WO2009007419A1 - Novel bronchodilating isoquinoline carbamates - Google Patents

Novel bronchodilating isoquinoline carbamates Download PDF

Info

Publication number
WO2009007419A1
WO2009007419A1 PCT/EP2008/058993 EP2008058993W WO2009007419A1 WO 2009007419 A1 WO2009007419 A1 WO 2009007419A1 EP 2008058993 W EP2008058993 W EP 2008058993W WO 2009007419 A1 WO2009007419 A1 WO 2009007419A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound according
alkylene
fluoroalkyl
alkoxy
Prior art date
Application number
PCT/EP2008/058993
Other languages
French (fr)
Inventor
Maria Dalence
Martin Johansson
Viveca Thornqvist Oltner
Jörgen TOFTERED
David Wensbo
Original Assignee
Respiratorius Ab
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 Respiratorius Ab filed Critical Respiratorius Ab
Publication of WO2009007419A1 publication Critical patent/WO2009007419A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • C07D217/06Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines with the ring nitrogen atom acylated by carboxylic or carbonic acids, or with sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to novel bronchorelaxing compounds, pharmaceutical compositions comprising such compounds, and a method of treating or alleviating conditions accompanied by bronchoconstriction and/or inflammation of the respiratory tract, and/or vasoconstriction, by use of such compounds.
  • Hypertension i.e. high blood pressure
  • Medications presently used for the treatment of hypertension include the administration of beta-blockers, calcium channel blockers, diuretics, angiotensin-converting enzyme inhibitors and angiotensin II receptor antagonists.
  • Vasoconstriction results in an increase in the blood pressure.
  • WO 98/17648 discloses 3,4-dihydro-6,7-dihydroxy-2(lH)- isoquinolinecarboxylic acid phenylmethyl ester as a synthetic intermediate in the synthesis of anthranilic acid derivatives, which derivatives are drug resistance modulators.
  • various embodiments of the present invention seek to mitigate, alleviate, circumvent or eliminate one or more of the above-identified deficiencies identified herein.
  • such a compound or a composition comprising such a compound may be used in medicine and therapy.
  • Such compounds or compositions may also be used to manufacture a medicament.
  • a medicament comprising a compound according to formula I may be used to treat or prevent a disease or disorder of the respiratory apparatus characterized by bronchoconstriction.
  • a medicament comprising a compound according to formula I may be used to treat or prevent a disease or disorder characterized by inflammation or vasoconstriction.
  • Diseases and disorders, which compounds according to formula I may be used to treat comprise asthma, chronic obstructive pulmonary disease, which comprises chronic bronchitis and emphysema, bronchiectasis, cystic fibrosis, bronchiolitis or bronchopulmonary dysplasia.
  • a compound according to formula I or a medicament comprising such a compound may also be used in a method to treat or prevent pulmonary disease characterized by bronchoconstriction.
  • a method comprises the administration to a person in need of a bronchoconstriction relaxing dose of a compound according to formula I.
  • addition salt is intended to mean salts formed by the addition of a pharmaceutical acceptable acid, such as organic or inorganic acids, or a pharmaceutical acceptable base.
  • a pharmaceutical acceptable acid such as organic or inorganic acids, or a pharmaceutical acceptable base.
  • the organic acid may be, but is not limited to, acetic, propanoic, methanesulfonic, benzenesulfonic, lactic, malic, citric, tartaric, succinic or maleic acid.
  • the inorganic acid may be, but is not limited to, hydrochloric, hydrobromic, sulfuric, nitric acid or phosphoric acid.
  • the base may be, but is not limited to, ammonia and hydroxides of alkali or alkaline earth metals.
  • addition salt also comprises the hydrates and solvent addition forms, such as hydrates and alcoholates.
  • halo or “halogen” refers to fluoro, chloro, bromo, and iodo.
  • alkyl used alone or as a suffix or prefix, is intended to include both branched and straight chain saturated aliphatic hydrocarbon groups having from 1 to 12 carbon atoms or if a specified number of carbon atoms is provided then that specific number is intended.
  • C 1-6 alkyl denotes alkyl having 1, 2, 3, 4, 5 or 6 carbon atoms.
  • the specific number denoting the alkyl-group is the integer 0 (zero)
  • a hydrogen-atom is intended as the substituent at the position of the alkyl-group.
  • N(CO alkyl)2 is equivalent to "NH2" (amino).
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, sec-butyl, t-butyl, pentyl, and hexyl.
  • alkylenyl or “alkylene” used alone or as a suffix or prefix, is intended to mean straight chain saturated aliphatic hydrocarbon groups having from 1 to 12 carbon atoms or if a specified number of carbon atoms is provided then that specific number is intended.
  • C 1-6 alkylene or “C 1-6 alkylenyl” denotes “alkylene” or “alkylenyl” having 1, 2, 3, 4, 5 or 6 carbon atoms.
  • the specific number denoting the alkylenyl or alkylene-group is the integer 0 (zero)
  • a bond is intended to link the groups onto which the alkylenyl or alkylene-group is substituted.
  • NH(CO alkylene)NH2 is equivalent to "NHNH2" (hydrazino).
  • the groups linked by an alkylene or alkylenyl-group are intended to be attached to the first and to the last carbon of the alkylene or alkylenyl-group. In the case of methylene, the first and the last carbon is the same.
  • H2N(C2 alkylene)NH2 is equivalent to 1,2-diamino ethane, 1,3-diamino propane, pyrrolidinyl, piperidinyl and piperazinyl, respectively.
  • alkylene or alkylenyl examples include, but are not limited to, methylene, ethylene, propylene, and butylene.
  • alkoxy or "alkyloxy” is intended to mean an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge.
  • alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy, n-pentoxy, isopentoxy, cyclopropylmethoxy, allyloxy and propargyloxy.
  • alkylthio or “thioalkoxy” represent an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge.
  • fluoroalkyl refers to groups in which one, two, or three of the hydrogen(s) attached to any of the carbons of the corresponding alkyl, alkylene and alkoxy-groups are replaced by fluoro.
  • fluoroalkyl include, but are not limited to, trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 2-fluoroethyl and 3- fluoropropyl.
  • fluoroalkylene examples include, but are not limited to, difluoromethylene, fluoromethylene, 2,2-difluorobutylene and 2,2,3-trifluorobutylene.
  • fluoroalkoxy examples include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, 3,3,3-trifluoropropoxy and 2,2-difluoropropoxy.
  • substituted refers to an atom to which a hydrogen is covalently attached, and to which another substituent may be attached instead of the hydrogen, directly or indirectly through synthesis.
  • substitutable atoms include the carbon-atoms of pyridine. The nitrogen-atom of pyridine is not substitutable according to this definition.
  • aryl refers to a ring structure, comprising at least one aromatic ring, made up of from 5 to 14 carbon atoms. Ring structures containing 5, 6, 7 and 8 carbon atoms would be single-ring aromatic groups, for example phenyl. Ring structures containing 8, 9, 10, 11, 12, 13, or 14 would be polycyclic, for example naphthyl. The aromatic ring may be substituted at one or more ring positions.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein at least one of the rings is aromatic, for example, the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.
  • ortho, meta and para apply to 1,2-, 1,3- and 1 ,4-disubstituted benzenes, respectively.
  • the names 1 ,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
  • heteroaryl refers to a heteroaromatic heterocycle, having at least one ring with aromatic character, (e.g. 6 delocalized electrons) or at least two conjugated rings with aromatic character, (e.g. 4n + 2 delocalized electrons where "n" is an integer), and comprising up to about 14 carbon atoms, and having at least one heteroatom ring member such as sulfur, oxygen, or nitrogen.
  • Heteroaryl groups include monocyclic and bicyclic (e.g., having 2 fused rings) systems.
  • heteroaryl groups include without limitation, pyridyl (i.e., pyridinyl), pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl (i.e.
  • furanyl quinolyl, tetrahydroquinolyl, isoquinolyl, tetrahydroisoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrryl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, benzimidazolyl, indolinyl, and the like.
  • substituted refers to an atom to which a hydrogen atom may be covalently attached, and to which another substituent may be present instead of the hydrogen atom.
  • substitutable atoms include the carbon-atoms of pyridine.
  • the nitrogen-atom of pyridine is not substitutable according to this definition.
  • the imine nitrogen at position 3 in imidazole is not substitutable, while the amine nitrogen at position 1 is.
  • Rl and R2 are, independently of each other, selected from H and methyl and at least one of them is H; R3 and R4 are, independently of each other, selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl; G is a CO-2 alkylene aryl or CO-2 alkylene heteroaryl; said alkylene is optionally substituted with 1 or 2 substituent(s) independently selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl; said aryl or heteroaryl is optionally substituted with 1 or 2 substituent(s) independently selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO- 3 alkyleneN(Cl-5 alkyl)2, in which the C 1-5 alky
  • a compound according to formula (I) as disclosed herein may be present as a free base, an acid in its non-charged protonated form or a pharmaceutically acceptable addition salt, solvate or solvate of a salt thereof;
  • a compound according to formula (I) as disclosed herein may be present as pure stereoisomer, a racemic-, diastereomeric- or scalemic mixture.
  • Rl and R2 are, independently of each other, selected from H and methyl and at least on of them is H;
  • R3 and R4 are, independently of each other, selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl;
  • G is selected from Gl to G5;
  • Ll is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "m” independently selected substituent(s) R5; "m” represents an integer number of 0, 1 or 2, i.e. if "m” is 0, then said alkylene is unsubstituted; R5 is selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl; Q is selected from phenyl and nitrogen containing heteroaryls, such as pyridyl and pyrimidinyl; Xl, X2, X3, X4 and X5 are independently selected from N and C and no more than two of Xl, X2, X3, X4 and X5 are N, the residue being C; Q is optionally substituted with a maximum of "n” independently selected substituent(s) R6 at any substitutable ring carbon atom; "n” represent an integer number of 0, 1 or 2, i.e.
  • R6 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the C 1 -5 alkyl may be the same or different, C 1 -5 alkylthio, S(O)C 1 -5 alkyl, SO2C 1 -5 alkyl, C 1-5 fluoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (CO)NH2, (CO)NHC 1-5 alkyl, (CO)N(C 1-5 alkyl)2,
  • R7 is selected from C 1-5 alkyl and C 1-5 fluoroalkyl
  • the condensed rings Wl and W2 together represent a bicyclic aromatic system, wherein X6, X7 and X8 are independently selected from N and C and no more than one of X6, X7 and X8 are N, the residue being C; said bicyclic aromatic system is optionally substituted with a maximum of "p" independently selected substituent(s) R8, at any substitutable ring carbon atom of any of the rings Wl and W2; "p” represents an integer number of 0, 1 or 2, i.e.
  • R8 is selected from Cl-5 alkyl, Cl- 5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the Cl-5 alkyl may be the same or different, Cl-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, Cl-5 fluoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, in which
  • v is O, then said alkylene is unsubstituted;
  • RI l is selected from C 1-5 alkyl and C 1-5 fluoroalkyl;
