WO2006138218A1 - Biphenyl compounds useful as muscarinic receptor antagonists - Google Patents

Biphenyl compounds useful as muscarinic receptor antagonists Download PDF

Info

Publication number
WO2006138218A1
WO2006138218A1 PCT/US2006/022829 US2006022829W WO2006138218A1 WO 2006138218 A1 WO2006138218 A1 WO 2006138218A1 US 2006022829 W US2006022829 W US 2006022829W WO 2006138218 A1 WO2006138218 A1 WO 2006138218A1
Authority
WO
WIPO (PCT)
Prior art keywords
ethyl
biphenyl
ester
piperidin
ylcarbamic acid
Prior art date
Application number
PCT/US2006/022829
Other languages
French (fr)
Inventor
Yu-Hua Ji
Mathai Mammen
Craig Husfeld
Li Li
Yongqi Mu
Aaron Kushner
Eric L. Stangeland
Trevor Mischki
Adam Hughes
Sarah Dunham
Original Assignee
Theravance, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Theravance, Inc. filed Critical Theravance, Inc.
Priority to EP06772935A priority Critical patent/EP1893577A1/en
Priority to JP2008516980A priority patent/JP2008546695A/en
Publication of WO2006138218A1 publication Critical patent/WO2006138218A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/46Oxygen atoms attached in position 4 having a hydrogen atom as the second substituent in position 4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/12Drugs for genital or sexual disorders; Contraceptives for climacteric disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems

Definitions

  • the present invention relates to novel biphenyl compounds having muscarinic receptor antagonist or anticholinergic activity.
  • the invention also relates to pharmaceutical compositions comprising these compounds, processes and intermediates for preparing these compounds and methods of using these compounds to treat pulmonary disorders.
  • Pulmonary or respiratory disorders such as chronic obstructive pulmonary disease
  • Muscarinic receptor antagonists are known to provide bronchoprotective effects and therefore are useful for treating respiratory disorders, such as COPD and asthma. Muscarinic receptor antagonists are typically administered by inhalation to treat these disorders. However, even when administered by inhalation, a significant amount of the antagonist is often absorbed into the systemic circulation resulting in systemic side effects, such as dry mouth, mydriasis and cardiovascular side effects. In addition, many inhaled muscarinic receptor antagonists have a relatively short duration of action requiring that they be administered several times per day. This multiple- daily dosing regime is inconvenient and creates a significant risk of inadequate treatment due to patient non-compliance with the required frequent dosing schedule.
  • muscarinic receptor antagonists in particular, those having high potency and reduced systemic side effects when administered by inhalation.
  • inhaled muscarinic receptor antagonists having a long duration of action thereby allowing for once-daily or even once-weekly dosing.
  • Such compounds are expected to be particularly effective for treating pulmonary disorders, such as COPD and asthma, while reducing or eliminating side effects, such as dry-mouth and constipation.
  • the present invention provides novel biphenyl compounds having muscarinic receptor antagonist or anticholinergic activity.
  • compounds of the invention are expected to possess high potency and reduced systemic side effects when administered by inhalation and are also expected to possess a long duration of action.
  • One aspect of the invention relates to a compound of formula I:
  • each R 1 is independently selected from -C 1-5 alkyl, -C 2-5 alkenyl, -C 2-5 alkynyl, -C 3-6 cycloalkyl, -cyano, -halo, -OR la , -C(O)OR lb , -SR lc , -S(O)R ld , -S(O) 2 R 16 , -NR lf R lg 5 -NR lh S(O) 2 R lj , and -NR lj C(O)R lk ; where each of R la'lk is independently -H, -C 1-5 alkyl or -phenyl-C 1-5 alkyl; b is 0 or an integer of from 1 to 4; each R 2 is independently selected from -C 1-5 alkyl, -C 2-5 alkenyl, -C 2-5 alkyn
  • Ar is a phenylene group or a C 3-5 heteroarylene group containing 1 or 2 heteroatoms independently selected from oxygen, nitrogen or sulfur; wherein the phenylene or C 3-5 heteroarylene group is substituted with (R 5 ) q where q is 0 or an integer from 1 to 4 and each R 5 is independently selected from -halo, -OH, -Q.salkyl or -C ⁇ salkoxy; t is 0, 1 or 2;
  • R 6 is selected from -H, -C 1-5 alkyl, and -X 6 R 6a ; where X 6 is -C 1-5 alkylene and R 6a is selected from -OH, -C 1-5 alkoxy and -heteroaryl; and R 7 is selected from -H, -C 1-5 alkyl, -C 3-6 cycloalkyl, and -X 7 R 7a ; where X 7 is selected from -C 1-5 alkylene, -C(O)-, -C 1-5 alkylene-C(O)-, -S(O 2 )-, -C 1-5 alkylene-S(O 2 )-, and -S(O 2 )-C 1-5 alkylene; and R 7a is selected from -OH, -Q ⁇ alkyl, -Cs- ⁇ cycloalkyl, -C ⁇ alkoxy, heteroaryl, -NR 7b R 7c , where R 7b and R 7c
  • R 9 is selected from -H, -C 1-5 alkyl, hydroxyphenyl, heteroaryl, and -X 9 R 9a ; where X 9 is selected from -C 1-5 alkylene, -C(O)-, -C 1-5 alkylene-C(O)-, -C(O)-C 1-5 alkylene, -S(O 2 )-, -C 1-5 alkylene-S(O 2 )-, and -S(O 2 )-C 1-5 alkylene; and R 9a is selected from -H, -OH, -C 1-5 alkyl, -C 1-5 alkoxy, aryl, heteroaryl, heterocyclyl, and -NR 9b R 9 °, where R 9b and R 9c are independently -H or -C 1-5 alkylene
  • compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof.
  • compositions comprising a compound of formula I in combination with one or more other therapeutic agents.
  • the invention is directed to compositions comprising (a) a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof; and (b) a therapeutically effective amount of an agent selected from a steroidal anti-inflammatory agent such as a corticosteroid; a ⁇ 2 adrenergic receptor agonist; a phos ⁇ hodiesterase-4 inhibitor; or a combination thereof; wherein the compound of formula I and the agent are formulated together or separately.
  • a pharmaceutically acceptable carrier may be included.
  • Compounds of the invention possess muscarinic receptor antagonist activity. Accordingly, compounds of formula I are expected to be useful for treating pulmonary disorders, such as chronic obstructive pulmonary disease and asthma.
