WO2011085990A1 - Anti - infective pyrido (1,2 -a) pyrimidines - Google Patents

Anti - infective pyrido (1,2 -a) pyrimidines Download PDF

Info

Publication number
WO2011085990A1
WO2011085990A1 PCT/EP2011/000135 EP2011000135W WO2011085990A1 WO 2011085990 A1 WO2011085990 A1 WO 2011085990A1 EP 2011000135 W EP2011000135 W EP 2011000135W WO 2011085990 A1 WO2011085990 A1 WO 2011085990A1
Authority
WO
WIPO (PCT)
Prior art keywords
nmr
mhz
cdc1
pyrido
heteroaryl
Prior art date
Application number
PCT/EP2011/000135
Other languages
English (en)
French (fr)
Other versions
WO2011085990A8 (en
Inventor
Zaesung No
Jaeseung Kim
Priscille Brodin
Min Jung Seo
Eunjung Park
Jonathan Cechetto
Heekyoung Jeon
Auguste Genovesio
Saeyeon Lee
Sunhee Kang
Fanny Anne Ewann
Ji Youn Nam
Denis Philippe Cedric Fenistein
Thierry Christophe
Monica Contreras Dominguez
Eunhye Kim
Jamung Heo
Original Assignee
Institut Pasteur Korea
Institut National De La Sante Et De La Recherche Medicale (Inserm) (Epst)
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 Institut Pasteur Korea, Institut National De La Sante Et De La Recherche Medicale (Inserm) (Epst) filed Critical Institut Pasteur Korea
Priority to JP2012548380A priority Critical patent/JP2013517233A/ja
Priority to EP11700392A priority patent/EP2523956A1/en
Priority to AU2011206761A priority patent/AU2011206761A1/en
Priority to CN2011800059743A priority patent/CN102812023A/zh
Priority to KR1020127018472A priority patent/KR20130088002A/ko
Priority to BR112012017473A priority patent/BR112012017473A2/pt
Priority to US13/518,292 priority patent/US20130012506A1/en
Priority to CA2786329A priority patent/CA2786329A1/en
Publication of WO2011085990A1 publication Critical patent/WO2011085990A1/en
Publication of WO2011085990A8 publication Critical patent/WO2011085990A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to small molecule compounds and their use in the treatment of bacterial infections, in particular Tuberculosis.
  • Tuberculosis (TB) as a disease continues to result in millions of deaths each year.
  • Inadequate use of chemotherapy has led to an increasing number of drug resistant cases. This situation is likely to worsen with the emergence of extremely resistant strains to all currently known drugs (Van Rie and Enarson, 2006).
  • the internationally recommended TB control strategy also referred to as directly observed short-course chemotherapy (DOTS), relies on a combination of five antibacterial agents to be taken for a protracted period of more than six months (http://www.who.int/tb/dots/en/).
  • the bacillus mainly localizes inside phagocytic cells, such as macrophages and dendritic cells, and it has clearly been established that the tubercle bacillus adopts a different phenotype in the host macrophage's phagosome compared to growth in extracellular conditions (Rohde et al., 2007; Schnappinger et al, 2003).
  • phagocytic cells such as macrophages and dendritic cells
  • a phenotypic cell-based assay suitable for high throughput screening, that allows for the search of compounds that would prevent M. tuberculosis multiplication inside the host macrophage was utilized.
  • the present invention relates to compounds having the general formula I:
  • n 0, 1, 2, or 3;
  • n 1, 2, 3,or, 4;
  • o 1, 2, 3,or, 4;
  • A is C 5 -Ci 2 heteroaryl
  • R 1 is selected from the group consisting of hydrogen, halogen, Ci-C 10 alkyl, C 3 -Cio cycloalkyl, C 2 -Ci 0 alkenyl, C3-Ci 0 cycloalkenyl, C 3 -Ci 5 cycloalkylalkoxy, C 3 -Ci 5
  • R 2 is selected from the group consisting of hydrogen, halogen, Cj-Cio alkyl, C 3 -C 10 cycloalkyl, C 2 -C 10 alkenyl, C 3 -C 10 cycloalkenyl, C3-C15 cycloalkylalkyl, -NH 2 , -N(R 6 ) 2 , - C(0)R 6 , -C(0)OR 6 , -C(0)N(R 6 ) 2 , -S(0)R 6 , -S(0) 2 R 6 , -S(0) 2 N(R 6 ) 2 , aryl, benzyl, heteroaryl, or heterocyclyl, or two groups of R and R are connected with each other to make a five or six membered cyclic or heterocyclic ring, any of which is optionally substituted;
  • R 3 is selected from the group consisting of hydrogen, halogen, Cj-Cio alkyl, C 3 -Cio cycloalkyl, hydroxyl, -OR 6 , -CN, -N0 2 , -NH 2 , -N(R 6 )C(0)R 6 , -C(0)R 6 , -C(0)OR 6 , - C(0)N(R 6 ) 2j -S(0)R 6 , -S(0) 2 R 6 , -S(0) 2 N(R 6 ) 2 , aryl, benzyl, heteroaryl, heterocyclyl, or two groups of R 3 are connected with each other to make a five or six membered cyclic or heterocyclic ring, any of which is optionally substituted;
  • R 4 is independently, at each occurrence, selected from the group consisting of hydrogen, halogen, C,-C 10 alkyl, C 3 -Ci 0 cycloalkyl, hydroxyl, -OR 6 , -CN, -N0 2 , -NH 2 , -N(R 6 )C(0)R 6 , - C(0)R 6 , -C(0)OR 6 , -C(0)N(R 6 ) 2, -S(0)R 6 , -S(0) 2 R 6 , -S(0) 2 N(R 6 ) 2 , aryl, benzyl, heteroaryl, heterocyclyl, or two groups of R 4 are connected with each other to make five or six membered cyclic or heterocyclic ring, any of which is optionally substituted;
  • R 5 and R 6 are independently, at each occurrence, selected from the group consisting from hydrogen, CpCio alkyl, C3-C10 cycloalkyl, C 2 -Ci 0 alkenyl, C 3 -Cio cycloalkenyl, C 2 -C 10 alkynyl, C1-C10 haloalkyl, aryl, benzyl, heteroaryl, or heterocyclyl, any of which is optionally substituted;
  • HetA refers to "heteroaryl”;
  • the term “optionally substituted” as used herein is meant to indicate that a hydrogen atom attached to a member atom within a group is possibly replaced by group, such as halogen including fluorine, Ci-Ci 0 alkyl, CrC 3 haloalkyl, C 3 -C 7 cycloalkyl, oxo, -OH, -OR 7 , - OC(0)R 7 -CN, ⁇ N0 2
  • R 7 is independently, at each occurrence, selected from the group consisting of hydrogen, aryl, benzyl, heteroaryl, heterocyclyl, Ci-C 8 alkyl, or C 3 -C 7 cycloalkyl;
  • alkyl refers to a monovalent straight or branched chain, saturated aliphatic hydrocarbon radical having a number of carbon atoms in the specified range.
  • Ci-C 6 alkyl refers to any of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec-, and t-butyl, n- and isopropyl, ethyl and methyl.
  • alkoxy means a group having the formula -O-alkyl, in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom.
  • the alkyl portion of an alkoxy group can have 1 to 20 carbon atoms (i.e., C ! -C 20 alkoxy), 1 to 12 carbon atoms (i.e., Ci-C 12 alkoxy), or 1 to 6 carbon atoms (i.e., Ci-C 6 alkoxy).
  • alkoxy groups include, but are not limited to, methoxy (-0-CH 3 or OMe), ethoxy (-OCH 2 CH 3 or - OEt), t-butoxy (-0-C(CH 3 ) 3 or -OtBu) and the like.
  • alkenyl refers to a monovalent straight or branched chain aliphatic hydrocarbon radical containing one carbon-carbon double bond and having a number of carbon atoms in the specified range.
  • C 2 -C 6 alkenyl refers to all of the hexenyl and pentenyl isomers as well as 1-butenyl, 2-butenyl, 3-butenyl, isobutenyl, 1-propenyl, 2- propenyl, and ethenyl (or vinyl).
  • alkynyl refers to a monovalent straight or branched chain aliphatic hydrocarbon radical containing one carbon-carbon triple bond and having a number of carbon atoms in the specified range.
  • C 2 -C 6 alkynyl refers to all of the hexynyl and pentynyl isomers as well as 1-butynyl, 2-butynyl, 3-butynyl, 1-propynyl, 2-propynyl, and ethynyl.
  • alkylene refers to a saturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms.
  • Typical alkylene radicals include, but are not limited to, methylene (-CH 2 -), 1,1 -ethyl (- CH(CH 3 )-), 1,2-ethyl (-CH 2 CH 2 -), 1,1 -propyl (-CH(CH 2 CH 3 )-), 1,2-propyl (-CH 2 CH(CH 3 )-), 1,3-propyl (-CH 2 CH 2 CH 2 -), 1,4-butyl (-CH 2 CH 2 CH 2 CH 2 -), and the like.
  • alkenylene refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of parent alkene.
  • an alkenylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms.
  • alkynylene refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of parent alkyne.
  • an alkynylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms or 1 to 6 carbon atoms.
  • Typical alkynylene radicals include, but are not limited to, acetylene (-C ⁇ C-), propargyl (-CH 2 C ⁇ C-), and 4-pentynyl (-CH 2 CH 2 CH 2 C ⁇ CH-).
  • cycloalkyl refers to a group, such as optionally substituted or non-substituted cyclic hydrocarbon, having from three to eight carbon atoms, unless otherwise defined.
  • C3-C8 cycloalkyl refers to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • haloalkyl refers to an alkyl group, as defined herein that is substituted with at least one halogen.
  • straight or branched chained “haloalkyl” groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, H-butyl, and t-butyl substituted independently with one or more halogens.
  • haloalkyl should be interpreted to include such substituents such as -CHF 2 , -CF 3 , -CH 2 -CH 2 -F, -CH 2 -CF 3 , and the like.
  • heteroalkyl refers to an alkyl group where one or more carbon atoms have been replaced with a heteroatom, such as, O, N, or S.
  • a heteroatom e.g., O, N, or S
  • the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g., -OCH 3 , etc.), an amine (e.g., -NHCH 3 , -N(CH 3 ) 2 , etc.), or thioalkyl group (e.g., -SCH3, etc.).