  • the condensed rings Tl and T2 together represent a bicyclic aromatic system, wherein Xl 1, Xl 2 and Xl 3 are independently selected from N and C and no more than one of Xl 1, Xl 2 and Xl 3 is N, the residue being C; said bicyclic aromatic system is optionally substituted with a maximum of "s” independently selected substituent(s) R12, at any substitutable ring carbon atom of any of the rings Tl and T2; "s" represents an integer number of O, 1 or 2, i.e.
  • R12 is selected from Cl-5 alkyl, Cl-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the Cl-5 alkyl may be the same or different, Cl-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, Cl-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C
  • Rl and R2 should be hydrogen in formula (I).
  • Compounds, in which at least one of Rl and R2 is hydrogen was shown to be more potent, in at least one of the herein described in- vitro methods, than the corresponding di-methoxy derivatives. In a preferred embodiment both are hydrogen.
  • R3 and R4 are, independently of each other, selected from H, fluoro, chloro, bromo, and methyl. In another embodiment they are independently of each other, selected from H, chloro and bromo and more preferred from chloro and bromo.
  • both R3 and R4 are the same and are selected from H, fluoro, chloro, bromo, and methyl. In another embodiment they are selected from H, chloro and bromo, more preferred from chloro and bromo and most preferred from chloro.
  • the advantage of a compound wherein R3 is identical to R4 is, among others, that the purification of synthetic intermediates to produce such a compound is easier.
  • Ll is preferably selected from methylene and ethylene, such as methylene. Further, if G is Gl, then according to on embodiment none or only one of Xl to X5 is N. According to another embodiment at least one of Xl to X5 is N. If one of Xl to X5 is N, when it is preferred if Xl or X2 is N, especially if only of Xl to X5 is N. According to another embodiment X2 is N.
  • G Gl
  • m is preferably 0 (zero), i.e. it is preferred if Ll is unsubstituted.
  • R5 is selected from methyl and trifluoromethyl.
  • Q it is preferred for Q to be substituted, i.e. it is preferred if "n” is 1 or 2. If “n” is 1 or 2, then it is preferred if one substituent is positioned at X3. If “n” is 2, then it is preferred if one substituent is positioned at X3 and the other at Xl or X5. In another embodiment "n” is 0 (zero).
  • R6 is preferably selected, independently of each other if “n” is 2, from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alkylene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N-morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
  • G is selected from G2 to G5.
  • "o" is 0 (zero), i.e. L2 is unsubstituted.
  • L2 is methylene.
  • X6, X7 and X8 are all C.
  • one of X6, X7 and X8 is N.
  • R8 is preferably selected, independently of each other if "p" is 2, from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
  • R8 is preferably selected, independently of each other if “p” is 2, from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N-morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
  • G is G3 it is preferred if X9 and XlO both are C.
  • R9 is preferably selected from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
  • R9 is preferably selected from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N- morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
  • RlO is preferably selected from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
  • RlO is preferably selected from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N- morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
  • G is G5
  • "v” is 0 (zero), i.e. L3 is unsubstituted.
  • Xl l, X12 and Xl 2 may all be C.
  • one of Xl 1, X12 and X12 may be N.
  • G is G5 it is preferred if L3 is methylene.
  • R12 is preferably selected, independently of each other if "s" is 2, from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
  • Rl 2 is preferably selected, independently of each other if “s” is 2, from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N-morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
  • a compound according to formula (I) may be used in therapy. It may be used to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being. Further it might be used to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by inflammation or vasoconstriction of the respiratory apparatus, or other organs or parts of the body, of a mammal, such as a human being.
  • a method to treat, revoke, mitigate, alleviate or prevent bronchoconstriction in a mammal, such as a human being, in need thereof is also disclosed.
  • a method to treat, revoke, mitigate, alleviate or prevent inflammation or vasoconstriction of the respiratory apparatus, or other organs or parts of the body, of a mammal, such as a human being, in need thereof includes the administration of therapeutically effective amount of a compound according to formula (I).
  • a compound according to formula (I) may be used for the prevention and/or treatment of a disease or condition selected from the group consisting of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia
  • a compound according to formula (I) may be used to manufacture a medicament, such as pharmaceutical composition.
  • a medicament may be useful to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being. It may also be useful to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by inflammation or vasoconstriction of the respiratory apparatus or other organs or parts of the body of a mammal, such as a human being.
  • a compound according to formula (I) may further be used to manufacture a medicament for the prevention and/or treatment of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia.
  • prevent/preventing should not be construed to mean that a condition and/or a disease never might occur again after use of a compound or medicament according to embodiments disclosed herein to achieve prevention. Further, the term should neither be construed to mean that a condition not might occur, at least to some extent, after such use to prevent said condition. Rather, “prevent/preventing” is intended to mean that the condition to be prevented, if occurring despite such use, will be less severe than without such use.
  • the compounds below may be used, as described above for other compounds of the invention, in the treatment of various diseases, such as diseases characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being. Further, these compounds may also be used in the manufacture of a medicament for the treatment of various diseases, such as diseases characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being.
  • the usefulness of the compounds, as defined in the preceding embodiments, in treating, pretreating, revoking, mitigating, alleviating and/or preventing a condition of the respiratory apparatus characterized by bronchoconstriction, were evaluated in a complex and relevant in vitro model.
  • the in vitro model was in accordance with the in vitro model disclosed in US 2006-0040254 Al and by Skogvall, S., Berglund, M., Dalence-Guzman, M. F., Svensson, K., J ⁇ nsson, P., Persson, C. G. A and Sterner, O., in Pulmonary Pharmacology and Therapeutics, vol 20:3, 2007, p. 273-280 All references disclosed herein are hereby incorporated in their entirety by reference.
  • lung tissue was obtained from patients undergoing lobectomia or pulmectomia due to lung carcinoma. From the bronchus of this tissue were rectangular oblong preparations obtained. The contraction induced by inflammatory mediators, such as Leukotriene D4, histamine, prostaglandin D2 or acetylcholine, in the presence and absence of the compound to be evaluated, were compared.
  • inflammatory mediators such as Leukotriene D4, histamine, prostaglandin D2 or acetylcholine
  • Capsazepine one of the first reported TRPVl -antagonists, has been shown to have an effect of human airways (Skogvall, S., Berglund, M., Dalence-Guzman, M. F., Svensson, K., J ⁇ nsson, P., Persson, C. G. A and Sterner, O., Pulmonary Pharmacology and Therapeutics, vol 20:3, 2007, p. 273-280), but is also known to posses a range of other biological effects. Consequently capsazepine is not selective towards one target and accordingly its usefulness as a molecular tool has been questioned (Gunthorpe, M. J., Neurpharmacology, 2004, 46, 133). According to one embodiment, preferred compounds according to any of the embodiments disclosed herein are those being at least comparably active to Capsazepine, in the in vitro model referred to herein.
  • preferred compounds according to any of the embodiments disclosed herein are those being at least comparably active to Res-4- 95, disclosed in Pulmonary Pharmacology & Therapeutics, 2007, 21(1), 125-133 as a potent analogue to Capsazepine, in the in vitro model referred to herein.
  • LTB4 leukotriene B4
  • MCP-I monocyte chemotactic protein- 1
  • MCP-I attracts monocytes that can differentiate into macrophages. Macrophages are generally believed to be responsible for the continued protolytic activity in the lungs of COPD-patients, as well as driving the inflammatory process in the same by recruitment of neutrophils. The fact that increased levels of various inflammatory mediators, or associated receptors, correlates with the diagnosis of COPD, is indicative of their relevance in disease severity and progression. Comparative studies between COPD-patients and non-COPD subjects have, for example, shown that the former group has increased levels of MCP-I in the sputum (Traves, L. S. et al, Thorax, 2002, 57, 590-595), increased mRNA expression of MCP-I in lung tissue (Tomaki, M.
  • LTB4 is an aracidonic acid metabolite involved in leukocyte recruitment. LTB4 is a potent chemoattractant and activator for neutrophils. The LTB4-receptors BLTl and PPAR are upregulated in peripheral lung of COPD patients (Marian, E. et al, 2006, 129, 1523-1530). Higher sputum- (Profita, M. et al, Allergy, 2005, 60, 1361-
  • the anti-inflammatory effect i.e. the usefulness in treating, pretreating, revoking, mitigating, alleviating and/or preventing an inflammation, such as an inflammation of the airways, of the compounds, as defined in the embodiments herein, may be assessed in an in vitro human peripheral blood mononuclear cell (PBMC) model. Further, the anti-inflammatory effect may be compared to the effect of dexamethasone, a known potent anti-inflammatory glucocorticoid.
  • PBMC peripheral blood mononuclear cell
  • preferred compounds according to any of the preceding embodiments are those being at least comparably active to dexamethaonse in such an anti-inflammatory in- vitro model.
  • a medicament e.g. a pharmaceutical composition, as has been described herein above may further comprise pharmaceutically acceptable carriers, diluents, stabilisers and/or excipients.
  • “Pharmaceutically acceptable” means a carrier, stabiliser, diluent or excipient that, at the dosage and concentrations employed, does not cause any unwanted effects in the patients to whom it is administered.
  • Such pharmaceutically acceptable carriers, stabilisers, dilutents or excipients are well-known in the art, and examples of such are for example disclosed in Remington's Pharmaceutical Sciences, 18th edition, A.R Gennaro, Ed., Mack Publishing Company (1990) and handbook of Pharmaceutical Excipients, 3rd edition, A. Kibbe, Ed., Pharmaceutical Press (2000).
  • a medicament according to embodiments herein maybe administered to a patient in a pharmaceutically effective dose.
  • pharmaceutically effective dose is meant a dose that is sufficient to produce the desired effects in relation to the condition for which it is administered. The exact dose may depend on the activity of the compound, manner of administration, nature and severity of the disorder and/or disease and the general conditions, such as age and body weight of the patient.
  • a medicament according embodiments herein may be administered alone or in combination with other therapeutic agents, such as anti-asthmatics. These agents may be incorporated as part of the same medicament or may be administered separately. It is well known in the art that a combination of mechanistically unrelated therapeutic agents in the same medicament may have beneficial effects in the treatment of conditions or diseases characterized by bronchoconstriction, as described in, for example, M. F. Fitzgerald and J. C. Fox, Drug Discovery Today, 2007, 12 (11/12), p. 472-478.