  • Yet another aspect of the invention is directed to methods for treating a pulmonary disorders comprising administering to a patient a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Still another aspect of the invention pertains to methods of producing bronchodilation in a patient comprising administering to a patient a bronchodilation- producing amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the invention is also directed to methods of treating chronic obstructive pulmonary disease or asthma comprising administering to a patient a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the invention is directed to methods for antagonizing a muscarinic receptor in a mammal comprising administering to the mammal a therapeutically effective amount of the compound of formula I. Since compounds of the invention possess muscarinic receptor antagonist activity, such compounds are also useful as research tools. Accordingly, in yet another embodiment, the invention is directed to methods for using a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof as a research tool for studying a biological system or sample, or for discovering new chemical compounds having muscarinic receptor antagonist activity.
  • the invention is also directed to processes and intermediates useful for preparing compounds of formula I and pharmaceutically acceptable salts or solvates or stereoisomers thereof. Accordingly, in another embodiment, the invention is directed to a process of preparing a compound of formula I, the process comprising: (a) reacting a compound of formula II with a compound of formula III; or (b) coupling a compound of formula IVa with a compound of formula Va or coupling a compound of formula IVb with a compound of formula Vb; or (c) reacting a compound of formula VI with a compound of formula VII; or (d) reacting a compound of formula II with a compound of formula VIII in the presence of a reducing agent; or (e) reacting a compound of formula IX with a compound of formula VII in the presence of a reducing agent; and then removing any protecting groups that may be present to provide a compound of formula I, and optionally, forming a pharmaceutically acceptable salt thereof, wherein compounds of formula I-IX, are as defined herein.
  • the invention is also directed to a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof, for use in therapy or as a medicament. Additionally, the invention is directed to the use of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof, for the manufacture of a medicament; especially for the manufacture of a medicament for the treatment of a pulmonary disorder or for antagonizing a muscarinic receptor in a mammal.
  • the invention is directed to novel biphenyl compounds of formula I, and pharmaceutically acceptable salts or solvates or stereoisomers thereof.
  • the compounds may contain one or more chiral centers and therefore, the invention is also directed to racemic mixtures, pure stereoisomers (i.e., enantiomers or diastereomers), stereoisomer-enriched mixtures, and the like unless otherwise indicated.
  • stereoisomer is shown or named herein, it will be understood by those skilled in the art that minor amounts of other stereoisomers may be present in the compositions of the invention unless otherwise indicated, provided that the desired utility of the composition as a whole is not eliminated by the presence of such other isomers.
  • the compounds of formula I also contain several basic groups (e.g., amino groups) and therefore, the compounds of formula I can exist as the free base or in various salt forms. AU such salt forms are included within the scope of the invention. Furthermore, solvates of compounds of formula I or salts thereof are included within the scope of the invention. Additionally, where applicable, all cis-trans or E/Z isomers (geometric isomers), tautomeric forms and topoisomeric forms of the compounds of formula I are included within the scope of the invention unless otherwise specified.
  • the compounds of formula I may also include isotopically-labeled compounds, i.e., where one or more atoms have been enriched with atoms having an atomic mass different from the atomic mass predominately found in nature.
  • isotopes that may be incorporated into the compounds of formula I include, but are not limited to, 2 H, 3 H 5 13 C, 14 C, 15 N, 18 O and 17 O.
  • each R 1 may be at the 2, 3, 4, 5 or 6-position of the phenyl ring to which it is attached.
  • Each R 1 is independently selected from -Ci-salkyl, -C 2-5 alkenyl,
  • each R 2 may be at the 3, 4, 5 or 6-position on the phenylene ring to which it is attached (where the carbon atom on the phenylene ring attached to the nitrogen atom is position 1).
  • Each R 2 is independently selected from -C 1-5 alkyl, -C 2- salkenyl, -C 2-5 alkynyl, -C 3-6 CyClOaIkVl, -cyano, -halo, -OR 2a , -C(O)OR 2b , -SR 2c , -S(O)R 2d , -S(O) 2 R 26 , -NR 2f R 2g , -NR 2h S(O) 2 R 2i , and -NR 2j C(O)R 2k , examples of which include methyl, fluoro, chloro, bromo, hydroxy, methoxy, amino, methylamino, dimethylamino and the like.
  • R 2 are fluoro or chloro.
  • each alkyl and alkoxy group in R 1 , R la'lk , R 2 , and R 2a"2k is optionally substituted with 1 to 5 fluoro substituents.
  • W is -O-.
  • W is -NW a -
  • W a is -H or -C 1-4 alkyl, examples of which include hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl.
  • W a is hydrogen or -C 1-3 alkyl.
  • W a is hydrogen, methyl or ethyl, particularly hydrogen or methyl
  • W a is hydrogen and -NW a - is -NH-.
  • each R 3 independently represents -d ⁇ alkyl or two R 3 groups that are joined to form C 1-3 alkylene, C 2 . 3 alkenylene or oxiran-2,3-diyl.
  • each R 3 is independently -C h alky!, such as methyl, ethyl, «-propyl, isopropyl, o-butyl, sec-butyl, isobutyl and tert-butyl.
  • each alkyl group in R 3 is optionally substituted with 1 to 5 fluoro substituents.
  • each R 3 is independently -C 1-3 alkyl, and in another embodiment, each R 3 is independently methyl or ethyl.
  • each R 3 is at the 3, 4 or 5-position on the piperidine ring
  • R 3 is at the 4-position on the piperidine ring
  • R 3 is at the 1-position of the piperidine ring, i.e., on the nitrogen atom of the piperidine ring thus forming a quaternary amine salt.
  • two R 3 groups are joined to form a C 1-3 alkylene or
  • R 3 groups at the 2 and 6-positions on the piperidine ring can be joined to form an ethylene bridge (i.e., the piperidine ring and the R 3 groups form an 8-azabicyclo[3.2.1]octane ring); or two R 3 groups at the 1 and 4-positions on the piperidine ring can be joined to form an ethylene bridge (i.e., the piperidine ring and the R 3 groups form an l-azabicyclo[2.2.2]octane ring), hi this embodiment, other R 3 groups as defined herein may also be present.
  • two R 3 groups are joined to form a oxiran-2,3-diyl group.
  • two R 3 groups at the 2 and 6-positions on the piperidine ring can be joined to form a 3-oxatricyclo[3.3.1.0 2>4 ]nonane ring).
  • other R 3 groups as defined herein may also be present.