  • a non-terminal carbon atom of the alkyl group which is not attached to the parent molecule is replaced with a heteroatom (e.g., O, N, or S) and the resulting heteroalkyl groups are, respectively, an alkyl ether (e.g., -CH 2 CH 2 -0-CH 3 , etc.), alkyl amine (e.g., -CH 2 NHCH 3 , -CH 2 N(CH 3 ) 2 , etc.), or thioalkyl ether (e.g., -CH 2 -S-CH 3 ).
  • an alkyl ether e.g., -CH 2 CH 2 -0-CH 3 , etc.
  • alkyl amine e.g., -CH 2 NHCH 3 , -CH 2 N(CH 3 ) 2 , etc.
  • thioalkyl ether e.g., -CH 2 -S-CH 3
  • halogen refers to fluorine, chlorine, bromine, or iodine.
  • aryl refers to (i) optionally substituted phenyl, (ii) optionally substituted 9- or 10 membered bicyclic, fused carbocyclic ring systems in which at least one ring is aromatic, and (iii) optionally substituted 1 1- to 14-membered tricyclic, fused carbocyclic ring systems in which at least one ring is aromatic.
  • Suitable aryls include, for example, phenyl, biphenyl, naphthyl, tetrahydronaphthyl (tetralinyl), indenyl, anthracenyl, and fluorenyl.
  • phenyl as used herein is meant to indicate that optionally substituted or non- substituted phenyl group.
  • benzyl as used herein is meant to indicate that optionally substituted or non- substituted benzyl group.
  • heteroaryl refers to (i) optionally substituted 5- and 6-membered heteroaromatic rings and (ii) optionally substituted 9- and 10-membered bicyclic, fused ring systems in which at least one ring is aromatic, wherein the heteroaromatic ring or the bicyclic, fused ring system contains from 1 to 4 heteroatoms independently selected from N, O, and S, where each N is optionally in the form of an oxide and each S in a ring which is not aromatic is optionally S(O) or S(0) 2 .
  • Suitable 5- and 6-membered heteroaromatic rings include, for example, pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thienyl, furanyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isooxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, and thiadiazolyl.
  • Suitable 9-and 10-membered heterobicyclic, fused ring systems include, for example, benzofuranyl, indolyl, indazolyl, naphthyridinyl, isobenzofuranyl, benzopiperidinyl, benzisoxazolyl, benzoxazolyl, chromenyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, isoindolyl, benzodioxolyl, benzofuranyl, imidazo[l,2-a]pyridinyl, benzotriazolyl, dihydroindolyl, dihydroisoindolyl, indazolyl, indolinyl, isoindolinyl, quinoxalinyl, quinazolinyl, 2,3-dihydrobenz
  • heterocyclyl refers to (i) optionally substituted 4- to 8-membered, saturated and unsaturated but non-aromatic monocyclic rings containing at least one carbon atom and from 1 to 4 heteroatoms, (ii) optionally substituted bicyclic ring systems containing from 1 to 6 heteroatoms, and (iii) optionally substituted tricyclic ring systems, wherein each ring in (ii) or (iii) is independent of fused to, or bridged with the other ring or rings and each ring is saturated or unsaturated but nonaromatic, and wherein each heteroatom in (i), (ii), and (iii) is independently selected from N, O, and S, wherein each N is optionally in the form of an oxide and each S is optionally oxidized to S(O) or S(0) 2 .
  • Suitable 4- to 8-membered saturated heterocyclyls include, for example, azetidinyl, piperidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, oxazolidonyl, isoxazolidinyl, pyrrolidinyl, imidazolidinyl, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolidinyl,
  • Suitable unsaturated heterocyclic rings include those corresponding to the saturated heterocyclic rings listed in the above sentence in which a single bond is replaced with a double bond. It is understood that the specific rings and ring systems suitable for use in the present invention are not limited to those listed in this and the preceding paragraphs. These rings and ring systems are merely representative.
  • the present invention relates to compounds having the general formula II:
  • p 0, 1, 2, or 3;
  • q 1, 2, 3,or, 4;
  • r is 1, 2, 3,or, 4;
  • X is alkyl or aryl
  • B is C 5 -Ci 2 aryl
  • R 8 is selected from the group consisting of hydrogen, halogen, Ci-C 10 alkyl, C3-C10
  • cycloalkyl hydroxyl, -OR 10 , -CN, -N0 2 , -NH 2 , -N(R 10 )C(O)R 10 , -C(0)R 10 , -C(0)-OR 10 , - C(O)N(R 10 ) 2, -S(0)R 10 , -S(0) 2 R 10 , -S(O) 2 N(R 10 ) 2 , aryl, benzyl, heteroaryl, hetero-cyclyl, or two groups of R 8 are connected with each other to make a five or six membered cyclic or heterocyclic ring, any of which is optionally substituted;
  • R 9 is selected from the group consisting of hydrogen, halogen, C 1-C 10 alkyl, C 3 -C 10 cycloalkyl, C 2 -Ci 0 alkenyl, C 3 -C 1 ocycloalkenyl, C3-C15 cycloalkylalkoxy, C 3 -Ci 5
  • R 10 and R u are independently, at each occurrence, selected from the group consisting from hydrogen, Q-do alkyl, C3-C 10 cycloalkyl, C 2 -Cioalkenyl, C 3 -Cio cycloalkenyl, C 2 -Cio alkynyl, Ci-Cio haloalkyl, aryl, benzyl, heteroaryl, or heterocyclyl, any of which is optionally substituted;
  • halogen including fluorine, C i-Cio alkyl, C1-C3 haloalkyl, C 3 -C 7 cycloalkyl, oxo, -OH, -OR 12 , - OC(0)R 12 -CN, -N0 2 , -N(R 12 ) 2 , -N(R 12 )C(0)R 12 , -C(0)R 12 , -C(0)OR 12 , -C(0)N(R ,2 ) 2 , - S(0)R 12 , -S(0) 2 R 12 , -S(0) 2 N(R 12 ) 2 , phenyl, benzyl, heteroaryl,or heterocyclyl, as also defined further above;
  • R 12 is independently, at each occurrence, selected from the group consisting of hydrogen, aryl, benzyl, heteroaryl, heterocyclyl, Ci-C 8 alkyl, or C 3 -C 7 cycloalkyl;
  • HetA refers to "heteroaryl”
  • alkyl has the same meaning as defined above.
  • alkoxy has the same meaning as defined above.
  • alkenyl has the same meaning as defined above.
  • alkynyl has the same meaning as defined above.
  • alkylene has the same meaning as defined above.
  • alkenylene has the same meaning as defined above.
  • alkynylene has the same meaning as defined above.
  • cycloalkyl alone or in combination with any other term, has the same meaning as defined above.
  • haloalkyl has the same meaning as defined above.
  • heteroalkyl has the same meaning as defined above.
  • halogen has the same meaning as defined above.
  • phenyl has the same meaning as defined above.
  • heteroaryl has the same meaning as defined above.
  • heterocyclyl has the same meaning as defined above.
  • Another embodiment of the present invention is compounds of general formula I and II, or pharmaceutically acceptable salts thereof.
  • the present invention relates to compounds having the general formula VIII:
  • n 0, 1, 2, or 3;
  • X 3 is selected from the group comprising CH 2 , O, S and NH;
  • X4 is selected from the group comprising halide, alkyl, OR 23 , SR 24 and NR 25 R 26 ;
  • R 20 is selected from the group comprising acyl, alkoxy, alkyl, alkylamino, alkylcarboxylic acid, arylcarboxylic acid, alkylcarboxylic alkylester, alkylene, alkylether, alkylhydroxy, alkylthio, alkynyl, amido, amino, aryl, arylalkoxy, arylamino, arylthio, carboxylic acid, cyano, cycloalkyl, carboxylic acid, ester, halo, haloalkoxy, haloalkyl, haloalkylether, heteroaryl, heteroarylamino, heterocycloalkyl and hydrogen, any of which is optionally substituted;
  • R 21 and R 22 are each independently selected from the group comprising alkoxy, alkyl, alkylamino, alkylene, alkylether, alkylthio, alkynyl, amido, amino, aryl, arylether, arylalkoxy, arylamino, arylthio, carboxy, cyano, cycloalkyl, ester, halo, haloalkoxy, haloalkyl, heteroaryl, heteroarylamino, heterocycloalkyl, hydroxyl, hydrogen, nitro, thio, sulfonate, sulfonyl and sulfonylamino, any of which is optionally substituted;
  • R 23 is selected from the group comprising acyl, alkyl, alkylamino, alkylene, alkynyl, aryl, arylalkoxy, arylamino, arylthio, carboxy, cycloalkyl, ester, ether, haloalkyl, heteroaryl, heteroarylamino, heterocycloalkyl, hydrogen, thio, sulfonate, and sulfonylamino, any of which is optionally substituted;
  • R 2 4 is selected from the group comprising alkyl, alkylaryl, alkylene, alkynyl, aryl, cycloalkyl, ester, halo, haloalkyl, heteroaryl, heterocycloalkyl, and hydrogen, any of which is optionally substituted; and
  • R 25 and R 26 are each independently selected from the group comprising acyl, alkyl, aminoalkyl, alkylene, alkylthio, alkynyl, aryl, arylalkoxy, arylamino, arylthio, carboxy, cycloalkyl, ester, ether, halo, haloalkoxy, haloalkyl, haloalkylether, heteroaryl, heteroarylamino, heterocycloalkyl and hydrogen, any of which is optionally substituted.
  • the term "optionally substituted” as used herein is meant to indicate that a group, such as alkyl, alkylene, alkynyl, aryl, cycloalkyl, heterocycloalkyl, or heteroaryl, may be unsubstituted or substituted with one or more substituents as also defined further above. "Substituted” in reference to a group indicates that a hydrogen atom attached to a member atom within a group is replaced as also defined further above.
  • the present invention relates to compounds having the general formula
  • o 0, 1 , 2, or 3;
  • Zi and Z 2 are each independently selected from the group comprising hydrogen, halogen, C ⁇ - Cio alkyl, C3-C 1 0 cycloalkyl, C 2 -C 10 alkenyl, C 3 -C!o cycloalkenyl, C 3 -Ci 5 cycloalkylalkoxy, Cj-Cis cycloalkylalkyl, hydroxyl, haloalkyl, oxo, -OR 31 , -OC(0)R 31 , -OC(0)N(R 31 ) 2 , - C(0)OR 31 , -C(0)R 31 , -C(0)N(R 31 ) 2 , -CN, -N0 2 , -NH 2 , -N(R 3 1 ) 2 , -N(R 3 1 )C(0)R 31 , - N(R 31 )C(0)N(R 31 ) 2 , -OR 31 HetA, -OR 31 N(R 31