  • such other therapeutic agents to be administered in combination with a medicament according to embodiments of the invention are selected from therapeutic agents known to the one skilled in the art to prevent bronchoconstriction or revoke, fully or partly, any present bronchoconstriction.
  • therapeutic agents known to the one skilled in the art to prevent bronchoconstriction or revoke, fully or partly, any present bronchoconstriction.
  • agents are, but not limited to, ⁇ 2-agonists, anticholinergics calcium antagonists, and other agents suitable for the treatment of asthma and/or COPD and related diseases and/or disorders.
  • Preferred agents in this aspect are ⁇ 2-agonists and anticholinergics.
  • such other therapeutic agents to be administered in combination with the medicament of the invention may also comprise therapeutic agents known to the one skilled in the art to be useful to treat, revoke, mitigate, alleviate or prevent inflammation associated with diseases and disorders of respiratory tract. Examples of such agents are corticosteroids.
  • a therapeutically effective dose of said medicament may comprise 1 to 10 times less than the respective established therapeutically effective dose of the components, i.e. a compound according to embodiments disclosed herein and the therapeutic agent, when administered alone for prevention or treatment of the same disease or condition of each. Accordingly, by combining a compound according to the present invention with another therapeutic agent, such as an anti-asthmatic, it may be possible to achieve synergistic effects compared to if only a compound according to the present invention, or the other therapeutic agent, were administrated alone. Furthermore, it may be possible to improve both the underlying cause, e.g.
  • a method to treat, revoke, mitigate, alleviate or prevent bronchoconstriction and/or an inflammatory condition in a mammal, such as a human being, in need thereof, by the administration of a compound or medicament, such as a pharmaceutical composition, according to embodiments disclosed herein may also include the simultaneous or consecutive administration a therapeutic agent, such as an anti- asthmatic.
  • the therapeutically effective dose of said compound, medicament or pharmaceutical composition and said therapeutic agent may comprise 1 to 10 times less than the respective established therapeutically effective dose when administered alone for prevention or treatment of the same disease or condition. The advantageous of such co-administration are discussed above.
  • a medicament according to embodiments disclosed herein may be administered through different routes such as, but not limited to, intravenously, intraperitoneal, intramuscularly, intranasaleously, subcutaneously, sublingually, rectally, orally or through inhalation or insufflation.
  • Particular suitable formulations of the medicament of the invention are formulations suitable to be taken orally or to be administrated through inhalation or insufflation.
  • inhalation or insufflation will allow a high proportion of the delivered dose to reach the site of action, that is, the bronchi and the lung in general. Furthermore the systemic effects may be lower if the medicament is administrated through inhalation or insufflation compared to other administration routes.
  • Inhalation may be by the oral or the nasal route.
  • Conventional pulmonary applicators may be employed, such as pressurized spray containers comprising suitable propellants for aerosols and powder spray devices for preparations in form of fine powders.
  • Pharmaceutical compositions suitable for administration by the inhalation or insufflation route are known in the art.
  • the compound may be dissolved in a suitable vehicle or employed as a fine powder, such as a micronized powder of a particle size from about 2 ⁇ m to about 20 ⁇ m.
  • An indicated daily dose for administration by inhalation may be 10 times and more lower than the corresponding oral dose. Satisfactory doses, preferably metered by using a device capable of metering, or by single doses of predetermined size, may easily be determined by experimentation.
  • Compounds according to embodiments of the present invention may also be useful in treatment or prevention of hypertension.
  • oral administration is the preferred route of administration.
  • compounds according to formula I may also be useful as pharmacological tools in the development and standardisation of in vitro and in vivo test systems for the evaluation of other compounds with similar activity.
  • compounds of formula I may be used as molecular probes to identify and/or locate the target of their action, such as a target within the airways, as well as employed as a diagnostic tool for diagnosis of a disease or condition in vivo, ex vivo or in vitro, or as synthetic precursors to such probes.
  • Molecular probes of formula I may include reactive, labeled, i.e. compounds of formula I wherein one or several of the composing atoms have been enriched with a radioactive or by other means detectable isotope, and fluorescent compounds as well known to the one skilled in the art.
  • inventions of the present invention relates to processes for preparing a compound according to formula I as a free base, acid, or salts thereof. Further, additionally embodiments relate to synthetic intermediates, which are useful in the synthesis of a compound of formula I as a free base, acid, or salts thereof. Specific and generic examples of such intermediates are given below. Further, such intermediates may include compounds according to formula I, which may be used to produce another compound according to formula I. Throughout the following description of such processes it is to be understood that, where appropriate, suitable protecting groups will be attached to, and subsequently removed from, the various reactants and intermediates in a manner that will be readily understood by one skilled in the art of organic synthesis.
  • room temperature and “ambient temperature” shall mean, unless otherwise specified, a temperature between 16 and 25 0 C.
  • flash chromatography or flash column chromatography shall mean preparative chromatography on silica using an organic solvent, or mixtures thereof, as mobile phase.
  • CDI 1 , 1 '-carbonyl diimidazole equiv. equivalents; quant. quantitatively.
  • Formation of compounds of formula I may be accomplished by coupling of intermediates II and III (wherein depicted groups Rl to R4 and G are the same as the corresponding groups in compounds of formula I, and "LG" is a leaving group) under standard conditions, such as in the presence of an amine, such as triethylamine, in a solvent, such as dichloromethane or DMF.
  • suitable leaving groups LG in intermediates of formula III include chloro and N-imidazolyl.
  • Methyl groups may be present in II, or else introduced by treatment with a methyl halide in the presence of a base according to standard procedures. Said methyl groups may be removed, preferably before coupling with III, by treatment with, for example, hydrogen bromide or boron tribromide (Hall et al, Bioorg. Med. Chem, 2005, 13, 1409-1413). Other aromatic methyl ethers, optionally present in the molecule, are then simultaneously cleaved off.
  • Intermediates of formula III may readily be prepared by standard methods known in the art.
  • alcohols V may be reacted with activated carbonyl compounds IV, wherein both leaving groups LG might be different or the same, such as 1,1 '-carbonyldiimidazole, phosgene and diphosgene, in the presence of a base, such as triethylamine, in a solvent, such as dichloromethane or DMF, as described in, for example, Konakahara et al. SYNTHESIS, 1993, 103-106.
  • Scheme 2 Methods of preparation of intermediates of formula II (Scheme 2)
  • Examples of two non- limiting methods for the preparation of intermediates of formula II, by assembly of the tetrahydroisoquinoline ring, include pathway A and pathway B.
  • pathway A involves a Pictet-Spengler reaction in which readily available phenylethylamine VI is reacted with formaldehyde to yield VII, followed by cyclisation under acidic conditions (Yokoyama et al., J Org Chem, 1999, 64, 611-617; for a modified procedure allowing cyclisation onto electron poor aromatics see for example Stokker et al., Tetrahedron Lett, 1996, 37, 5453-5456).
  • pathway B involves a Pomerantz-Fritsch reaction under reductive conditions in which readily available benzaldehydes VIII are reacted with aminoacetales IX (depicted "alkyl” is preferably short alkyls such as ethyl) to yield X, followed by cyclisation under acidic and reductive conditions to yield II (see for example: Bobbit et al., J Org Chem, 1965, 30, 2247-2250; Bobbit et al., J Org Chem, 1968, 33, 856-858).
  • Additional methods for the preparation of intermediates II include, for example, the direct introduction of the substituents R3 and R4 by electrophilic aromatic substitution, such as chlorination by treatment with sulphuryl chloride in acetic acid, or bromination as described in Okano et al., Tetrahedron, 2006, 128, 7136-7137.
  • NMR spectra (in CDC13, CD3OD or DMSO-d6) were recorded on a Bruker DRX 400 or on a Bruker Ultrashield 400 spectrometer at 400 MHz. All chemical shifts are in ppm on the delta-scale ( ⁇ ) relative to TMS using the residual CHC13 peak in CDC13, or the residual CD2HOD peak in CD3OD, or the residual CD3SOCD2H peak in (CD3)2SO as internal standard (7.26, 3.31 or 2.50 ppm respectively relative to TMS) and the fine splitting of the signals as appearing in the recordings (s: singlet, d: doublet, t: triplet, q: quartet, m: multiplet, br: broad signal). Flash chromatography was performed using 6 ⁇ A 35-70 ⁇ m Davisil silica gel. TLC analyses were made on Silica Gel 60 F254 (Merck) plates and visualised under a 254/365 nm UV-lamp.
  • 6-(Thiophen-3-yl)pyridine-2-carboxaldehyde (515.0 mg, 2.7 mmol) was suspended in methanol (10 ml). The suspension was cooled to 0 °C and sodium borohydride was added. The mixture was stirred at rt for 40 minutes. The solvent was removed and the residue partitioned between water and EtOAc. The organic phase was dried (MgSO 4 ), filtered and concentrated. No further purification was done. (515.0 mg, quant).
  • PBMC peripheral blood mononuclear cell
  • PBMC viability 96%).
  • Cells are then resuspended to 1 x 106 cells/ml in culture media and 0.5 ml plated into 24 well culture plates (5 x 105 cells/well) before incubation for 30 minutes at 37°C with 5% CO2 prior to addition of a compound to be assessed (10 ⁇ M) or dexamethasone (1 ⁇ M).
  • a compound to be assessed (10 ⁇ M) or dexamethasone (1 ⁇ M).
  • LPS 0.1 ⁇ g/ml, Salmonella abortus equi, Sigma, # L 1887
  • the cells are incubated for another 24h before collection of the cell culture supernatants, which are assayed for the presence of MCP-I and LTB4.
  • MCP-I levels are quantified employing a Luminex-based assay according the manufacturer's instructions. Data are collected using a Luminex 100 (Luminex Corporation, Austin, TX). Standard curves are generated using a 5-parameter logistic curve fitting equation weighted by 1/y (StarStation V 2.0; Applied Cytometry Systems, Sacramento, CA). Each sample reading are interpolated from the appropriate standard curve. Calculated concentrations are multiplied by the appropriate dilution factor when necessary. LTB4 levels are quantified by ELISA following the manufacturer's instructions. Absorbance readings are detected using a ThermoMax microplate reader (Molecular Devices). Standard curves are generated using a 4-parameter logistic curve fitting equation (SoftMax Pro 4.7.1; Molecular Devices). Each sample reading are interpolated from the appropriate standard curve. Duplicate interpolated sample values are averaged and standard deviations are calculated. Calculated concentrations are multiplied by the appropriate dilution factor.