  • Z is selected from -C(O)N(R 4 )- and -N(R 4 )C(O)-.
  • R 4 is -H, -C 1-5 alkyl, or -C 3-5 CyClOaIkVl.
  • -C 1-5 alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, -fee-butyl, isobutyl, tert-butyl, and pentyl.
  • Examples of -C 3-5 cycloalkyl groups include cyclopropyl, cyclobutyl, and cyclopentyl.
  • R 4 is hydrogen or -C 1-3 alkyl, in particular hydrogen or methyl.
  • exemplary embodiments of Z include -NHC(O)-, -N(CH 3 )C(O)-, and -C(O)NH-.
  • the value for s is 0, 1 or 2.
  • One particular value for s is 0; in another embodiment, s is 1; and in yet another embodiment, the value for s is 2.
  • Ar is a phenylene group or a C 3-5 heteroarylene group containing 1 or 2 heteroatoms independently selected from oxygen, nitrogen or sulfur.
  • the phenylene or heteroarylene group may be unsubstituted (q is 0) or substituted with 1, 2, 3, or 4 (q is 1, 2, 3, or 4) R 5 substituents, which are independently selected from -halo, -OH, -C 1-5 alkyl or -C 1-5 alkoxy.
  • each alkyl and alkoxy group in R 5 is optionally substituted with 1 to 5 fluoro substituents.
  • q is 0, 1, 2 or 3; in another embodiment, q is 0, 1 or 2.
  • q is 1 and R 5 is methoxy.
  • the point of attachment for Ar is at any available carbon or heteroatom ring atom.
  • Ar is a phenylene group attached at the meta or para position.
  • Ar is phen-l,3-ylene or phen-l,4-ylene wherein the phenylene group is unsubstituted or substituted with 1, 2 or 3 R 5 substituents.
  • R 5 substituents include fluoro, chloro, bromo, methyl, ethyl, «-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, isopropoxy, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl and trifluoromethoxy.
  • Ar 1 groups in this embodiment include 2-fluorophen-l,4-ylene, 3-fluorophen-l,4-ylene, 2- chlorophen-l,4-ylene, 3-chlorophen-l,4-ylene, 2-methylphen-l,4-ylene, 3-methylphen-l,4- ylene, 2-methoxyphen-l,4-ylene, 3-methoxyphen-l,4-ylene, 2-trifluoromethoxyphen-l,4- ylene, 3-trifluoromethoxyphen-l,4-ylene, 2,3-difluorophen-l,4-ylene, 2,5-difluorophen- 1,4-ylene, 2,6-difluorophen-l,4-ylene, 2,3-dichlorophen-l,4-ylene, 2,5-dichlorophen-l,4- ylene, 2,6-dichlorophen- 1,4-ylene, 2-chloro-5-methoxyphen- 1,4-
  • Ar is a C 3-5 heteroarylene group containing 1 or 2 heteroatoms, and is unsubstituted or substituted with 1 or 2 R 5 substituents.
  • Representative C 3-5 heteroarylene groups include divalent species of pyrrole, imidazole, thiazole, oxazole, furan, thiophene, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine and pyrimidine, where the point of attachment is at any available carbon or nitrogen ring atom.
  • Ar groups include: pyridylene such as 2,5-pyridylene and 2,6-pyridylene; thienylene such as 2,4-thienylene and 2,5-thienylene; pyrrolylene such as 2,4-pyrrolylene and 2,5-pyrrolylene; and furylene such as 2,5-furylene.
  • R 5 substituents include fluoro, chloro, methyl, ethyl, w-propyl, isopropyl, ⁇ -butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, isopropoxy, difluoromethyl, trifluoromethyl, 2,2,2- trifluoroethyl and trifluoromethoxy.
  • Particular examples of substituted Ar groups include 3-fluoro-2,5-thienylene, 3-chloro-2,5-thienylene, 3-methyl-2,5-thienylene, 3-methoxy-2,5- thienylene, and 3-methoxy-6-chloro-2,5-pyridylene.
  • Ar represents phen-l,3-ylene, phen-l,4-ylene, 2-methoxyphen-l,4-ylene, 2,5-pyridylene, 2,5-thienylene, 2,4-pyrrolylene, 2,5-pyrrolylene, or 2,5-furylene.
  • the value for t is 0, 1 or 2.
  • a particular value for t is 1.
  • R 6 is selected from -H, -C 1-5 alkyl, and -X 6 R 6a , where X 6 is -C 1-5 alkylene and R 6a is selected from -OH, -C 1-5 alkoxy and -heteroaryl.
  • R 6 is -H.
  • R 6 is -C 1-5 alkyl such as methyl or ethyl.
  • R 6 is -X 6 R 6a , where X 6 is -C 1-5 alkylene such as -(CBb) 2 -, and R 6a is -OH.
  • R 6 is -X 6 R 6a , where X 6 is -C 1-5 alkylene such as -(CH 2 ) 2 -, and R 6a is -Q-salkoxy such as -OCH 3 or -OCH 2 CH 3 .
  • each alkyl and alkoxy group in R 6 and R a is optionally substituted with 1 to 5 fluoro substituents.
  • R 7 is selected from -H, -C 1-5 alkyl, -C 3-6 cycloalkyl, and -X 7 R 7a .
  • X 7 is selected from
  • R 7a is selected from -OH, -C 1-5 alkyl, -C 3-6 CyClOaIkVl, -C 1-5 alkoxy, heteroaryl, -NR 7b R 7c (R 7b and R 7c are independently -H or -C 1-5 alkyl), and aryl (which may be substituted with 1 or 2 -C 1-5 alkyl or halo groups).
  • each alkyl and alkoxy group in R 7 and R 7a'c is optionally substituted with 1 to 5 fluoro substituents.
  • R can be -CH 2 CH 2 CF 3 or -S(O 2 )CF 3 .
  • R is -H.
  • R 7 is -C 1-5 alkyl such as methyl, ethyl, isopropyl, isobutyl, and tert-butyl.
  • R 7 is -C 3-6 cycloalkyl such as cyclopropyl, cyclobutyl, or cyclopentyl.
  • R 7 is -X 7 R 7a , where X 7 is -C 1-5 alkylene such as -CH 2 -, and R 7a is: -C 3-6 cycloalkyl such as cyclopropyl; heteroaryl such as 2-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 5-benzo[l,3]dioxole, or 7-lH-indole.
