: microbial adhesion to host tissues, protection from host defense mechanisms, acquisition of nutrients for bacterial growth, and secretion of toxins and invasins. These virulence factors carry out important roles in the infection process. The possibility of human interventions interrupting this process through interference with virulence is a possibility, but not yet a reality. Gram-positive bacteria such as, but not limited to, Staphylococci, are endowed with a multitude of cell-wall anchored proteins that serve as an interface between the microbe and its host. Bacterial sortases are cysteine transpeptidases that participate in secretion and anchoring of many cell wall proteins by a mechanism highly conserved in almost the entire class of Gram-positive bacteria. Sortases and sortase homologs have also been identified in certain Gram-negative bacteria and appear to use the same mechanism. Sortases, because of their control over the cellular location of multiple virulence factors, is an attractive potential target for interrupting bacterial virulence and thereby reducing the infectivity of bacteria towards a host. Surface proteins can be attached to bacterial surface in one of several fashions (Navarre, et al., Microbiol MoI Biol Rev 1999, 63, (1), 174-229; Cossart, et al., Proc Natl Acad Sci US A 2000, 97, (10), 5013-5). Proteins that are covalently attached to the cell wall share conserved regions known as the "sorting signal" or cell wall anchors (Fischetti, V. et al., MoI Microbiol 1990, 4, (9), 1603-5; 15; Schneewind, et al., Cell 1992, 70, (2), 267-81). Sortases from Gram- positive bacteria are currently grouped into four, sometimes five, families designated Sortase (Srt) A, SrtB, SrtC (Family 3), and SrtD (Families 4 and 5), with each family recognizing a slightly different sorting signal. The sortases found in gram-negative bacteria have been grouped into another separate family (Family 6). The sorting signal for SrtA includes a conserved amino acid motif, usually LPXaaTG (SEQ ID NO: 1), where Xaa can. be any one of the naturally occurring L-amino acids, while the sorting signal for SrtB includes the conserved amino acid motif NPQTXaaN (SEQ ID NO: 2), where Xaa can be any one of the naturally occurring L- amino acids. Additional sorting signals are also known to be present in Gram-positive bacteria.
In terms of the mechanism of covalent attachment of the cell surface proteins, precursor proteins are directed into a secretory pathway by their N-terminal signal peptides (Schneewind, et al., Cell 1992, 70, (2), 267-81; Navarre, et al., MoI Microbiol 1994, 14, (1), 115-21; Navarre, et al.,. J Biol Chem 1998, 273, (44), 29135-42; Perry, et al., J Biol Chem 2002, 277, (18), 16241- 8; Schneewind, et al., Science 1995, 268, (5207), 103-6; Schneewind, et al., Embo J 1993, 12, (12), 4803-11; Ton-That, et al., J Biol Chem 1997, 272, (35), 22285-92; Ton-That, et al., J Biol Chem 1998, 273, (44), 29143-9). They are translocated across the membrane and the signal peptide is cleaved (Schneewind, et al., Cell 1992, 70, (2), 267-81; Navarre, et al., MoI Microbiol 1994, 14, (1), 115-21; Navarre, et al., J Biol Chem 1998, 273, (44), 29135-42; Perry, et al., J Biol Chem 2002, 277, (18), 16241-8; Schneewind, et al., Science 1995, 268, (5207), 103-6; Schneewind, et al., Embo J 1993, 12, (12), 4803-11). Then, the C-terminal sorting signal retains the protein in the secretory pathway. The enzyme sortase acts at this point to cleave the protein at a point in the sorting signal (for example, SrtA cleaves between the threonine (T) and the glycine (G) of SEQ ID NO: 1) (Navarre, et al., MoI Microbiol 1994, 14, (1), 115-21). The end residue of the sorting signal is then covalently linked to a cell wall component of the bacteria. The bacterial cell wall target can vary depending on the bacterial species and the sortase involved. For example, in the reaction catalyzed by SrtA in Staphylococcus, the carboxyl group of the Thr is then amide-linked to the amino group of the pentaglycine cross bridges in the staphylococcal cell wall (Perry, et al, J Biol Chem 2002, 277, (18), 16241-8). This transpeptidation is also carried out by sortase enzyme (see FIG. 1).
Thus, sortase is an enzyme that recognizes a specialized cell wall "sorting signals" in proteins destined for surface attachment and covalently links them to the bacterial cell wall (Navarre, et al., Microbiol MoI Biol Rev 1999, 63, (1), 174-229). Sortase-defective strains of various pathogens were shown to be faulty in the display of surface proteins and are less virulent (Jonsson, et al., Microbes Infect 2003, 5, (9), 775-80; Mazmanian, et al., Proc Nαtl Acαd Sci US A 2000, 97, (10), 5510-5). In SrtA gene knock-out mutants of Staphylococci, surface protein precursors were found to be accumulated in the cytoplasm or to be mis-attached to the cell membrane and cell wall (mis-sorted phenotype) (Mazmanian, et al., Proc Natl Acad Sci U S A 2000, 97, (10), 5510-5). In a number of studies, individual sortase genes have been deleted and the loss-of-sortase function resulted in less virulence in several animal models of disease (S. aureus, S. gordonii, Listeria, group A streptococci, S. pneumoniae) (Jonsson, et al, Microbes Infect 2003, 5, (9), 775-80; Barnett, et al., J Bacteriol 2002, 184, (8), 2181-91 ; Bierne, et al., MoI Microbiol 2002, 43, (4), 869-81 ; Bolken, et al., Infect Immun 2001, 69, (1), 75-80; Garandeau, et al., Infect Immun 2002, 70, (3), 1382-90; Jonsson, let al., J Infect Dis 2002, 185, (10), 1417-24).
Two sortases from Staphylococcus aureus, SrtA and SrtB have been cloned, sequenced, expressed and purified (Mazmanian, et al., Science 1999, 285, (5428), 760-3; Mazmanian, et al., Proc Natl Acad Sci U S A 2002, 99, (4), 2293-8). S. aureus SrtA is the best-characterized enzyme from the family of bacterial sortases. SrtA is a 206-amino acid protein with a cysteine residue at position 184, which was shown to be critical for its function (Ton-That, et al, Proc Natl Acad Sci USA 1999, 96, (22), 12424-9). SrtB, exhibiting 23% primary sequence homology with SrtA, is the second identified S. aureus sortase. SrtA is responsible for anchoring all LPXTG- containing surface proteins including CIfA, CIfB, FnbpA, FnbpB, SdrC, and Cna. The amino acid sequence of SrtA (SEQ ID NO: 3) and SrtB (SEQ ID NO: 4) are shown in FIGS. IB and 1C, respectively. Despite an overall homology of 20-30% among different bacterial sortases, the homology in the active site regions is significantly higher.
All sortases share various degrees of homology to the archetypal sortase, SrtA of S. aureus; however, all known SrtA homologs (all sortases) contain structurally conserved active- site residues of histidine, arginine, and cysteine (SrtA residues Hisl20, Argl97, and Cysl84) (Marraffmi, et al., Microbiol MoI Biol Rev 2006, 70, (1), 192-221). This absolute conservation includes the SrtB family, which has the most divergent consensus sorting signal of all the sortase families (Maresso, et al., J Bacteriol 2006, 188, (23), 8145-52). The absolute conservation of these residues in the active-site, along with data demonstrating that the sulfhydryl moiety in the active-site is required for the transpeptidation reaction suggests that compounds interacting with or blocking access to the Cysl84 residue (or appropriate cysteine residue in the SrtA homologs) would inhibit the enzymatic activity of any SrtA homolog. Furthermore, the close proximity of the absolutely conserved Argl97 residue to the Cysl84 residue, along with data demonstrating that arginine to alanine substitutions at the 197 residue in S. aureus greatly impaired transpeptidation activity (Marraffini, et al., J Biol Chem 2004, 279, (36), 37763-70) suggests that compounds interacting with or blocking access to the Argl97 residue (or appropriate arginine residue in the SrtA homologs) would inhibit the enzymatic activity of any SrtA homolog. Sortases are virtually universal among Gram-positive bacteria. However, some examples are also found in limited number of archaeal and Gram-negative bacteria (Pallen, et al., Trends Microbiol 2001, 9, (3), 97-102). Due to their universal presence in Gram-positive bacteria and other bacterial species, sortases are attractive pharmacotherapeutic targets (Cossart, et al., Proc Natl Acad Sci U S A 2000, 97, (10), 5013-5). The sorting signal of proteins (SEQ ID NO: 1) recognized by the main sortase is nearly identical among Gram-positive bacteria (Pallen, et al., Trends Microbiol 2001, 9, (3), 97-102; Comfort, et al., Infect Immun 2004, 72, (5), 2710-22).
Altering the targeting and/ distribution of virulence factors, such as, but not limited to, surface proteins, as opposed to bacterial growth is a novel approach for reducing bacterial virulence and infectivity towards a host. As more and more pathogens become resistant to antibiotics, altering the targeting and/or distribution of cell surface proteins on the cell wall offers a novel strategy against bacterial infections. Additionally, sortase inhibitors have the potential to be powerful therapeutic agents when used in conjunction with existing traditional antibiotics as a combination therapy since these function by a complementary mechanism.
Currently there have only been a few reports of specific sortase inhibitors (Kang, et al., Biol Pharm Bull 2006, 29, (8), 1751-5; Kim, et al., Biotechnol Biochem 2003, 67, (11), 2477-9; Kim, et al., Biosci Biotechnol Biochem 2002, 66, (12), 2751-4; Scott, et al., Biochem J 2002, 366, (Pt 3), 953-8). Recently, Oh et al. identified a small molecule reversible inhibitor with a low micromolar ICs0 value by structurally modifying a lead compound identified by random screening of a group of small molecules (Oh, et al., J Med Chem 2004, 47, (10), 2418-21). None of the existing reports have utilized the structural information of the sortase active site for the design of their inhibitors.
Therefore, there remains a need in the art to identify new inhibitors of the bacterial sortase enzymes. The crystal structures of recombinant S. aureus SrtAΔ59 and SrtBΔ30 were recently determined (Zong, et al., J Biol Chem 2004, 279, (30), 31383-9; Zong, et al., Structure 2004, 12, (1), 105-12). SrtAΔ59 and SrtBΔ30 are fully active variants of SrtA and SrtB with 59 and
30 residues shorter from the NH-terminus, respectively. The present disclosure described the identification of several new low micromolar sortase inhibitors identified through structure based inhibitor design studies. The present disclosure also described pharmaceutical compositions containing these inhibitors methods of using the identified inhibitors and pharmaceutical compositions.
BRIEF DESCRIPTION OF THE FIGURES
FIG. IA illustrates the reaction catalyzed by the S. aureus Sortase A.
FIG. IB shows the amino acid sequence of the Sortase A enzyme from S. aureus. FIG. 1C shows the amino acid sequence of the Sortase B enzyme from S. aureus.
FIG. 2 shows the structure of compound 1.
FIG. 3 shows a FlexX model of compound 1 in the SrtA^sg active site.
FIG. 4 shows the interactions of compound 1 with amino acid residues in the SrtAΛ59 active site.
FIG. 5 shows an exemplary synthesis of compounds 1, 13, 12a and 12b. FIG. 6 shows an exemplary synthesis of compounds 14, 15, 16 and 17.
FIG. 7 shows an exemplary synthesis of compounds 18a, 18b, 19a and 19b.
FIG. 8 shows an exemplary synthesis of compound 20.
FIG. 9 shows an exemplary synthesis of compounds 21-24.
FIG. 10 shows an exemplary synthesis of compounds 25-28. FIG. 11 shows an exemplary synthesis of compounds 29-30.
FIG. 12 shows an exemplary synthesis of compounds 31-34.
FIG. 13 shows an exemplary synthesis of compounds 43-46.
FIG. 14 shows an exemplary synthesis of compounds 51-56.
FIG. 15 shows an exemplary synthesis of compound 57.
DETAILED DESCRIPTION
Definitions
As used in this specification, the followings words and phrases have the meanings as defined below, unless otherwise limited in specific instances, either individually or as part of a larger group.
As used herein, the term "alkyl", whether used alone or as part of a substituent group, includes straight hydrocarbon groups comprising from one to twenty carbon atoms. Thus the phrase includes straight chain alkyl groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -CH(CHj)2, -CH(CH3)(CH2CH3), -CH(CH2CH3)2, -C(CH3)3, - C(CH2CH3)3, -CH2 CH(CH3)2) -CH2CH(CH3)(CH2CH3), -CH2CH(CH2CH3);,, -CH2C(CH3)3, - CH2C(CH2CH3);,-, -CH(CH3)CH(CH3)(CH2CH3), -CH2CH2CH(CH3)2, CH2CH2CH(CH3)(CH2CH3), -CH2CH2CH(CH2CH3)Z, -CH2CH2C(CH3)3,
CH2CH2C(CH2CH3)3, -CH(CH3)CH2CH(CH3)2, -CH(CH3)CH(CH3)CH(CH3)CH(CH3)2, -CH(CH2 CH3)CH(CH3)CH(CH3)(CH2CH3), and others. The phrase also includes cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above. The phrase also includes polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above.
As used herein, the term "alkenyl", whether used alone or as part of a substituent group, includes an alkyl group having at least one double bond between any two adjacent carbon atoms. As used herein, the term "alkynyl", whether used alone or as part of a substituent group, includes an alkyl group having at least one triple bond between any two adjacent carbon atoms.
As used herein, the term "unsubstituted alkyl", "unsubstituted alkenyl", and "unsubstituted alkynyl" refers to alkyl, alkenyl and alkynyl groups that do not contain heteroatoms. The phrase "substituted alkyl", "substituted alkenyl", and "substituted alkynyl" refers to alkyl, alkenyl and alkynyl groups as defined above in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen or non-carbon atoms such as, but not limited to, a halogen atom, such as F, Cl, Br, and I; and oxygen atom in groups such as carbonyl, carboxyl, hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, enamines, imines, oximes, hydrazones, and nitriles; a silicon atom in groups such as in trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. Other alkyl groups include those in which one or more bonds to a carbon or hydrogen atom is replaced by a bond to an oxygen atom such that the substituted alkyl group contains a hydroxyl, alkoxy, aryloxy group, or heterocyclyloxy group. Still other alkyl groups include alkyl groups that have an amine, alkylamine, dialkylamine, arylamine, (alkyl)(aryl)amine, diarylamine, heterocyclylamine, (alkyl)(heterocyclyl)amine, (aryl)(heterocyclyl)amine, or diheterocyclylamine group.
As used herein, the term "unsubstituted aryl" refers to monocyclic or bicyclic aromatic hydrocarbon groups having 6 to 12 carbon atoms in the ring portion, such as, but not limited to, phenyl, naphthyl, anthracenyl, biphenyl and diphenyl groups, that do not contain heteroatoms. Although the phrase "unsubstituted aryl" includes groups containing condensed rings such as naphthalene, it does not include aryl groups that have other groups such as alkyl or halogen groups bonded to one of the ring members, as aryl groups such as tolyl are considered herein to be substituted aryl groups as described below. Unsubstituted aryl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
As used herein, the term "substituted aryl group" has the same meaning with respect to unsubstituted aryl groups that substituted alkyl groups had with respect to unsubstituted alkyl groups. However, a substituted aryl group also includes aryl groups in which one of the aromatic carbons is bonded to one of the non-carbon or non-hydrogen atoms described above and also includes aryl groups in which one or more aromatic carbons of the aryl group is bonded to a substituted and/or unsubstituted alkyl, alkenyl, or alkynyl group as defined herein. This includes bonding arrangements in which two carbon atoms of an aryl group are bonded to two atoms of an alkyl, alkenyl, or alkynyl group to define a fused ring system (e.g. dihydronaphthyl or tetrahydronaphthyl). Thus, the phrase "substituted aryl" includes, but is not limited to tolyl, and hydroxyphenyl among others. Substituted aryl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
As used herein, the term "unsubstituted aralkyl" refers to unsubstituted alkyl, alkenyl or alkyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkyl, alkenyl or alkyl group is replaced with a bond to an unsubstituted or substituted aryl group as defined above. For example, methyl (CH3) is an unsubstituted alkyl group. If a hydrogen atom of the methyl group is replaced by a bond to a phenyl group, such as if the carbon of the methyl were bonded to a carbon of benzene, then the compound is an unsubstituted aralkyl group (i.e., a benzyl group). Unsubstituted aralkyl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
As used herein, the term "substituted aralkyl" has the same meaning with respect to unsubstituted aralkyl groups that substituted aryl groups had with respect to unsubstituted aryl groups. However, a substituted aralkyl group also includes groups in which a carbon or hydrogen bond of the alkyl, alkenyl or alkyl part of the group is replaced by a bond to a non-carbon or a non-hydrogen atom.
As used herein, the term "unsubstituted heterocyclyl" refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S. Although the phrase "unsubstituted heterocyclyl" includes condensed heterocyclic rings such as benzimidazolyl, it does not include heterocyclyl groups that have other groups such as alkyl or halogen groups bonded to one of the ring members, as compounds such as 2-methylbenzimidazolyl are "substituted heterocyclyl" groups as defined below. Examples of heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl; saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, oxazolyl, isoxazolyl, oxadiazolyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, morpholinyl; unsaturated condensed heterocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, benzoxazolyl, benzoxadiazolyl, benzoxazinyl (e.g. 2H-l,4-benzoxazinyl etc.); unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g. 1,2,3- thiadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolodinyl; saturated and unsaturated 3 to 8 membered rings containing 1 to 2 sulfur atoms such as, but not limited to, thienyl, dihydrodithiinyl, dihydrodithionyl, tetrahydrothiophene, tetrahydrothiopyran; unsaturated condensed heterocyclic rings containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, benzothiazolyl, benzothiadiazolyl, benzothiazinyl (e.g. 2H-l,4-benzothiazinyl, etc.), dihydrobenzothiazinyl (e.g. 2H-3,4- dihydrobenzothiazinyl, etc.), unsaturated 3 to 8 membered rings containing oxygen atoms such as, but not limited to furyl; unsaturated condensed heterocyclic rings containing 1 to 2 oxygen atoms such as benzodioxolyl (e.g. 1,3-benzodioxoyl, etc.); unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1 ,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing a sulfur atom and 1 to 2 oxygen atoms such as benzoxathiinyl. Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones). For example, heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1 -dioxide. Preferred heterocyclyl groups contain 5 or 6 ring members.