Abstract

The invention relates to novel compounds having the general formula (I), and which molecules are useful to manufacture a medicament to treat a disorder or disease characterized by bronchoconstriction, e.g. COPD and asthma, and vasoconstriction, e.g. hypertension.

Description

NOVEL BRONCHODILATING ISOQUINOLINE CARBAMATES
Field of the invention
The present invention relates to novel bronchorelaxing compounds, pharmaceutical compositions comprising such compounds, and a method of treating or alleviating conditions accompanied by bronchoconstriction and/or inflammation of the respiratory tract, and/or vasoconstriction, by use of such compounds.
Background Asthma and chronic obstructive pulmonary disease (COPD) are diseases affecting the respiratory system, which millions of people suffer from. These diseases are today regarded as inflammatory diseases and the symptoms comprise constriction of the airways. Common treatment of the associated bronchoconstriction involves use of beta-agonists, such as terbutalin and formoterol, and anticholinergics, such as ipratropium bromide and thiotropium bromide.
Hypertension, i.e. high blood pressure, increases the risk of stroke, heart attacks, heart failure and kidney disease. Medications presently used for the treatment of hypertension include the administration of beta-blockers, calcium channel blockers, diuretics, angiotensin-converting enzyme inhibitors and angiotensin II receptor antagonists. Vasoconstriction results in an increase in the blood pressure.
The treatments for prevention or reduction of bronchoconstricion, inflammation, such inflammation of the respiratory tract, and vasoconstriction are in many ways insufficient and there is a need for alternative treatments. WO 98/17648 discloses 3,4-dihydro-6,7-dihydroxy-2(lH)- isoquinolinecarboxylic acid phenylmethyl ester as a synthetic intermediate in the synthesis of anthranilic acid derivatives, which derivatives are drug resistance modulators.
Acta Chimica Academiae Scientiarium Hungaricae (1974), 80(1), 111-118, discloses the use of 3,4-dihydro-6-hydroxy-7-methoxy-2(lH)-isoquinolinecarboxylic acid phenylmethyl ester and 3,4-dihydro-7-hydroxy-6-methoxy-2(lH)- isoquinolinecarboxylic acid phenylmethyl ester in the synthesis of tetrahydroiosquino line-O-glucosides . Summary
Accordingly, various embodiments of the present invention seek to mitigate, alleviate, circumvent or eliminate one or more of the above-identified deficiencies identified herein.
According to one aspect of the present invention, there is provided a compound of the general formula (I),
Figure imgf000003_0001
wherein Rl is selected from H and methyl; R2 is selected from H and methyl; R3 is selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl; R4 is selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl; G is a CO-2 alkylene aryl or CO-2 alkylene heteroaryl; said alkylene is optionally substituted with 1 or 2 substituent(s) independently selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl; said aryl or heteroaryl is optionally substituted with 1 or 2 substituent(s) independently selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO- 3 alkyleneN(Cl-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fluoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2Cl-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHC 1-5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); as a free base or a pharmaceutically acceptable addition salt, solvate or solvate of a salt thereof and as a pure stereoisomer, a racemic-, diastereomeric- or scalemic mixture; with the provisos that not both Rl and R2 is methyl and that the following compounds are excluded:
Figure imgf000004_0001
According to another aspect such a compound or a composition comprising such a compound may be used in medicine and therapy. Such compounds or compositions may also be used to manufacture a medicament.
According to another aspect such a medicament comprising a compound according to formula I may be used to treat or prevent a disease or disorder of the respiratory apparatus characterized by bronchoconstriction. Furthermore, a medicament comprising a compound according to formula I may be used to treat or prevent a disease or disorder characterized by inflammation or vasoconstriction. Diseases and disorders, which compounds according to formula I may be used to treat comprise asthma, chronic obstructive pulmonary disease, which comprises chronic bronchitis and emphysema, bronchiectasis, cystic fibrosis, bronchiolitis or bronchopulmonary dysplasia.
According to another aspect a compound according to formula I or a medicament comprising such a compound, may also be used in a method to treat or prevent pulmonary disease characterized by bronchoconstriction. Such a method comprises the administration to a person in need of a bronchoconstriction relaxing dose of a compound according to formula I.
Further features of the invention are defined in the dependent claims and embodiments disclosed herein.
Detailed description of example embodiments
Definitions: In the context of the present application and invention, the following definitions apply:
The term "addition salt" is intended to mean salts formed by the addition of a pharmaceutical acceptable acid, such as organic or inorganic acids, or a pharmaceutical acceptable base. The organic acid may be, but is not limited to, acetic, propanoic, methanesulfonic, benzenesulfonic, lactic, malic, citric, tartaric, succinic or maleic acid. The inorganic acid may be, but is not limited to, hydrochloric, hydrobromic, sulfuric, nitric acid or phosphoric acid. The base may be, but is not limited to, ammonia and hydroxides of alkali or alkaline earth metals. The term "addition salt" also comprises the hydrates and solvent addition forms, such as hydrates and alcoholates.
As used herein, "halo" or "halogen" refers to fluoro, chloro, bromo, and iodo. As used herein, "alkyl" used alone or as a suffix or prefix, is intended to include both branched and straight chain saturated aliphatic hydrocarbon groups having from 1 to 12 carbon atoms or if a specified number of carbon atoms is provided then that specific number is intended. For example "C 1-6 alkyl" denotes alkyl having 1, 2, 3, 4, 5 or 6 carbon atoms. When the specific number denoting the alkyl-group is the integer 0 (zero), a hydrogen-atom is intended as the substituent at the position of the alkyl-group. For example, "N(CO alkyl)2" is equivalent to "NH2" (amino). Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, sec-butyl, t-butyl, pentyl, and hexyl.
As used herein, "alkylenyl" or "alkylene" used alone or as a suffix or prefix, is intended to mean straight chain saturated aliphatic hydrocarbon groups having from 1 to 12 carbon atoms or if a specified number of carbon atoms is provided then that specific number is intended. For example "C 1-6 alkylene" or "C 1-6 alkylenyl" denotes "alkylene" or "alkylenyl" having 1, 2, 3, 4, 5 or 6 carbon atoms. When the specific number denoting the alkylenyl or alkylene-group is the integer 0 (zero), a bond is intended to link the groups onto which the alkylenyl or alkylene-group is substituted. For example, "NH(CO alkylene)NH2" is equivalent to "NHNH2" (hydrazino). As used herein, the groups linked by an alkylene or alkylenyl-group are intended to be attached to the first and to the last carbon of the alkylene or alkylenyl-group. In the case of methylene, the first and the last carbon is the same. For example, "H2N(C2 alkylene)NH2", "H2N(C3 alkylene)NH2", "N(C4 alkylene)", "N(C5 alkylene)" and "N(C2 alkylene)2NH" is equivalent to 1,2-diamino ethane, 1,3-diamino propane, pyrrolidinyl, piperidinyl and piperazinyl, respectively.
Examples of alkylene or alkylenyl include, but are not limited to, methylene, ethylene, propylene, and butylene.
As used herein, "alkoxy" or "alkyloxy" is intended to mean an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy, n-pentoxy, isopentoxy, cyclopropylmethoxy, allyloxy and propargyloxy. Similarly, "alkylthio" or "thioalkoxy" represent an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge. As used herein, "fluoroalkyl", "fluoroalkylene" and "fluoroalkoxy", used alone or as a suffix or prefix, refers to groups in which one, two, or three of the hydrogen(s) attached to any of the carbons of the corresponding alkyl, alkylene and alkoxy-groups are replaced by fluoro. Examples of fluoroalkyl include, but are not limited to, trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 2-fluoroethyl and 3- fluoropropyl.
Examples of fluoroalkylene include, but are not limited to, difluoromethylene, fluoromethylene, 2,2-difluorobutylene and 2,2,3-trifluorobutylene.
Examples of fluoroalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, 3,3,3-trifluoropropoxy and 2,2-difluoropropoxy.
As used herein, the term "substitutable" refers to an atom to which a hydrogen is covalently attached, and to which another substituent may be attached instead of the hydrogen, directly or indirectly through synthesis. A non-limiting example of substitutable atoms include the carbon-atoms of pyridine. The nitrogen-atom of pyridine is not substitutable according to this definition.
As used herein, the term "aryl" refers to a ring structure, comprising at least one aromatic ring, made up of from 5 to 14 carbon atoms. Ring structures containing 5, 6, 7 and 8 carbon atoms would be single-ring aromatic groups, for example phenyl. Ring structures containing 8, 9, 10, 11, 12, 13, or 14 would be polycyclic, for example naphthyl. The aromatic ring may be substituted at one or more ring positions. The term "aryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein at least one of the rings is aromatic, for example, the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls. The terms ortho, meta and para apply to 1,2-, 1,3- and 1 ,4-disubstituted benzenes, respectively. For example, the names 1 ,2-dimethylbenzene and ortho-dimethylbenzene are synonymous. As used herein, "heteroaryl" refers to a heteroaromatic heterocycle, having at least one ring with aromatic character, (e.g. 6 delocalized electrons) or at least two conjugated rings with aromatic character, (e.g. 4n + 2 delocalized electrons where "n" is an integer), and comprising up to about 14 carbon atoms, and having at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include monocyclic and bicyclic (e.g., having 2 fused rings) systems. Examples of heteroaryl groups include without limitation, pyridyl (i.e., pyridinyl), pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl (i.e. furanyl), quinolyl, tetrahydroquinolyl, isoquinolyl, tetrahydroisoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrryl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, benzimidazolyl, indolinyl, and the like.
As used herein, the term "substitutable" refers to an atom to which a hydrogen atom may be covalently attached, and to which another substituent may be present instead of the hydrogen atom. A non- limiting example of substitutable atoms include the carbon-atoms of pyridine. The nitrogen-atom of pyridine is not substitutable according to this definition. Further, according to the same definition, the imine nitrogen at position 3 in imidazole is not substitutable, while the amine nitrogen at position 1 is.
According to one embodiment of the present invention there is disclosed a compound according to formula (I)
Figure imgf000007_0001
wherein Rl and R2 are, independently of each other, selected from H and methyl and at least one of them is H; R3 and R4 are, independently of each other, selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl; G is a CO-2 alkylene aryl or CO-2 alkylene heteroaryl; said alkylene is optionally substituted with 1 or 2 substituent(s) independently selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl; said aryl or heteroaryl is optionally substituted with 1 or 2 substituent(s) independently selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO- 3 alkyleneN(Cl-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fluoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2Cl-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHC 1-5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); with the proviso that the following compounds are excluded:
Figure imgf000008_0001
A compound according to formula (I) as disclosed herein may be present as a free base, an acid in its non-charged protonated form or a pharmaceutically acceptable addition salt, solvate or solvate of a salt thereof;
Further a compound according to formula (I) as disclosed herein may be present as pure stereoisomer, a racemic-, diastereomeric- or scalemic mixture.
As disclosed below, various embodiments of the present invent are drawn to compounds according to the general formula (I), as disclosed above, wherein the various generic groups (Rl to R4 and G) are elaborately disclosed.
In another embodiment of the invention Rl and R2 are, independently of each other, selected from H and methyl and at least on of them is H; R3 and R4 are, independently of each other, selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl; G is selected from Gl to G5;
Figure imgf000008_0002