  • X 7 can also be -C 1-5 alkylene such as -(CH 2 ) 2 -, and R 7a can be -OH, or -Ci ⁇ alkoxy such as -OCH 3 and -OCH 2 CH 3 .
  • R 7 can also be -X 7 R 7a , where X 7 is -C(O)-, and R 7a is -C 1-5 alkyl such as methyl, -C 3-6 cycloalkyl such as cyclopropyl or cyclobutyl, heteroaryl such as 2-furyl, 2-thienyl, or 4-pyridyl.
  • R 7 is -X 7 R 7a , where X 7 is -C 1-5 alkylene-C(O)- such as -CH 2 C(O)-, and R 7a is: -OH; or -NR 7b R 7c such as -NH 2 .
  • R 7 can also be -X 7 R 7a , where X 7 is -S(O 2 )-, and R 7a is: -Ci-salkyl such as methyl, ethyl, w-propyl, isopropyl, and 7j-butyl; heteroaryl such as 2-thienyl and 8-quinolinyl; or aryl such as 4-trifluoromethylphenyl,
  • R 7 is -X 7 R 7a , where X 7 is -C 1-5 alkylene-S(O 2 )- such as -(CH 2 ) 2 S(O 2 )-, and R 7a is -C 1-5 alkyl such as methyl.
  • R 7 is -X 7 R 7a , where X 7 is -S(O 2 )-C 1-5 alkylene such as -S(O 2 )(CH 2 ) 2 -, and R 7a is -NR 7b R 7c such as -NHCH 3 .
  • R 6 /R 7 combination is where R 6 is -H and R 7 is -H, -C 1-5 alkyl (e.g., methyl, ethyl, isopropyl, isobutyl, or tert-buty ⁇ ), or -C 3-6 CyClOaUCyI (e.g., cyclopropyl, cyclobutyl, or cyclopentyl).
  • R 6 is -H and R 7 is -H, -C 1-5 alkyl (e.g., methyl, ethyl, isopropyl, isobutyl, or tert-buty ⁇ ), or -C 3-6 CyClOaUCyI (e.g., cyclopropyl, cyclobutyl, or cyclopentyl).
  • R 6 is -H and R 7 is -X 7 R 7a , where X 7 is -C 1-5 alkylene (e.g., -CH 2 -), and R 7a is -C 3-6 cycloalkyl (e.g., cyclopropyl) or heteroaryl (e.g., 2-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 5-benzo[l,3]dioxole, or 7-lH-indole).
  • X 7 may also be -Q.salkylene such as -(CH 2 ) 2 -, and R 7a is -OH or -C 1-5 alkoxy such as
  • R 6 /R 7 combination is where R 6 is -H, and R 7 is -X 7 R 7a , where X 7 is -C(O)-, and R 7a is: -Q-salkyl such as methyl; -Cs- ⁇ Cycloalkyl such as cyclopropyl or cyclobutyl; or heteroaryl such as 2-furyl, 2-thienyl, or 4-pyridyl.
  • R /R combination is where R is -H, and R is -X R , where X is -C 1-5 alkylene-C(O)- such as -CH 2 C(O)-, and R 7a is -NR 7b R 7c such as -NH 2 .
  • R 6 /R 7 combination is where R 6 is -H, and R 7 is -X 7 R 7a , where X 7 is -S(O 2 )-, and R 7a is -Q.salkyl (e.g., methyl, ethyl, ra-propyl, isopropyl, or n-butyl), heteroaryl (e.g., thiophen-2-yl or 8-quinolinyl), or aryl (e.g., 4-trifluoromethylphenyl, 2-fluorophenyl, 2,6- dichlorophenyl, 4-methylphenyl, or phenyl).
  • R 6 is -H
  • R 7 is -X 7 R 7a , where X 7 is -S(O 2 )-, and R 7a is -Q.salkyl (e.g., methyl, ethyl, ra-propyl, isopropyl, or n-but
  • R 6 /R 7 combination is where R 6 is -H, and R 7 is -X 7 R 7a , where X 7 is -S(O 2 )-C 1-5 alkylene such as -S(O 2 )(CH 2 ) 2 -, and R 7a is -NR 7b R 7c such as -NHCH 3 .
  • R 6 ZR 7 combination is where R 6 is -C 1-5 alkyl such as methyl, and R 7 is: -X 7 R 7a , where X 7 is -C 1-5 alkylene such as -(CH 2 ) 2 -, and R 7a is -OH; -X 7 R 7a , where X 7 is -C ⁇ alkylene such as -CH 2 -, and R 7a is heteroaryl such as 3-pyridyl, 4-pyridyl, or 2-furyl; X 7 R 7a , where X 7 is -C 1-5 alkylene-C(O)- such as -CH 2 C(O)-, and R 7a is -OH; or X 7 R 7a , where X 7 is -C 1-5 alkylene-S(O 2 )- such as -(CH 2 ) 2 S(O 2 )-, and R 7a is -C 1-5 alkyl such as methyl.
  • R 6 ZR 7 combination is where R 6 and R 7 are the same.
  • R 6 and R 7 can both be a -C 1-5 alkyl group such as methyl and ethyl.
  • R 6 and R 7 can also both be a -C ⁇ salkylene-d-salkoxy group (X 6 and X 7 are -Q.salkylene, R 6a and R 7a are -C 1-5 alkoxy), examples of which include -(CH 2 ) 2 OCH 3 and -(CH 2 ) 2 OCH 2 CH 3 .
  • R 6 and R 7 can also both be a -C 1-5 alkylene-OH group (X 6 and X 7 are -C 1-5 alkylene, R 6a and R 7a are -OH), an example of which includes -(CH 2 ) 2 OH.
  • R 6 and R 7 may be taken together to form a thiomorpholine 1,1 -dioxide ring:
  • R 6 and R 7 may also be taken together to form morpholin-4-yl:
  • R 6 and R 7 may also be taken together to form:
  • d is 1 or 2. In one embodiment, d is 1. The value for e is 0, 1 or 2. In one embodiment, e is 0 and in another embodiment e is 1. In one embodiment, d is 1 and e is 0 or 1. The value for f is 0, 1, 2 or 3. In one embodiment, f is 0.
  • R 9 is selected from -H, -C 1-5 alkyl, hydroxyphenyl, heteroaryl, and -X 9 R 9a .
  • X 9 is selected from -C 1-5 alkylene, -C(O)-, -C 1-5 alkylene-C(O)-, -C(O)-C 1-5 alkylene, -S(O 2 )-, -C 1-5 alkylene-S(O 2 )-, and -S(O 2 )-C 1-5 alkylene.