As used herein, the term "substituted heterocyclyl" refers to an unsubstituted heterocyclyl group as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples, include, but are not limited to, 2-methylbenzimidazolyl, 5-methylbenzimidazolyl, 5- chlorobenzthiazolyl, 1 -methyl piperazinyl, and 2-chloropyridyl among others.
The phrase "unsubstituted heterocyclylalkyl" refers to unsubstituted alkyl, alkenyl or alkynyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkyl, alkenyl or alkynyl group is replaced with a bond to a substituted or unsubstituted heterocyclyl group as defined above. For example, methyl (-CH3) is an unsubstituted alkyl group. If a hydrogen atom of the methyl group is replaced by a bond to a substituted or unsubstituted heterocyclyl group, such as if the carbon of the methyl were bonded to carbon 2 of pyridine (one of the carbons bonded to the N of the pyridine) or carbons 3 or 4 of the pyridine, then the compound is an unsubstituted heterocyclylalkyl group.
The phrase "substituted heterocyclylalkyl" refers to substituted alkyl, alkenyl or alkynyl groups as defined above in which a hydrogen or carbon bond of the substituted alkyl, alkenyl or alkynyl group is replaced with a bond to a substituted or unsubstituted heterocyclyl group as defined above. However, a substituted heterocyclylalkyl group also includes groups in which a non-hydrogen atom is bonded to a heteroatom in the heterocyclyl group of the heterocyclylalkyl group such as, but not limited to, a nitrogen atom in the piperidine ring of a piperidinylalkyl group.
The phrase "halogen" or "halo" refers to fluorine, chlorine, bromine and iodine. The term "pharmaceutically acceptable salts" isv meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present invention contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, oxalic, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galacrunoric acids and the like (see, for example, Berge, S. M., et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
The term "prodrug" is meant to include functional derivatives of the compounds disclosed which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present disclosure, the term "administering" shall encompass the treatment of the various disease states/conditions described with the compound specifically disclosed or with a prodrug which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985. The terms "prevent", "preventing", "prevention" "suppress", "suppressing" and
"suppression" as used herein refer to administering a compound prior to the onset of clinical symptoms of a disease state/condition so as to prevent any symptom, aspect or characteristic of the disease state/condition. Such preventing and suppressing need not be absolute to be useful.
The terms "treat", "treating" and "treatment" as used herein refers to administering a compound after the onset of clinical symptoms of a disease state/condition so as to reduce or eliminate any symptom, aspect or characteristic of the disease state/condition. Such treating need not be absolute to be useful.
The term "in need of treatment" as used herein refers to a judgment made by a caregiver that a patient requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, and may include the knowledge that the patient is ill as the result of a disease state/condition that is treatable by a compound or pharmaceutical composition of the disclosure.
The term "in need of prevention" as used herein refers to a judgment made by a caregiver that a patient requires or will benefit from prevention. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, and may include the knowledge that the patient may become ill as the result of a disease state/condition that is treatable by a compound or pharmaceutical composition of the disclosure.
The term "individual" or "patient" as used herein refers to any animal, including mammals, such as, but not limited to, mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, or humans. The term may specify male or female or both, or exclude male or female; and
The term "therapeutically effective amount", in reference to the treating, preventing or suppressing of a disease state/condition, refers to an amount of a compound either alone or as contained in a pharmaceutical composition that is capable of having any detectable, positive effect on any symptom, aspect, or characteristics of the disease state/condition. In one embodiment, a therapeutically effective amount is a tumor growth inhibiting amount. Such effect need not be absolute to be beneficial. Summary of the Disclosure The present disclosure provides compounds of the general formula I (including compounds having the formula Ia, Ib and Ic) through XII, specifically compounds 1-8, 12-34 and 43-67. In one embodiment, these compounds are useful as inhibitors of bacterial sortase enzymes. In a particular embodiment, these compounds are useful as inhibitors of bacterial SrtA, SrtB, SrtC and/or SrtD, or homologs thereof. In a further particular embodiment, the compounds are useful as inhibitors of bacterial SrtA. The compounds of the present disclosure have utility in treating and/or preventing a variety of disease states and/or conditions involving bacterial infection and virulence, wherein said bacteria causing or contributing to the bacteria infection and virulence express a sortase enzyme.
In an alternate embodiment, the present disclosure provides a method of treating a disease state and/or condition in a subject which is caused by, at least in part, bacterial infection and virulence, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure. In a further alternate embodiment, the present disclosure provides a method of preventing or suppressing a disease state and/or condition in a subject which is caused by, at least in part, bacterial infection and virulence, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
In yet a further alternate embodiment, the present disclosure provides for a method of reducing or eliminating bacterial virulence in a subject, the method comprising the administration to said subject a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure. In still a further embodiment, the present disclosure provides for methods of treating bacterial infection in a subject, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
In still a further embodiment, the present disclosure provides for methods of preventing or suppressing bacterial infection in a subject, the method comprising the administration to said subject of a therapeutically effective amount of a compound of the present disclosure or a pharmaceutical composition containing a compound of the present disclosure.
Other embodiments and aspects of the invention will be apparent according to the description provided below. Sortase Inhibitors
This present disclosure provides compounds of the general formula I (including compounds having the formula Ia, Ib and Ic) through XII and pharmaceutical compositions comprising the same or pharmaceutically acceptable salts thereof, or esters thereof, or prodrugs thereof and tautomers and polymorphic variants of any of the foregoing. In one embodiment, the compounds disclosed are useful as inhibitors of bacterial sortase enzymes.
With regard to compounds having the general formula (I), the structure below is provided:
(D
Figure imgf000015_0001
Where
Ring Z is selected from substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; Ring Y is selected from substituted or unsubstituted aryl or substituted or unsubstituted heterocycle;
Ring X is optional and when present is selected from substituted or unsubstituted aryl or substituted or unsubstituted heterocycle;
B is selected from S or O or NRi, where Ri is H or a substituted or unsubstituted alkyl; Li and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and
L in Li and L2 is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; and
The linking group I, as indicated, is a C2 to C5 substituted or unsubstituted alkyl, C2 to C5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C2 to C5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
In a specific embodiment, ring Z is a substituted or unsubstituted 5 membered ring, including but not limited to, heterocylic compounds containing oxygen (such as furan), sulfur
(such as thiophene) or nitrogen (such as pyrrole) or a substituted or unsubstituted 6 member ring, including but not limited to, phenyl, heterocylic compounds containing oxygen (such as pyran), sulfur (such as thiapyrane) or nitrogen (such as pyridine) phenyl, or selected from substituted or unsubstituted phenyl, benzothiophene and indole and ring X is morpholine. In a specific embodiment of compounds having the general formula (I), compounds of the structure Ia are provided:
(Ia)
Figure imgf000016_0001
Where
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRj, where R1 is H or a substituted or unsubstituted alkyl;
E is selected from the group consisting of: CH2, O and N;
Li and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and L in Li and L2 is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; The linking group I, as indicated, is a C2 to C5 substituted or unsubstituted alkyl, C2 to C5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C2 to C5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
R2 through R5 are each independently selected from the group consisting of: H, OH, CORn, COORi4, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl.
R6 through R8 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl.
Ri3 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi5Ri6; Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl. Ri5 and Ri6 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl.
In a specific embodiment, R2 through R5 are each independently selected from the group consisting of: H, CORJ3, COORH and substituted or unsubstituted alkyl. In a further specific embodiment, R2 through R5 are each independently selected from the group consisting of: COOCH3, COOH, CHO, CH2OH and CONH2.
In a specific embodiment, R2 is selected from the group consisting of: H, COR13,
COORH and substituted or unsubstituted alkyl while R3 through R8 are H, I is a C2 alkenyl, B is
NH and 1 and 2 = 0. In a further specific embodiment, R2 is selected from the group consisting of: COOCH3, COOH, CHO, CH2OH and CONH2, while R3 through R8 are H, I is a C2 alkenyl, B is NH and 1 and 2 = O.
In certain specific embodiment of the structure of compound Ia, the structures below are provided:
A is S, B is NH, E is O, R2 is COOH, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 1.
Figure imgf000017_0001
A is S, B is NH, E is O, R2 is COOCH3, R3 through R8 are H, Lj and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 12a.
Figure imgf000017_0002
A is O, B is NH, E is O, R2 is COOCH3, R3 through R8 are H, L1 and L2 are not present (i.e., 1 and 2 each equal 0), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 12b.
Figure imgf000018_0001
A is O, B is NH, E is O, R2 is COOH, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 13.
Figure imgf000018_0002
A is S, B is NH, E is O, R2 is CH2OH, R3 through R8 are H, L1 and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 18a.
Figure imgf000018_0003
A is O, B is NH, E is O, R2 is CH2OH, R3 through R8 are H, L1 and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 18b.
Figure imgf000018_0004
A is S, B is NH, E is O, R2 is CHO, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal 0), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 19a.
Figure imgf000019_0001
A is O, B is NH, E is O, R2 is CHO, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 19b.
Figure imgf000019_0002
A is S, B is NH, E is O, R2 is CONH2, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 20.
Figure imgf000019_0003
A is S, B is NH, E is O, R2 is COOCH3, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkynyl and the compound has the structure of compound 21.
Figure imgf000019_0004
A is S, B is NH, E is O, R2 is COOH, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal 0), the linking group I is a C2 alkynyl and the compound has the structure of compound 22.
Figure imgf000020_0001
A is S, B is NH, E is O, R2 is COOCH3, R3 through R8 are H, L1 and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the cis configuration and the compound has the structure of compound 23.
Figure imgf000020_0002
A is S, B is NH, E is O, R2 is COOH, R3 through R8 are H, L1 and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the cis configuration and the compound has the structure of compound 24.
Figure imgf000020_0003
A is S, B is NH, E is O, R2 is COOH, R3 through R8 are H, Lj and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 29.
Figure imgf000020_0004
A is S, B is NH, E is CH2, R2 is COOCH3, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal 0), the linking group I is a C3 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 31.
Figure imgf000021_0001
A is O, B is NH, E is CH2, R2 is COOCH3, R3 through R8 are H, Lj and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 32.
Figure imgf000021_0002
A is S, B is NH, E is CH2, R2 is COOH, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 33.
Figure imgf000021_0003
A is S, B is NH, E is O, R2 is COOCH2CH3, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 43.
Figure imgf000021_0004
A is O, B is NH, E is O, R2 is COOCH2-C6Hi2, R3 through R8 are H, Lj and L2 are not present (i.e., 1 and 2 each equal 0), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 44.
Figure imgf000022_0001
A is O, B is NH, E is O, R2 is COOCH2CH2CH3, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 45.
Figure imgf000022_0002
A is O, B is NH, E is O, R2 is COOCH2CH2CH2CH3, R3 through R8 are H, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 46.
Figure imgf000022_0003
A is S, B is NH, E is O, R2 is COOCH3, R3 through R8 are H, Li is not present (i.e., 1 is equal to O) and L2 is CH3, the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 47.
Figure imgf000022_0004
A is S, B is NH, E is O, R2 is COOH, R3 through R8 are H, Li is not present (i.e., 1 is equal to O) and L2 is CH3, the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 48.
Figure imgf000023_0001
In another specific embodiment of compounds having the general formula (I), compounds of the structure Ib are provided:
(Ib)
Figure imgf000023_0002
Where
A is selected from the group consisting of: S, CH2, O and N; B is selected from the group consisting of: S, O and NRi, where Ri is H or a substituted or unsubstituted alkyl;
E is a heteroatom selected from the group consisting of: CH2, O and N;
F is a single, optional N heteroatom; when present, F may be in a para, meta or ortho position with respect to the point of attachment; Li and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and
L in Li and L2 is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
The linking group I, as indicated, is a C2 to C5 substituted or unsubstituted alkyl, C2 to C5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C2 to C5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
R2 through R5 are each independently selected from the group consisting of: H, OH, CORn, COOR15, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
R9 through R)3 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl; provided that when F is present at a given position (i.e., para, meta or ortho position), one of the R9 through Ri3 substituent groups will be absent;
R14 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR16R17;
R15 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl;
Rj6 and Ri7 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl.
In a specific embodiment, R2 through R5 are each independently selected from the group consisting of: H, CORi4, COOR15 and substituted or unsubstituted alkyl. In a further specific embodiment, R2 through R8 are each independently selected from the group consisting of:
COOCH3, COOH, CHO, CH2OH and CONH2.
In a specific embodiment, R2 is selected from the group consisting of: H, CORi4,
COORi5 and substituted or unsubstituted alkyl, while R3 through R5 and R9 through Ri3 are H, I is a C2 alkenyl, B is NH and 1 and 2 = O. In a further specific embodiment, R2 is selected from the group consisting of: COOCH3, COOH, CHO, CH2OH and CONH2, while R3 through R5 and
R9 through Ri3 are H, I is a C2 alkenyl, B is NH and 1 and 2 = O.
In certain specific embodiment of the structure of compound Ib, the structures below are provided (see Table 2): F is N and is in the para position, B is NH, E is O, R2 is COOCH3, R3 through R5 and R9,
Rio, Ri2 and Ri3 are H, Rn is absent, Lj and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 51.
Figure imgf000025_0001
F absent, B is NH, E is O, R2 is COOCH3, R3 through R5 and R9 through R13 are H, Li and L2 are not present (i.e., 1 and 2 each equal 0), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 52.
Figure imgf000025_0002
F is N and is in the meta position, B is NH, E is O, R2 is COOCH3, R3 through R5 and R9, Rn, Ri2 and Rj3 are H, R10 is absent, L1 and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 55.
Figure imgf000025_0003
F is N and is in the meta position, B is NH, E is O, R2 is COOH, R3 through R5 and R9, Rn, Ri2 and Rj3 are H, Ri0 is absent, Li and L2 are not present (i.e., 1 and 2 each equal O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 56.
Figure imgf000025_0004
In another specific embodiment of compounds having the general formula (I), compounds of the structure Ic are provided:
(Ic)
Figure imgf000026_0001
Where
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRj, where R1 is H or a substituted or unsubstituted alkyl;
L1 is an optional linking groups, where 1 is selected from 0 or 1 and L in L1 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