when G is Gl then Ll is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "m" independently selected substituent(s) R5; "m" represents an integer number of 0, 1 or 2, i.e. if "m" is 0, then said alkylene is unsubstituted; R5 is selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl; Q is selected from phenyl and nitrogen containing heteroaryls, such as pyridyl and pyrimidinyl; Xl, X2, X3, X4 and X5 are independently selected from N and C and no more than two of Xl, X2, X3, X4 and X5 are N, the residue being C; Q is optionally substituted with a maximum of "n" independently selected substituent(s) R6 at any substitutable ring carbon atom; "n" represent an integer number of 0, 1 or 2, i.e. if "n" is 0, then Q is unsubstituted; R6 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the C 1 -5 alkyl may be the same or different, C 1 -5 alkylthio, S(O)C 1 -5 alkyl, SO2C 1 -5 alkyl, C 1-5 fluoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (CO)NH2, (CO)NHC 1-5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); when G is G2 then L2 is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "o" independently selected substituent(s) R7, wherein "o" represent an integer number of 0, 1 or 2, i.e. if "o" is 0, then said alkylene is unsubstituted; R7 is selected from C 1-5 alkyl and C 1-5 fluoroalkyl; the condensed rings Wl and W2 together represent a bicyclic aromatic system, wherein X6, X7 and X8 are independently selected from N and C and no more than one of X6, X7 and X8 are N, the residue being C; said bicyclic aromatic system is optionally substituted with a maximum of "p" independently selected substituent(s) R8, at any substitutable ring carbon atom of any of the rings Wl and W2; "p" represents an integer number of 0, 1 or 2, i.e. if "p" is 0, then Wl and W2 is unsubstituted; R8 is selected from Cl-5 alkyl, Cl- 5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the Cl-5 alkyl may be the same or different, Cl-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, Cl-5 fluoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, in which the Cl-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); when G is G3 then Y represents a fused benzene or pyridine ring, wherein X9 and XlO are independently selected from N and C and no more than one of X9 and XlO is N, the residue being C; Y is optionally substituted with one substituent R9 at any substitutable aromatic ring carbon atom; R9 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fiuoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); when G is G4 then Z represents a phenyl which is optionally substituted with one substituent RlO at any substitutable aromatic ring carbon; RlO is selected from Cl- 5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C 1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHC 1-5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); when G is G5 then L3 is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "v" independently selected substituent(s) Rl 1; "v" represents an integer number of O, 1 or 2, i.e. if "v" is O, then said alkylene is unsubstituted; RI l is selected from C 1-5 alkyl and C 1-5 fluoroalkyl; the condensed rings Tl and T2 together represent a bicyclic aromatic system, wherein Xl 1, Xl 2 and Xl 3 are independently selected from N and C and no more than one of Xl 1, Xl 2 and Xl 3 is N, the residue being C; said bicyclic aromatic system is optionally substituted with a maximum of "s" independently selected substituent(s) R12, at any substitutable ring carbon atom of any of the rings Tl and T2; "s" represents an integer number of O, 1 or 2, i.e. when "s" is 0, then said alkylene is unsubstituted; R12 is selected from Cl-5 alkyl, Cl-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, in which the Cl-5 alkyl may be the same or different, Cl-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, Cl-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, in which the C 1-5 alkyl may be the same or different, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido).
As disclosed above, either of Rl and R2 should be hydrogen in formula (I). Compounds, in which at least one of Rl and R2 is hydrogen was shown to be more potent, in at least one of the herein described in- vitro methods, than the corresponding di-methoxy derivatives. In a preferred embodiment both are hydrogen.
In another embodiment R3 and R4 are, independently of each other, selected from H, fluoro, chloro, bromo, and methyl. In another embodiment they are independently of each other, selected from H, chloro and bromo and more preferred from chloro and bromo.
In another embodiment both R3 and R4 are the same and are selected from H, fluoro, chloro, bromo, and methyl. In another embodiment they are selected from H, chloro and bromo, more preferred from chloro and bromo and most preferred from chloro. The advantage of a compound wherein R3 is identical to R4 is, among others, that the purification of synthetic intermediates to produce such a compound is easier.
In an embodiment wherein G is Gl, Ll is preferably selected from methylene and ethylene, such as methylene. Further, if G is Gl, then according to on embodiment none or only one of Xl to X5 is N. According to another embodiment at least one of Xl to X5 is N. If one of Xl to X5 is N, when it is preferred if Xl or X2 is N, especially if only of Xl to X5 is N. According to another embodiment X2 is N.
Furthermore, if G is Gl then m is preferably 0 (zero), i.e. it is preferred if Ll is unsubstituted. In an embodiment, wherein "m" is 1 or 2, such as being 1, R5 is selected from methyl and trifluoromethyl. Further it is preferred for Q to be substituted, i.e. it is preferred if "n" is 1 or 2. If "n" is 1 or 2, then it is preferred if one substituent is positioned at X3. If "n" is 2, then it is preferred if one substituent is positioned at X3 and the other at Xl or X5. In another embodiment "n" is 0 (zero).
If "n" is 1 or 2, then R6 is preferably selected, independently of each other if "n" is 2, from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alkylene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N-morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
In another embodiment G is selected from G2 to G5. In an embodiment wherein G is G2, "o" is 0 (zero), i.e. L2 is unsubstituted. In an embodiment wherein G is G2, it is preferred if L2 is methylene. Furthermore, in an embodiment wherein G is G2, X6, X7 and X8 are all C. In another embodiment wherein G is G2, one of X6, X7 and X8 is N. In embodiment wherein G is G2 and "p" is 1 or 2, R8 is preferably selected, independently of each other if "p" is 2, from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
In an embodiment wherein "p" is 1 or 2, R8 is preferably selected, independently of each other if "p" is 2, from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N-morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
In embodiment wherein G is G3 it is preferred if X9 and XlO both are C. In embodiment wherein G is G3 and wherein Y is substituted, R9 is preferably selected from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
In an embodiment Y is substituted, R9 is preferably selected from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N- morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
In embodiment wherein G is G4 and wherein Z is substituted, RlO is preferably selected from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
In an embodiment Z is substituted, RlO is preferably selected from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N- morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
In an embodiment wherein G is G5, "v" is 0 (zero), i.e. L3 is unsubstituted. In an embodiment wherein G is G5, Xl l, X12 and Xl 2 may all be C. In embodiment wherein G is G5, one of Xl 1, X12 and X12 may be N. In embodiment wherein G is G5 it is preferred if L3 is methylene. Additionally, in embodiment wherein G is G5 and "s" is 1 or 2, R12 is preferably selected, independently of each other if "s" is 2, from methyl, trifluoromethyl, fluoro, chloro, methoxy, SO2Me, cyano, thienyl and nitro, and more preferably from trifluoromethyl, fluoro and thienyl.
In an embodiment wherein "s" is 1 or 2, Rl 2 is preferably selected, independently of each other if "s" is 2, from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, such as fluoro and chloro, hydroxy, N(C4-5 alky lene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl, such as N-morpholinyl, and NMe2, and more preferably from methyl, trifluoromethyl, fluoro, chloro, NMe2 and thienyl, such as methyl, trifluoromethyl, fluoro, chloro and NMe2.
In another embodiment a compound according to formula I is selected from the group consisting of:
Figure imgf000013_0001
In another embodiment a compound according to formula I is selected from the group consisting of:
Figure imgf000013_0002
A compound according to formula (I) may be used in therapy. It may be used to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being. Further it might be used to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by inflammation or vasoconstriction of the respiratory apparatus, or other organs or parts of the body, of a mammal, such as a human being. A method to treat, revoke, mitigate, alleviate or prevent bronchoconstriction in a mammal, such as a human being, in need thereof is also disclosed. Furthermore a method to treat, revoke, mitigate, alleviate or prevent inflammation or vasoconstriction of the respiratory apparatus, or other organs or parts of the body, of a mammal, such as a human being, in need thereof is also disclosed. Such method includes the administration of therapeutically effective amount of a compound according to formula (I). Furthermore a compound according to formula (I) may be used for the prevention and/or treatment of a disease or condition selected from the group consisting of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia
Additionally, a compound according to formula (I) may be used to manufacture a medicament, such as pharmaceutical composition. Such a medicament may be useful to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being. It may also be useful to treat, revoke, mitigate, alleviate or prevent a disease or condition characterized by inflammation or vasoconstriction of the respiratory apparatus or other organs or parts of the body of a mammal, such as a human being. A compound according to formula (I) may further be used to manufacture a medicament for the prevention and/or treatment of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia. When used in herein, "prevent/preventing" should not be construed to mean that a condition and/or a disease never might occur again after use of a compound or medicament according to embodiments disclosed herein to achieve prevention. Further, the term should neither be construed to mean that a condition not might occur, at least to some extent, after such use to prevent said condition. Rather, "prevent/preventing" is intended to mean that the condition to be prevented, if occurring despite such use, will be less severe than without such use.
The compounds below may be used, as described above for other compounds of the invention, in the treatment of various diseases, such as diseases characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being. Further, these compounds may also be used in the manufacture of a medicament for the treatment of various diseases, such as diseases characterized by bronchoconstriction of the respiratory apparatus of a mammal, such as a human being.
Figure imgf000015_0001
The usefulness of the compounds, as defined in the preceding embodiments, in treating, pretreating, revoking, mitigating, alleviating and/or preventing a condition of the respiratory apparatus characterized by bronchoconstriction, were evaluated in a complex and relevant in vitro model. The in vitro model was in accordance with the in vitro model disclosed in US 2006-0040254 Al and by Skogvall, S., Berglund, M., Dalence-Guzman, M. F., Svensson, K., Jόnsson, P., Persson, C. G. A and Sterner, O., in Pulmonary Pharmacology and Therapeutics, vol 20:3, 2007, p. 273-280 All references disclosed herein are hereby incorporated in their entirety by reference.
In short, lung tissue was obtained from patients undergoing lobectomia or pulmectomia due to lung carcinoma. From the bronchus of this tissue were rectangular oblong preparations obtained. The contraction induced by inflammatory mediators, such as Leukotriene D4, histamine, prostaglandin D2 or acetylcholine, in the presence and absence of the compound to be evaluated, were compared.
Capsazepine, one of the first reported TRPVl -antagonists, has been shown to have an effect of human airways (Skogvall, S., Berglund, M., Dalence-Guzman, M. F., Svensson, K., Jόnsson, P., Persson, C. G. A and Sterner, O., Pulmonary Pharmacology and Therapeutics, vol 20:3, 2007, p. 273-280), but is also known to posses a range of other biological effects. Consequently capsazepine is not selective towards one target and accordingly its usefulness as a molecular tool has been questioned (Gunthorpe, M. J., Neurpharmacology, 2004, 46, 133). According to one embodiment, preferred compounds according to any of the embodiments disclosed herein are those being at least comparably active to Capsazepine, in the in vitro model referred to herein.
According to another embodiment, preferred compounds according to any of the embodiments disclosed herein are those being at least comparably active to Res-4- 95, disclosed in Pulmonary Pharmacology & Therapeutics, 2007, 21(1), 125-133 as a potent analogue to Capsazepine, in the in vitro model referred to herein.
Inflammation is closely associated with COPD. New drugs that reduce pulmonary inflammation by modulation of inflammatory pathways involving inflammatory mediators, such as leukotriene B4 (LTB4) and monocyte chemotactic protein- 1 (MCP-I), is believed to provide effective and disease-modifying therapies and is therefore much desired (Friedman et al, Clinical Cornerstone, 2003, 5, 45-51).
MCP-I attracts monocytes that can differentiate into macrophages. Macrophages are generally believed to be responsible for the continued protolytic activity in the lungs of COPD-patients, as well as driving the inflammatory process in the same by recruitment of neutrophils. The fact that increased levels of various inflammatory mediators, or associated receptors, correlates with the diagnosis of COPD, is indicative of their relevance in disease severity and progression. Comparative studies between COPD-patients and non-COPD subjects have, for example, shown that the former group has increased levels of MCP-I in the sputum (Traves, L. S. et al, Thorax, 2002, 57, 590-595), increased mRNA expression of MCP-I in lung tissue (Tomaki, M. et al, Pulmonary Pharmacology & Therapeutics, 2007, 20, 596-605), and increased lipopolysaccharide (LPS) stimulated release of MCP-I from isolated blood monocytes (Aldonyte, R. et al, Respiratory Research, 2003, http://respiratory- research.com/content/4/1/11).