  • R 7 and R 28 are each independently selected from the group comprising alkoxy, alkyl, alkylamino, alkylene, alkylether, alkylthio, alkynyl, amido, amino, aryl, arylether, arylalkoxy, arylamino, arylthio, carboxy, cyano, cycloalkyl, ester, halo, haloalkoxy, haloalkyl, heteroaryl, heteroarylamino, heterocycloalkyl, hydroxyl, hydrogen, nitro, thio, sulfonate, sulfonyl and sulfonylamino, any of which is optionally substituted;
  • R 2 9 and R30 are each independently selected from the group comprising alkoxy, alkyl, alkylamino, alkylene, alkylether, alkylthio, alkynyl, amido, amino, aryl, arylether, arylalkoxy, arylamino, arylthio, carboxy, cyano, cycloalkyl, ester, halo, haloalkoxy, haloalkyl, heteroaryl, heteroarylamino, heterocycloalkyl, hydroxyl, hydrogen, nitro, thio, sulfonate, sulfonyl and sulfonylamino, or two groups of R 29 and R30 are connected with each other to make a five or six membered cyclic, heterocyclic, aryl, or heteroaryl ring, any of which is optionally substituted;
  • R 31 is independently, at each occurrence, selected from the group consisting from hydrogen, Ci-Cio alkyl, C 3 -Ci 0 cycloalkyl, C 2 -Ci 0 alkenyl, C3-C 10 cycloalkenyl, C 2 -Cio alkynyl, Cj-Cio haloalkyl, aryl, benzyl, heteroaryl, or heterocyclyl, any of which is optionally substituted.
  • alkyl as used herein is meant to indicate that a group, such as substituted or non- substituted CI -CJO alkyl group which has the straight or branched chain.
  • cycloalkyl as used herein is meant to indicate that a group, such as substituted or non-substituted cyclic compound of C3-C8 ring structure.
  • heteroaryl as used herein is meant to indicate that a group, such as substituted or non-substituted 5- to 9-membered aromatic compounds which have more than one heteroatom of N, O, and S in the ring structure itself.
  • R 31 and R 32 are each independently selected from the group comprising hydrogen, alkyl, alkyloxy, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylcarbonyloxy, alkylaminocarbonyl, alkyloxycarbonyl, cycloalkyl, cycloalkyloxy, cycloalkylamino, cycloalkylcarbonyl, cycloalkylcarbonylamino, cycloalkylcarbonyloxy, cycloalkylaminocarbonyl, cycloalkyloxycarbonyl, heteroaryl, heteroaryloxy, heteroaryl amino, heteroaryl carbonyl, heteroaryl carbonylamino, heteroaryl carbonyloxy, heteroaryl aminocarbonyl, heteroaryl oxycarbonyl, heteroaryl alkyl, heteroaryl alkyloxy, heteroaryl alkylamino, heteroaryl alkylcarbonyl, heteroaryl alkylcarbonyl, heteroaryl
  • the present invention relates to compounds having one of the formulas 1- 120, as shown in Example 6, 125-359 as shown in Example 7, preferably 4, 5, 13, 61 , 65, 71, 74, 78, 97, 102-105,1 17, 132-135, 137, 139-140, 147, 151-152, 160, 163, 173, 180, 184-185, 193, 195, 199-201, 204, 206-222, 224, 226, 229, 231-243, 245-278, 280-286, 290-305, 316, 324, 337, 340, 341, 355 and 356 as shown in Tables 1 or 2.
  • Particularly preferred compounds are compounds having one of the formulas 4, 5, 13, 61 , 65, 71, 74, 78, 97, 102-105, 1 17, 133, 206-210, 220, 231, 232, 235, 236, 257-259, 261, 264, 265, 267, 270, 273, 278, 295, 299-305, 337, 340 and 356 as shown in Tables 1-4.
  • the compounds as defined above have an inhibitory activity, preferably an inhibitory activity above 65%, on bacterial growth, preferably on the growth of M. tuberculosis, inside a host cell, preferably a macrophage, at a concentration between 5-20 ⁇ , preferably less than 5 ⁇ .
  • compositions in accordance with the present invention are also contemplated herein.
  • such pharmaceutically acceptable salts may be acid addition salts.
  • a compound in accordance with the present invention treated with an inorganic acid such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, and the like, or an organic acid such as an acetic, propionic, glycolic, pyruvic, oxalic, malic, malonic, succinic, maleic, fumaric, tataric, citric, benzoic, cinnamic, mandelic, methanesulfonic, ethanesulfonic, p-toluenesulfonic, salicyclic and the like, to provide a water soluble salt of the compound is suitable for use in the invention.
  • the present invention relates to compounds as defined above for use in the treatment of bacterial infections.
  • the present invention relates to compounds as defined above for use in the treatment of Tuberculosis.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound as defined above.
  • the present invention relates to a method of treatment of Tuberculosis, comprising the application of a pharmaceutically suitable amount of a compound as defined above to a person in need thereof.
  • the present invention relates to compounds having one of the general formulas/scaffolds I, II, VIII and Villa, or pharmaceutically acceptable salts thereof, as defined further above.
  • the present invention relates to a compound listed in Table 1.
  • the present invention relates to a compound listed in Table 2.
  • the present invention relates to compounds as defined above for use in the treatment of bacterial infections.
  • the present invention relates to compounds as defined above for use in the treatment of Tuberculosis.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound as defined above.
  • the present invention relates to a method of treatment of a bacterial infection, in particular tuberculosis, said method comprising the application of a pharmaceutically suitable amount of a compound as defined above to a patient in need thereof.
  • the patient is a non-human animal, in another embodiment, the patient is a human.
  • compositions of the invention are suitable for use in a variety of drug delivery systems. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia, PA, 17th ed. (1985). For a brief review of methods for drug delivery, see, Langer, Science 249: 1527-1533 (1990).
  • the pharmaceutical compositions may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, intravenous, intracranial, intraperitoneal, subcutaneous or intramuscular administration.
  • the carrier preferably comprises water, saline, alcohol, a fat, a wax or a buffer.
  • any of the above carriers or a solid carrier such as mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, glucose, sucrose, and magnesium carbonate, may be employed.
  • Biodegradable microspheres e.g., polylactate polyglycolate
  • suitable biodegradable microspheres are disclosed, for example, in U.S. Patent Nos. 4,897,268 and 5,075,109.
  • compositions for parenteral administration which comprise the compound dissolved or suspended in an acceptable carrier, preferably an aqueous carrier, e.g., water, buffered water, saline, PBS and the like.
  • an acceptable carrier e.g., water, buffered water, saline, PBS and the like.
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents and the like.
  • compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered.
  • the resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8.
  • the compounds of the invention can be incorporated into liposomes formed from standard vesicle-forming lipids.
  • a variety of methods are available for preparing liposomes, as described in, e.g., Szoka et ah, Ann. Rev. Biophys. Bioeng. 9: 467 (1980), U.S. Pat. Nos. 4,235,871, 4,501,728 and 4,837,028.
  • the targeting of liposomes using a variety of targeting agents is well known in the art ⁇ see, e.g., U.S. Patent Nos. 4,957,773 and 4,603,044).
  • the dosage ranges for the administration of the compounds of the invention are those large enough to produce the desired anti-infective effect.
  • the dosage should not be so large as to cause adverse side effects.
  • the dosage will vary with the age, condition, sex and extent of the disease in the animal/patient and can be determined by one of skill in the art.
  • the dosage can be adjusted by the individual physician in the event of any counterindications.
  • Controlled release preparations may be achieved by the use of polymers to conjugate, complex or adsorb the compound.
  • the controlled delivery may be exercised by selecting appropriate macromolecules (for example, polyesters, polyamino carboxymethylcellulose, and protamine sulfate) and the concentration of macromolecules as well as the methods of incorporation in order to control release.
  • Another possible method to control the duration of action by controlled release preparations is to incorporate the compound into particles of a polymeric material such as polyesters, polyamino acids, hydrogels, poly (lactic acid) or ethylene vinylacetate copolymers.
  • Figure 1 shows the monitoring of tubercle bacillus intracellular growth inside macrophages by automated confocal microscopy:
  • Figure 2 shows the pharmacological validation and MIC (minimal inhibitory concentration) comparison of the reference drugs in the in vitro growth fluorescence assay and the phenotypic cell-based assay:
  • Figure 3 shows assay automation validation of the phenotypic cell-based assay:
  • Figure 4 shows primary screening results for the phenotypic cell-based assay and the in vitro growth assay for 26500 compounds: (a) Percent inhibition based on infection ratio relative to each compound and distribution, (b) Percent inhibition based on RFU relative to each compound and distribution, (c) Comparison of inhibition percentage for the phenotypic cell- based assay and the in vitro growth assay for each compound;
  • Figure 5 shows serial dilution results from the in vitro growth fluorescence assay and the phenotypic cell-based assay: Typical curves for compounds inhibiting (a,b,c) in vitro bacterial growth (d,e,f) both in vitro and intracellular growth and (g,h,i) intracellular growth only. (a,d,g) Infection ratio relative to compound concentration. (b,e,h) Cell number relative to compound concentration. (c,f,i) Relative fluorescence intensity relative to compound concentration. Compound concentration is given in M;
  • Figure 6 shows (a) a scheme of assay automation, (b) a 384-plate format description; (c) a 384-plate dose-response curve description, A to P and a to b correspond to 2-fold serial dilution of INH and Rifampin respectively with a starting concentration of 20 mg/mL in well A or a; RIF: Rifampin 5 ⁇ g/mL, Cpd: compound, INH100 1 ⁇ £/ ⁇ ,, INH50 0.05 ⁇ g/mL;
  • FIG. 7 illustrates the colony forming units (CFUs) recovered from macrophages at different time points after infection with M. tuberculosis H37Rv.
  • CFUs colony forming units
  • FIG 8 illustrates the colony forming units (CFUs) recovered from macrophages at different time points after infection with M. tuberculosis H37Rv. Cells were infected and treated with the indicated amount of pyridopyrimidione compound 71 (4 to 20 ⁇ );
  • Table 2 pyridopyrimidinone derivatives (general scaffold VIII and Villa) with their respective inhibitory activities, wherein the numbers in bold print refer to the compounds listed in Example 7;
  • Table 3 shows the cytotoxicity and antibacterial spectrum of pyridopyrimidinone compound 133 (see Table 2);
  • Table 4 shows the frequency of spontaneous resistance for representative pyridopyrimidinone compound 264 (see Table 2).
  • H37Rv-GFP green fluorescent protein
  • tuberculosis H37Rv-GFP were prepared from 400 mL of a 15 days old Middlebrook 7H9 culture (Difco, Sparks MD, USA) supplemented with albumin-dextrose-catalase (ADC, Difco, Sparks MD, USA), glycerol and 0.05% Tween 80.
  • Bacilli were harvested by centrifugation at 3000 g for 20 min, washed twice with H 2 0 at room temperature, and resuspended in 1-2 mL of 10% glycerol at room temperature after recentrifugation. 250 ⁇ of bacilli were mixed with green fluorescent protein encoding plasmid and electroporated using a Biorad Gene Pulser (Biorad).
  • bacilli were resuspended in medium and left one day at 37°C.
  • Transformants were selected on Middlebrook 7H1 1 medium (Difco, Sparks MD, USA) supplemented with oleic acid- albumin-dextrose-catalase (OADC, Difco, Sparks MD, USA) and 50 ⁇ g/mL hygromycin (Invitrogen, Carlsbad, CA USA). The selected hygromycin-resistant and green fluorescent colonies appeared after 3 weeks.
  • a 100 mL culture of the H37Rv-GFP strain was grown in Middlebrook 7H9-ADC medium supplemented with 0.05% Tween 80 and 50 ⁇ g/mL of hygromycin.
  • Bacteria were harvested, washed twice and suspended in 50 mM sodium phosphate buffer (pH 7.5). The bacteria were then sonicated and allowed to stand for 1 hour to allow residual aggregates to settle. The bacterial suspensions were then aliquoted and frozen at -80°C. A single defrosted aliquot was used to quantify the CFUs (colony forming units) prior to inoculation and typical stock concentrations were about 2 to 5 x 10° CFU/mL.
  • CFUs colony forming units
  • the small synthetic molecules from the screening libraries were suspended in pure DMSO (Sigma, D5879-500 mL) at a concentration of 10 mM (Master plates) in Corning 96 well clear V-bottom polypropylene plates (Corning, #3956). The compounds were then reformatted in Greiner 384 well V-shape polypropylene plates (Greiner, #781280) and diluted to a final concentration of 2 mM in pure DMSO. The compounds were kept frozen until use. For screening, compound plates were incubated at room temperature until thawed.
  • the compounds were directly added into the assay plates from the DMSO stock using an EVObird liquid handler (Evotec Technologies), which transfers 250 nl of compound twice to reach a final dilution of 1 :100. This one-step dilution reduces the risk of compound precipitation in intermediate plates and allows for a low final DMSO concentration (1%).
  • Cells were first seeded in 50 ⁇ at a density of 20,000 cells per well of a 384- well plate (Evotec technologies #781058) for 16 hours and then infected with bacterial suspensions at a multiplicity of infection (MOI) varying from 10: 1 to 1 :1 (bacteria:host cells). After 2 hours, cells were washed three times with phosphate buffered saline (PBS) and the compounds diluted in fresh culture medium were added. Cells were incubated at 37 °C, 5% C0 2 for up to seven days. Macrophage batch infection assay scale-up
  • MOI multiplicity of infection
  • Cells (1.5 x 10 8 cells) were infected with H37Rv-GFP suspension at a MOI of 1 : 1 in 300 mL for 2 hours at 37 °C with shaking (100 rpm). After two washes by centrifugation at 1 100 rpm (Beckman SX4250, 165 g) for 5 min., the remaining extracellular bacilli from the infected cells suspension were killed by a 1 hour amykacin (20 ⁇ , Sigma, A2324-5G) treatment. After a final centrifugation step, cells were dispensed with the Wellmate (Matrix) into 384- well Evotec plates (#781058) preplated with 10 ⁇ of the respective compound diluted in cell medium.
  • Infected cells were then incubated in the presence of the compound for 5 days at 37 °C, 5% C0 2 . After five days, macrophages were stained with SYTO 60 (Invitrogen, SI 1342) followed by plate sealing and image acquisition. During screening, staining of the live cells was carried out on a set of three plates every two hours to limit cell death due to prolonged incubation with cell chemical stain.
  • Confocal images were recorded on an automated fluorescent confocal microscope OperaTM (Evotec Technologies) using a 20X-water objective (NA 0.70), 488-nm and 635-nm lasers and a 488/635 primary dichroic mirror. Each image was then processed using dedicated in- house image analysis software (IM). Parameters determined were the total cell number and the number of infected cells. Briefly, the algorithm first segments the cells on the red channel using a sequence of processing steps as described elsewhere (Fenistein et ah, 2008).
  • Infected cells are then defined as those having at least a given number of pixels (usually 3) whose intensity in the green channel is above a given intensity threshold. The ratio of infected cells to the total number of cells is the measure of interest (named infection ratio). For each well, 4 pictures were recorded and for each parameter, the mean of the four images was used.
  • IDBS ActivityBase
  • a frozen aliquot of M. tuberculosis H37Rv-GFP was diluted at 1.5 x 10 6 CFU /mL in Middlebrook 7H9-ADC medium supplemented with 0.05% Tween 80.
  • Greiner ⁇ clear-black 384-well plates (Greiner, #781091) were first preplated with 0.5 ⁇ of compound dispensed by EVOBird (Evotec) in 10 ⁇ of Middlebrook 7H9-ADC medium supplemented with 0.05% Tween 80.
  • 40 ⁇ of the diluted H37Rv-GFP bacterial suspension was then added on top of the diluted compound resulting in a final volume of 50 ⁇ containing 1% DMSO. Plates were incubated at 37 °C, 5% C0 2 for 10 days after which GFP-fluorescence was recorded using a Victor 3 reader (Perkin-Elmer Life Sciences).
  • Raw 264.7 (ATCC # TIB-71) (1.5* 10 8 cells) were infected with H37Rv-GFP (Abadie et al, 2005, Cremer et al, 2002) in suspension at a MOI of 1 : 1 for 2 hours at 37 °C with shaking. After two washes by centrifugation, the remaining extracellular bacilli from the infected cell suspension were killed by a 1 hour Amikacin (20 ⁇ , Sigma, A2324) treatment. After a final centrifugation step, cells were dispensed into 384-well Evotec plates (#781058) preplated with compounds and controls. Infected cells were then incubated for 5 days at 37°C, 5% C0 2 .
  • BMDM Murine Bone Marrow-Derived Macrophages
  • FCS heat-inactivated fetal calf serum
  • PBMC Peripheral Blood Mononuclear Cells
  • Buffy coat diluted in PBS supplemented with 1% > FCS was treated with 15 ml of Ficoll-Paque Plus (Amersham Biosciences, Sweden) and centrifuged at 2500 x g for 20 min.
  • PBMC were obtained by CD14 + bead separation (Miltenyi Biotec, Germany), washed 3 -times with PBS (1% FCS) and transferred to 75 cm culture flasks containing RPMI 1640 media, 10%) FCS and 50 ng/ml of recombinant-human macrophage colony stimulating factor (R & D systems, Minneapolis).
  • Mycobacteria-GFP were detected using a 488-nm laser coupled with a 535/50 nm detection filter and cells labeled with a 635-nm laser coupled with a 690/40 nm detection filter. Four fields were recorded for each plate well and each image was then processed using dedicated in-house image analysis software (IM) as described elsewhere (Fenistein et al, 2008).
  • IM in-house image analysis software