The linking group I, as indicated, is a C2 to C5 substituted or unsubstituted alkyl, C2 to C5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C2 to C5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration; R2 through R6 are each independently selected from the group consisting of: H, OH, CORi0,
COORi l, NR]4R]5, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, C1 to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl; R7 through R9 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl;
Ri0 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR]2R]3;
Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl;
Ri2 and R]3 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl; and Ri4 and R15 are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl and substituted or unsubstituted aralkyl.
In a specific embodiment, R2 through R6 are each independently selected from the group consisting of: H, OH, CORio, COORn, NRJ4RI 5 and substituted or unsubstituted alkyl. In a further specific embodiment, R2 through R6 are each independently selected from the group consisting of: NHCH2-Phe, NH-Phe, N(CH2-Phe)2, N(CH2CH2OH)2, COOCH3, COOH, CHO, CH2OH and CONH2.
In a specific embodiment, R2 and R3 are selected from the group consisting of: H, CORi0, COORi i, NR14Ri5 and substituted or unsubstituted alkyl, while R3 through R9 are H, I is a C2 alkenyl, B is NH and 1 and 2 = 0. In a further specific embodiment, R2 and R3 are selected from the group consisting of: NH-CH2-Phe, NH-Phe, N-(CH2-Phe)2, N-(CH2CH2OH)2, COOCH3, COOH, CHO, CH2OH and CONH2, while R4 through R9 are H, I is a C2 alkenyl, B is NH and 1 and 2 = 0.
In certain specific embodiment of the structure of compound Ib, the structures below are provided (see Table 2):
A is S, B is NH, R2 is NH-CH2-Ph, R3 is COOCH3, R4 through R9 are H, Li is not present (i.e., 1 equals O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 58.
Figure imgf000027_0001
A is S, B is NH, R2 is NH-Ph, R3 is COOCH3, R4 through R9 are H, Li is not present (i.e., 1 equals O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 60.
Figure imgf000027_0002
A is S, B is NH, R2 is N-(CH2-Ph)2, R3 is COOCH3, R4 through R9 are H, L1 is not present (i.e., 1 equals 0), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 61.
Figure imgf000028_0001
A is S, B is NH, R2 is N-(CH2-Ph)2, R3 is COOH, R4 through R9 are H, Li is not present (i.e., 1 equals O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 62.
Figure imgf000028_0002
A is S, B is NH, R2 is N-(CH2CH2OH)2, R3 is COOH, R4 through R9 are H, L, is not present (i.e., 1 equals O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 63.
Figure imgf000028_0003
A is S, B is NH, R2 is N-(CH2-Phe)2, R3 through R9 are H, Lj is not present (i.e., 1 equals O), the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 67.
Figure imgf000028_0004
With regard to compounds having the general formula II through VIII, the following structures are provided:
Figure imgf000029_0001
Figure imgf000029_0002
Figure imgf000029_0003
Figure imgf000029_0004
Figure imgf000029_0005
Figure imgf000029_0006
Figure imgf000029_0007
Figure imgf000030_0001
Figure imgf000030_0002
Figure imgf000030_0003
With regard to compounds having the general formula (XII), the structure below is provided:
Figure imgf000030_0004
Where
A is selected from the group consisting of: S, CH2, O and NRi8, where Ri8 is H or a substituted or unsubstituted alkyl
The linking group I, as indicated, is a C2 to C5 substituted or unsubstituted alkyl, C2 to C5 substituted or unsubstituted alkenyl containing a carbon-carbon double bond (in either the cis or trans configuration) or C2 to C5 substituted or unsubstituted alkynyl containing a carbon-carbon triple bond; in one embodiment, the linking group I is unsubstituted; in an alternate embodiment, the linking group I is a C2 unsubstituted alkenyl with the carbon-carbon double bond in the trans configuration;
R1 through R9 are each independently selected from the group consisting of: H, OH, NRi6R17,
CORi2, COOR13, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, C1 to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
R10 and Rn are each independently selected from the group consisting of: H substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl; R12 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR14Rj 5;
Ri3 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; Ri4 and Rn are each independently selected from the group consisting of: H, O and substituted or unsubstituted alkyl.
In a specific embodiment, Ri through R9 are each independently selected from the group consisting of: H, NRi6Ri7, COR12, COORi3 and substituted or unsubstituted alkyl. In a further specific embodiment, Ri is NO2 or NH2. In a specific embodiment, Ri is selected from the group consisting of: H, NRi6Ri7,
COR)2, COOR]3 and substituted or unsubstituted alkyl, while R2 through R9 are at H, Rj0 through
Rn are CH3, A is O and I is a C2 alkenyl. In a further specific embodiment, Ri is NO2 or NH2, while R2 through R9 are at H, Ri0 through Ri 1 are CH3, A is O and I is a C2 alkenyl.
In certain specific embodiment of the structure of compound XII, the structures below are provided:
A is O, Ri is NO2, R2 through R9 are H, the linking group I is a C2 alkenyl with the carbon-carbon double bond in the trans configuration and the compound has the structure of compound 57.
Figure imgf000031_0001
Any of the foregoing compounds may be provided as pharmaceutically acceptable salts, esters, prodrugs, tautomers or polymorphic variants. Compound Identification
The structures of compound 1 (FIG. 2), compound 57 (Table 2) and compounds 2-8 (Table 1) were identified as inhibitor of bacterial sortase enzymes by in-silico virtual screening against the S. Aureus SrtAΔS9 enzyme. Virtual screening of commercial small molecule libraries from Maybridge and ChemBridge (-150,000 compounds) against the S. aureus SrtAΛ59 active site were conducted using FlexX software package integrated in SYBYL 7.0. FlexX is a fast, flexible docking method that uses an incremental construction algorithm to place ligands into the active site of the receptor. FlexX predicts the geometry of the complex as well as the free energy of binding for a given protein and a ligand. The high resolution X-ray crystal structure of both the apo enzyme and its complex with the peptide substrate were available, thus giving a clear description of the substrate binding site. In this case, FlexX is an ideal tool for screening large libraries of ligands to find leads for drug design. FlexX has the feature of allowing conformational flexibility of the ligand in complementing the active site, when active site is used as a constraint, while docking. FlexX allows complete specification of the receptor active site, including oxidation states, metal ions, side chain protonation states and automatic ligand positioning, while docking. FlexX allows the ability to dock and score small combinatorial or related libraries of compounds to identify lead structural scaffolds.
In FlexX docking, the enzyme active site environment is identified by a receptor description file (RDF). A RDF for SrtAΔ59 was created using the receptor PDB file, the "active site file" which is constituted of all residues within 6.5 A distance from the center of the active site and a "pocket file" which is composed of a few hand-picked residues that are directly interacting with the substrate peptide. 2D compound data sets from commercial sources were transformed into three dimensional molecular structures using COMPARE program. All compounds were generated in the protonated state that can be assumed under physiological conditions. The following limits were applied for the virtual screening: molecular weight >150 <500, ClogP <5. In addition, compounds bearing metal ions were omitted from the data set. All calculations were performed on Silicon Graphics Octane (2.2 GHz RlOOOO processor) computer. Docking was carried out to obtain a population of possible conformations and orientations of the inhibitors in the active site. For each inhibitor, docking runs were performed with a maximum allowed number of 10 poses. Docking position of highest scoring conformation is individually analyzed. The structures of top scoring compounds were manually examined for their drug like properties based on Lipinski's rule45 and for synthetic versatility.
The best scoring compounds from the in-silico virtual screen were purchased and screened against the SrtAΛ59 protein by monitoring the effect of the compounds on the steady state cleavage of a model substrate peptide using a FRET assay, from which IC50 values were determined (see experimental section). Nine compounds, compound 1 (FIG. 2), compound 57 (Table 2) and compounds 2-8 (Table 1) exhibited in vitro inhibition of the catalytic activity of the SrtAΔ59 enzyme with IC50 values ranging from about 31 to 400 μM. These compounds were obtained in milligram quantities from commercial sources. The most active compounds identified from this virtual screening were compound 1 (FIG. 2) which had an IC50 value of 75 μM, compound 57 (Table 2) which had an ICs0 value of 31 μM and compound 2 (Table 1) which had an IC50 value of 97 μM. The IC50 values of compounds 3 - 8 are given in Table 1.
A FlexX docking model of inhibitor 1 in SrtAΛ59 active site showed that the inhibitor fits well in the active site with multiple contacts (Figure 3). Inhibitor 1 has a three-ring structure. The left end is a thiophene ring (ring A), the middle ring is phenyl ring (ring B) and the right end is a morpholine ring (ring C). Ring A and B are connected by an acrylamido linkage in which the double bond has trans stereochemistry. Middle ring B bears a carboxylic acid group at a position meta to the point of attachment of acrylamido linkage. Ring C is connected to the middle ring B at the ortho position to COOH group through the morpholine ring N atom.
FlexX docking model of inhibitor 1 revealed several interactions of the inhibitor within the active site (Figure 4). Morpholine ring oxygen atom has a hydrogen bonding interaction with amide backbone NH of Trpl94. The cysteine residue (Cysl84) is at 2.94A distance from the morpholine ring carbon atom next to the oxygen. If the morpholine ring is appropriately substituted with a hydrogen bond acceptor or donor the modification may result in additional hydrogen bonding interactions. The carboxylic acid on the middle phenyl ring has a direct salt bridge interaction with the guanidine side chain of Argl97. The amide NH group of the linker has a hydrogen bonding interaction with the carboxylic acid side chain of GIu 105 and the amide carbonyl oxygen of the linker has a hydrogen bonding interaction with the OH side chain of Serl lό. The thiophene ring (ring A) of inhibitor 1 is in close proximity to Glnl72 (3.2A) and Asnl 14 (3.1 A). Appropriate substitution to the thiophene ring may result in additional hydrogen bonding interactions with Glnl72 and Asnl 14. Besides, a wide range of hydrophobic residues (Val201, Phe200, Ilel99, Leul69, Ilel58, VaIl 68, VaIl 64, Leul79, Ilel l5, Leul81, Ilel82, Ilel 17, Leu97 and PhelO3) surround the middle phenyl ring B on either side. The structural features of the lead compound along with its critical interactions with active site residues and the presence of key active site amino acid residues in close proximity to the inhibitor provide us plenty of opportunities for SAR studies and lead optimization (FIG. 4).
FlexX docking model of inhibitor 57 also revealed several interactions of the inhibitor within the active site. The carbonyl group present in the molecule showed an interaction with an arginine residue (Arg 197) that is conserved in the active site of sortase family enzymes. The nitro group present in the molecule showed critical interactions with several residues in the active site (Asp 170, GIu 171, GIn 172 and GIn 178). Furthermore, the nitrogen of dimethylamino group was found to be in close proximity to Serl 16 and Glul05. Synthesis and Biological Activity
Based on these initial studies, structure-activity relationship (SAR) studies were carried out. These studies were initially conducted using compound 1 and compound 57 as model compounds.
For SAR studies a general method for the synthesis of compound 1 was developed. The synthetic procedures for synthesis of the various analogs described were modeled around this synthesis. Chemistry employed for the synthesis of compound 1 is outlined in Scheme 1 (FIG. 5).
Commercially available methyl 2-(N-morpholino)-5-nitrobenzoate (9) was reduced using hydrogen in the presence of PaVC in anhydrous ethyl acetate to afford the corresponding amino compound 10. The compound 10 was coupled with commercially available trøns-3-(thiophene- 2-yl)acrylic acid (lla) in the presence of ethyl(N,N-dimethylaminopropyl)carbodiimide (EDAC) and iVyV-dimethylaminopyridine (DMAP) in 1 ,2-dichloroethane to form the amide compound 12a. Basic hydrolysis of the ester methyl group present in compound 12a afforded the compound 1 as a white crystalline solid. The synthesized product was found to be exactly identical to the compound obtained from commercial sources according to 1H-NMR, 13C-NMR, MS and enzyme inhibition data.
Derivatives of compound 1 were synthesized in order to derive preliminary structure activity relationship data. The activities of the derivatives against the activity of the SrtAΔ59 protein were determined using the FRET assay as described. Structures and IC50 values of the newly synthesized derivative compounds are given in Table 2.
Compound 13 is a furan analog of compound 1. Design of this structure was inspired by the comparison of activities of thiophene and furan compounds 3 and 6 (Table 1). These two compounds have the same structure except that compound 3 contained a furan ring and compound 6 contained a thiophene ring. Furan compound 3 is more than two fold more active than the thiophene compound, 6. Therefore, a furan substitution of the thiophene ring of compound 1 may increase its activity. Compound 13 was synthesized by a similar procedure as described for the synthesis of compound 1 (Scheme 1, FIG. 5). However, in the synthesis of compound 13, a trøM_?-3-(furan-2-yl) acrylic acid (lib) instead of trøns-3-(thiophene-2-yl) acrylic acid (Ha) was used. Compound 10 was coupled with commercially available trans-3- (furan-2-yl) acrylic acid (lib) in the presence of EDAC and DMAP in 1 ,2-dichloroethane to form the amide compound 12b. Basic hydrolysis of the ester methyl group present in compound 12b afforded compound 13.
Compound 13 along with the synthetic intermediate methyl esters 12a and 12b were evaluated for their inhibitory activity using the SI±AΛ59 protein. Compound 13 did not show an enhancement of activity as expected. Instead compound 13 was found to be less active compared to compound 1 in the enzymatic assay (ICs0 = 181 μM). However, the methyl ester intermediates (12a and 12b) showed improved inhibition as compared to corresponding acid derivatives 1 and 13. The methyl ester derivative of thiophene compound (12a) showed an IC50 value of 71 μM and methyl ester derivative of furan compound (12b) showed an IC5O value of 58 μM.
Compounds 14-17 were synthesized to evaluate the importance of the double bond present in compounds 1 and 13 for their inhibitory activity. Compounds 14 and 16 are the saturated analogs of the carboxylic acid derivatives 1 and 13, respectively. Compounds 15 and 17 are the saturated analogs of the methyl ester derivatives 12a and 12b, respectively. The SrtAΛ59 protein assay showed that all four saturated compounds 14-17 were inactive up to a concentration of 600 μM, indicating that the double bond is necessary for the activity of the disclosed compounds. Synthesis of compounds 14 - 17 is outlined in Scheme 2 (FIG. 6).
Hydrogenation of the thiophene derivatives 1 and 12a using Pd/C as catalyst was not effective, possibly due to the catalyst poisoning effect of the thiophene ring present in these molecules. Use of a stronger catalyst like Pd black resulted in the hydrogenation of 1 and 12a to corresponding hydrogenated compounds 14 and 15. Hydrogenation of 13 and 12b were carried out under H2 in the presence of Pd/C catalyst to afford compounds 16 and 17.
Several additional derivatives of compound 1 were created by incorporating different substituents in the place of the carboxylic acid group of compounds 1. Derivative included the following substitutions: CH2OH (18a), CHO (19a) or CONH2 (20). Substitution with CH2OH and CHO groups did not result in a major change in the activity (18a, IC50 = 73 μM and 19a, IC5O = 77 μM), while substitution with CONH2 group resulted in a decrease in activity as compared to compound 1 (20, IC50 = 105 μM). Derivatives of the furan compound 13 were also created by incorporating different substituents in the place of the carboxylic acid group of compounds 13. Derivative included the following substitutions: CH2OH (18b) and CHO (19b). These compounds showed improved inhibition as compared to parent furan compound 13. Compound 18b showed an ICs0 value of 111 μM and compound 19b showed an IC50 value of 107 μM. Synthesis of compounds 18a, 18b, 19a and 19b is outlined in Scheme 3 (FIG. 7). Compounds 12a and 12b were reduced using DIBAL in a mixture of anhydrous dichloromethane and THF to afford the alcohol derivatives 18a and 18b. Oxidation of alcohols using PCC in anhydrous THF afforded the aldehydes 19a and 19b. Synthesis of amide compound 20 is outlined in Scheme 4 (FIG. 8). Compound 20 was prepared from compound 1 by treatment with SOCl2 followed by ammonia.
Synthesis of compounds 21-24 is outlined in Scheme 5 (FIG. 9). These compounds were designed to evaluate the nature of the linking group I on the activity of the compounds. Generally, the compounds containing triple bonds showed less activity than compounds containing double bonds. Compounds 21 and 22 showed IC50 values of 165 and 183 μM, respectively (Table 2). Compounds containing a double bond in the cis configuration were more effective, with compounds 23 and 24 showing ICs0 values of 61 and 154 μM, respectively (Table 2). The synthesis started from the known 3-(thiophen-2-yl)propiolic acid 35. The acid 35 was coupled with the amino compound 36 in the presence of EDAC and DMAP in 1,2- dichloroethane afforded the alkyne ester 21. The ester group present in compound 21 was hydrolyzed using NaOH in MeOH to afford the acetylenic acid 22. The partial hydrogenation of the acetylenic ester 21 using Lindlar Pd catalyst did not work as expected possibly due to the catalytic poisoning effect of the thiophene ring already present in the molecule. However, reduction worked smoothly to a cis double bonded product 23 when it was hydrogenated with H2 in the presence of Pd/C in ethyl acetate. The ester group present in compound 23 was hydrolyzed using NaOH in MeOH to afford the cis acid 24.