LTB4 is an aracidonic acid metabolite involved in leukocyte recruitment. LTB4 is a potent chemoattractant and activator for neutrophils. The LTB4-receptors BLTl and PPAR are upregulated in peripheral lung of COPD patients (Marian, E. et al, 2006, 129, 1523-1530). Higher sputum- (Profita, M. et al, Allergy, 2005, 60, 1361-
1369) and serum- (Segger, J.S. et al, Chest, 1991, 99, 289-291) concentrations of LTB4 is found in COPD-patients as compared to healthy controls. Reduction of serum inflammatory mediator levels, including LTB4-levels, by a dietary supplement containing omega-3 polyunsaturated fatty acids, correlated significantly to a clinical improvement in COPD-patients (Matsuyama, W. et al, Chest, 2005, 128, 3817-3827).
Accordingly, the anti-inflammatory effect, i.e. the usefulness in treating, pretreating, revoking, mitigating, alleviating and/or preventing an inflammation, such as an inflammation of the airways, of the compounds, as defined in the embodiments herein, may be assessed in an in vitro human peripheral blood mononuclear cell (PBMC) model. Further, the anti-inflammatory effect may be compared to the effect of dexamethasone, a known potent anti-inflammatory glucocorticoid.
According to one embodiment, preferred compounds according to any of the preceding embodiments are those being at least comparably active to dexamethaonse in such an anti-inflammatory in- vitro model. A medicament, e.g. a pharmaceutical composition, as has been described herein above may further comprise pharmaceutically acceptable carriers, diluents, stabilisers and/or excipients.
"Pharmaceutically acceptable" means a carrier, stabiliser, diluent or excipient that, at the dosage and concentrations employed, does not cause any unwanted effects in the patients to whom it is administered. Such pharmaceutically acceptable carriers, stabilisers, dilutents or excipients are well-known in the art, and examples of such are for example disclosed in Remington's Pharmaceutical Sciences, 18th edition, A.R Gennaro, Ed., Mack Publishing Company (1990) and handbook of Pharmaceutical Excipients, 3rd edition, A. Kibbe, Ed., Pharmaceutical Press (2000).
A medicament according to embodiments herein maybe administered to a patient in a pharmaceutically effective dose. By "pharmaceutically effective dose" is meant a dose that is sufficient to produce the desired effects in relation to the condition for which it is administered. The exact dose may depend on the activity of the compound, manner of administration, nature and severity of the disorder and/or disease and the general conditions, such as age and body weight of the patient.
According to one embodiment, a medicament according embodiments herein may be administered alone or in combination with other therapeutic agents, such as anti-asthmatics. These agents may be incorporated as part of the same medicament or may be administered separately. It is well known in the art that a combination of mechanistically unrelated therapeutic agents in the same medicament may have beneficial effects in the treatment of conditions or diseases characterized by bronchoconstriction, as described in, for example, M. F. Fitzgerald and J. C. Fox, Drug Discovery Today, 2007, 12 (11/12), p. 472-478. In one embodiment of the invention such other therapeutic agents to be administered in combination with a medicament according to embodiments of the invention are selected from therapeutic agents known to the one skilled in the art to prevent bronchoconstriction or revoke, fully or partly, any present bronchoconstriction. Examples of such agents are, but not limited to, β2-agonists, anticholinergics calcium antagonists, and other agents suitable for the treatment of asthma and/or COPD and related diseases and/or disorders. Preferred agents in this aspect are β2-agonists and anticholinergics. Furthermore such other therapeutic agents to be administered in combination with the medicament of the invention may also comprise therapeutic agents known to the one skilled in the art to be useful to treat, revoke, mitigate, alleviate or prevent inflammation associated with diseases and disorders of respiratory tract. Examples of such agents are corticosteroids.
When a compound according to embodiments disclosed herein is combined with at least another therapeutic agent, such as an anti-asthmatic, in a medicament, such as a pharmaceutical composition, a therapeutically effective dose of said medicament may comprise 1 to 10 times less than the respective established therapeutically effective dose of the components, i.e. a compound according to embodiments disclosed herein and the therapeutic agent, when administered alone for prevention or treatment of the same disease or condition of each. Accordingly, by combining a compound according to the present invention with another therapeutic agent, such as an anti-asthmatic, it may be possible to achieve synergistic effects compared to if only a compound according to the present invention, or the other therapeutic agent, were administrated alone. Furthermore, it may be possible to improve both the underlying cause, e.g. the inflammation, and the clinical signs, e.g. airflow obstruction and exacerbations. A method to treat, revoke, mitigate, alleviate or prevent bronchoconstriction and/or an inflammatory condition in a mammal, such as a human being, in need thereof, by the administration of a compound or medicament, such as a pharmaceutical composition, according to embodiments disclosed herein may also include the simultaneous or consecutive administration a therapeutic agent, such as an anti- asthmatic. In such a method the therapeutically effective dose of said compound, medicament or pharmaceutical composition and said therapeutic agent may comprise 1 to 10 times less than the respective established therapeutically effective dose when administered alone for prevention or treatment of the same disease or condition. The advantageous of such co-administration are discussed above. A medicament according to embodiments disclosed herein may be administered through different routes such as, but not limited to, intravenously, intraperitoneal, intramuscularly, intranasaleously, subcutaneously, sublingually, rectally, orally or through inhalation or insufflation.
Particular suitable formulations of the medicament of the invention are formulations suitable to be taken orally or to be administrated through inhalation or insufflation.
Administration by inhalation or insufflation will allow a high proportion of the delivered dose to reach the site of action, that is, the bronchi and the lung in general. Furthermore the systemic effects may be lower if the medicament is administrated through inhalation or insufflation compared to other administration routes. Inhalation may be by the oral or the nasal route. Conventional pulmonary applicators may be employed, such as pressurized spray containers comprising suitable propellants for aerosols and powder spray devices for preparations in form of fine powders. Pharmaceutical compositions suitable for administration by the inhalation or insufflation route are known in the art. The compound may be dissolved in a suitable vehicle or employed as a fine powder, such as a micronized powder of a particle size from about 2 μm to about 20 μm. An indicated daily dose for administration by inhalation may be 10 times and more lower than the corresponding oral dose. Satisfactory doses, preferably metered by using a device capable of metering, or by single doses of predetermined size, may easily be determined by experimentation.
Compounds according to embodiments of the present invention may also be useful in treatment or prevention of hypertension. In the treatment of conditions or diseases characterized by hypertension, by employment of the compounds of the present invention, oral administration is the preferred route of administration. In addition to their use in therapeutic medicine, compounds according to formula I may also be useful as pharmacological tools in the development and standardisation of in vitro and in vivo test systems for the evaluation of other compounds with similar activity. Furthermore, compounds of formula I may be used as molecular probes to identify and/or locate the target of their action, such as a target within the airways, as well as employed as a diagnostic tool for diagnosis of a disease or condition in vivo, ex vivo or in vitro, or as synthetic precursors to such probes. Molecular probes of formula I may include reactive, labeled, i.e. compounds of formula I wherein one or several of the composing atoms have been enriched with a radioactive or by other means detectable isotope, and fluorescent compounds as well known to the one skilled in the art.
Methods of preparation
Other embodiments of the present invention relates to processes for preparing a compound according to formula I as a free base, acid, or salts thereof. Further, additionally embodiments relate to synthetic intermediates, which are useful in the synthesis of a compound of formula I as a free base, acid, or salts thereof. Specific and generic examples of such intermediates are given below. Further, such intermediates may include compounds according to formula I, which may be used to produce another compound according to formula I. Throughout the following description of such processes it is to be understood that, where appropriate, suitable protecting groups will be attached to, and subsequently removed from, the various reactants and intermediates in a manner that will be readily understood by one skilled in the art of organic synthesis. Conventional procedures for using such protecting groups, as well as examples of suitable protecting groups, are well known within the art. Further such procedures and groups are described in the literature, such as, in "Protective Groups in Organic Synthesis", 3rd ed., T.W. Green, P.G.M. Wuts, Wiley-Interscience, New York (1999).
It is also to be understood that a transformation of a group or substituent into another group or substituent by chemical manipulation can be conducted on any intermediate or final product on the synthetic path toward the final product, in which the possible type of transformation is limited only by inherent incompatibility of other functionalities carried by the molecule at that stage to the conditions or reagents employed in the transformation. Such inherent incompatibilities, and ways to circumvent them by carrying out appropriate transformations and synthetic steps in a suitable order, will be readily understood to the one skilled in the art of organic synthesis.
Examples of transformations are given below, and it is to be understood that the described transformations are not limited only to the generic groups or substituents for which the transformations are exemplified.
References and descriptions on other suitable transformations are for example given in "Comprehensive Organic Transformations - A Guide to Functional Group Preparations", 2nd ed., R. C. Larock, Wiley- VCH, New York (1999). References and descriptions of other suitable reactions are described in textbooks of organic chemistry well known to the one skilled in the art, such as "March's Advanced Organic
Chemistry", 5th ed., M. B. Smith, J. March, John Wiley & Sons (2001) or, "Organic Synthesis", 2nd ed., M. B. Smith, McGraw-Hill, (2002).
Techniques for purification of intermediates and final products include for example, straight and reversed phase chromatography on column or rotating plate, recrystallisation, distillation and liquid-liquid or solid-liquid extraction, which will be readily understood by the one skilled in the art.
The terms "room temperature" and "ambient temperature" shall mean, unless otherwise specified, a temperature between 16 and 25 0C. The term "reflux" shall mean, unless otherwise stated, in reference to an employed solvent using a temperature at or slightly above the boiling point of the named solvent. It is understood that microwaves can be used for the heating of reaction mixtures.
The terms "flash chromatography" or "flash column chromatography" shall mean preparative chromatography on silica using an organic solvent, or mixtures thereof, as mobile phase.
Abbreviations: aq. aqueous;
DMF N,N-dimethylformamide; DMSO dimethyl sulfoxide;
EtOAc ethyl acetate;
EtOH ethanol;
Et20 diethylether; h hour(s); Pet. Ether petroleum ether; r.t. or rt room temperature;
THF tetrahydrofurane;
TMS tetramethylsilane;
CDI 1 , 1 '-carbonyl diimidazole; equiv. equivalents; quant. quantitatively.
Methods of preparation of final compounds of formula I by coupling of intermediates II and III (Scheme 1)
Figure imgf000022_0001
Figure imgf000022_0002
IV
Scheme 1
Formation of compounds of formula I, may be accomplished by coupling of intermediates II and III (wherein depicted groups Rl to R4 and G are the same as the corresponding groups in compounds of formula I, and "LG" is a leaving group) under standard conditions, such as in the presence of an amine, such as triethylamine, in a solvent, such as dichloromethane or DMF. Examples of suitable leaving groups LG in intermediates of formula III include chloro and N-imidazolyl.
Methyl groups (Rl and R2 in intermediates of formula II) may be present in II, or else introduced by treatment with a methyl halide in the presence of a base according to standard procedures. Said methyl groups may be removed, preferably before coupling with III, by treatment with, for example, hydrogen bromide or boron tribromide (Hall et al, Bioorg. Med. Chem, 2005, 13, 1409-1413). Other aromatic methyl ethers, optionally present in the molecule, are then simultaneously cleaved off.