  • Mycobacterium tuberculosis H37Rv, H37Ra and BCG Pasteur were used as reference strains. All strains were diluted at 1.5 x 10 6 CFU /mL in Middlebrook 7H9-ADC medium supplemented with 0.05% Tween 80. 384- well plates (Greiner, #781091) were first preplated with 0.5 ⁇ of compound dispensed by EVOBird (Evotec) in 10 ⁇ of Middlebrook 7H9-ADC medium supplemented with 0.05% Tween 80. Forty microliters of the diluted H37Rv-GFP bacterial suspension was then added to the diluted compound resulting in a final volume of 50 ⁇ containing 1% DMSO.
  • the frequency of spontaneous mutations was determined on 7H10 plates containing increasing concentrations of dintirobenzamide (0.2, 0.8, 1.6 and 3.2 ⁇ g/ml) or pyridopyrimidinone (0.4, 0.8, 1.6 and 3.2 ⁇ g/ml) compounds.
  • 10 6 , 10 7 and 10 8 CFU containing bacterial suspensions were spread on compound containing agar plates. After 5-6 weeks at 37°C, colonies were counted and frequency of mutation was evaluated as the ratio of colonies relative to the original inoculum.
  • DMSO and ⁇ were used as negative and positive controls, respectively.
  • Example 1 Phenotvpic macrophage-based assay set-up and automated image quantification
  • Raw264.7 macrophages were first infected with mycobacteria that constitutively express green fluorescent protein (GFP) at different multiplicities of infection (MOI) followed by kinetic analysis.
  • GFP green fluorescent protein
  • MOI multiplicities of infection
  • the host live cells were daily labeled with the red chemical fluorescent dye Syto60, and confocal images of live samples were acquired using an automated confocal microscope. Typical images are displayed in Figure la.
  • a few discrete weakly fluorescent bacteria localized within the cells.
  • the average number of cells had increased and mycobacteria had started to spread into neighboring cells leading to zones of strongly fluorescent bacteria.
  • an in-house image analysis script was developed. This script enables the automated quantification of the number of cells and the percentage of infected cells, whereby an infected cell is a cell containing at least three green pixels with an intensity above a defined threshold ( Figure lb). 2 hours after infection, between 2 and 10% of Raw264.7 cells were found to harbor a low number of bacilli ( Figure lc). The percentage of infected cells, hereafter named infection ratio, continued to increase from 72 hours post-infection reaching up to 70% at seven days post infection. This increase in infection ratio correlated with an increase in cell mortality ( Figure ld/e).
  • Example 2 Comparative minimal inhibitory concentration of known anti-tubercular drugs
  • macrophages were infected in batch with M. tuberculosis before being dispensed onto the compounds.
  • the batch infection was carried out with macrophages in suspension at 37°C under mild shaking. Free unbound mycobacteria were removed by washing three times with PBS and differential centrifugation, as well as by an additional one-hour incubation step with amykacin, an antibiotic known to selectively kill extracellular microbes (Figure 6a).
  • M. tuberculosis infected macrophages were then seeded in plates that had been previously dispensed with the compounds, DMSO or antibiotic controls. The day-to-day as well as plate-to plate reproducibility was first tested.
  • Example 4 Primary screening of a large library of small synthetic compounds using the phenotypic cell-based assay
  • a 26500 small molecule compound library that was selected for its high chemical diversity and drug-like properties according to the Lipinski rules (Lipinski et al., 2001), was chosen as the first library to be screened using the validated phenotypic cell-based assay.
  • the primary screen was carried out with compounds at 20 ⁇ in singleton.
  • the throughput was set to about 6000 compounds per working day encompassing 25 plates.
  • the screening was performed with Raw264.7 cells that had been expanded from frozen stocks for ten days before infection with M. tuberculosis H37Rv-GFP.
  • the MICs obtained from 2 serial dilutions of ⁇ and Rifampin processed at the beginning and at the end of the screening day should show similar results compared to the values obtained during the validation (see above).
  • Each screened plate is then accepted by the quality control procedure if the window between DMSO and INH ( ⁇ g/ml) is higher than 3 and the CV calculated for the 320 compounds present in each plate is lower than 25.
  • Such quality control criteria allow the identification of hits with an activity higher than 75%.
  • the percent inhibition for each compound was determined relative to the corresponding mean infection ratio between 1 ⁇ g/mL INH (100%) and DMSO (0%) in the same 384-well plate. The percent inhibition distribution is centered around -20% of inhibition ( Figure 4a). It was decided to select compounds that have an inhibitory effect greater than 65% which corresponds to a little less than 1.5 % of the total compounds.
  • cell cytotoxicity An important parameter that can be measured during image analysis is the total cell number, also referred to as cell cytotoxicity.
  • a low cell number can be the result of two independent phenomena, the compound toxicity and M. tuberculosis growth mediated cell toxicity. Indeed, at day 5 after infection with M. tuberculosis, the cell number decreased to less than 100 cells per image compared to more than 500 cells per image for uninfected cells ( Figure le). In contrast, a high cell number is obtained only when the compound is not toxic and prevents mycobacterial growth. This turns out to be a second relevant measurement of a compound's anti-mycobacterial activity.
  • the 657 selected hits were first confirmed at 3 different concentrations, 20 ⁇ , 2 ⁇ and 0.2 ⁇ .
  • the activity was confirmed either at 20 ⁇ or 2 ⁇ , on the intracellular or the in vitro assay. From this latter list, 121 compounds demonstrated an inhibitory activity above 65% at 2 ⁇ without any apparent cell toxicity at 20 ⁇ and consequently were selected for further confirmation by ten 3-fold serial dilutions. All 121 compounds were confirmed by serial dilution with a MIC ranging between 250 nM and 20 ⁇ .
  • the results shown in Figure 5 are representative of the three types of behavior observed: most of the compounds exhibited a clear dose response curve when activity was measured as infection ratio (Figure 5b/e/h).
  • the 121 confirmed hits can be clustered as various independent/general scaffolds.
  • the number of compounds for each scaffold varied, ranging from 1 to 69 molecules.
  • the molecules from the 69-compound scaffold share a common structure which is similar to ⁇ thereby validating the screening results.
  • the pyridopyrimidinone general scaffold is the focus of the present invention.
  • Example 6 Derivatization of the pyridopyrimidinone compounds
  • the pyridopynmidinone compounds (scaffolds I and II) underwent derivatization according to the methods outlined below. (Schemes 1-6). Resulting derivatives were examined for inhibitory activity using the assay described above and the results are summarized in Table 1.
  • aldehyde 0.060 mmol
  • THF 500 uL
  • alkyl or phenylmagnessium bromide 3.0 M solution in ether, 0.070 mmol
  • the reaction mixture was quenched with water (3.0 mL) and extracted with
  • pyridopyrimidinone compounds (scaffold VIII and Villa) underwent derivatization according to the methods outlined below (Schemes 7-24). Resulting derivatives were examined for inhibitory activity using the assay described above and the results are summarized in Table 2.
  • G3 (36.6 ⁇ ) was dissolved in 760 ⁇ of tert-butyl alcohol and 180 ⁇ of 2- methyl-2- butene.
  • a solution of sodium chlorite (335 ⁇ ) and sodium dihydrogenphosphate (253 ⁇ ) in 300 ⁇ 1 of water was added dropwise. The reaction mixture was stirred at room temperature overnight. Volatile components were then removed under vacuum and the residue was dissolved in 10 ml of water and extracted with two 10 ml portions of hexane. The aqueous layer was acidified to pH 3 with HCl(aq) and extracted with 10 ml portions of methylene chloride. The combined organic layers were washed with 20 ml of cold water, dried and concentrated to give G5.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)
PCT/EP2011/000135 2010-01-13 2011-01-13 Anti - infective pyrido (1,2 -a) pyrimidines WO2011085990A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
JP2012548380A JP2013517233A (ja) 2010-01-13 2011-01-13 抗感染ピリド(1,2−a)ピリミジン
EP11700392A EP2523956A1 (en) 2010-01-13 2011-01-13 Anti - infective pyrido (1,2 -a) pyrimidines
AU2011206761A AU2011206761A1 (en) 2010-01-13 2011-01-13 Anti - infective pyrido (1,2 -a) pyrimidines
CN2011800059743A CN102812023A (zh) 2010-01-13 2011-01-13 抗感染吡啶并(1,2-a)嘧啶类
KR1020127018472A KR20130088002A (ko) 2010-01-13 2011-01-13 항-감염성 피리도(1,2-a)피리미딘
BR112012017473A BR112012017473A2 (pt) 2010-01-13 2011-01-13 pirido(1,2-a) pirimidinas, seu uso, bem como composição farmacêutica compreendendo tais compostos
US13/518,292 US20130012506A1 (en) 2010-01-13 2011-01-13 Anti-infective pyrido (1,2-a) pyrimidines
CA2786329A CA2786329A1 (en) 2010-01-13 2011-01-13 Anti - infective pyrido (1,2-a) pyrimidines