  • R 9a is selected from -H, -OH, -C 1-5 alkyl, -C 1-5 alkoxy, aryl, heteroaryl, heterocyclyl, and -NR 9b R 9c (R 9b and R 9c are independently -H or -C ⁇ aUcyl).
  • each alkyl and alkoxy group in R 9 and R 9a"c is optionally substituted with 1 to 5 fluoro substituents.
  • R 9 is -H or hydroxyphenyl.
  • R 9 can also be -C 1-5 alkyl such as ethyl and isopropyl.
  • R 9 is heteroaryl such as 4-pyridyl.
  • R 9 is -X 9 R 9a , where X 9 is -C 1-5 alkylene such as -CH 2 -, and R 9a is heteroaryl such as 4-pyridyl, or where X 9 is -Q-salkylene such as -(CH 2 ) 2 -, and R 9a is -C 1-S aIkOXy such as -OCH 3 .
  • R 9 can also be -X 9 R 9a , where X 9 is -C(O)-, and R 9a is: -H; -C 1-5 alkyl such as methyl; -C 1-5 alkoxy such as -OCH 3 ; heterocyclyl such as 1-pyrrolidinyl, 1-piperidyl, and 4-morpholinyl; or -NR 9b R 9c such as -N(CH 3 ) 2 .
  • R 9 is -X 9 R 9a , where X 9 is -C 1-5 alkylene-C(O)- such as -CH 2 C(O)-, and R 9a is heterocyclyl such as 1- pyrrolidinyl, or -NR 9b R 9 ° such as -N(CH 3 ) 2 .
  • R 9 can also be -X 9 R 9a , where X 9 is -S(O 2 )-, and R 9a is -C 1-5 alkyl such as methyl and ethyl.
  • d is 1, e is O, and f is 1, i.e., R 6 and R 7 are taken together to form a piperidine ring.
  • d is 1, and e and f are O, i.e., R 6 and R 7 are taken together to form a piperidine ring.
  • Particular embodiments of the piperidine ring include those where R 9 is: -Q-salkyl such as ethyl and isopropyl; hydroxyphenyl; heteroaryl such as 4-pyridyl; -X 9 R 9a , where X 9 is -C 1-5 alkylene such as -CH 2 -, and R 9a is heteroaryl such as 4-pyridyl; -X 9 R 9a , where X 9 is -C 1-5 alkylene such as -(CH 2 ) 2 -, and R 9a is -C 1-5 alkoxy such as -OCH 3 ; -X 9 R 9a , where X 9 is -C(O)-, and R 9a is -H, -C 1-5 alkyl such as methyl, -C 1-5 alkoxy
  • d is 2, and e and f are O, i.e., R 6 and R 7 are taken together to form a [l,4]diazepane ring.
  • Particular embodiments of the [l,4]diazepane ring include those where R 9 is -X 9 R 9a , where X 9 is -C(O)-, and R 9a is -C 1-5 alkyl such as methyl, hi another embodiment, d and e are 1, and f is O, i.e., R 6 and R 7 are taken together to form a 2,5-diaza-bicyclo[2.2.1]heptane ring.
  • Particular embodiments ofthe 2,5-diaza- bicyclo[2.2.1]heptane ring include those where R 9 is: -H; or -X 9 R 9a , where X 9 is -C 1-5 alkylene such as -CH 2 -, and R 9a is aryl such as phenyl.
  • a particular group of compounds of interest are compounds of formula I wherein a, b, and c are 0. Another group of compounds of interest are compounds of formula I wherein W is -O-. A particular group of compounds of interest are compounds of formula I where m is 0 and t is 1. A particular group of compounds of interest are compounds of formula I where Z is -NHC(O)-, -N(CH 3 )C(O)-, or -C(O)NH-.
  • Ar is phenylene, phenylene substituted with methoxy, pyridylene, thienylene, pyrrolylene, or furylene, and in particular where Ar is phen-1,3- ylene, phen-l,4-ylene, 2-methoxyphen-l,4-ylene, 2,5-pyridylene, 2,5-thienylene, 2,4-pyrrolylene, 2,5-pyrrolylene, or 2,5-furylene.
  • a, b, c, s, R 1"3 , R 6"7 , Z, and Ar 3 are as defined for formula I.
  • Another example of a group of compounds of interest are compounds of formula Ia wherein a, b, and c are 0; Z is selected from -NHC(O)-, -N(CH 3 )C(O)-, and -C(O)NH-; and Ar is selected from phenylene, phenylene substituted with methoxy, pyridylene, thienylene, pyrrolylene, and furylene.
  • particular compounds of formula I that are of interest include: biphenyl-2-yl-carbamic acid l-(2- ⁇ [4-(4-acetylpiperazin-l-ylmethyl)benzoyl] methylamino ⁇ ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l- ⁇ 2-[(4-aminomethylbenzoyl)methylamino]ethyl ⁇ piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- ⁇ 2-[(4- ⁇ [(furan-2-carbonyl)amino]methyl ⁇ benzoyl) methylamino] ethyl ⁇ piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- ⁇ [4-(acetylaminomethyl)benzoyl]methylamino ⁇ ethyl)piperidin-4-yl ester; biphenyl-2
  • alkyl means a monovalent saturated hydrocarbon group which may be linear or branched. Unless otherwise defined, such alkyl groups typically contain from 1 to 10 carbon atoms. Representative alkyl groups include, by way of example, methyl, ethyl, r ⁇ -propyl, isopropyl, ⁇ -butyl, sec-butyl, isobutyl, tert-bntyl, «-pentyl, r ⁇ -hexyl, n-heptyl, Tz-octyl, n-nonyl, n-decyl and the like.
  • alkylene means a divalent saturated hydrocarbon group which may be linear or branched. Unless otherwise defined, such alkylene groups typically contain from 1 to 10 carbon atoms. Representative alkylene groups include, by way of example, methylene, ethane- 1,2-diyl ("ethylene”), propane- 1,2-diyl, propane- 1, 3 -diyl, butane- 1,4- diyl, pentane-l,5-diyl and the like.
  • ethylene ethane- 1,2-diyl
  • propane- 1,3 -diyl propane- 1, 3 -diyl
  • butane- 1,4- diyl pentane-l,5-diyl and the like.
  • alkoxy means a monovalent group of the formula (alkyl)-O-, where alkyl is as defined herein.