Intermediates of formula III may readily be prepared by standard methods known in the art. For example, alcohols V may be reacted with activated carbonyl compounds IV, wherein both leaving groups LG might be different or the same, such as 1,1 '-carbonyldiimidazole, phosgene and diphosgene, in the presence of a base, such as triethylamine, in a solvent, such as dichloromethane or DMF, as described in, for example, Konakahara et al. SYNTHESIS, 1993, 103-106. Methods of preparation of intermediates of formula II (Scheme 2)
Figure imgf000023_0001
Scheme 2
Examples of two non- limiting methods for the preparation of intermediates of formula II, by assembly of the tetrahydroisoquinoline ring, include pathway A and pathway B.
The synthesis according to pathway A involves a Pictet-Spengler reaction in which readily available phenylethylamine VI is reacted with formaldehyde to yield VII, followed by cyclisation under acidic conditions (Yokoyama et al., J Org Chem, 1999, 64, 611-617; for a modified procedure allowing cyclisation onto electron poor aromatics see for example Stokker et al., Tetrahedron Lett, 1996, 37, 5453-5456).
The synthesis corresponding to pathway B involves a Pomerantz-Fritsch reaction under reductive conditions in which readily available benzaldehydes VIII are reacted with aminoacetales IX (depicted "alkyl" is preferably short alkyls such as ethyl) to yield X, followed by cyclisation under acidic and reductive conditions to yield II (see for example: Bobbit et al., J Org Chem, 1965, 30, 2247-2250; Bobbit et al., J Org Chem, 1968, 33, 856-858).
Additional methods for the preparation of intermediates II include, for example, the direct introduction of the substituents R3 and R4 by electrophilic aromatic substitution, such as chlorination by treatment with sulphuryl chloride in acetic acid, or bromination as described in Okano et al., Tetrahedron, 2006, 128, 7136-7137.
Compound examples
General Methods
All materials were obtained from commercial sources, unless not stated otherwise, and were used without further purification unless otherwise noted. DMF was dried over molecular sieves (4A). THF was distilled from sodium and benzophenone. HRMS (ESI) spectra were recorded with a micromass Q-TOF Micro spectrometer.
NMR spectra (in CDC13, CD3OD or DMSO-d6) were recorded on a Bruker DRX 400 or on a Bruker Ultrashield 400 spectrometer at 400 MHz. All chemical shifts are in ppm on the delta-scale (δ) relative to TMS using the residual CHC13 peak in CDC13, or the residual CD2HOD peak in CD3OD, or the residual CD3SOCD2H peak in (CD3)2SO as internal standard (7.26, 3.31 or 2.50 ppm respectively relative to TMS) and the fine splitting of the signals as appearing in the recordings (s: singlet, d: doublet, t: triplet, q: quartet, m: multiplet, br: broad signal). Flash chromatography was performed using 6θA 35-70 μm Davisil silica gel. TLC analyses were made on Silica Gel 60 F254 (Merck) plates and visualised under a 254/365 nm UV-lamp.
Preparation of intermediates
Below follows non- limiting examples on the synthesis of intermediates useful for the preparation of compounds of formula I.
5,8-Dichloro-6,7-dimethoxy-l,2,3,4-tetrahydroisoquinoline hydrochloride
Figure imgf000024_0001
6,7-Dimethoxy-l,2,3,4-tetrahydroisoquinoline hydrochloride (200 mg, 0.870 mmol) was suspended in glacial acetic acid (5 mL) and sulphuryl chloride (154 μL, 1.92 mmol) was added slowly. The resulting mixture was stirred at room temperature for 3 hours and then evaporated to give the title compound (quant) as a yellowish mass.
1H NMR (400 MHz, CD3OD) 54.35 (br s, 2H), 3.90 (br d, 6H), 3.50 (br, 2H), 3.05 (br, 2H). 5,8-Dichloro-6,7-dihydroxy-l,2,3,4-tetrahydroisoquinoline hydrobromide
Figure imgf000025_0001
5, 8-Dichloro-6,7-dimethoxy-l, 2, 3,4-tetrahydroisoquino line hydrochloride (1.0 g, 3.35 mmol) was suspended in aqueous HBr (48%, 10 mL) and refluxed for 5 hours before evaporation. The remaining residue was evaporated twice from toluene to give 1.05 g (quant) of the title compound as a pale solid.
1H NMR (400 MHz, CD3OD) 56.60 (s, IH) 6.57 (s, IH) 4.26 (s, 2H) 3.50 (t, 2H, J=6 Hz) 3.01 (t, 2H, J=6 Hz).
2-(4-Fluorophenyl)-ethyl chloroformate
Figure imgf000025_0002
2-Phenylethanol (2.0 ml, 14.3 mmol) was dissolved in dichloromethane (50 ml) and cooled to 0 0C. Diphosgene (2.37 ml, 19.6 mmol) was slowly added followed by slow addition of diisopropylethylamine (2.45 ml, 14.3 mmol). The reaction was stirred at 0 0C for 1 h, then at rt for 21 h and finally refluxed for 2 h. The solvent was evaporated and the residue was diluted with THF (20 mL). The resulting suspension was filtered and evaporated into a yellowish liquid. This was suspended in Et2O, filtered through a plug of silica and evaporated to give 3.57 g of crude 2-(4-fluorophenyl)-ethyl chloroformate contaminated with diisopropylethylamine hydrochloride. This material was used in subsequent reactions without further purification.
1H NMR (CDCl3) δ 7.25-7.18 (m, 2H) 7.08-7.01 (m, 2H) 4.54-7.47 (m, 2H) 3.07-3.00 (m, 2H).
(6-(Thiophen-3-yl)pyridin-2-yl)methanol
6-(Thiophen-3-yl)pyridine-2-carboxaldehyde (515.0 mg, 2.7 mmol) was suspended in methanol (10 ml). The suspension was cooled to 0 °C and sodium borohydride was added. The mixture was stirred at rt for 40 minutes. The solvent was removed and the residue partitioned between water and EtOAc. The organic phase was dried (MgSO4), filtered and concentrated. No further purification was done. (515.0 mg, quant).
1H NMR (CDCl3) δ 7.94 (dd, J=3.0 Hz, J=I.2 Hz, IH) 7.70 (t, J=7.6 Hz, IH) 7.68 (dd, J=4.9 Hz, J=I.2 Hz, IH) 7.53 (dd, J=7.6 Hz, J=0.8 Hz, IH) 7.41 (dd, J=4.9 Hz, J=3.0 Hz, IH) 7.10 (dd, J=7.6 Hz, J=0.8 Hz, IH) 4.79 (bs, 2H) 4.15 (t, J=4.9 Hz, IH).
Preparation of final compounds The following non- limiting examples of compounds of formula I did all show less than 80% remaining contraction, at a concentration of lOμM, of human bronchiols after LTD4 induced contraction according to the method described herein.
Example 1 N-CPhenylmethoxycarbonylJ-S^-dichloro-ό^-dihydroxy-l,!^^- tetrahydroisoquinoline
Figure imgf000026_0001
5,8-Dichloro-6,7-dihydroxy-l,2,3,4-tetrahydroisoquinoline hydrobromide (150 mg, 0.476 mmol) and benzyl chloroformate (68 μL, 0.476 mmol) were suspended in dimethylformamide (5 mL). Triethylamine (134 μL, 0.952 mmol) was added and the resulting mixture was stirred at rt for 8 hours. The reaction mixture was then diluted with water (40 mL) and extracted with ethyl acetate (3x25 mL). The combined organics were dried over magnesium sulphate, filtered and evaporated into a reddish mass. Purification with flash chromatography (petroleum ether/ethyl acetate 2/1 → 1/1) yielded 46 mg (26%) of the title compound as an yellowish mass.
1H-NMR (CD3OD) 57.45-7.30 (m, 5H), 5.21 (s, 2H), 4.57 (s, 2H), 3.72 (t, J=6 Hz, 2H), 2.78 (br s, 2H).
TLC (pet.ether/EtOAc 1/1) Rf 0.55.
HRMS (ESI) calc for Ci7Hi6NO4Cl2 [M+H] 368.0456, found 368.0437.
General procedure for the synthesis of compounds of formula I (Scheme 1, R1=R2=H) via coupling in the presence of CDI and triethyl amine. CDI (1.0 equiv.) was suspended in anhydrous dichloromethane (3.0 mL/mmol CDI) under a nitrogen atmosphere. The solution was cooled to 0 °C and an alcohol of general formula V (1.0 equiv.) was added drop-wise as a solution in anhydrous dichloromethane (2.4 mL/mmol alcohol). The mixture was stirred until the alcohol was consumed as indicated by TLC. 5,8-dichloro-6,7-dihydroxy-l,2,3,4- tetrahydroisoquinoline hydrobromide (1.0 equiv.) and triethyl amine (3.0 equiv.) were added. The mixture was stirred at rt until the starting materials were consumed as indicated by TLC. EtOAc (10 mL) was added and the mixture was washed with aq. HCl (10%). The organic phase was dried (MgSO4), filtered and concentrated. Purification was done by flash chromatography (solvent system, yield and analytical data given for each intermediate).
Example 2 N-(Pyridin-2-ylmethoxycarbonyl)-5,8-dichloro-6,7-dihydroxy-l,2,3,4- tetrahydroisoquinoline
Figure imgf000027_0001
(EtOAc/Pet. Ether 2/1 → 3/1) Yield: 23%
1H NMR (CD3OD) δ 8.53 (d, J=5.1 Hz, IH) 7.86 (t, J=7.6 Hz, IH) 7.48 (bs, IH), 3.49 (dt, J=7.6 Hz, J=5.1 Hz, IH) 5.26 (s, 2H) 4.58(bs, 2H) 3.75 (bs, 2H) 2.79 (bs, 2H).
HRMS (ESI) calc for Ci6Hi5Cl2N2O4 [M+H] 369.0409, found 369.0396.
Example 3 N-(Naphthalen-l-ylmethoxycarbonyl)-5,8-dichloro-6,7-dihydroxy-l,2,3,4- tetrahydroisoquinoline
Figure imgf000027_0002
(Pet. Ether/EtOAc 2/1) Yield: 6%
1H NMR (DMSO-J6) δ 9.57 (bs, 2H) 8.07 (d, J=8.8 Hz, IH) 7.98 (d, J=8.8 Hz, IH) 7.93 (d, J=8.8 Hz, IH) 7.58-7.48 (m, 4H) 5.60 (s, 2H) 4.45 (s, 2H) 3.60 (s, 2H) 2.63 (bs, 2H).
HRMS (ESI) calc for C2IHi8Cl2NO4 [M+H] 418.0613, found 418.0630.
Example 4
N-(Naphthalen-2-ylmethoxycarbonyl)-5,8-dichloro-6,7-dihydroxy-l,2,3,4- tetrahydroisoquinoline
(Pet. Ether/EtOAc 2/1) Yield: 9%
1H NMR (DMSO-J6) δ 9.59 (bs, 2H) 7.92 (m, 4H) 7.52 (m, 3H) 5.30 (s, 2H) 4.49 (bs, 2H) 3.67 (s, 2H) 2.69 (t, J=6.0 Hz, 2H).
HRMS (ESI) calc for C2IHi8Cl2NO4 [M+Na] 440.0432, found 440.0382.
Example 5
4-(Trifluoromethyl)benzyl 5,8-dichloro-6,7-dihydroxy-3,4- dihydroisoquinoline-2(lH)-carboxylate
(Pet. Ether/EtOAc 2/1) Yield: 56 %
1H NMR (CD3OD) δ 1.61 (d, J=6.0 Hz, 2H) 7.57 (bs, 2H) 5.26 (s, 2H) 4.56 (bs, 2H) 3.72 (bs, 2H) 2.77 (t, J=6.0 Hz, 2H).
HRMS (ESI) calc for Ci8Hi5Cl2F3NO4 [M+H] 436.0330, found 436.0354.
Example 6
(6-(Trifluoromethyl)pyridin-3-yl)methyl 5,8-dichloro-6,7-dihydroxy-3,4- dihydroisoquinoline-2(lH)-carboxylate
Figure imgf000029_0001
(Pet. ether/EtOAc, gradient elution: 2/1 → 3/2) Yield: 30 %
1H NMR (CD3OD) δ 8.75 (s, IH) 8.07 (bs, IH) 7.83 (d, J=8.0 Hz, IH) 5.32 (s, 2H) 4.56 (bs, 2H) 3.72 (bs, 2H) 2.78 (t, J=6.0 Hz, 2H).
HRMS (ESI) calc for Ci7Hi4Cl2F3N2O4 [M+H] 437.0383, found 437.0237.
Example 7
(6-(Thiophen-3-yl)pyridin-2-yl)methyl 5,8-dichloro-6,7-dihydroxy-3,4- dihydroisoquinoline-2(lH)-carboxylate
(Pet. Ether/EtOAc 2/1) Yield: 37 %
1H NMR (CD3OD) δ 7.93 (bs, 1Η) 7.80 (bs, 1Η) 7.66 (m, 2Η) 7.42 (bs, IH) 7.27 (bs, IH) 5.31 (s, 2H) 4.63 (bs, 2H) 3.76 (bs, 2H) 2.80 (bs, 2H)
HRMS (ESI) calc for C20Hi7SCl2N2O4 [M+H] 451.0286, found 451.0284.
Example 8
2-(4-Fluorophenyl)-ethyl 6,7-dihydroxy-3,4-dihydroisoquinoline-2(lH)- carboxylate
Figure imgf000029_0002
l,2,3,4-Tetrahydroisoquinoline-6,7-diol hydrobromide (100 mg, 0.406 mmol) was dissolved together with triethyl amine (114 μl, 0.812 mmol) in DMF (1 ml). 2-(4- Fluorophenyl)-ethyl chloroformate was slowly added and the reaction mixture was stirred at rt for 16 h before evaporation into a brownish mass. The product was purified by flash chromatography using Pet. Ether/EtOAc (2/l→l/l) as eluent to give 58 mg (43 %) of 2-(4-fluorophenyl)-ethyl 6,7-dihydroxy-3 ,4-dihydroisoquinoline-2( 1 H)- carboxylate as an yellowish residue.
1U NMR (CDCl3) δ 7.25-7.17 (br, 2H) 7.02-6.90 (br, 2H) 6.62 (br d, 2H) 4.50- 4.38 (br, 2H) 4.32 (t, J= 7 Hz, 2H) 3.67-3.53 (br, 2H) 2.94 (t, J = IO Hz, 2H) 2.66 (t, J = 7 Hz).
TLC (Pet. Ether/EtOAc 1/1) Rf 0.44.
Biological examples
Biological example 1
The remaining contraction, after pre-treatment with various compound examples at a concentration of lOμM, of human bronchiols after Leukotriene D4 (1OnM) induced contraction according to the in vitro method described herein above are tabulated below.
Figure imgf000031_0001
Biological example 2
The remaining contraction, after pre-treatment with various compound examples at a concentration of 10 μM, of human bronchiols after the induction of contraction by treatment with a mixture of contractile agents consisting of acetylcholine (10OuM), leukotriene D4 (LTD4) (O.OluM) and histamine (10OuM), according to the in vitro method described herein above, are tabulated below:
CONTRACTI .LE .. C /OI-XM/ ΛP ivO /inU.N I-D DCM Λ IM IM/-
. _,_. .._. r REMA N NG
AGENT [concentration (EXAMPLE CONTRACTION (%)
(UM)] NUMBER) UON I KAU I IUN ( /o)
Mixture of 6 13
Acetylcholine
(10OuM), LTD4
(O.Ol uM) and histamine (10OuM) 7 39
Human peripheral blood mononuclear cell (PBMC) in vitro model
Cryopreserved PBMCs (SeraCare, # 72001) are thawed, washed with culture media (RPMI- 1640 from Invitrogen, # 61870-036 + 10% heat inactivated fetal bovine serum from Invitrogen, # 10082-147 + 100 U/ml penicillin + lOOμg/ml streptomycin) and tested for viability using Trypan blue (PBMC viability = 96%). Cells are then resuspended to 1 x 106 cells/ml in culture media and 0.5 ml plated into 24 well culture plates (5 x 105 cells/well) before incubation for 30 minutes at 37°C with 5% CO2 prior to addition of a compound to be assessed (10 μM) or dexamethasone (1 μM). One hour thereafter, LPS (0.1 μg/ml, Salmonella abortus equi, Sigma, # L 1887) are added and the cells are incubated for another 24h before collection of the cell culture supernatants, which are assayed for the presence of MCP-I and LTB4.
MCP-I levels are quantified employing a Luminex-based assay according the manufacturer's instructions. Data are collected using a Luminex 100 (Luminex Corporation, Austin, TX). Standard curves are generated using a 5-parameter logistic curve fitting equation weighted by 1/y (StarStation V 2.0; Applied Cytometry Systems, Sacramento, CA). Each sample reading are interpolated from the appropriate standard curve. Calculated concentrations are multiplied by the appropriate dilution factor when necessary. LTB4 levels are quantified by ELISA following the manufacturer's instructions. Absorbance readings are detected using a ThermoMax microplate reader (Molecular Devices). Standard curves are generated using a 4-parameter logistic curve fitting equation (SoftMax Pro 4.7.1; Molecular Devices). Each sample reading are interpolated from the appropriate standard curve. Duplicate interpolated sample values are averaged and standard deviations are calculated. Calculated concentrations are multiplied by the appropriate dilution factor.