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29455210P 2010-01-13 2010-01-13
US61/294,552 2010-01-13

Publications (2)

Publication Number Publication Date
WO2011085990A1 true WO2011085990A1 (en) 2011-07-21
WO2011085990A8 WO2011085990A8 (en) 2012-10-26

Family

ID=43610218

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/000135 WO2011085990A1 (en) 2010-01-13 2011-01-13 Anti - infective pyrido (1,2 -a) pyrimidines

Country Status (9)

Country Link
US (1) US20130012506A1 (pt-PT)
EP (1) EP2523956A1 (pt-PT)
JP (1) JP2013517233A (pt-PT)
KR (1) KR20130088002A (pt-PT)
CN (1) CN102812023A (pt-PT)
AU (1) AU2011206761A1 (pt-PT)
BR (1) BR112012017473A2 (pt-PT)
CA (1) CA2786329A1 (pt-PT)
WO (1) WO2011085990A1 (pt-PT)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013119916A3 (en) * 2012-02-10 2013-10-24 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
WO2016086261A1 (en) * 2014-12-02 2016-06-09 Prana Biotechnology Limited 4H-PYRIDO[1,2-a]PYRIMIDIN-4-ONE COMPOUNDS
US9371336B2 (en) 2012-03-01 2016-06-21 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US9914722B2 (en) 2012-03-23 2018-03-13 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US10195202B2 (en) 2013-12-19 2019-02-05 Ptc Therapeutics, Inc. Methods for modulating the amount of RNA transcripts
US10668171B2 (en) 2015-05-30 2020-06-02 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US10874672B2 (en) 2015-12-10 2020-12-29 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
CN114106000A (zh) * 2021-12-15 2022-03-01 中国科学院新疆理化技术研究所 一种咪唑并[4,5-d]嘧啶酮三环类衍生物及抗肿瘤用途
US11382918B2 (en) 2017-06-28 2022-07-12 Ptc Therapeutics, Inc. Methods for treating Huntington's Disease
US11395822B2 (en) 2017-06-28 2022-07-26 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US11407753B2 (en) 2017-06-05 2022-08-09 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11608501B2 (en) 2017-06-14 2023-03-21 Ptc Therapeutics, Inc. Methods for modifying RNA splicing
US11685746B2 (en) 2018-06-27 2023-06-27 Ptc Therapeutics, Inc. Heteroaryl compounds for treating Huntington's disease
US11702646B2 (en) 2016-11-28 2023-07-18 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US11780839B2 (en) 2018-03-27 2023-10-10 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11858941B2 (en) 2018-06-27 2024-01-02 Ptc Therapeutics, Inc. Heterocyclic and heteroaryl compounds for treating Huntington's disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105153151A (zh) * 2015-08-28 2015-12-16 张文莲 一种嘧啶酮类化合物的合成方法