  • Representative alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, isobutoxy, tert-butoxy and the like.
  • alkenyl means a monovalent unsaturated hydrocarbon group which may be linear or branched and which has at least one, and typically 1, 2 or 3, carbon-carbon double bonds. Unless otherwise defined, such alkenyl groups typically contain from 2 to 10 carbon atoms. Representative alkenyl groups include, by way of example, ethenyl, n-propenyl, isopropenyl, «-but-2-enyl, «-hex-3-enyl and the like.
  • alkenylene means a divalent alkenyl group.
  • alkynyl means a monovalent unsaturated hydrocarbon group which may be linear or branched and which has at least one, and typically 1, 2 or 3, carbon- carbon triple bonds. Unless otherwise defined, such alkynyl groups typically contain from 2 to 10 carbon atoms. Representative alkynyl groups include, by way of example, ethynyl, 7?-propynyl, w-but-2-ynyl, n-hex-3-ynyl and the like.
  • alkynylene means a divalent alkynyl group.
  • aryl means a monovalent aromatic hydrocarbon having a single ring (i.e., phenyl) or fused rings (i.e., naphthalene). Unless otherwise defined, such aryl groups typically contain from 6 to 10 carbon ring atoms. Representative aryl groups include, by way of example, phenyl and naphthalene- 1-yl, naphthalene-2-yl, and the like.
  • arylene means a divalent aryl group.
  • cycloalkyl means a monovalent saturated carbocyclic hydrocarbon group. Unless otherwise defined, such cycloalkyl groups typically contain from 3 to 10 carbon atoms. Representative cycloalkyl groups include, by way of example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • cycloalkylene means a divalent cycloalkyl group.
  • halo means fluoro, chloro, bromo and iodo.
  • heteroaryl means a monovalent aromatic group having a single ring or two fused rings and containing in the ring at least one heteroatom (typically 1 to 3 heteroatoms) selected from nitrogen, oxygen or sulfur. Unless otherwise defined, such heteroaryl groups typically contain from 5 to 10 total ring atoms.
  • heteroaryl groups include, byway of example, monovalent species of pyrrole, imidazole, thiazole, oxazole, furan, thiophene, triazole, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine, pyrimidine, triazine, indole, benzofuran, benzothiophene, benzimidazole, benzthiazole, quinoline, isoquinoline, quinazoline, quinoxaline and the like, where the point of attachment is at any available carbon or nitrogen ring atom.
  • heteroarylene means a divalent heteroaryl group.
  • heterocyclyl or “heterocyclic” means a monovalent saturated or unsaturated (non-aromatic) group having a single ring or multiple condensed rings and containing in the ring at least one heteroatom (typically 1 to 3 heteroatoms) selected from nitrogen, oxygen or sulfur. Unless otherwise defined, such heterocyclic groups typically contain from 2 to 9 total ring carbon atoms.
  • Representative heterocyclic groups include, byway of example, monovalent species of pyrrolidine, imidazolidine, pyrazolidine, piperidine, 1,4-dioxane, morpholine, thiomorpholine, piperazine, 3-pyrroline and the like, where the point of attachment is at any available carbon or nitrogen ring atom.
  • heterocyclene means a divalent heterocyclyl or heterocyclic group.
  • (l-5C)alkyl or "C ⁇ alkyl” means an alkyl group having from 1 to 5 carbon atoms.
  • salts means a salt which is acceptable for administration to a patient, such as a mammal (e.g., salts having acceptable mammalian safety for a given dosage regime).
  • Such salts can be derived from pharmaceutically acceptable inorganic or organic bases and from pharmaceutically acceptable inorganic or organic acids.
  • Salts derived from pharmaceutically acceptable inorganic bases include ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like. Particularly preferred are ammonium, calcium, magnesium, potassium and sodium salts.
  • Salts derived from pharmaceutically acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, iV.N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, 7V-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperadine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
  • arginine betaine
  • caffeine choline
  • Salts derived from pharmaceutically acceptable acids include acetic, ascorbic, benzenesulfonic, benzoic, camphosulfonic, citric, ethanesulfonic, edisylic, fumaric, gentisic, gluconic, glucuronic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, lactobionic, maleic, malic, mandelic, methanesulfonic, mucic, naphthalenesulfonic, naphthalene- 1, 5 -disulfonic, naphthalene-2,6-disulfonic, nicotinic, nitric, orotic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, xinafoic and the like. Particularly preferred are citric, hydrobromic, hydrochloric, isethionic,
  • salt thereof means a compound formed when the hydrogen of an acid is replaced by a cation, such as a metal cation or an organic cation and the like.
  • the salt is a pharmaceutically acceptable salt, although this is not required for salts of intermediate compounds that are not intended for administration to a patient.
  • solvate means a complex or aggregate formed by one or more molecules of a solute, i.e. a compound of formula I or a pharmaceutically acceptable salt thereof, and one or more molecules of a solvent.
  • solvates are typically crystalline solids having a substantially fixed molar ratio of solute and solvent.
  • Representative solvents include, by way of example, water, methanol, ethanol, isopropanol, acetic acid and the like. When the solvent is water, the solvate formed is a hydrate.
  • a pharmaceutically acceptable salt or solvate or stereoisomer thereof is intended to include all permutations of salts, solvates and stereoisomers, such as a solvate of a pharmaceutically acceptable salt of a stereoisomer of a compound of formula I.
  • therapeutically effective amount means an amount sufficient to effect treatment when administered to a patient in need of treatment.
  • a therapeutically effective amount for antagonizing a muscarinic receptor is that amount which will achieve the desired antagonizing effect.
  • a therapeutically effective amount for treating a pulmonary disorder is the amount that will achieve the desired therapeutic result, which may be disease prevention, amelioration, suppression or alleviation, as described below.
  • treating means the treating or treatment of a disease or medical condition (such as COPD) in a patient, such as a mammal (particularly a human) that includes: (a) preventing the disease or medical condition from occurring, i.e., prophylactic treatment of a patient; (b) ameliorating the disease or medical condition, i.e., eliminating or causing regression of the disease or medical condition in a patient; (c) suppressing the disease or medical condition, i.e., slowing or arresting the development of the disease or medical condition in a patient; or (d) alleviating the symptoms of the disease or medical condition in a patient.
  • a disease or medical condition such as COPD
  • a mammal particularly a human
  • leaving group means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction.
  • representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; and acyloxy groups, such as acetoxy, trifluoroacetoxy and the like.