Claims

Claims
1. A compound according to formula I,
Figure imgf000034_0001
wherein
Rl is selected from H and methyl;
R2 is selected from H and methyl;
R3 is selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl;
R4 is selected from H, fluoro, chloro, bromo, C 1-3 alkyl and CH2 phenyl;
G is a CO-2 alkylene aryl or CO-2 alkylene heteroaryl; said alkylene is optionally substituted with 1 or 2 substituent(s) independently selected from benzyl, Cl- 5 alkyl and C 1-5 fluoroalkyl; said aryl or heteroaryl is optionally substituted, at any substitutable ring atom, with 1 or 2 substituent(s) independently selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1 -5 alkyl, NH(CO)C 1 -5 alkoxy, NHSO2C 1 -5 alkyl, (CO)C 1 -5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHC 1-5 alkyl, (CO)N(C 1-5 alkyl)2, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); as a free base or a pharmaceutically acceptable addition salt, solvate or solvate of a salt thereof and as a pure stereoisomer, a racemic-, diastereomeric- or scalemic mixture;
with the provisos that
not both of Rl and R2 is methyl; and
the following compounds are excluded:
Figure imgf000035_0001
2. A compound according claim 1, wherein G is selected from Gl to G5;
Figure imgf000035_0002
(R1 1 )V
Figure imgf000035_0003
and wherein, Ll is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "m" independently selected substituent(s) R5, wherein "m" represent an integer number;
the integer number "m" is 0 to 2;
R5 is selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl;
Xl, X2, X3, X4 and X5 are, independently of each other, selected from N and C; 0 (zero), 1 or 2 of Xl to X5 are N; Q is optionally substituted with a maximum of "n" independently selected substituent(s) R6 at any substitutable ring carbon atom, wherein "n" represent an integer number;
the integer number "n" is 0 (zero) to 2;
R6 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C 1 -5 alkyl, (CO)C 1 -5 alkyl, COOH, (CO)C 1 -5 alkoxy, (C0)NH2, (CO)NHC 1 - 5 alkyl, (CO)N(C 1-5 alkyl)2, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido);
L2 is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "o" independently selected substituent(s) R7, wherein "o" represent an integer number;
the integer number "o" is 0 (zero) to 2;
R7 is selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl;
the condensed rings Wl and W2 together represent a bicyclic aromatic system, in which X6, X7 and X8, independently of each other, are selected from N and C; 0 (zero) or 1 of X6 to X8 is N; said bicyclic aromatic system is optionally substituted with a maximum of "p" independently selected substituent(s) R8, at any substitutable ring carbon atom(s) of any of the rings Wl and W2, wherein "p" represent an integer number;
the integer number "p" is 0 (zero) to 2;
R8 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C 1 -5 alkyl, (CO)C 1 -5 alkyl, COOH, (CO)C 1 -5 alkoxy, (C0)NH2, (CO)NHC 1 - 5 alkyl, (CO)N(C 1-5 alkyl)2, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido);
X9 and XlO are independently selected from N and C; O (zero) or 1 of X9 and XlO are N; Y is optionally substituted with one substituent R9 at any substitutable aromatic ring carbon atom.
R9 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido);
Z is optionally substituted with one substituent RlO at any substitutable aromatic ring carbon;
RlO is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido); L3 is C 1-2 alkylene, wherein said alkylene is optionally substituted with a maximum of "v" independently selected substituent(s) Rl 1, wherein "v" represent an integer number;
the integer number "v" is 0 (zero) to 2;
Rl 1 is selected from benzyl, C 1-5 alkyl and C 1-5 fluoroalkyl;
the condensed rings Tl and T2 together represent a bicyclic aromatic system, in which Xl 1, Xl 2 and Xl 3, independently of each other, are selected from N and C; 0 (zero) or 1 of said Xl 1, Xl 2 and Xl 3 is N; said bicyclic aromatic system is optionally substituted with a maximum of "s" independently selected substituent(s) R12, at any substitutable ring carbon atom(s) of any of the rings Tl and T2, wherein "s" represent an integer number;
the integer number "s" is 0 (zero) to 2;
Rl 2 is selected from C 1-5 alkyl, C 1-5 fluoroalkyl, halo, hydroxy, CO-3 alkyleneOCO-5 alkyl, CO-3 alkyleneOCl-5 fluoroalkyl, CO-3 alkyleneNH2, CO-3 alkyleneNHCl-3 alkyl, CO-3 alkyleneN(Cl-5 alkyl)2, C 1-5 alkylthio, S(O)C 1-5 alkyl, SO2C1-5 alkyl, C 1-5 fiuoroalkylthio, NH(CO)C 1-5 alkyl, NH(CO)C 1-5 alkoxy, NHSO2C1-5 alkyl, (CO)C 1-5 alkyl, COOH, (CO)C 1-5 alkoxy, (C0)NH2, (CO)NHCl- 5 alkyl, (CO)N(C 1-5 alkyl)2, cyano, SO2NHC0-5 alkyl, nitro, aryl, heteroaryl, morpholinyl, N(C4-5 alkylene) and N3 (azido);
3. The compound according to claim 1 or 2, wherein Rl and R2 is H.
4. The compound according to claim 1, 2 or 3, wherein R3 and R4, independent of each other, are selected from H, fluoro, chloro, bromo, and methyl.
5. The compound according to claim 4, wherein R3 and R4, independent of each other, are selected from H, chloro and bromo.
6. The compound according to claim 5, wherein R3 and R4 are chloro.
7. The compound according to any of the claims 2 to 6, wherein G is Gl.
8. The compound according to claim 7, wherein at least one of Xl to X5 is N.
9. The compound according to claim 8, wherein Xl or X2 is N and only one of Xl to X5 is N.
10. The compound according to any of the claims 7 to 9, wherein m is 0 (zero) and n is 1 or 2 and R6 is selected from C1-C3 alkyl, such as methyl, trifluoromethyl, halo, hydroxy, N(C4-5 alkylene), methoxy, SO2Me, cyano, thienyl, nitro, phenyl, morpholinyl and NMe2.
11. The compound according to claim 10, wherein R6 is selected from methyl, trifluoromethyl, fluoro, chloro and NMe2.
12. The compound according to any of the claims 2 to 6, wherein G is G2 and L2 is methylene or wherein G is G5 and L3 is methylene.
13. The compound according to claim 1, said compound being selected from the group consisting of:
Figure imgf000039_0001
14. A compound according to claim 1, said compound being selected from the group consisting of:
Figure imgf000040_0001
15. A pharmaceutical composition comprising a compound according to any of the claims 1 to 14 and at least one pharmaceutically acceptable carrier.
16. The pharmaceutical composition according to claim 15, comprising an antiasthmatic.
17. The pharmaceutical composition according to claim 16, wherein the principal mechanism of action of the anti-asthmatic is selected from the group consisting of β2-agonist, anticholinergic and calcium antagonist, or wherein the antiasthmatic is a corticosteroid.
18. The compound according to any of the claims 1 to 14 or a composition according to any of the claims 15 to 17 for use in therapy.
19. The compound according to any of the claims 1 to 14 or a composition according to any of the claims 15 to 17 for use in the prevention and/or treatment of a disease or condition characterized by bronchoconstriction of the respiratory apparatus.
20. The compound according to any of the claims 1 to 14 or a composition according to any of the claims 15 to 17 for use in the prevention and/or treatment of a disease or condition characterized by inflammation of the respiratory apparatus.
21. The compound or composition according to claim 19 or 20, wherein said disease or condition is selected from the group consisting of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia.
22. A method of prevention and/or treatment a disease or condition characterized by bronchoconstriction and/or inflammatory conditions of the respiratory apparatus, such as of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia, comprising administering to a mammal, including man in need of such prevention and/or treatment, a therapeutically effective amount of a compound according to any of the claims 1 to 14 or a composition according to any of claim 15 to 17 comprising a therapeutically effective amount of a compound according to any of the claims 1 to 14.
23. A method of prevention and/or treatment of a disease or condition characterized by bronchoconstriction and/or inflammatory conditions of the respiratory apparatus, such as of asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, bronchiectasis, cystic fibrosis, bronchiolitis and bronchopulmonary dysplasia, comprising the simultaneous or consecutive administration therapeutically effective amounts of a compound according to any of the claims 1 to 14 and an antiasthmatic.
24. The method according to claim 23, wherein the administered dose of the anti-asthmatic is 1 to 10 times less than the established therapeutically effective dose when administered alone for prevention or treatment of the same disease or condition.
25. The method according to claim 23 or 24, wherein the administered dose of a compound according to any one of claims 1 to 14 is 1 to 10 times less than the established therapeutically effective dose when administered alone for prevention or treatment of the same disease or condition.
26. The method according to any of the claims 23 to 25, wherein the principal mechanism of action of the anti-asthmatic is selected from the group consisting of β2- agonist, anticholinergic and calcium antagonist, or wherein the anti-asthmatic is a corticosteroid.
27. The compound according to any of the claims 1 to 14 or a composition according to claim 15 for use in the prevention and/or treatment of a disease or condition characterized by systemic or respiratory vasoconstriction.
28. A method of prevention and/or treatment of a disease or condition characterized by systemic or respiratory vasoconstriction, comprising administrering to a mammal, including man in need of such prevention and/or treatment, a therapeutically effective amount of a compound according to any of the claims 1 to 14 or a composition according to claim 15 comprising a therapeutically effective amount of a compound according to any of the claims 1 to 14.
PCT/EP2008/058993 2007-07-12 2008-07-10 Novel bronchodilating isoquinoline carbamates WO2009007419A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US95923307P 2007-07-12 2007-07-12
SE0701696 2007-07-12
US60/959,233 2007-07-12
SE0701696-7 2007-07-12

Publications (1)

Publication Number Publication Date
WO2009007419A1 true WO2009007419A1 (en) 2009-01-15

Family

ID=39748174

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/058993 WO2009007419A1 (en) 2007-07-12 2008-07-10 Novel bronchodilating isoquinoline carbamates

Country Status (1)

Country Link
WO (1) WO2009007419A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8318768B2 (en) 2007-07-12 2012-11-27 Respiratorius Ab Bronchodilating alpha, beta-unsaturated isoquinoline amides
US11680054B2 (en) 2018-07-06 2023-06-20 Arcede Pharma Ab Bronchodilating hetero-linked amides

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1288202A1 (en) * 2000-05-11 2003-03-05 Banyu Pharmaceutical Co., Ltd. N-acyltetrahydroisoquinoline derivatives
WO2005070887A1 (en) * 2004-01-22 2005-08-04 Respiratorius Ab Bronchorelaxing compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1288202A1 (en) * 2000-05-11 2003-03-05 Banyu Pharmaceutical Co., Ltd. N-acyltetrahydroisoquinoline derivatives
WO2005070887A1 (en) * 2004-01-22 2005-08-04 Respiratorius Ab Bronchorelaxing compounds

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8318768B2 (en) 2007-07-12 2012-11-27 Respiratorius Ab Bronchodilating alpha, beta-unsaturated isoquinoline amides
US11680054B2 (en) 2018-07-06 2023-06-20 Arcede Pharma Ab Bronchodilating hetero-linked amides

Similar Documents

Publication Publication Date Title
CA2901022C (en) Substituted pyridine compounds as inhibitors of histone demethylases
JP5405316B2 (en) Cycloalkylamine substituted isoquinoline derivatives
PT1606283E (en) P38 inhibitors and methods of use thereof
JP2001505585A (en) Novel amide compounds and their use as nitric oxide synthase inhibitors
AU2008345681A1 (en) Imidazo [1,2-a] pyridine compounds
WO2011023081A1 (en) 5,6-bicyclic heteroaryl-containing urea compounds as kinase inhibitors
JP2020516642A (en) Opioid Receptor (MOR) Agonist Salt, Fumarate Salt I Crystalline Form Thereof, and Process for Producing the Same
PL119501B1 (en) Process for manufacturing novel,condensed pyrimidine derivatives pirimidina
MX2013001202A (en) Condensed ring pyridine compound.
JP2006503799A (en) Guanidino compounds
WO2009007418A1 (en) Novel bronchodilating isoquinoline amides
WO2016140501A1 (en) Pyridine n-oxide for enhancer of zeste homolog 2 inhibitors
WO2008095852A1 (en) Bronchorelaxing arylamides
WO2012007008A1 (en) Inhibitors of hdme
ES2236288T3 (en) 6-HETEROARILFENANTRIDINAS.
US8318768B2 (en) Bronchodilating alpha, beta-unsaturated isoquinoline amides
WO2009007419A1 (en) Novel bronchodilating isoquinoline carbamates
JP2022528109A (en) Quinoline-containing compounds, pharmaceutical compositions and their use
EP3818055B1 (en) Novel bronchodilating hetero-linked amides
JPS61167663A (en) Morphanthridine derivative
CN115368306B (en) HDAC inhibitor containing tetrahydroisoquinoline structure, composition and application thereof
WO2023109802A1 (en) Tricyclic fused heterocyclic pde3/4 dual inhibitor and use thereof
CA2301074A1 (en) Crystalline 10,10-bis((2-fluoro-4-pyridinyl)methyl)-9(10h)-anthracenone and an improved process for preparing the same
CN117642396A (en) Alpha V beta 6 and alpha V beta 1 integrin inhibitors and uses thereof
WO2017107877A1 (en) Biphenyl derivative and preparation method and medical use thereof

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: 08774960

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08774960

Country of ref document: EP

Kind code of ref document: A1