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4603044A (en) 1983-01-06 1986-07-29 Technology Unlimited, Inc. Hepatocyte Directed Vesicle delivery system
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
US4957773A (en) 1989-02-13 1990-09-18 Syracuse University Deposition of boron-containing films from decaborane
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
EP1262477A1 (en) * 2000-02-09 2002-12-04 Daiichi Pharmaceutical Co., Ltd. Anti-acid-fast bacterial agents containing pyridonecarboxylic acids as the active ingredient
US20090163545A1 (en) * 2007-12-21 2009-06-25 University Of Rochester Method For Altering The Lifespan Of Eukaryotic Organisms
WO2010001366A1 (en) * 2008-07-04 2010-01-07 Institut National De La Sante Et De La Recherche Medicale (Inserm) Piperazines derivatives as proteasome modulators
WO2010003533A2 (en) * 2008-06-17 2010-01-14 Institut Pasteur Korea Anti-infective compounds

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4603044A (en) 1983-01-06 1986-07-29 Technology Unlimited, Inc. Hepatocyte Directed Vesicle delivery system
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
US4957773A (en) 1989-02-13 1990-09-18 Syracuse University Deposition of boron-containing films from decaborane
EP1262477A1 (en) * 2000-02-09 2002-12-04 Daiichi Pharmaceutical Co., Ltd. Anti-acid-fast bacterial agents containing pyridonecarboxylic acids as the active ingredient
US20090163545A1 (en) * 2007-12-21 2009-06-25 University Of Rochester Method For Altering The Lifespan Of Eukaryotic Organisms
WO2010003533A2 (en) * 2008-06-17 2010-01-14 Institut Pasteur Korea Anti-infective compounds
WO2010001366A1 (en) * 2008-07-04 2010-01-07 Institut National De La Sante Et De La Recherche Medicale (Inserm) Piperazines derivatives as proteasome modulators

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACE PUBLISHING COMPANY
ABADIE, V.; BADELL, E.; DOUILLARD, P.; ENSERGUEIX, D.; LEENEN, P. J.; TANGUY, M.; FIETTE, L.; SAELAND, S.; GICQUEL, B.; WINTER, N.: "Neutrophils rapidly migrate via lymphatics after Mycobacterium bovis BCG intradermal vaccination and shuttle live bacilli to the draining lymph nodes", BLOOD, vol. 106, 2005, pages 1843 - 1850
ABASS, MOHAMED; ISMAIL, MOSTAFA M.; ABDEL-MONEM, WAFAA R.; MAYAS, AISHA S.: "Substituted pyridopyrimidinones. Part 5. Behavior of 2-hydroxy-4-oxo-4H-pyrido[1,2-a]pyrimidine-3-carbaldehyde in nucleophilic condensation reactions", CHEMICAL PAPERS, vol. 64, no. 1, 28 November 2009 (2009-11-28), pages 72 - 83, XP002626421, ISSN: 0366-6352 *
ANDRIES, K.; VERHASSELT, P.; GUILLEMONT, J.; GOHLMANN, H. W.; NEEFS, J. M.; WINKLER, H.; VAN GESTEL, J.; TIMMERMAN, P.; ZHU, M.; L: "A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis", SCIENCE, vol. 307, 2005, pages 223 - 227, XP002358962, DOI: doi:10.1126/science.1106753
ARAIN, T. M.; RESCONI, A. E.; SINGH, D. C.; STOVER, C. K.: "Reporter gene technology to assess activity of antimycobacterial agents in macrophages", ANTIMICROB AGENTS CHEMOTHER, vol. 40, 1996, pages 1542 - 1544
BRODIN, P.; MAJLESSI, L.; MARSOLLIER, L.; DE JONGE, M.I.; BOTTAI, D.; DEMANGEL, CL.; HINDS, J.; NEYROLLES, O.; BUTCHER, P.D.; LECL: "Dissection of ESAT-6 system 1 of Mycobacterium tuberculosis and impact on immunogenicity and virulence", INFECT IMMUN, vol. 74, 2006, pages 88 - 98, XP002474771, DOI: doi:10.1128/IAI.74.1.88-98.2006
C. PLUEG, B. WALLFISCH, P.V. BERNHARDT, L-J. BAKER, G.R. CLARK, M. W. WONG, C. WENTRUP: "Mesoionic pyridopyrimidinylium and pyridooxazinylium olates and non-mesoionic pyridopyrimidinones. Structures in the solid state, solution, and matrices", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANS. 2, no. 10, 21 September 2000 (2000-09-21), pages 2096 - 2108, XP002626412, ISSN: 0300-9580, DOI: 10.1039/B003933K *
CHO, S. H.; WARIT, S.; WAN, B.; HWANG, C. H.; PAULI, G. F.; FRANZBLAU, S. G.: "Low- oxygen-recovery assay for high-throughput screening of compounds against nonreplicating Mycobacterium tuberculosis", ANTIMICROB AGENTS CHEMOTHER, vol. 51, 2007, pages 1380 - 1385
CREMER, I.; DIEU-NOSJEAN, M. C.; MARECHAL, S.; DEZUTTER-DAMBUYANT, C.; GODDARD, S.; ADAMS, D.; WINTER, N.; MENETRIER-CAUX, C.; SAU: "Long-lived immature dendritic cells mediated by TRANCE-RANK interaction", BLOOD, vol. 100, 2002, pages 3646 - 3655, XP002444107, DOI: doi:10.1182/blood-2002-01-0312
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 10 April 2006 (2006-04-10), XP002626417, Database accession no. 879955-40-5 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 10 April 2006 (2006-04-10), XP002626418, Database accession no. 879947-89-4 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 10 November 2010 (2010-11-10), XP002626420, Database accession no. 1252331-94-4 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 14 April 2006 (2006-04-14), XP002626416, Database accession no. 880395-82-4 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 2 January 2004 (2004-01-02), XP002626419, Database accession no. 633299-82-8 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 22 December 2008 (2008-12-22), XP002626415, Database accession no. 1088210-57-4 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 3 February 2009 (2009-02-03), XP002626414, Database accession no. 1100208-88-5 *
FENISTEIN, D.; LENSEIGNE, B.; CHRISTOPHE, T.; BRODIN, P.; GENOVESIO, A.: "A fast fully automated cell segmentation algorithm for high throughput and high content screening", CYTOMETRY, vol. 73, 2008, pages 958 - 64
H. G. ANDERSEN, U. MITSCHKE, C. WENTRUP: "Ketenes and mesoions. Interconversion of mesoionic pyridopyrimidinium olates and pyridopyrimidinones. (2-Pyridyl)iminopropadienone. Part 2", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 2, no. 4, 1 March 2001 (2001-03-01), pages 602 - 607, XP002626413, ISSN: 1472-779X, DOI: 10.1039/b007298m *
HOUBEN, E. N.; NGUYEN, L.; PIETERS, J.: "Interaction of pathogenic mycobacteria with the host immune system", CURR OPIN MICROBIOL, vol. 9, 2006, pages 76 - 85, XP028033898, DOI: doi:10.1016/j.mib.2005.12.014
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LENAERTS, A. J.; HOFF, D.; ALY, S.; EHLERS, S.; ANDRIES, K.; CANTARERO, L.; ORME, I. M.; BASARABA, R. J.: "Location of persisting mycobacteria in a Guinea pig model of tuberculosis revealed by r207910", ANTIMICROB AGENTS CHEMOTHER, vol. 51, 2007, pages 3338 - 3345
LIPINSKI, C. A.; LOMBARDO, F.; DOMINY, B. W.; FEENEY, P. J.: "Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings", ADVANCED DRUG DELIVERY REVIEWS, vol. 46, 2001, pages 3 - 26
NEYROLLES, 0.; HERNANDEZ-PANDO, R.; PIETRI-ROUXEL, F.; FORNES, P.; TAILLEUX, L.; BARRIOS PAYAN, J. A.; PIVERT, E.; BORDAT, Y.; AGU: "Is adipose tissue a place for Mycobacterium tuberculosis persistence?", PLOS ONE, vol. 1, 2006, pages E43
ROHDE, K. H.; ABRAMOVITCH, R. B.; RUSSELL, D. G.: "Mycobacterium tuberculosis invasion of macrophages: linking bacterial gene expression to environmental cues", CELL HOST MICROBE, vol. 2, 2007, pages 352 - 364
SALOMON, J. A.; LLOYD-SMITH, J. 0.; GETZ, W. M.; RESCH, S.; SANCHEZ, M. S.; PORCO, T. C.; BORGDORFF, M. W.: "Prospects for advancing tuberculosis control efforts through novel therapies", PLOS MED, vol. 3, 2006, pages E273
SCHNAPPINGER, D.; EHRT, S.; VOSKUIL, M. I.; LIU, Y.; MANGAN, J. A.; MONAHAN, I. M.; DOLGANOV, G.; EFRON, B.; BUTCHER, P. D.; NATHA: "Transcriptional Adaptation of Mycobacterium tuberculosis within Macrophages: Insights into the Phagosomal Environment", J EXP MED, vol. 198, 2003, pages 693 - 704
SZOKA ET AL., ANN. REV. BIOPHYS. BIOENG., vol. 9, 1980, pages 467
VAN RIE, A.; ENARSON, D.: "XDR tuberculosis: an indicator of public-health negligence", LANCET, vol. 368, 2006, pages 1554 - 1556, XP025093602, DOI: doi:10.1016/S0140-6736(06)69575-5