  • the biphenyl compounds of the invention can be prepared from readily available starting materials using the following general methods, the procedures set forth in the Examples, or by using other methods, reagents, and starting materials that are readily available to those of ordinary skill in the art. Although a particular embodiment of the present invention may be shown or described herein, those skilled in the art will recognize that all embodiments or aspects of the present invention can be prepared using the methods described herein or by using other methods, reagents and starting materials known to those skilled in the art. It will also be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. While the optimum reaction conditions may vary depending on the particular reactants or solvent used, such conditions can be readily determined by one skilled in the art by routine optimization procedures.
  • protecting groups may be necessary or desired to prevent certain functional groups from undergoing undesired reactions.
  • the choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection of such functional groups are well-known in the art.
  • Functional groups which may be protected include, by way of example, carboxylic acid groups, amino groups, hydroxyl groups, thiol groups, carbonyl groups and the like. Protecting groups other than those illustrated in the procedures described herein may be used, if desired.
  • protecting groups for carboxylic acids include esters, amides and hydrazides; for amino groups, carbamates and amides; for hydroxyl groups, ethers and esters; for thiol groups, thioethers and thioesters; for carbonyl groups, acetals and ketals; and the like.
  • Exemplary protecting groups are listed below, and other groups as well as details of their introduction and removal, are described in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein.
  • Amino-protecting groups are suitable for preventing undesired reactions at an amino group, and include, but are not limited to, tert-butoxycarbonyl (BOC), trityl (Tr), benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc), formyl, trimethylsilyl (TMS), fert-butyldimethylsilyl (TBS), and the like.
  • Carboxy-protecting groups are suitable for preventing undesired reactions at a carboxy group, and include, but are not limited to, esters, such as methyl, ethyl, tert-butyl, benzyl (Bn),p-methoxybenzyl (PMB), 9- fluroenylmethyl (Fm), trimethylsilyl (TMS), tert-butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like.
  • esters such as methyl, ethyl, tert-butyl, benzyl (Bn),p-methoxybenzyl (PMB), 9- fluroenylmethyl (Fm), trimethylsilyl (TMS), tert-butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like.
  • Hydroxyl-protecting groups are suitable for preventing undesirable reactions at a hydroxyl group, and include, but are not limited to, silyl groups including tri(l-6C)alkylsilyl groups, such as trimethylsilyl (TMS), triethylsilyl (TES), tert- butyldimethylsilyl (TBS) and the like; esters (acyl groups) including (l-6C)alkanoyl groups, such as formyl, acetyl and the like; arylmethyl groups, such as benzyl (Bn), p- methoxybenzyl (PMB), 9-fluorenylmethyl (Fm), diphenylmethyl (benzhydryl, DPM) and the like. Additionally, two hydroxyl groups can also be protected as an alkylidene group, such as prop-2-ylidine, formed, for example, by reaction with a ketone, such as acetone.
  • the compounds of formula I can be prepared by a process comprising:
  • Vb or a reactive derivative thereof
  • a salt of one of the starting materials is used in the processes described above, such as an acid addition salt
  • the salt is typically neutralized before or during the reaction process. This neutralization reaction is typically accomplished by contacting the salt with one molar equivalent of a base for each molar equivalent of acid addition salt.
  • the reaction between the compounds of formula II and III, the leaving represented by L 1 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
  • the reaction is conveniently performed in the presence of a base, for example, a tertiary amine such as diisopropylethylamine.
  • Convenient solvents include nitriles, such as acetonitrile.
  • the reaction is conveniently conducted at a temperature in the range of from 0 0 C to 100 °C.
  • P 1 represents an amino-protecting group, such as a benzyl group.
  • Benzyl groups are conveniently removed by reduction, using a hydrogen or ammonium formate and a Group VIII metal catalyst, such as palladium.
  • W represents NW a
  • the hydrogenation is conveniently performed using Pearlman's catalyst (Pd(OH) 2 ).
  • Compounds of formula III can be prepared starting from a corresponding compound in which L 1 represents a hydroxyl group, for example, by reaction of a halogenating agent, such as thionyl chloride, to afford a compound of formula III in which L 1 represents halo, such as chloro.
  • a halogenating agent such as thionyl chloride
  • L 1 represents halo
  • Compounds in which L 1 represents a hydroxyl group may be prepared, for example, by reacting a compound of formula Vb with an appropriate amino-substituted alcohol, such as 2-aminoethanol or 3-aminopropan-l-ol.
  • the term "reactive derivative" of compound IVa or Vb is intended to mean that the carboxylic acid is activated, for example, by forming an anhydride or carboxylic acid halide, such as a carboxylic acid chloride.
  • the carboxylic acid can be activated using conventional carboxylic acid/amine coupling reagents, such carbodiimides, O-(7-azabenzotriazol-l-yl-iV, ⁇ N',N' tetramethyluronium hexafluorophosphate (HATU) and the like.
  • This reaction is conveniently performed under conventional amide bond-forming conditions.
  • the process is conveniently conducted at a temperature in the range of from -10 °C to 100 °C.
  • L 3 represents a leaving group including, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate; and P 2 represents a hydrogen atom or a carboxyl-protecting group, such as a (l-4C)alkyl group. If necessary, the carboxyl-protecting group P 2 , is then removed, for example, by hydrolysis under conventional conditions, such as by using lithium hydroxide.
  • P represents hydrogen or an amino-protecting group, such as fert-butoxycarbonyl
  • L 4 represents a leaving group including, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate; followed if necessary, by removing an amino-protecting group P 3 .
  • such compounds can be prepared by reductive amination of a compound of formula XV:
  • the reducing agent may be, for example, hydrogen in the presence of a Group VDI metal catalyst, such as palladium, or a metal hydride reducing agent, such as a borohydride, including sodium triacetoxyborohydride.
  • a Group VDI metal catalyst such as palladium
  • a metal hydride reducing agent such as a borohydride, including sodium triacetoxyborohydride.
  • Convenient solvents include alcohols, such as methanol. The reaction is conveniently performed at a temperature in the range of from 0 0 C to 100 °C.
  • P 4 represents hydrogen or an amino-protecting group, such as benzyl, in the presence of a reducing agent, such as sodium triacetoxyborohydride, followed if necessary by removing the amino-protecting group P 4 by, for example, hydrogenation in the presence of palladium.