Synthesis of compounds 25-28 is outlined in Scheme 6 shown in FIG. 10. These compounds were designed to evaluate substitutes at the at the nitrogen group. Generally, replacement of the H with a methyl group reduced the effectiveness of the compounds (IC50 values fro compounds 25-28 were over 500 μM, Table 2). Compound 37 and 38 were methylated using methyl iodide in the presence of NaH in DMF to afford the N-methylated esters 25 and 26. Basic hydrolysis of 25 and 26 using IN. NaOH in MeOH afforded the final acid products 27 and 28, respectively.
Synthesis is of compound 29-30 is outlined in Scheme 7 shown in FIG. 11. These compounds were designed to evaluate the nature of the linking group I on the activity of the compounds. Compounds 29-30 contain a C3 alkenyl as the linking group as compared to a C2 alkenyl linking group in the previous compounds. Generally, the compounds containing a C3 alkenyl as the linking group I showed less activity than compounds containing a C2 alkenyl as the linking group I. Compounds 29 and 30 showed IC50 values of about 250 μM and greater (Table 2). Reductive amination of the known aldehyde 39 with the amine 36 in the presence of NaCNBH3 and ZnCl2 in methanol afforded the ester compound 29. Basic hydrolysis of compound 29 using NaOH in MeOH afforded the final acid product 30.
Synthesis of the compounds 31-34 is outlined in Scheme 8 shown in FIG. 12. These compounds were designed to evaluate substitution of the morpholine group the activity of the compounds. Compounds 31-34 contain a piperidine group rather than a morpholine group present in the previous compounds. Generally, the compounds containing a piperidine group showed similar activity to compounds containing a morpholine group. Compounds 31-34 showed IC50 values of 92 μM, 131 μM, 181 μM and 463 μM, respectively (Table 2). Commercially available methyl 2-fluoro-5-nitro benzoate (40) was treated with piperidine in DMF to afford the compound 41, which upon hydrogenation using H2 in the presence of Pd/C gave the amine 42. Compound 42 was coupled with trøn.s-3-(thiophene-2-yl) acrylic acid or trø«s-3-(furan-2-yl) acrylic acid in the presence of EDAC and DMAP in 1 ,2-dichloroethane to form the amides compound 31 or 32. Basic hydrolysis of the ester methyl group present in compound 31 and 32 afforded the compounds 33 and 34.
Synthesis of compounds 43-46 is outlined in Scheme 9 shown in FIG. 13. These compounds were designed to evaluate substitution of the compounds at the R2 position on the activity of the compounds. Compounds 43-46 contain various alkyl and aralkyl substituents at the R2 position in place of the esters and acids generally present on the parent compounds. Generally, the compounds 43-46 showed similar IC50 values to previous compounds (see for example, 18a,b and 19a,b). Compounds 43-46 showed IC50 values of 95 μM, 104 μM, 80 μM and 78 μM, respectively (Table 2). Compound 13 (1 mmol) was alkylated with alkyl bromides, R-Br (1.2 mmol) in the presence Of K2CO3 (2 mmol) in anhydrous N,N-dimethylformamide (5 mL) at room temperature for 12 h. TLC examination (50% EtOAc in CHCl3) showed that the reaction is complete. The solvent was completely removed under high vacuum and the residue obtained was dissolved in EtOAc (20 mL). The solution was washed with IM. NaHCO3 (2 x 15 mL), water (2 x 15 mL), brine (1 x 15 mL) and dried over anhydrous Na2SO4. The drying agent was filtered off and the solvent was evaporated to obtain the crude product. It was purified by column chromatography over silica gel using EtOAc / hexanes (1 :1) as eluent to afford the pure products 43-46.
Synthesis of compounds 51-56 is outlined in Scheme 10 shown in FIG. 14. These compounds were designed to evaluate substitution of the compounds at the position of the 5- membered (left most) ring on activity of the compounds. Compounds 51-56 contain benzyl and piperidine in place of thiophene and furan groups generally present on the parent compounds. Generally, the compounds 51-56 showed similar IC50 values to previous compounds. Compounds 51-56 showed IC50 values of 208 μM, 245 μM, >300 μM, n.d.,204 μM and 242 μM, respectively (Table 2). To a solution of 10 (1 mmol) in 1 ,2-dichloroethane (20 mL) aryl acrylic acid (1.2 mmol), EDAC (2 mmol), and DMAP (0.1 mmol) were added and stirred for 16 h at room temperature. TLC analysis (50% EtOAc in CHCl3) revealed that the reaction is complete. The reaction mixture was diluted with 1 ,2-dichloroethane (50 mL) and the solution was washed with saturated NaHCO3 solution (2 x 25 mL), water (1 x 25 mL), brine (1 x 25 mL) and dried over anhydrous Na2SO4. The drying agent was filtered off and the solvent was concentrated in vacuo to afford the crude product which was purified by column chromatography over silica gel using EtOAc / hexanes (1 : 1) as eluent to afford the pure products 51, 52 and 55. Compounds 51, 52 and 55 (1 mmol) in a mixture of THF (3 mL) and MeOH (3 mL) were treated with 3N. NaOH solution (2.5 mL) at room temperature and the resulting mixture was refluxed gently for 45 min. TLC analysis (50% EtOAc in CHCl3) revealed that the reaction is complete. The solvent was completely removed in vacuo and the residue obtained was taken up in water (3 mL). The resulting mixture was cooled to 0 °C and acidified to pH ~2 by carefully adding 3N HCl. The resulting white solid was filtered and dried under vacuum to furnish pure compounds 53, 54 and 56.
Synthesis of compound 57 is outlined in Scheme 11 showin in FIG. 15. This compounds represents a new strucutral formula for sortase inhibitors different from the strcuture of the compounds in formulas Ia-Ic. Commercially available 4-hydroxy acetophenone (68) is refluxed with 4-chloronitrobenzene in the presence of KOH in DMF afforded the ether derivatice 69. Coupling of ether 69 with N,N-dimethylformamide dimethyl acetal in DMF afforded the compound 57.
Compounds 58, 60-63 and 67 are presented in Table 2. These compounds were generated as a result of the SAR studies discussed above. Compounds 58, 60-63 and 67 are represented by the general formula Ic and share structural similarity to the general formulas provided in structures Ia and Ib. Specifically, these compounds provide additional variation along the phenyl group to provide configurations similar to the morpholine and piperidine groups present in many of the previous compounds. Compounds 59, 64 and 66 also share similarity with the compounds discussed above in that they share a thiophene group linked to an N group via a C2-C3 alkenyl linkage containing at least one double bond. Although compounds 58-64 and 66- 67 have not yet been tested against sortase enzymes in vitro, they can be expected to display inhibitory activity based on the similarity to the compounds disclosed herein. Pharmaceutical Compositions and Modes of Administration
The present disclosure provides compounds of the general formula (I), (Ia), (Ib), (Ic) and II-XII and pharmaceutical compositions comprising the same. In a specific embodiment, the present disclosure provides compounds 12-13, 18-20, 23-24, 31-32 and 43-46 and pharmaceutical compositions comprising the same. In one embodiment, these compounds are useful as inhibitors of bacterial sortase enzymes. In a particular embodiment, these compounds are useful as inhibitors of bacterial SrtA, SrtB, SrtC and/or SrtD or homologs thereof. In a further particular embodiment, the compounds are useful as inhibitors of bacterial SrtA. The compounds of the present disclosure have utility in treating and/or preventing a variety of disease states and/or conditions involving bacterial infection and virulence, wherein said bacteria causing or contributing to the bacteria infection and virulence express a sortase enzyme.
The present disclosure provides for a pharmaceutical composition comprising a pharmaceutically effective amount of at least one compound of the present disclosure as described herein. Such pharmaceutical compositions may be used in the manufacture of a medicament for treating and/or preventing a disease or condition in which it is desirable to inhibit involving bacterial infection and virulence. Such pharmaceutical compositions and medicaments may also comprise a pharmaceutically acceptable carrier. The compounds of the disclosure are useful in both free form and in the form of pharmaceutically acceptable salts.
The pharmaceutically acceptable carriers described herein, including, but not limited to, vehicles, adjuvants, excipients, or diluents, are well-known to those who are skilled in the art. Typically, the pharmaceutically acceptable carrier is chemically inert to the active compounds and has no detrimental side effects or toxicity under the conditions of use. The pharmaceutically acceptable carriers can include polymers and polymer matrices.
The compounds described in the instant disclosure can be administered by any conventional method available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in combination with additional therapeutic agents.
The compounds described are administered in therapeutically effective amount. The therapeutically effective amount of the compound and the dosage of the pharmaceutical composition administered will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration, the age, health and weight of the recipient; the severity and stage of the disease state or condition; the kind of concurrent treatment; the frequency of treatment; and the effect desired. A daily dosage of active ingredient can be expected to be about 0.001 to 1000 milligrams (mg) per kilogram (kg) of body weight. In one embodiment, the total amount is between about 0.1 mg/kg and about 1000 mg/kg of body weight; in an alternate embodiment between about 1.0 mg/kg and about 100 mg/kg of body weight; in yet another alternate embodiment between 0.1 mg/kg and about 30 mg/kg of body weight. The above described amounts may be administered as a series of smaller doses over a period of time if desired. As would be obvious, the dosage of active ingredient may be given other than daily if desired.
The total amount of the compound administered will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side effects that might accompany the administration of the compound and the desired physiological effect. It will be appreciated by one skilled in the art that various conditions or disease states, in particular chronic conditions or disease states, may require prolonged treatment involving multiple administrations.
Dosage forms of the pharmaceutical compositions described herein (forms of the pharmaceutical compositions suitable for administration) contain from about 0.1 mg to about 500 mg of active ingredient (i.e. the compounds disclosed) per unit. In these pharmaceutical compositions, the active ingredient will ordinarily be present in an amount of about 0.5-95% weight based on the total weight of the composition. Multiple dosage forms may be administered as part of a single treatment. The active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups and suspensions. It can also be administered parenterally, in sterile liquid dosage forms. The active ingredient can also be administered intranasally (nose drops) or by inhalation via the pulmonary system, such as by nebulizers, propellant based metered dose inhalers or dry powders inhalation devices. Other dosage forms are potentially possible such as administration transdermally, via a patch mechanism or an ointment.
Formulations suitable for oral administration can consist of (a) liquid solutions, such as a pharmaceutically effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined pharmaceutically effective amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, propylene glycol, glycerin, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms can include one or more of the following: lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acadia, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti -oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the patient, and aqueous and non- aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound can be administered in a physiologically acceptable diluent in a pharmaceutically acceptable carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol such as poly(ethyleneglycol) 400, glycerol ketals, such as 2,2-dimethyl-l,3-dioxolane-4-methanol, ethers, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants.
Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters. Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyldialkylammonium halides, and alkylpyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl .beta.-aminopropionates, and 2-alkylimidazoline quaternary ammonium salts, and (e) mixtures thereof.
The parenteral formulations typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5% to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
Pharmaceutically acceptable excipients are also well-known to those who are skilled in the art. The choice of excipient will be determined in part by the particular compound, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention. The following methods and excipients are merely exemplary and are in no way limiting. The pharmaceutically acceptable excipients preferably do not interfere with the action of the active ingredients and do not cause adverse side-effects. Suitable carriers and excipients include solvents such as water, alcohol, and propylene glycol, solid absorbants and diluents, surface active agents, suspending agent, tableting binders, lubricants, flavors, and coloring agents.
The compounds of the present disclosure, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, and nitrogen. Such aerosol formulations may be administered by metered dose inhalers. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
The formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets. The requirements for effective pharmaceutically acceptable carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J.B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, Eds., 238- 250 (1982) and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., 622-630 (1986).
Formulations suitable for topical administration include pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, as well as creams, emulsions, and gels containing, in addition to the active ingredient, such carriers as are known in the art.
Additionally, formulations suitable for rectal administration may be presented as suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
One skilled in the art will appreciate that suitable methods of administering a compound of the present invention to an patient are available, and, although more than one route can be used to administer a particular compound, a particular route can provide a more immediate and more effective reaction than another route. Methods of Treatment and Prevention
In one embodiment, the teachings of the present disclosure provide for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in a method of treating a bacterial infection or a disease state and/or condition which is caused by, at least in part, bacterial infection and/or virulence. In one embodiment, such disease states and conditions include, but are not limited to, hospital-acquired bacterial infections, endocarditis, osteomyelitis, septic arthritis pneumonia, toxic shock syndrome, skin infections, meningitis, anthrax, B. cereus food poisoning, tetanus, clostridial myonecrosis, pneumonia, toxic shock syndrome, skin infections, meningitis, ear infections, strep throat, rheumatic fever, scarlet fever, puerperal fever, listeriosis, perinatal septicemia, encephalitis, intrauterine infections sinusitis and severe invasive infections; however, other disease states and conditions may be treated as would be obvious to one of ordinary skill in the art.
The method of treatment comprises the steps of providing such composition or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection and/or virulence in a patient in need of such treatment. One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure. By decreasing, at least in part, the activity of a bacterial sortase enzyme, the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject and/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the disease state and/or condition which is caused by, at least in part, bacterial infection and/or virulence is thereby treated in the subject. In an alternate embodiment, the teachings of the present disclosure provide for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in a method of preventing or suppressing a bacterial infection or a disease state and/or condition which is caused by, at least in part, bacterial infection and/or virulence. In one embodiment, such disease states and conditions include, but are not limited to, hospital- acquired bacterial infections, endocarditis, osteomyelitis, septic arthritis pneumonia, toxic shock syndrome, skin infections, meningitis, anthrax, B. cereus food poisoning, tetanus, clostridial myonecrosis, pneumonia, toxic shock syndrome, skin infections, meningitis, ear infections, strep throat, rheumatic fever, scarlet fever, puerperal fever, listeriosis, perinatal septicemia, encephalitis, intrauterine infections sinusitis and severe invasive infections; however, other disease states and conditions may be treated as would be obvious to one of ordinary skill in the art.
The method of preventing or suppressing comprises the steps of providing such compound or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection and/or virulence in a patient in need of such preventing or suppressing. One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure. By decreasing, at least in part, the activity of a bacterial sortase enzyme, the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject and/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the disease state and/or condition which is caused by, at least in part, bacterial infection and virulence is thereby prevented or suppressed in the subject.