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA029542B1 (ru) * 2012-02-10 2018-04-30 ПиТиСи ТЕРАПЬЮТИКС, ИНК. ПРОИЗВОДНЫЕ 4Н-ПИРИДО[1,2-а]ПИРИМИДИН-4-ОНА ДЛЯ ЛЕЧЕНИЯ СПИНАЛЬНОЙ МЫШЕЧНОЙ АТРОФИИ
EA037123B1 (ru) * 2012-02-10 2021-02-09 ПиТиСи ТЕРАПЬЮТИКС, ИНК. Соединения для лечения спинальной мышечной атрофии
US11753407B2 (en) 2012-02-10 2023-09-12 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
WO2013119916A3 (en) * 2012-02-10 2013-10-24 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US9586955B2 (en) 2012-02-10 2017-03-07 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
JP2017122097A (ja) * 2012-02-10 2017-07-13 ピーティーシー セラピューティクス, インコーポレイテッド 脊髄性筋萎縮症を治療するための化合物
US9879007B2 (en) 2012-02-10 2018-01-30 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US10851101B2 (en) 2012-02-10 2020-12-01 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
JP2015508075A (ja) * 2012-02-10 2015-03-16 ピーティーシー セラピューティクス, インコーポレイテッド 脊髄性筋萎縮症を治療するための化合物
US9371336B2 (en) 2012-03-01 2016-06-21 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US9914722B2 (en) 2012-03-23 2018-03-13 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US10195202B2 (en) 2013-12-19 2019-02-05 Ptc Therapeutics, Inc. Methods for modulating the amount of RNA transcripts
US10688099B2 (en) 2013-12-19 2020-06-23 Ptc Therapeutics, Inc. Methods for modulating the amount of RNA transcripts
WO2016086261A1 (en) * 2014-12-02 2016-06-09 Prana Biotechnology Limited 4H-PYRIDO[1,2-a]PYRIMIDIN-4-ONE COMPOUNDS
US10287285B2 (en) 2014-12-02 2019-05-14 Prana Biotechnology Limited 4H-pyrido[1,2-A]pyrimidin-4-one compounds
US10738050B2 (en) 2014-12-02 2020-08-11 Prana Biotechnology Limited 4H-pyrido[1,2-A]pyrimidin-4-one compounds
EA031505B1 (ru) * 2014-12-02 2019-01-31 Прана Байотекнолоджи Лимитед СОЕДИНЕНИЯ 4Н-ПИРИДО[1,2-а]ПИРИМИДИН-4-ОНА
US10668171B2 (en) 2015-05-30 2020-06-02 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US11602567B2 (en) 2015-05-30 2023-03-14 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US10874672B2 (en) 2015-12-10 2020-12-29 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US11638706B2 (en) 2015-12-10 2023-05-02 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US10881658B2 (en) 2015-12-10 2021-01-05 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US11702646B2 (en) 2016-11-28 2023-07-18 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US11407753B2 (en) 2017-06-05 2022-08-09 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11608501B2 (en) 2017-06-14 2023-03-21 Ptc Therapeutics, Inc. Methods for modifying RNA splicing
US11382918B2 (en) 2017-06-28 2022-07-12 Ptc Therapeutics, Inc. Methods for treating Huntington's Disease
US11395822B2 (en) 2017-06-28 2022-07-26 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US11780839B2 (en) 2018-03-27 2023-10-10 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11685746B2 (en) 2018-06-27 2023-06-27 Ptc Therapeutics, Inc. Heteroaryl compounds for treating Huntington's disease
US11858941B2 (en) 2018-06-27 2024-01-02 Ptc Therapeutics, Inc. Heterocyclic and heteroaryl compounds for treating Huntington's disease
CN114106000A (zh) * 2021-12-15 2022-03-01 中国科学院新疆理化技术研究所 一种咪唑并[4,5-d]嘧啶酮三环类衍生物及抗肿瘤用途
CN114106000B (zh) * 2021-12-15 2023-11-24 中国科学院新疆理化技术研究所 一种咪唑并[4,5-d]嘧啶酮三环类衍生物及抗肿瘤用途

Also Published As

Publication number Publication date
CA2786329A1 (en) 2011-07-21
WO2011085990A8 (en) 2012-10-26
KR20130088002A (ko) 2013-08-07
CN102812023A (zh) 2012-12-05
EP2523956A1 (en) 2012-11-21
AU2011206761A1 (en) 2012-07-12
US20130012506A1 (en) 2013-01-10
JP2013517233A (ja) 2013-05-16
BR112012017473A2 (pt) 2019-09-24
AU2011206761A2 (en) 2012-12-13

Similar Documents

Publication Publication Date Title
WO2011085990A1 (en) Anti - infective pyrido (1,2 -a) pyrimidines
AU2009267519B2 (en) Pyridopyrimidine compounds as anti-tubercular agents
KR101732212B1 (ko) 항감염성 화합물
EP2054422B1 (en) Acrylamide derivatives as fab i inhibitors
WO2007067416A2 (en) Heterocyclylacrylamide compounds as fabi inhibitors and antibacterial agents
WO2007053131A2 (en) Acrylamide derivatives as antibiotic agents
CA3204171A1 (en) Bicyclic derivatives
EP3668856B1 (en) Novel tetrazole compounds and their use in the treatment of tuberculosis
AU2006228142A1 (en) Imidazopyridine derivatives useful as iNOS inhibitors
JP2015500295A (ja) ヒト結核菌を標的とする医薬としてのベンゾトリアジンオキサイド類
RU2800930C2 (ru) Новые производные тетразола и их применение в лечении туберкулеза
NZ539646A (en) Method of preparing inhibitors of phosphodiesterase-4 and intermediates thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180005974.3

Country of ref document: CN

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

Ref document number: 11700392

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011206761

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2786329

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2011700392

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 6040/CHENP/2012

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2011206761

Country of ref document: AU

Date of ref document: 20110113

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20127018472

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012548380

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 13518292

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012017473

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012017473

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120713