In a specific embodiment, the disease state or condition caused, at least in part, by a bacterial infection or virulence is endocarditis. Endocarditis may be related infection of the endocardium and/or the heart valves by bacteria. Staphylococcus aureus is a common cause of endocarditis. Through inhibition of the sortase enzyme, such as, but not limited to SrtA, the ability of bacteria, such as, but not limited to, Staphylococcus aureus, to bind to and infect the endocardium and/or heart valves may be reduced; furthermore, pre-existing bacterial infections may be eliminated. The foregoing leads to a decrease in disease. In yet another alternate embodiment, the present disclosure provides for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in methods of reducing or eliminating bacterial virulence in a subject in need of such reduction. The method of reducing or eliminating comprises the steps of providing such compound or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to reduce or eliminate bacterial virulence in a patient in need of such reducing or eliminating. One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure. By reducing or eliminating, at least in part, the activity of a bacterial sortase enzyme, the compounds of the present disclosure reduce the virulence of the bacteria towards the host. As a result, health of the subject is improved.
In yet a further alternate embodiment, the present disclosure, provides for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in methods of treating bacterial infection.
The method of treatment comprises the steps of providing such composition or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection in a patient in need of such treatment. One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure. By decreasing, at least in part, the activity of a bacterial sortase enzyme, the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject an/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the bacterial infection is thereby treated in the subject. In still a further alternate embodiment, the present disclosure provides for the use of a compound of the present disclosure or pharmaceutical compositions containing a compound of the present disclosure in methods of preventing or suppressing bacterial infection.
The method of preventing or suppressing comprises the steps of providing such compound or pharmaceutical composition and administering such compound or pharmaceutical composition in a therapeutically effective amount to inhibit or reduce bacterial infection in a patient in need of such preventing or suppressing. One or more compounds of the present disclosure may be used or the pharmaceutical composition may contain one or more compounds of the present disclosure. By decreasing, at least in part, the activity of a bacterial sortase enzyme, the compounds of the present disclosure reduce the ability of the bacteria to infect the cells of the subject an/or reduce the virulence of the bacteria towards the host. Furthermore, the viability of the bacteria is also reduced. As a result, the bacterial infection is thereby prevented or suppressed in the subject.
In the foregoing methods of use, the bacteria may be any bacteria or other organism that contains and expresses a sortase enzyme, hi one embodiment, the sortase enzyme is SrtA, SrtB, SrtC, and/or SrtD, and/or homologues of any of the foregoing. In a specific embodiment, the sortase enzyme is SrtA. In another embodiment, the bacteria is Staphylococcus aureus. Furthermore, the bacteria may one that is resistant to a commonly used antibiotic, such as methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococcus (VRE), vancomycin-intermediate Staphylococcus aureus (VISA) and vancomycin-resistant Staphylococcus aureus (VRSA). The bacteria may be Gram-positive bacteria, Gram-negative bacteria or combinations of the foregoing that expresses a sortase enzyme; in one embodiment, the bacteria is a Gram-positive bacteria that expresses the sortase enzyme. Representative bacteria that have been documented to express a sortase enzyme, include, but are not limited to, Acidothermus cellulolyticus, Actinomyces naeslundii, Actinomyces viscosus, Arthrobacter aurescens, Arthrobacter sp. FB24, Bacillus anthracis, Bacillus cereus, Bacillus clausii, Bacillus halodurans, Bacillus licheniformis, Bacillus subtilis, Bacillus thuringiensis, Bacillus weihenstephanensis, Bifidobacterium longum, Bradyrhizobium japonicum, Chloroflexus aurantiacus, Clostridium acetobutylicum, Clostridium difficile.Clostridium limosum, Clostridium perfringens, Clostridium tetani, Congregibacter litoralis, Corynebacterium diphtheriae, Corynebacterium efficiens, Corynebacterium glutamicum, Corynebacterium jeikeium, Desulfitobacterium hafniense, Enterococcus faecalis, Enterococcus faecium, Exiguobacterium sibiricum, Exiguobacterium sp. 255-15, Frankia sp. CcB, Fran/da sp. EAN] pec, Geobacillus kaustophilus, Hahella chejuensis, Kineococcus radiotolerans, Lactobacillus acidophilus, Lactobacillus brevis, Lactobacillus casei, Lactobacillus delbrueckii, Lactobacillus johnsonii, Lactobacillus plantarum, Lactobacillus reuteri, Lactobacillus sakei, Lactobacillus salivarius, Lactococcus lactis, Leifsonia xyli, Leuconostoc mesenteroides, Listeria innocua, Listeria monocytogenes, Listeria welshimeri, Marinobacter hydrocarbonoclasticus, Nocardioides sp. JS614, Oceanobacillus iheyensis, Pediococcus pentosaceus, Pseudoalteromonas atlantica, Pseudomonas mendocina, Rhodobacter sphaeroides, Rubrobacter xylanophilus, Saccharophagus degradans, Shewanella amazonensis, Shewanella baltica, Shewanella frigidimarina, Shewanella oneidensis, Shewanella sp. ANA-3, Shewanella sp. MR-4, Shewanella sp. MR-7, Shewanella sp. W3-18-1, Solibacter usitatus, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus haemolyticus, Staphylococcus saprophyticus, Streptococcus agalactiae, Streptococcus gordonii, Streptococcus mutans, Streptococcus oralis, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus sanguinis, Streptococcus suis, Streptococcus thermophilus, Streptomyces avermitilis, Streptomyces coelicolor, Symbiobacterium thermophilum, Thermobifida fusca and Tropheryma whipplei. Representative other organisms that have been demonstrated to contain a sortase enzyme include, but are not limited to, Methanothermobacter thermautotrophicus. However, any bacteria or organism that expresses a sortase enzyme may be subject to the methods of the present disclosure.
Furthermore, the methods of use described herein may further comprise the administration of a therapeutically effective amount of one or more additional agents to further improve the efficacy of the methods described herein. The additional agents may be administered at the same time or a different time than a compound of the present disclosure. In one embodiment, the additional agent has a mechanism of action that is distinct from the compounds of the present disclosure (i.e., the second compound is not an inhibitor of a sortase enzyme). As the mechanisms of action the additional agent and the compounds of the present disclosure are different, the combination of the compounds of the present disclosure and the additional agent are expected to be effective in the methods described herein. The identity and nature of the additional agent may be determined by the healthcare provider based on his/her knowledge of the subject's condition.
In one embodiment, the additional agent is an antibiotic. Any antibiotic or antibiotic combinations may be used as the additional agent. As the compounds of the present disclosure operate in a manner that is different and distinct from the mechanisms of action of currently known antibiotics, the combination of the compounds of the present disclosure and antibiotics are expected to be effective in the methods described herein. Representative antibiotics may be selected from the classes of aminoglycosides, ansamycins, carbacephem, carbapenems, cephalosporins, glycopeptides, macrolides, monobactams, penicillins, polypeptides, quinolones, sulfonamides and/or tetracyclines; other classes of antibiotics may also be used. Exemplary antibiotics include, but are not limited to, Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Streptomycin, Tobramycin, Geldanamycin, Herbimycin, Loracarbef, Ertapenem, Imipenem/Cilastatin, Meropenem, Cefadroxil, Cefazolin, Cephalexin, Cefaclor, Cefamandole, Cefoxitin, Cefprozil, Cefuroxime, Cefixime, Cefdinir, Cefditoren, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefepime, Teicoplanin, Vancomycin, Methicillin, Azithromycin, Clarithromycin, Dirithromycin, Erythromycin, Roxithromycin, Troleandomycin, Aztreonam, Amoxicillin, Ampicillin, Azlocillin, Carbenicillin, Cloxacillin, Dicloxacillin, Flucloxacillin, Mezlocillin, Nafcillin, Penicillin, Piperacillin, Ticarcillin, Bacitracin, Trimethoprim Colistin, Polymyxin B, Ciprofloxacin, Enoxacin, Gatifloxacin, Levofloxacin, Lomefloxacin, Moxifloxacin, Norfloxacin, Ofloxacin, Trovafloxacin, Mafenide, Prontosil, Sulfacetamide, Tobramycin Sulfamethizole, Sulfanilimide, Sulfasalazine, Sulfisoxazole, Trimethoprim, Trimethoprim-Sulfamethoxazole (Co-trimoxazole) (TMP-SMX), Demeclocycline, Doxycycline, Minocycline, Oxytetracycline, Tetracycline, Arsphenamine, Chloramphenicol, Clindamycin, Ethambutol, Fosfomycin, Fusidic acid, Furazolidone, Isoniazid, Linezolid, Metronidazole, Mupirocin, Nitrofurantoin, Platensimycin, Pyrazinamide, Quinupristin/Dalfopristin, Rifampin, Spectinomycin and/or Telithromycin. In a specific embodiment, the antibiotic is methicillin and/or vancomycin. In this embodiment, the co-administration of the foregoing agents with a sortase inhibitor of the present disclosure increases the efficacy of the foregoing antibiotics against their bacterial targets.
The foregoing description illustrates and describes the compounds of the present disclosure. Additionally, the disclosure shows and describes only certain embodiments of the compounds but, as mentioned above, it is to be understood that the teachings of the present disclosure are capable of use in various other combinations, modifications, and environments and is capable of changes or modifications within the scope of the inventive concept as expressed herein, commensurate with the above teachings and/or the skill or knowledge of the relevant art. The embodiments described hereinabove are further intended to explain best modes known of practicing the invention and to enable others skilled in the art to utilize the invention in such, or other, embodiments and with the various modifications required by the particular applications or uses of the invention. Accordingly, the description is not intended to limit the invention to the form disclosed herein. The claim below is appended for purposes of foreign priority only, if required. All references cited herein are incorporated by reference as if fully set forth in this disclosure. METHODS
Fluorescence Resonance Energy Transfer (FRET") assay
To determine IC50 values each potential inhibitor, the previously published fluorescenceresonance energy transfer (FRET) assay was used. Kruger, R. G.; Dostal, P.; McCafferty, D. G., Development of a high-performance liquid chromatography assay and revision of kinetic parameters for the Staphylococcus aureus sortase transpeptidase SrtA. Anal Biochem 2004, 326, (1), 42-8; Ton-That, H.; Mazmanian, S. K.; Faull, K. F.; Schneewind, O., Anchoring of surface proteins to the cell wall of Staphylococcus aureus. Sortase catalyzed in vitro transpeptidation reaction using LPXTG peptide and NH(2)-Gly(3) substrates. J Biol Chem 2000, 275, (13), 9876-81. This FRET assay employs the use of the donor and quencher pair, EDANS and Dabcyl, respectively. The peptide Dabcyl-QALPETGEE-EDANS was purchased from a commercial source. The EDANS fluorophore has an excitation wavelength, λex>d = 336 nm and an emission maximum, λemjd = 490 nm. The fluorescence emission spectra for EDANS overlaps very well with the absorption spectra of Dabcyl, where Dabcyl has an absorption maximum, λmax,a = 472 nm. As such, when the two molecules are spatially close, as they are in the intact peptide, the efficiency of transfer from EDANS to Dabcyl is high. Upon excitation of EDANS little emission from the donor is observed because most of the energy from fluorescence is transferred to the dabcyl acceptor and lost through non radiative decay pathways. In contrast, upon cleavage the newly formed fragments will diffuse apart and the two peptide fragments will become spatially separated and the FRET efficiency will diminish. Thus, the emission from the EDANS fluorophore will precipitously increase. Because of these properties, this FRET pair has been successfully employed to monitor SrtA catalyzed peptide cleavage.
Traditionally, this assay is performed in a fluorometer under steady-state or multiple turnover conditions, where steady-state conditions are defined as the substrate being in large excess over the enzyme. In this experimental design a constant enzyme concentration is titrated with increasing concentrations of the peptide substrate. The EDANS fluorophore is excited at 350 nm and fluorescence from EDANS is observed at 495 nm over a period of time. To reduce the correction for the inner filter effect the reaction was monitored at an emission of 590 nm. At the beginning of the observation, fluorescence is low due to a high FRET efficiency, but, as the peptide is cleaved the fluorescence increases linearly. The slope of this linear increase is taken to be the initial velocity, V0. In principle, the initial velocity as a function of peptide concentration will exhibit Michaelis-Menten kinetics, i.e. a hyperbolic dependence on substrate concentration, and can be analyzed to yield the steady-state kinetic constants Kn, and kcat. Because of the large inner filter correction at high concentrations of substrate, the effect of inhibitors on V0 was monitored at select enzyme and substrate concentration. These experiments are performed by mixing 5 μM SrtA with 20 μM Dabcyl-EDANS labeled peptide in a 0.5 cm cuvette. This mixture is excited at λex = 350 nm and fluorescence is observed at λem = 590 nm. From this experiment, a time course of fluorescence increase as a function of time is acquired. The slope of this line is considered the maximum velocity in the absence of inhibitor. The identical experiment is then performed in the presence of increasing concentrations of inhibitor and the slopes are compared until a 50 % decrease in the slope is observed. The concentration of inhibitor where 50 % of this maximum velocity is observed is taken to be the IC50. Each experiment was repeated at least three times to ensure the reproducibility and calculate standard deviation of the reported IC50 values. General Methods for Synthesis
Solvent evaporations were carried out in vacuo with a rotary evaporator. Analytical samples were prepared by drying the samples in vacuo (0.2 mmHg) in an Abderhalden drying apparatus over P2O5 and ethyl acetate at reflux temperature. Thin layer chromatography (TLC) was performed on silica gel plates with fluorescent indicator (Whatmann, silica gel, UV254, 25 μm plates). Spots were visualized by UV light (254 and 365 ran). AU analytical samples were single spots on TLC in at least two different solvent systems. Purification by column and flash chromatography was carried out using 'BAKER' silica gel (40 μm) in the solvent systems indicated. The amount (weight) of silica gel for column chromatography was in the range of 50- 100 times the amount (weight) of the crude compounds being separated. Melting points were determined on a Mel-Temp II melting point apparatus and are uncorrected. Proton nuclear magnetic resonance (1H NMR) and carbon nuclear magnetic resonance (13C NMR) spectra were recorded on a Brucker DPX-300 spectrometer using TMS as internal standard. The values of chemical shifts (δ) are given in ppm and coupling constants (J) in Hz. The chemical shift values are reported as parts per million (ppm) relative to tetramethylsilane as internal standard. Mass spectra were recorded on a Micromass Platform LCC instrument. Elemental analyses were performed by Atlantic Microlab, Norcross, Georgia and the results indicated by symbols for the elements were within ±0.4% of theoretical values. Anhydrous solvents used for reactions were purchased in Sure-Seal™ bottles from Aldrich Chemical Company. Other reagents were purchased from Aldrich, Lancaster or Acros chemical companies and used as received. Methyl 5-amino-2-morpholinobenzoate (Compound 10)
To a solution of Methyl 5-nitro-2-morpholinobenzoate 9 (1.0 g, 3.76 mmol) in EtOAc (35 mL) 10% PdVC (100 mg) was added and stirred under an atmosphere of H2 from a balloon (~1 atm) for 12 h. TLC analysis (1 :1 EtOAcZCHCl3) revealed that the reaction was complete. The catalyst was removed by filtration through a celite bed and the filtrate was concentrated on vacuum to furnish 10 as a solid (0.88 g, 99 %): mp 121 - 122 0C; 1H NMR (CDCl3) δ 2.91 - 2.97 (m, 4H), 3.60 (bs, 2H), 3.79 - 3.85 (m, 4H), 3.87 (s, 3H), 6.78 (dd, IH, J1 = 3.0 Hz, J2 = 8.7 Hz), 6.95 (d, IH, J = 8.4 Hz) and 7.05 (d, IH, J = 2.7 Hz); 13C NMR (CDCl3) δ 51.7, 53.3, 67.1, 116.7, 118.8, 120.8, 127.0, 141.9, 143.7 and 168.0; MS (ES) m/z 237 (M + H); Anal. (C12H16N2O3) C5 H5 N. Methyl 5-((EV3-(thiophen-2-yl)acrylaniido)-2-rnorpholinobenzoate (Compound 12a) To a solution of 10 (0.430 g, 1.82 mmol) in 1 ,2-dichloroethane (20 mL) 3-(2- thienyl)acrylic acid (0.337 g, 2.18 mmol), EDAC (1.12 g, 5.82 mmol), and DMAP (0.022 mg, 0.182 mmol) were added and stirred for 16 h at room temperature. TLC analysis (1 :1 EtOAc/CHCl3) revealed that the reaction was complete. The reaction mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated NaHCO3 solution (2 x 25 mL), water (1 x 25 mL), brine (1 x 25 mL) and dried over anhydrous Na2SO4. The drying agent was filtered off and the solvent was concentrated in vacuo to afford the crude product which was crystallized form a mixture of CHCl3 and hexanes to furnish 12a as a yellow solid (0.62 g, 91%): mp 196 - 197 0C; 1H NMR (CDCl3) δ 3.00 - 3.05 (m, 4H), 3.83 - 3.88 (m, 4H), 3.89 (s, 3H), 6.31 (d, IH, J = 15.3 Hz), 7.03 - 7.08 (m, 2H), 7.24 - 7.28 (m, 2H), 7.33 - 7.38 (m,lH), 7.80 (d, IH, J = 8.7 Hz), 7.83 - 7.90 (m, IH) and 7.92 (d, IH, J = 2.3 Hz); 13C NMR (CDCl3) δ 52.2, 53.1, 67.2, 1 19.3, 119.8, 123.2, 124.6, 124.9, 127.9, 128.2, 130.9, 132.5, 135.1, 139.8, 149.0, 163.7 and 167.5; MS (ES) m/z 373 (M + H); Anal. (Ci9H20N2O4S) C, H, N. Methyl 5-((iπ-3-(furan-2-y0acrylamido)-2-morpholinobenzoate (Compound 12b) Compound 12b was synthesized, following a similar procedure as the one used for preparation of 12a starting from compound 10 (0.426 g, 1.8 mmol), 3-(2-furyl)acrylic acid (0.298 g, 2.16 mmol), EDAC (1.10 g, 5.76 mmol), and DMAP (0.022 mg, 0.182 mmol) to obtain the pure product (0.639 g, 99 %): mp 172 - 173 °C; 1H NMR (CDCl3) δ 2.98-3.07 (m, 4H), 3.82-3.88 (m, 4H), 3.89 (s, 3H), 6.42 (d, IH, J = 15.0 Hz), 6.48 (dd, IH, J1 = 1.8 Hz, J2 = 3.3 Hz), 6.60 (d, IH, J = 3.6 Hz), 7.04 (d, IH, J = 9.0 Hz), 7.40 (bs, IH), 7.46 (d, IH, J = 1.5 Hz), 7.52 (d, IH, J = 15.0 Hz), 7.82 ( d, IH, J= 7.2 Hz) and 7.91 (s, IH); 13C NMR (CDCl3) δ 52.1, 53.0, 67.1, 112.3, 114.4, 118.3, 119.6, 123.2, 124.6, 128.8, 132.7, 144.2, 148.8, 151.1 (2C), 164.1 and 167.6; MS (ES) m/z 357 (M + H); Anal. (C19H20N2O5) C, H, N. 5-((E)-3-(Thiophen-2-yl)acrylamido)-2-morpholinobenzoic acid (Compound 1) To a solution of compound 12a (0.300 g, 0.81 mmol) in a mixture of THF (1.5 mL) and methanol (1.5 mL), 3N. NaOH solution (2.5 mL) was added at room temperature and the resulting mixture was refluxed gently for 45 min. TLC analysis (1 :1 ΕtOAc/CHCl3) revealed that the reaction was complete. The solvent was completely removed in vacuo and the residue obtained was taken up in water (3 mL). The resulting mixture was cooled to 0 0C and acidified carefully with 3N HCl (-2.5 mL). The resulting white solid was filtered and dried under vacuum to furnish pure compound 1 (0.283 g, 98%): mp 278-279 °C; 1H NMR (DMSO-d6) δ 3.00-3.09 (m, 4H), 3.74-3.85 (m, 4H), 6.55 (d, IH, J = 15.3 Hz), 7.14 (dd, IH, J, = 3.6 Hz, J2 = 5.1 Hz), 7.47 (d, IH, J = 3.3 Hz), 7.67 (s, IH), 7.68 (d, IH, J = 3.0 Hz), 7.77 (d, IH, J = 15.3 Hz), 7.99 (dd, IH, J1 = 3.6 Hz, J2 = 5.1 Hz), 8.29 (d, IH, J= 2.7 Hz) and 10.4 (bs, IH); 13C NMR (DMSO- d6) δ 52.7, 66.3, 120.3, 120.9, 123.8, 124.0, 125.4, 128.5, 128.7, 131.6, 133.7, 137.8, 139.6, 145.2, 163.4 and 166.4; MS (ES) m/z 357 (M- H); Anal. (Ci8H18N2O4S) C, H, N. 5-((E)-3-(Furan-2-yl)acrylamido)-2-moφholinobenzoic acid (Compound 13)
Compound 13 was synthesized following a similar procedure as the one used for preparation of compound 1 starting from 12b (0.200 g, 0.57 mmol) and 3N NaOH solution to afford the pure product as a white solid (0.192 g, 99%): mp 281-282 °C; 1H NMR (DMSO-d6) δ 3.01-3.11 (m, 4H), 3.75-3.86 (m, 4H), 6.59 (d, IH, J = 15.3 Hz), 6.60-6.69 (m, IH), 6.87 (d, IH, J = 3.6 Hz), 7.41 (d, IH, J = 15.6 Hz), 7.68 (d, IH, J = 9.0 Hz), 7.84 (d, IH, J = 1.5 Hz), 8.00 (dd, IH, J1 = 2.7 Hz, J2 = 8.7 Hz), 8.29 (d, IH, J = 2.7 Hz) and 10.4 (bs, IH); 13C NMR (DMSO-d6) δ 52.8, 66.4, 112.5, 115.0, 118.9, 120.9, 123.8, 124.0, 125.4, 127.8, 137.8, 145.2, 145.4, 150.8, 163.6 and 166.4; MS (ES) m/z 341 (M - H); Anal. (C18H18N2O5) C, H, N. 5-(3-(Thiophen-2-yl)propanamido)-2-morpholinobenzoic acid (Compound 14)
A solution of compound 1 (0.071 g, 0.20 mmol), ammonium formate (0.188 g, 2.97 mmol) and Pd black (40 mg) in a mixture of methanol (4 mL) and EtOAc (3 mL) was refluxed for 6 h. TLC analysis (1 :1 EtOAc/CHCl3) and mass spectral data revealed that the reaction was complete. The catalyst was removed by filtration through a celite bed, washed with a mixture of methanol and CHCl3, and the filtrate was concentrated under vacuum to afford the crude product. The crude product was purified by washing thoroughly with water to remove the inorganic salts, filtered and dried under vacuum to furnish pure product 14 (0.049 g, 69%): mp 226 - 227 °C; 1H NMR (DMSO-d6) δ 2.68 (t, 2H, J= 7.5 Hz), 3.13 (t, 2H, J= 7.5 Hz), 2.99 - 3.08 (m, 4H), 3.74 - 3.86 (m, 4H), 6.86 - 6.97 (m, 2H), 7.30 (dd, IH, J, = 1.0 Hz, J2 = 4.9 Hz), 7.65 (d, IH, J= 8.4 Hz), 7.90 (dd, IH, J1 = 2.7 Hz, J2 = 8.7 Hz), 8.21 (d, IH, J = 2.4 Hz) and 10.2 (bs, IH); 13C NMR (DMSO-d6) δ 24.9, 38.0, 52.7, 66.3, 120.8, 123.7, 123.8, 123.9, 124.7, 125.2, 126.9, 137.7, 143.4, 145.0, 166.3 and 170.1; MS (ES) m/z 361 (M + H); Anal. (Ci8H20N2O4S) C, H, N. Methyl 5-(3-(thiophen-2-yl)propanamido)-2-morpholinobenzoate (Compound 15)
A solution of compound 12a (0.100 g, 0.27 mmol), ammonium formate (0.255 g, 4.05 mmol) and Pd black (50 mg) in methanol (5 mL) was refluxed for 6 h. TLC analysis (1 :1 EtOAc/CHCl3) and mass spectrum revealed that the reaction was complete. The catalyst was removed by filtration through a celite bed, washed with chloroform, and the filtrate was concentrated under vacuum. The residue obtained was dissolved in CH2Cl2 (20 mL) and water (15 mL) was added. The organic layer was separated and washed with saturated NaHCO3 solution (2 x 15 mL), water (2 x 15 mL), brine (1 x 15 mL) and dried (Na2SO4). The drying agent was filtered off and the solvent was removed under vacuum to obtain the crude product. This crude product was purified by column chromatography over Si gel (4 x 2 cm) using 1 :5 EtOAc/CHCl3 as eluent to afford the pure compound 15 (0.082 g, 82 %): mp 115 - 116 "C; 1H NMR (CDCl3) δ 2.70 (t, 2H, J = 7.5 Hz), 2.97 - 3.05 (m, 4H), 3.27 (t, 2H, J = 7.2 Hz), 3.81 - 3.87 (m, 4H), 3.88 (s, 3H), 6.86 (dd, IH, J1 = 0.9 Hz, J2 = 3.3 Hz), 6.90 - 6.96 (m, IH), 7.01 (d, IH, J= 9.0 Hz), 7.11 (bs, IH), 7.14 (dd, IH, J1 = 1.2 Hz, J2 = 5.1 Hz), 7.64 (dd, IH, J1 = 2.7 Hz, J2 = 8.7 Hz) and 7.76 (d, IH, J = 8.7 Hz); 13C NMR (CDCl3) δ 25.5, 38.9, 52.0, 52.9, 67.0, 119.5, 123.2, 123.4, 124.6, 124.7 (2C), 126.8, 132.3, 143.0, 148.6, 167.4 and 170.1. MS (ES) m/z 375 (M + H). Anal. (Ci9H22N2O4S-O^SH2O) C, H, N. 5-(3-(Furan-2-yl)propanamido)-2-morpholinobenzoic acid (Compound 16)
To a solution of compound 13 (0.075 g, 0.22 mmol) in EtOAc (15 mL) 10% Pd/C (20 mg) was added and stirred under a hydrogen atmosphere from balloon (~1 atm) for 25 min. TLC analysis (1 :1 EtOAc: CHCl3) and 1H NMR of aliquot revealed that the reaction was complete. (Note: Prolonged reaction times lead to hydrogenation of furan ring). The catalyst was removed by filtration through a celite bed and the filtrate was concentrated on vacuum to afford the crude product which was purified by column chromatography over Si gel (4 x 2 cm) using EtOAc as eluent to afford pure compound 16 as a white solid (0.066 g, 88 %): mp 230 - 231 °C; 1H NMR (DMSO-d6) δ 2.65 (t, 2H, J= 7.5 Hz), 2.93 (t, 2H, J= 7.3 Hz), 3.00-3.07 (m, 4H), 3.73-3.83 (m, 4H), 6.11 (dd, IH, J1 = 1.2 Hz, J2 = 2.1 Hz), 6.34 (dd, IH, J1 = 1.8 Hz, J2 = 3.0 Hz), 7.51 - 7.52 (m, IH), 7.65 (d, IH, J = 8.7 Hz), 7.89 (dd, IH, J1 = 2.7 Hz, J2 = 8.7 Hz), 8.21 (d, IH, J = 2.7 Hz) and 10.2 (bs, IH); 13C NMR (DMSO-d6) δ 23.2, 34.5, 52.8, 66.4, 105.2, 1 10.4, 120.8, 123.8, 123.9, 125.3, 137.8, 141.5, 145.0, 154.4, 166.4 and 170.2; MS (ES) m/z 343 (M - H); Anal. (C18H20N2O5) C, H, N. Methyl 5-(3-(furan-2-yl)propanamido)-2-morpholinobenzoate (Compound 17)
Compound 17 was synthesized, following procedure used for the preparation of compound 16 starting from compound 12b (0.136 g, 0.38 mmol) in EtOAc (15 mL) and 10% Pd/C (20 mg) under a hydrogen atmosphere (0.106 g, 77 %): mp 119 - 120 °C; 1H NMR (CDCl3) δ 2.69 (t, 2H, J = 7.5 Hz), 2.97-3.03 (m, 4H), 3.07 (t, 2H, J = 7.3 Hz), 3.81-3.87 (m, 4H), 3.88 (s, 3H), 6.07 (d, IH, J= 3.3 Hz), 6.27-6.32 (m, IH), 7.01 (d, IH, J= 9.0 Hz), 7.16 (bs, IH), 7.32 (d, IH, J = 1.2 Hz), 7.65 (dd, IH, J1 = 2.7 Hz, J2 = 8.7 Hz) and 7.78 (d, IH, J = 2.4 Hz); 13C NMR (CDCl3) δ 23.7, 35.4, 52.0, 52.9, 67.1, 105.5, 110.2, 119.5, 123.2, 124.6, 124.7, 132.3, 141.1, 148.7, 154.0, 167.5 and 170.3; MS (ES) m/z 357 (M - H). Anal. (Ci9H22N2O5O^SH2O) C, H, N.
(EVN-(3-(Hvdroxymethyl)-4-moφholinophenyl)-3-(thiophen-2-vDacrylamide (Compound 18a) To a solution of compound 12a (0.500 g, 1.34 mmol) in a mixture of anhydrous CH2Cl2 (35 mL) and THF (5 mL), DIBAL-H (6.7 mL, IM in dichloromethane, excess) was added and stirred at 0 °C for 1 h. Then it was allowed to attain room temperature at which it was stirred for an additional 2 h. TLC analysis (1 :1 EtOAc/CHCl3) revealed that the reduction was complete. The reaction mixture was cooled to 0 °C and was quenched by careful addition of methanol (3 mL) and IN. HCl (0.5 mL). The solvent was removed under reduced pressure and the residue obtained was dissolved in CHCl3 (300 mL) and washed with water (2 x 75 mL), brine (1 x 150 mL) and dried (Na2SO4). The drying agent was filtered off and the solvent was removed under vacuum to afford the crude product which was purified by flash column chromatography on silica gel (20 x 3 cm) using EtOAc/hexanes (3: 1) as eluent to furnish compound 18a as a yellow solid (0.354 g, 77 %): mp 184 - 185 °C; 1H NMR (DMSOd6) δ 2.79 - 2.83 (m, 4H), 3.65 - 3.75 (m, 4H), 4.54 (d, 2H, J- 5.1 Hz), 5.12 (t, IH, J= 5.4 Hz), 6.58 (d, IH, J = 15.0 Hz), 7.02 (d, IH, J = 8.7 Hz), 7.13 (dd, IH, J1 = 3.6 Hz, J2 = 5.1 Hz), 7.42 (d, IH, J= 3.3 Hz), 7.60-7.74 (m, 4H) and 10.1 (bs, IH); 13C NMR (DMSOd6) δ 52.7, 58.5, 66.6, 118.1, 119.0, 119.1, 121.1, 128.3, 128.4, 131.1, 132.7, 135.0, 137.1, 139.8, 145.6 and 162.9; MS (ES) m/z 343 (M - H); Anal. (C18H20N2O3S) C5 H5 N. (E)-3-(Furan-2-vπ-N-(3-(hvdroxymethyl)-4-morpholinophenvπacrylamide (Compound 18b).
Compound 18b was synthesized, following the procedure used for preparation of 18a, by the reaction of 12b (0.500 g, 1.40 mmol) and DIBAL-H (7.02 mL, IM in dichloromethane) in dichloromethane (35 mL) and THF (5 mL) for 2 h at room temperature to furnish pure product (0.372 g, 81 %): mp 182 - 183 0C; 1H NMR (DMSO - d6) δ 2.74-2.83 (m, 4H), 3.65-3.74 (m, 4H)5 4.54 (d, 2H, J = 5.1 Hz), 5.12 (t, IH, J = 5.4 Hz)5 6.58-6.67 (m, 2H), 6.82 (d, IH5 J = 3.3 Hz), 7.02 (d, IH, J= 8.7 Hz), 7.35 (d, IH, J= 15.3 Hz), 7.61-7.71 (m, 4H)5 7.81 (d, IH5 J= 1.5 Hz) and 10.1 (bs, IH); 13C NMR (DMSO - d6) δ 53.1, 59.0, 67.1, 113.0, 114.8, 118.7, 119.4, 119.6, 120.1, 127.3, 135.4, 137.5, 145.5, 146.0, 151.4 and 163.5; MS (ES) m/z 327 (M - H); Anal. (C18H20N2O4) C5 H5 N. (E)-N-(3-Formyl-4-morpholinophenyl)-3-(thiophen-2-yl)acrylamide (Compound 19a)
To a solution of compound 18a (0.500 g, 0.44 mmol) in anhydrous THF at 0 0C, pyridinium chlorochromate (0.188 g, 0.87 mmol) was added and stirred for 30 min and then at room temperature for 1 h. TLC analysis (1 :1 ΕtOAc/CHCl3) revealed that the reaction was complete. The solvent was removed under vacuum and the crude product obtained was purified by column chromatography over Si gel using EtOAc/CHCl3 (1 :4) as the eluent to afford pure 19a (0.107 g, 72 %): mp 218 - 219 °C; 1H NMR (DMSO) δ 2.94-3.03 (m, 4H), 3.72-3.82 (m, 4H)5 6.53 (d, IH, J= 15.3 Hz), 7.11-7.17 (m, IH), 7.25 (d, IH, J= 8.7 Hz)5 7.45 (d, IH5 J= 3.3 Hz), 7.66 (d, IH, J= 5.1 Hz), 7.74 (d, IH5 J= 15.6 Hz)5 7.88 (dd, IH, J1 = 2.4 Hz, J2 = 8.7 Hz), 8.06 (d, IH, J = 2.7 Hz)5 10.2 (s, IH) and 10.3 (bs, IH). 13C NMR (CD3COCD3) δ 55.3, 67.4, 120.1, 121.0, 121.4, 126.8, 128.7, 129.1, 130.1, 131.7, 134.6, 136.1, 141.0, 152.5, 164.2 and 191.0. MS (ES) m/z 341 (M - H). Anal. (Ci8Hi8N2O3S) C, H, N. (E)-N-(3-Formyl-4-moφholinophenyl)-3-(fiiran-2-vπacrylamide (Compound 19b)
Compound 19b was synthesized, following the procedure used for preparation of 19a, by the reaction of 18b (0.188 g, 0.57 mmol) and pyridinium chlorochromate (0.247 g, 1.15 mmol) in anhydrous THF (10 mL) to furnish the pure product (0.115 g, 61 %): mp 231 - 232 0C. 1H NMR (DMSO) δ 2.94-3.03 (m, 4H), 3.73-3.82 (m, 4H), 6.53-6.65 (m, 2H), 6.86 (d, IH, J= 3.6 Hz), 7.24 (d, IH, J = 9.0 Hz), 7.38 (d, IH, J = 15.6 Hz), 7.82 (d, IH, J= 1.5 Hz), 7.89 (dd, IH, Ji = 2.5 Hz, J2 = 8.8 Hz), 8.06 (d, IH, J = 2.7 Hz), 10.2 (s, IH) and 10.3 (bs, IH). 13C NMR (DMSO) δ 54.1, 66.2, 112.6, 114.8, 119.0, 119.1, 120.2, 126.0, 127.4, 128.2, 134.6, 145.3, 150.9, 151.2, 163.4 and 190.7. MS (ES) m/z 325 (M - H). Anal. (Ci8Hi8N2O4) C, H, N. 5-((E)-3-(Thiophen-2-yl)acrylamido)-2-morpholinobenzamide (Compound 20)
To a solution of compound 1 (0.050 g, 0.14 mmol) in anhydrous dichloromethane (4 mL) at room temperature a mixture of thionyl chloride (0.2 mL) and anhydrous DMF (0.2 mL) was added and the resulting mixture was stirred for 4 h at room temperature. The solvent was removed under reduced pressure. The resulting gum was dissolved in anhydrous CH2Cl2 (3 mL) and excess 50% aqueous ammonium hydroxide (3 mL) was added to achieve basic pH (-12) and stirred at room temperature for 2 h. The solvent was removed under reduced pressure and the crude compound was purified by flash column chromatography (20 x 1 cm) on silica gel using EtOAc/CHCl3 (1 :1 ) as eluent to furnish the pure amide 20 (0.013 g, 26 %): 1H NMR (DMSO- d6) δ 2.86-2.94 (m, 4H), 3.71-3.78 (m, 4H), 6.55 (d, IH, J= 15.3 Hz), 7.14 (dd, IH, J1 = 3.6 Hz, J2 = 5.1 Hz), 7.20 (d, IH, J = 8.7 Hz), 7.45 (d, IH, J = 3.0 Hz), 7.57 (bs, IH), 7.66 (d, IH, J =
5.1 Hz), 7.73 (d, IH, J = 15.6 Hz), 7.84 (dd, IH, J1 = 2.7 Hz, J2 = 8.7 Hz), 7.99 (d, IH, J = 2.7 Hz), 8.64 (bs, IH) and 10.2 (bs, IH). 13C NMR (DMSO-d6) δ 52.9, 66.4, 120.4, 120.8, 121.1, 122.0, 128.5 (2C), 129.3, 131.3, 133.1, 135.1, 139.7, 146.2, 163.1 and 167.7; MS (ES) m/z 356 (M - H). Ethyl 5-((E)-3-(furan-2-yl)acrylamidoV2-moφholinobenzoate (Compound 43)
1H-NMR (CDCl3): δ 1.38 (t, 3H, J = 7.1 Hz ), 3.01 (t, 4H, J = 4.2 Hz), 3.84 (t, 4H, J =
4.2 Hz), 4.37 (q, 2H, J = 7.1 Hz), 6.38-6.47 (m, 2H), 6.58 (d, IH, J = 3.2 Hz), 7.03 (d, IH, J = 8.7 Hz), 7.45 (s, IH), 7.51 (d, IH, J = 15.1 Hz), 7.63(s, IH), 7.86 (m, 2H); 13C NMR (CDCl3) δ
14.3, 53.0, 61.2, 67.1, 112.3, 114.6, 118.2, 119.8, 122.9, 124.4, 125.4, 129.0, 132.6, 144.3,
148.7, 151.2, 163.9, 167.3; MS (ES") m/z 369 (M -H).
Benzyl 5-((E)-3-(raran-2-vl)acrvlamidoV2-morpholinobenzoate (44) 1H-NMR (CDCl3): δ 2.96 (t, 4H, J = 4.5 Hz), 3.71 (t, 4H, J = 4.5 Hz), 5.34 (s, 2H), 6.40 (d, IH, J = 15.3 Hz), 6.38-6.49 (m, IH), 6.61 (d, IH, J = 3.3 Hz), 7.05 (d, J = 8.7 Hz, IH), 7.32-7.44 (m, 4H), 7.45-7.7.56 (m, 4H), 7.81 (s, IH) 7.90 (bs, IH); MS (ES+) m/z 433 (M +H). Propyl 5-((EV3-(faran-2-yl)acrylamido)-2-morpholinobenzoate (Compound 45) 1H-NMR (CDCl3): δ 0.99 (t, 3H, J = 7.4 Hz), 1.74 (sextet, 2H, J = 7.2 Hz ), 2.98 (bs,
4H), 3.82 (bs, 4H), 4.21 (t, 2H, J = 6.4 Hz), 6.35 - 6.60 (m, 3H), 6.98 (d, IH, J = 8.8 Hz), 7.39 (s, IH), 7.48 (d, IH, J =15.2 Hz), 7.86 (d, IH, J = 8.2 Hz), 7.95 (s, IH), 8.54 (s, IH); 13C NMR (CDCl3) δ 11.0, 22.4, 53.4, 67.2, 67.5, 112.7, 114.8, 119.0, 120.1, 123.3, 124.8, 125.8, 129.1, 133.4, 144.7, 148.9, 151.6, 164.7, 168.0; MS (ES') m/z 383 (M -H). Butyl 5-((E)-3-(faran-2-yl)acrylamido)-2-morpholinobenzoate (Compound 46)
1H-NMR (CDCl3): δ 0.94 (t, 3H, J = 7.5 Hz), 1.43 (sextet, 2H, J = 7.5 Hz), 1.70 (quintet, 2H, J = 7.5 Hz), 2.97 (bs, 4H), 3.82 (bs, 4H), 4.26 (t, 2H, J = 6.8 Hz), 6.39-6.58 (m, 3H), 6.98 (d, IH, J = 8.7 Hz), 7.28 (s, IH), 7.49 (d, IH, J = 15.3 Hz), 7.89 (m, 2H), 8.47 (bs, IH); 13C NMR (CDCl3) δ 14.2, 19.6, 31.1, 53.4, 65.5, 67.5, 112.7, 114.7, 119.1, 120.1, 123.3, 124.9, 125.9, 129.0, 133.5, 144.6, 148.9, 151.6, 164.8, 168.0; MS (ES') m/z 397 (M-H). Methyl 5-(cinnamamido)-2-moφholinobenzoate (Compound 52)
1H-NMR (CDCl3): δ 3.03 (t, 4H, J = 4.5Hz), 3.86 (t, 4H, J = 4.5Hz), 3.89 (s, 3H), 6.52 (d, IH, J = 15.3 Hz), 7.06 (d, IH, J= 8.7Hz), 7.35-7.41 (m, 4H), 7.52-7.56 (m, 2H), 7.76(d, IH, J = 15.3Hz), 7.93 (bs, IH); 13C NMR (CDCl3) δ 52.2, 53.0, 67.1, 119.7, 121.0, 123.5, 124.5, 124.9, 127.9, 128.8,129.9, 132.8, 134.6, 142.1, 148.9, 164.7, 167.6.
Methyl 5-((E)-3-(pyridin-3-yl)aciΥlamido)-2-morpholinobenzoate (Compound 55)
1H-NMR (CDCl3): δ 3.00 (t, 4H, J = 4.5 Hz ), 3.84 (t, 4H, J = 4.5 Hz), 3.87 (s, 3H), 6.69 (d, IH, J = 15.6 Hz ), 7.03 (d, IH, J = 8.7 Hz), 7.29-7.38 (m, IH), 7.67-7.79 (m, 2H), 7.86 (d, IH, J = 7.2 Hz ), 8.00 (s, IH), 8.44 (s, IH), 8.59 (s, IH), 8.77 (s, IH); 13C NMR (CDCl3) δ 52.2, 53.0, 67.1, 119.8, 123.1, 123.4, 123.9, 124.8, 130.7, 132.4, 134.9, 138.5, 148.9, 149.1, 150.4, 163.3, 167.5; MS (ES+) m/z 368 (M+H). Methyl 5-((E)-3-(pyridin-4-yl)acrylamido)-2-morpholinobenzoate (Compound 51)
1H-NMR (DMSOd6): δ 2.91 (t, 4H, J = 4.5 Hz), 3.69 (t, 4H, J = 4.5Hz), 3.82 (s, 3H), 7.04 (d, IH, J = 15.9 Hz), 7.13 (d, IH, J = 9 Hz), 7.57 (d, IH, J = 15.9 Hz), 7.65 (d, IH, J = 5.1 Hz), 7.80 (dd, IH, J1 = 2.4 Hz, J2 = 8.7 Hz), 8.02 (d, IH, J = 2.7 Hz), 8.31 (s, IH), 8.68 (d, 2H, J = 4.2 Hz) ; 13C NMR (DMSOd6) δ 52.5, 53.0, 66.8, 79.6, 120.2, 122.1, 122.6, 124.0, 125.0, 127.8, 133.6, 137.6, 143.6, 148.1, 149.8, 163.0, 167.9; MS (ES+) m/z 368 (M+H). 5-(Cinnamamido)-2-morpholinobenzoic acid (53) 1H-NMR (DMSO-d6): δ 3.05 (t, 4H, J = 4.2 Hz), 3.80 (t, 4H, J = 4.2 Hz), 6.82 (d, IH, J = 15.9 Hz), 7.38-7.51 (m, 3H), 7.57-7.72 (m, 4H), 8.03 (dd, IH, J1 = 2.4Hz, J2, = 8.7 Hz), 8.31 (d, IH, J = 2.7 Hz ), 10.50 (s, IH).
5-((E)-3-(Pyridin-3-vDacrylamido)-2-morpholinobenzoic acid (Compound 56) 1H-NMR (DMSOd6): δ 3.09 (t, 4H, J = 4.5 Hz ), 3.81 (t, 4H, J = 4.5 Hz), 7.14 (d, IH, J
= 15.9 Hz), 7.71 (d, IH, J = 2.7 Hz), 7.75 (d, IH, J = 9.9 Hz ), 7.91 (dd, IH, J1 = 5.4 Hz, J2 = 8.1Hz), 8.05 (dd, IH, J1 = 2.7 Hz, J2 = 9.0 Hz), 8.37 (d, IH, J = 2.4 Hz ), 8.53 (d, IH, J = 7.0 Hz ), 8.81 (dd, IH, J1 = 1.5 Hz, J2 = 5.4 Hz), 9.07 (d, IH, J = 1.8 Hz ), 10.91 (s, IH; 13C NMR (DMSO-d6) δ 53.3, 66.8, 121.7, 124.2, 124.7, 125.8, 126.7, 127.0, 133.3, 135.6, 138.0, 140.2, 144.8, 145.5, 145.7, 163.3, 166.9; MS (ES+) m/z 354 (M+H).
5-((E)-3-(Pyridin-4-yDaciylamido)-2-morpholinoberizoic acid (Compound 54)
1H-NMR (DMSOd6): δ 3.06 (bs , 4H), 3.80 (bs, 4H), 7.28 (d, IH, J = 16.2 Hz), 7.65- 7.76 (m, 2H), 8.05 (bs, 3H), 8.34 (s, IH), 8.87 (d, 2H, J = 5.4 Hz), 10.94 (s, IH). (E)-I -(4-(4-nitrophenoxy)phenyl)-3-(dimethylamino)prop-2-en-l -one (Compound 57) 1H-NMR (CDCl3): δ 2.96 (s, 3H), 3.17 (s, 3H), 5.70 (d, IH, J = 12.3 Hz), 7.05-7.13 (m,
4H), 7.84 (d, IH, J = 12.3Hz), 7.98 (d, 2H, J = 8.7 Hz), 8.22 (d, 2H, J = 9.3 Hz; 13C NMR (CDCl3) δ 38.0, 46.0, 117.9 (2C), 120.4, 126.6 (2C), 130.3, 137.9, 143.0, 155.1, 157.2, 163.3; MS (ES+) m/z 313 (M+H).
Table 1
Compd. Structure No. IC50 (μM)fl
Figure imgf000058_0001
NH, >N'NγNH_
NH, 275 ± 11.8
H2NΛN'N<
Figure imgf000058_0002
" IC50Valueswere determined by fluorescence resonance energy transfer (FRET) assay Measurements were carried out in triplicate and average value with standard deviation is reported. Table 2: S. aureus SrtAa59 inhibition of the synthesized compounds.
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
aIC50Valueswere determined by fluorescence resonance energy transfer (FRET) assay. Measurements were carried out in triplicate and average value with standard deviation is reported.

Claims

CLAIMSWhat is claimed:
1. A method of inhibiting a bacterial sortase in a subject, the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000063_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRi, where Ri is H or a substituted or unsubstituted alkyl;
E is selected from the group consisting of: CH2, O and N;
L1 and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and L is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R5 are each independently selected from the group consisting of: H, OH, CORi3, COORi4, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclyl alkyl ; R6 through Rg are each independently selected from the group consisting of: H, OH, and substituted or unsubstituted alkyl;
R13 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR15R16;
RH is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; and
R15 and R16 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl, or a pharmaceutically acceptable salt thereof.
2. A method of inhibiting a bacterial sortase in a subject, the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000064_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NR1, where Ri is H or a substituted or unsubstituted alkyl;
E is a heteroatom selected from the group consisting of: CH2, O and N;
F is a single, optional N heteroatom; when present, F may be in a para, meta or ortho position with respect to the point of attachment; Li and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and L is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl; I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R5 are each independently selected from the group consisting of: H, OH, CORi4, COORi5, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl ;
R9 through Rn are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl;, provided that, when F is present at a given position, the respective R9-R13 substituent group will be absent
Ri4 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi6Ri7;
Ri5 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; and
Ri6 and Ri7 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl, or a pharmaceutically acceptable salt thereof.
3. A method of inhibiting a bacterial sortase in a subject, the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000066_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRi, where Ri is H or a substituted or unsubstituted alkyl;
Li is an optional linking groups, where 1 is selected from 0 or 1 and L is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R6 are each independently selected from the group consisting of: H, OH; CORi0, COORn, NRi4Ri5, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
R7 through R9 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl;
Rio is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR]2R)3;
Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl;
Ri2 and Rj3 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl; and RH and R15 are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl and substituted or unsubstituted aralkyl, or a pharmaceutically acceptable salt thereof.
4. A method of inhibiting a bacterial sortase in a subject, the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000067_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R1 through R9 are each independently selected from the group consisting of: H, OH, NR]6R17, CORi2, COORi3, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
Rio and Rn are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
Ri2 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi4RiS;
R13 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; and
RH through R17 are each independently selected from the group consisting of: H, O and substituted or unsubstituted alkyl, or a pharmaceutically acceptable salt thereof.
5. A method of inhibiting a bacterial sortase in a subject, the method comprising the steps of administering to the subject a compound having a structure selected from the group consisting of:
Figure imgf000068_0001
Figure imgf000068_0002
Figure imgf000068_0003
Figure imgf000068_0004
Figure imgf000068_0005
Figure imgf000069_0001
Figure imgf000069_0002
or a pharmaceutically acceptable salt thereof.
6. The method of any one of claims 1-5 where the sortase enzyme is SrtA, SrtB, SrtC, SrtD or a homolog of any of the foregoing.
7. The method of claim 6 where the sortase enzyme is SrtA.
8. The method of any one of claims 1-5 where said inhibition reduces, at least in part, the ability of a bacterium to infect a cell of the subject or reduces, at least in part, virulence of a bacterium towards the subject.
9. The method of claim 8 where said bacterium is a Gram-positive bacterium.
10. The method of claim 8 where said bacterium is Staphylococcus aureus.
11. The method of claim 8 where said bacterium is a Gram-negative bacterium.
12. The method of claim 8 where the bacterium is resistant to one or more antibiotics.
13. The method of claim 8 where the bacterium is a methicillin-resistant Staphylococcus aureus, a vancomycin-resistant enterococcus, a vancomycin-intermediate Staphylococcus aureus or a vancomycin-resistant Staphylococcus aureus.
14. The method of any one of claims 1-5 where said inhibition reduces, at least in part, a bacterial infection.
15. The method of any one of claims 1 -5 where said inhibition reduces, at least in part, a disease state or condition caused by a bacterial infection.
16. The method of claim 15 where the disease state or condition is selected from the group consisting of: endocarditis, osteomyelitis, septic arthritis pneumonia, toxic shock syndrome, skin infections, meningitis, anthrax, B. cereus food poisoning, tetanus, clostridial myonecrosis, pneumonia, toxic shock syndrome, skin infection, meningitis, ear infection, strep throat, rheumatic fever, scarlet fever, puerperal fever, listeriosis, perinatal septicemia, encephalitis, intrauterine infection, sinusitis, severe invasive infection, and a combination of the foregoing.
17. The method of claims 15 where the disease state or condition is endocarditis.
18. The method of any one of claims 1 -5 further comprising administering a secondary antibiotic.
19. A method of reducing bacterial infection or bacterial virulence in a subject the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000070_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRi, where Ri is H or a substituted or unsubstituted alkyl;
E is selected from the group consisting of: CH2, O and N;
Li and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and L is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R5 are each independently selected from the group consisting of: H, OH, CORi3, COORi4, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
R6 through R8 are each independently selected from the group consisting of: H, OH, and substituted or unsubstituted alkyl; Ri3 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR15Ri6;
RH is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; and
Ri 5 and Ri6 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl, or a pharmaceutically acceptable salt thereof.
20. A method of reducing bacterial infection or bacterial virulence in a subject bacterial virulence in a subject the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000071_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRj, where Ri is H or a substituted or unsubstituted alkyl;
E is a heteroatom selected from the group consisting of: CH2, O and N;
F is a single, optional N heteroatom; when present, F may be in a para, meta or ortho position with respect to the point of attachment;
Li and L2 are optional linking groups, where 1 and 2 are independently selected from 0 or 1 and L is each independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R5 are each independently selected from the group consisting of: H, OH, CORi4, COORi 5, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Cj to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclyl alkyl;
R9 through Rn are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl, provided that, when F is present at a given position, the respective R9-R13 substituent group will be absent;
Ri4 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRj6Ri7;
Ri5 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; and
Ri6 and Rj7 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl, or a pharmaceutically acceptable salt thereof.
21. A method of reducing bacterial infection or bacterial virulence in a subject the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000072_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N; B is selected from the group consisting of: S, O and NRi, where R1 is H or a substituted or unsubstituted alkyl; Li is an optional linking groups, where 1 is selected from 0 or 1 and L is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R6 are each independently selected from the group consisting of: H, OH; CORi0, COORn, NRi4R15, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl;
R7 through R9 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl;
Rio is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi2Ri3;
Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl;
Ri2 and Ri3 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl; and
Ri4 and Rj5 are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl and substituted or unsubstituted aralkyl, or a pharmaceutically acceptable salt thereof.
22. A method of reducing bacterial infection or bacterial virulence in a subject, the method comprising the steps of administering to the subject a compound having the structure:
Figure imgf000074_0001
where:
A is selected from the group consisting of: S, CH2, O and N; I is linking group and is a C2 to C5 alkenyl containing a single carbon- carbon double bond or C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R1 through R9 are each independently selected from the group consisting of: H, OH, NR16Rj7, COR12, COORi3, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl ;
Ri0 and Rn are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl and substituted or unsubstituted alkynyl;
Ri2 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi4Ri5;
Ri3 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl; and
Ri4 and Ri7 are each independently selected from the group consisting of: H, O and substituted or unsubstituted alkyl, or a pharmaceutically acceptable salt thereof.
23. A method of reducing bacterial virulence in a subject the method comprising the steps of administering to the subject a compound a structure selected from the group consisting of:
Figure imgf000075_0001
Figure imgf000075_0002
Figure imgf000075_0003
Figure imgf000075_0004
Figure imgf000075_0005
Figure imgf000075_0006
Figure imgf000075_0007
or a pharmaceutically acceptable salt thereof 24. The method of any one of claims 19-23 where the administration decreases, in whole or part, the activity of a bacterial sortase enzyme.
25. The method of claim 24 where the bacterial sortase enzyme is SrtA, SrtB, SrtC, SrtD or a homolog of any of the foregoing.
26. The method of claim 24 where the bacterial sortase enzyme is SrtA.
27. The method of claim 24 where said decrease in activity reduces, at least in part, the ability of a bacterium to infect the cells of the subject.
28. The method of any one of claims 19-23 where the bacterial infection or virulence is caused, at least in part, by a Gram-positive bacterium.
29. The method of claim 28 where said Gram-positive bacterium is Staphylococcus aureus.
30. The method of any one of claims 19-23 where the bacterial infection or virulence is caused, at least in part, by a Gram-negative bacterium.
31. The method of any one of claims 19-23 where the bacterial infection or virulence is caused, at least in part, by an antibiotic resistant bacterium.
32. The method of claim 31 where the antibiotic resistant bacterium is a methicillin-resistant Staphylococcus aureus, a vancomycin-resistant enterococcus, a vancomycin-intermediate Staphylococcus aureus or a vancomycin-resistant Staphylococcus aureus.
33. The method of any one of claims 19-23 further comprising administering a secondary antibiotic.
34. A compound of the formula Ia:
(Ia)
Figure imgf000076_0001
wherein:
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRi, wherein R] is H or a substituted or unsubstituted alkyl; E is selected from the group consisting of: CH2, O and N; Li and L2 are optional linking groups, wherein 1 and 2 are independently selected from 0 and 1, and L is independently selected from the group consisting of: substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, and substituted or unsubstituted alkynyl; I is a linking group, selected from the group consisting of a C2 to C5 alkenyl containing a single carbon-carbon double bond and a C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R5 are each independently selected from the group consisting of: H, OH,
COR13, COORi4, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, C1 to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl, wherein R13 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NR15Ri6; wherein R14 is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, and substituted or unsubstituted aralkyl; wherein R15 and R16 are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl; and R6 through R8 are each independently selected from the group consisting of: H, OH, and substituted or unsubstituted alkyl; provided that whenever A is S, R2 cannot be COOH when B is NH, E is O, I is a C2 alkenyl in trans configuration, Li and L2 are both O, and R3 through R8 are H; or a pharmaceutically acceptable salt thereof.
35. A compound according to claim 34, wherein R2 is COOCH3, or a pharmaceutically acceptable salt thereof.
36. A compound according to claim 35, wherein A is S, or a pharmaceutically acceptable salt thereof.
37. A compound according to claim 36, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both O, or a pharmaceutically acceptable salt thereof.
38. A compound according to claim 36, wherein: B is NH, E is O, I is a C2 alkynyl, R3 through R8 are H, and 1 and 2 of L1 and L2 are both 0, or a pharmaceutically acceptable salt thereof.
39. A compound according to claim 36, wherein: B is NH, E is O, I is a C2 alkenyl in the cis configuration, R3 through R8 are H, and 1 and 2 of Lj and L2 are both 0, or a pharmaceutically acceptable salt thereof.
40. A compound according to claim 36, wherein: B is NH, E is O, I is a C3 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
41. A compound according to claim 36, wherein: B is NH, E is CH2, 1 is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
42. A compound according to claim 36, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, 1 of Li is 0, 2 of L2 is 1 and L in L2 is CH2, or a pharmaceutically acceptable salt thereof.
43. A compound according to claim 35, wherein A is O, or a pharmaceutically acceptable salt thereof.
44. A compound according to claim 43, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Lj and L2 are both 0, or a pharmaceutically acceptable salt thereof.
45. A compound according to claim 43, wherein: B is NH, E is CH2, 1 is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
46. A compound according to claim 34, wherein R2 is COOH, or a pharmaceutically acceptable salt thereof.
47. A compound according to claim 46, wherein A is S, or a pharmaceutically acceptable salt thereof.
48. A compound according to claim 47, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Lj and L2 are both 0, or a pharmaceutically acceptable salt thereof.
49. A compound according to claim 47, wherein: B is NH, E is O, I is a C2 alkenyl in the cis configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
50. A compound according to claim 47, wherein: B is NH, E is CH2, 1 is a C2 alkenyl in the trans configuration, R3 through Rg are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
51. A compound according to claim 47, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, 1 Of L1 is 0, 2 of L2 is 1 and L in L2 is CH2, or a pharmaceutically acceptable salt thereof.
52. A compound according to claim 46, wherein A is O, or a pharmaceutically acceptable salt thereof.
53. A compound according to claim 52, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
54. A compound according to claim 34, wherein: R2 is C(=O)H, or a pharmaceutically acceptable salt thereof.
55. A compound according to claim 54, wherein: A is S, or a pharmaceutically acceptable salt thereof.
56. A compound according to claim 55, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
57. A compound according to claim 54, wherein: A is O, or a pharmaceutically acceptable salt thereof.
58. A compound according to claim 57, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
59. A compound according to claim 34, wherein: R2 is CH2OH, or a pharmaceutically acceptable salt thereof.
60. A compound according to claim 59, wherein: A is S, or a pharmaceutically acceptable salt thereof.
61. A compound according to claim 60, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
62. A compound according to claim 59, wherein: A is O, or a pharmaceutically acceptable salt thereof.
63. A compound according to claim 62, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
64. A compound according to claim 34, wherein: R2 is C(=0)NH2, or a pharmaceutically acceptable salt thereof.
65. A compound according to claim 64, wherein: A is S, or a pharmaceutically acceptable salt thereof.
66. A compound according to claim 65, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 are both 0, or a pharmaceutically acceptable salt thereof.
67. A compound according to claim 34, wherein: R2 is C(=O)-ORi4, wherein Ri4 is selected from a group consisting Of C2H5, C3H7, C4Hg, and C7H7, or a pharmaceutically acceptable salt thereof.
68. A compound according to claim 67, wherein: A is O, or a pharmaceutically acceptable salt thereof.
69. A compound according to claim 68, wherein: B is NH, E is O, I is a C2 alkenyl in the trans configuration, R3 through R8 are H, and 1 and 2 of Li and L2 are both 0, or a pharmaceutically acceptable salt thereof.
70. A compound according to claim 69, wherein: R]4 is C2H5, or a pharmaceutically acceptable salt thereof.
71. A compound according to claim 69, wherein: Ri4 is CH2-C6Hs, or a pharmaceutically acceptable salt thereof.
72. A compound according to claim 69, wherein: Ri4 is C3H7, or a pharmaceutically acceptable salt thereof.
73. A compound according to claim 69, wherein: Rj4 is C4H9, or a pharmaceutically acceptable salt thereof.
74. A compound of the formula Ib:
(Ib)
Figure imgf000081_0001
wherein:
A is selected from the group consisting of:, S, CH2, O and N;
B is selected from the group consisting of: S, O and NR1, wherein Ri is H or a substituted or unsubstituted alkyl;
E is selected from the group consisting of: CH2, O and N;
F is a single, optional N heteroatom; when present, F may be in a para, meta or ortho position with respect to the point of attachment;
L] and L2 are optional linking groups, wherein 1 and 2 are independently selected from 0 and 1, and L is independently selected from the group consisting of: substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, and substituted or unsubstituted alkynyl;
I is a linking group, selected from the group consisting of a C2 to C5 alkenyl containing a single carbon-carbon double bond and a C2 to C5 alkynyl containing a single carbon-carbon triple bond;
R2 through R5 are each independently selected from the group consisting of: H, OH, COR]4, COORi5, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl, wherein Ri4 is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRj6Ri7; wherein Ru is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, and substituted or unsubstituted aralkyl; wherein Rj6 and Rp are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl; and
R9 through Ri3 are each independently selected from the group consisting of: H, OH, and substituted or unsubstituted alkyl, provided that when F is present at a given position, the respective R9 through Rj3 substituent group will be absent; or a pharmaceutically acceptable salt thereof.
75. A compound according to claim 74, wherein: B is NH, E is O, and I is a C2 alkenyl in the trans configuration, or a pharmaceutically acceptable salt thereof.
76. A compound as in claim 75, wherein R2 is COOH, or a pharmaceutically acceptable salt thereof.
77. A compound according to claim 76, wherein: F is absent, 1 and 2 of Li and L2 are both O,
R3 through R5 and R9 through Ri3 are H, or a pharmaceutically acceptable salt thereof.
78. A compound according to claim 76, wherein: F is present in the meta position, 1 and 2 of Li and L2 are both 0, Ri0 is absent, R3 through R5, R9 and Rn through R13 are H, or a pharmaceutically acceptable salt thereof.
79. A compound according to claim 76, wherein: F is present in the para position, 1 and 2 of Li and L2 are both 0, Rn is absent, R3 through R5, R9 through Ri0 and Ri2 through Ri3 are H, or a pharmaceutically acceptable salt thereof.
80. A compound as in claim 75, wherein R2 is COOCH3, or a pharmaceutically acceptable salt thereof.
81. A compound according to claim 80, wherein: F is absent, 1 and 2 of Li and L2 are both 0,
R3 through R5 and R9 through Rj3 are H, or a pharmaceutically acceptable salt thereof.
82. A compound according to claim 80, wherein: F is present in the meta position, 1 and 2 of Li and L2 are both 0, Ri0 is absent, R3 through R5, R9 and Rn through R]3 are H, or a pharmaceutically acceptable salt thereof.
83. A compound according to claim 80, wherein: F is present in the para position,l and 2 of Li and L2 are both 0, Rn is absent, R3 through R5, R9 through Rj0 and R)2 through Rj3 are H, or a pharmaceutically acceptable salt thereof.
84. A compound of the formula Ic: (Ic)
Figure imgf000083_0001
Wherein
A is selected from the group consisting of: S, CH2, O and N;
B is selected from the group consisting of: S, O and NRi, wherein Rj is H or a substituted or unsubstituted alkyl;
Li is an optional linking groups, where 1 is selected from 0 or 1 and L is selected from the group consisting of substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl;
I is a linking group, s*eje^j^|frpm the group consisting of a C2 to C5 alkenyl containing a single carbon-carbon double bond in either the cis or trans configuration and a C2 to C5 alkynyl containing a single carbon-carbon triple bond; and
R2 through R6 are each independently selected from the group consisting of: H, OH, CORio, COORi i, NR)4Ri5, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, Ci to C7 alcohols, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aralkyl and substituted or unsubstituted heterocyclylalkyl, wherein R^ is selected from the group consisting of: H, substituted or unsubstituted alkyl, and NRi2Ri3, wherein Rn is selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl and substituted or unsubstituted aralkyl, wherein R]2 and Rn are each independently selected from the group consisting of: H and substituted or unsubstituted alkyl, and wherein Ri4 and R)5 are each independently selected from the group consisting of: H, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl and substituted or unsubstituted aralkyl; and
R7 through R9 are each independently selected from the group consisting of: H, OH and substituted or unsubstituted alkyl; or a pharmaceutically acceptable salt thereof.
85. A compound as in claim 84, wherein R3 is COOCH3, or a pharmaceutically acceptable salt thereof.
86. A compound as in claim 85, wherein: A is S, B is NH, I is a C2 alkenyl in the trans configuration, R2 is NH-CH2-C6H5, R4 through R9 are H, and 1 of Li is 0, or a pharmaceutically acceptable salt thereof.
87. A compound as in claim 85, wherein: A is S, B is NH, I is a C2 alkenyl in the trans configuration, R2 is NH-C6H5, R4 through R9 are H, and 1 of Li is 0, or a pharmaceutically acceptable salt thereof.
88. A compound as in claim 85, wherein: A is S, B is NH, I is a C2 alkenyl in the trans configuration, R2 is N-(CH2-C6H5)2, R4 through R9 are H, and 1 of Li is 0, or a pharmaceutically acceptable salt thereof.
89. A compound as in claim 84, wherein: R3 is COOH, or a pharmaceutically acceptable salt thereof.
90. A compound as in claim 89, wherein: A is S, B is NH, I is a C2 alkenyl in the trans configuration, R2 is NH-C6H5, R4 through R9 are H, and 1 of Lj is 0, or a pharmaceutically acceptable salt thereof.
91. A compound as in claim 89, wherein: A is S, B is NH, I is a C2 alkenyl in the trans configuration, R2 is N-(CH2-C6Hs)2, R4 through R9 are H, and 1 of Li is 0, or a pharmaceutically acceptable salt thereof.
92. A compound as in claim 89, wherein: A is S, B is NH, I is a C2 alkenyl in the trans configuration, R2 is N-(C2H4OH)2, R4 through R9 are H, and 1 of Li is 0, or a pharmaceutically acceptable salt thereof.
PCT/US2008/009577 2007-08-11 2008-08-11 Novel inhibitors of bacterial sortase enzymes and methods of using the same WO2009023160A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US95535307P 2007-08-11 2007-08-11
US60/955,353 2007-08-11

Publications (3)

Publication Number Publication Date
WO2009023160A2 true WO2009023160A2 (en) 2009-02-19
WO2009023160A3 WO2009023160A3 (en) 2009-05-07
WO2009023160A8 WO2009023160A8 (en) 2011-11-24

Family

ID=40351355

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/009577 WO2009023160A2 (en) 2007-08-11 2008-08-11 Novel inhibitors of bacterial sortase enzymes and methods of using the same

Country Status (1)

Country Link
WO (1) WO2009023160A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011084882A3 (en) * 2010-01-05 2011-10-20 Contrafect Corporation Methods and compositions for enhanced immunological therapy and targeting of gram-positive bacteria
WO2011104531A3 (en) * 2010-02-24 2012-01-05 London School Of Hygiene And Tropical Medicine Clostridium gene
EP2875816A1 (en) * 2013-11-21 2015-05-27 Latvian Biomedical Research and Study Centre N-(3-hydroxy-5,7-dimethyladamantan-1-yl)-2-(3-oxobenzo[d]isothiazol-2(3h)-yl)acetamide as a novel inhibitor for staphylococcus aureus sortase a
EP2875815A1 (en) * 2013-11-21 2015-05-27 Latvian Biomedical Research and Study Centre N'-(2-(3-oxobenzo[d]isothiazol-2(3h)-yl)acetyl)adamantane-1-carbohydrazide as a novel inhibitor for staphylococcus aureus sortase a
CN107904279A (en) * 2017-11-03 2018-04-13 中国农业科学院北京畜牧兽医研究所 A kind of screening technique of staphylococcus aureus inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003049702A2 (en) * 2001-12-10 2003-06-19 Amgen Inc. Vanilloid receptor ligands and their use in treatments
US7119200B2 (en) * 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003049702A2 (en) * 2001-12-10 2003-06-19 Amgen Inc. Vanilloid receptor ligands and their use in treatments
US7119200B2 (en) * 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BIAVA, M. ET AL.: 'Synthesis and antimycobacterial activity of new amido derivatives of ortho-, meta- and para-toluidine' MEDICINAL CHEMISTRY RESEARCH vol. 8, no. 9, 1998, ISSN 1054-2523 pages 523 - 541 *
CHENNA, B. C. ET AL.: 'Identification of novel inhibitors of bacterial surface enzyme Staphylococcus aureus Sortase A.' BIOORGANIC & MEDICINAL CHEMISTRY LETTERS vol. 18, no. 1, 2008, ISSN 0960-894X pages 380 - 385 *
PUBCHEMCOMPOUND, [Online] Retrieved from the Internet: <URL:http://pubchem.ncbi.nlm.nih.gov/summary/summary.cgi?cid=#######&loc=ec_rcs> *
RAHMAN, A.A. ET AL.: 'Biological Activities of a New Acrylamide Derivative from Ipomoea turpethum.' PARKISTAN JOURNAL OF BIOLOGICAL SCIENCES vol. 5, no. 9, 2002, ISSN 1028-8880 pages 968 - 969 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011084882A3 (en) * 2010-01-05 2011-10-20 Contrafect Corporation Methods and compositions for enhanced immunological therapy and targeting of gram-positive bacteria
US9005579B2 (en) 2010-01-05 2015-04-14 Contrafect Corporation Methods and compositions for enhanced immunological therapy and targeting of gram-positive bacteria
US9731010B2 (en) 2010-01-05 2017-08-15 The Rockefeller University Methods and compositions for enhanced immunological therapy and targeting of gram-positive bacteria
US10588971B2 (en) 2010-01-05 2020-03-17 The Rockefeller University Methods and compositions for enhanced immunological therapy and targeting of gram-positive bacteria
WO2011104531A3 (en) * 2010-02-24 2012-01-05 London School Of Hygiene And Tropical Medicine Clostridium gene
EP2875816A1 (en) * 2013-11-21 2015-05-27 Latvian Biomedical Research and Study Centre N-(3-hydroxy-5,7-dimethyladamantan-1-yl)-2-(3-oxobenzo[d]isothiazol-2(3h)-yl)acetamide as a novel inhibitor for staphylococcus aureus sortase a
EP2875815A1 (en) * 2013-11-21 2015-05-27 Latvian Biomedical Research and Study Centre N'-(2-(3-oxobenzo[d]isothiazol-2(3h)-yl)acetyl)adamantane-1-carbohydrazide as a novel inhibitor for staphylococcus aureus sortase a
CN107904279A (en) * 2017-11-03 2018-04-13 中国农业科学院北京畜牧兽医研究所 A kind of screening technique of staphylococcus aureus inhibitor

Also Published As

Publication number Publication date
WO2009023160A3 (en) 2009-05-07
WO2009023160A8 (en) 2011-11-24

Similar Documents

Publication Publication Date Title
EP3255045A1 (en) Salts, prodrugs and polymorphs of fab i inhibitors
WO2017181897A1 (en) Novel broad-spectrum β-lactamase inhibitor
EP3655405B1 (en) Chemical compounds
WO2009023160A2 (en) Novel inhibitors of bacterial sortase enzymes and methods of using the same
JP2017502970A (en) Nitrogen-containing compounds and uses thereof
Jin et al. Bioisosteric investigation of ebselen: Synthesis and in vitro characterization of 1, 2-benzisothiazol-3 (2H)-one derivatives as potent New Delhi metallo-β-lactamase inhibitors
JP7134177B2 (en) macrocyclic broad-spectrum antibiotic
Chenna et al. Synthesis and structure activity relationship studies of novel Staphylococcus aureus Sortase A inhibitors
EP1511472B1 (en) Compounds useful in the treatment of anthrax and inhibiting lethal factor
US20170088524A1 (en) Compositions, Methods of Use, And Methods of Treatment
Niu et al. Small molecule LpxC inhibitors against gram-negative bacteria: Advances and future perspectives
JP6411482B2 (en) Nitrogen-containing compounds and uses thereof
US7538085B2 (en) Peptide deformylase inhibitors as novel antibiotics
WO2012079164A1 (en) Activators of cylindrical proteases
CN107151236B (en) 2, 3-epoxysuccinyl derivative and preparation method and application thereof
AU2002314773B2 (en) Peptide deformylase activated prodrugs
KR20090033583A (en) Compounds having glysin aryl amide moiety as inhibitors of beta-secretase
ZA200209474B (en) Novel MMP-2/MMP-9 inhibitors.
Supuran et al. Protease inhibitors. Part 7: Inhibition of Clostridium histolyticum collagenase with sulfonylated derivatives of L-valine hydroxamate
WO2011060976A1 (en) Tryptamine-derived compounds as antibacterial agents
US20170369864A1 (en) Inhibition and diagnostics of emerging tetracycline resistance enzymes
KR20090031331A (en) Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety
AU2002314773A1 (en) Peptide deformylase activated prodrugs
US11555010B2 (en) Diamide antimicrobial agents
AU2003225047A1 (en) Peptide deformylase activated prodrugs

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: 08795186

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08795186

Country of ref document: EP

Kind code of ref document: A2