  • a reducing agent such as sodium triacetoxyborohydride
  • the reducing agent may be, for example, hydrogen in the presence of a Group VIII metal catalyst, such as palladium, or a metal hydride reducing agent, such as a borohydride, including sodium triacetoxyborohydride.
  • Convenient solvents include alcohols, such as methanol.
  • the reaction is conveniently performed at a temperature in the range of from 0 °C to lOO °C.
  • the leaving group represented by L 2 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
  • This reaction is conveniently performed in the presence of a base, for example, a tertiary amine such as d ⁇ sopropylethylamine.
  • a base for example, a tertiary amine such as d ⁇ sopropylethylamine.
  • Convenient solvents include nitriles, such as acetonitrile.
  • the reaction is conveniently conducted at a temperature in the range of from 0 0 C to 100 0 C.
  • the compounds of formula VI can be prepared by reacting a compound of formula IVa with a compound of formula XIX:
  • reaction is conveniently performed following, for example, the method of process (b) described herein.
  • Compounds of formula VII are generally known or can be prepared from readily available starting materials using well-known synthetic methods.
  • the reducing agent may be, for example, hydrogen in the presence of a Group VIII metal catalyst, such as palladium, or a metal hydride reducing agent, such as a borohydride, including sodium triacetoxyborohydride, optionally used in combination with a titanium tetraalkoxide, such as titanium tetraisopropoxide.
  • a Group VIII metal catalyst such as palladium
  • a metal hydride reducing agent such as a borohydride, including sodium triacetoxyborohydride
  • titanium tetraalkoxide such as titanium tetraisopropoxide.
  • Convenient solvents include alcohols, such as methanol and halogenated hydrocarbons, such as dichloromethane. The reaction is conveniently performed at a temperature in the range of from 0 0 C to 100 0 C.
  • the compound of formula VIII may be prepared by oxidizing a compound corresponding to formula III in which L 1 represents a hydroxyl group. Such oxidation reactions can be conducted, for example, using sulfur dioxide pyridine complex in dimethylsulfoxide in the presence of a tertiary amine, such as diisopropylethylamine. In process (e), the reduction is preformed as described for process (d).
  • Compounds of formula IX may be prepared by reacting a compound of formula rVb with a compound of formula XXI:
  • XXI in the presence of a carboxylic acid/amine coupling agent, such as l-(3- dimethylaminopropyl)-3-ethylcarbodiimide (EDC) and 1-hydroxybenzotriazole hydrate (HOBT) and the like.
  • a carboxylic acid/amine coupling agent such as l-(3- dimethylaminopropyl)-3-ethylcarbodiimide (EDC) and 1-hydroxybenzotriazole hydrate (HOBT) and the like.
  • compounds of formula I prepared by any of steps (a) to (f) herein may be further derivatized to form other compounds of formula I using methods and reagents well-known in the art.
  • a compound of formula I may be reacted with bromine to afford a corresponding compound of formula I in which R 2 , for example, represents a bromo group.
  • a compound of formula I in which R 4 represents a hydrogen atom may be alkylated to afford a corresponding compound of formula I in which R 4 represents a (1-4C) alkyl group.
  • compositions typically administered to a patient in the form of a pharmaceutical composition or formulation.
  • Such compositions may be administered by any acceptable route of administration including, but not limited to, inhaled, oral, nasal, topical (including transdermal) and parenteral modes of administration. It will be understood that any form of the compound, i.e., free base, pharmaceutically acceptable salt, solvate, etc., that is suitable for the particular mode of administration can be used in the compositions described herein.
  • the invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the pharmaceutical composition may contain other therapeutic and/or formulating agents if desired.
  • compositions of the invention typically contain a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof, as the active agent.
  • the composition will contain from about 0.01-95% by weight of the active agent; including, from about 0.01-30% by weight; such as from about 0.01-10% by weight of the active agent.
  • any conventional carrier or excipient may be used in the compositions of the invention.
  • the choice of a particular carrier or excipient, or combination of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease state.
  • the preparation of a suitable composition for a particular mode of administration is well within the scope of those skilled in the pharmaceutical arts.
  • the ingredients for such compositions are commercially available from, for example, Sigma, P.O. Box 14508, St. Louis, MO 63178.
  • conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20 th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7 th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
  • compositions which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar;) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (alginic acid; pyrogen-free water; is
  • compositions of the invention are typically prepared by thoroughly and intimately mixing or blending a compound of formula I with a pharmaceutically acceptable carrier and one or more optional ingredients. If necessary or desired, the resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills, canisters, cartridges, dispensers and the like using conventional procedures and equipment.
  • compositions of the invention are suitable for inhaled administration.
  • Suitable compositions for inhaled administration will typically be in the form of an aerosol or a powder, and are generally administered using well-known delivery devices, such as a nebulizer inhaler, a dry powder inhaler (DPI), a metered-dose inhaler (MDI) or similar delivery device.
  • DPI dry powder inhaler
  • MDI metered-dose inhaler
  • a pharmaceutical composition comprising the active agent is administered by inhalation using a nebulizer inhaler.
  • nebulizer devices typically produce a stream of high velocity air that causes the composition comprising the active agent to spray as a mist that is carried into the patient's respiratory tract.
  • the active agent when formulated for use in a nebulizer inhaler, is typically dissolved in a suitable carrier to form a solution.
  • the active agent can be micronized and combined with a suitable carrier to form a suspension of micronized particles of respirable size, where micronized is typically defined as having about 90% or more of the particles with a diameter of less than about 10 ⁇ m.
  • Suitable nebulizer devices are commercially available, for example, by PARI GmbH (Sternberg, German). Other nebulizer devices include Respimat (Boehringer Ingelheim) and those described, for example, in U.S. Patent No. 6,123,068 to Lloyd et al. and WO 97/12687 (Eicher et al.), the disclosures of which are incorporated herein by reference in their entirety.
  • a representative pharmaceutical composition for use in a nebulizer inhaler comprises an isotonic aqueous solution comprising from about 0.05 ⁇ g/mL to about 10 mg/niL of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • a pharmaceutical composition comprising the active agent is administered by inhalation using a DPI.
  • DPIs typically administer the active agent as a free-flowing powder that is dispersed in a patient's air- stream during inspiration.
  • the active agent is typically formulated with a suitable excipient such as lactose or starch.
  • Micronization is a common method of reducing crystal size to that suitable for pulmonary delivery.
  • the active agent is micronized and combined with a suitable carrier to form a • suspension of micronized particles of respirable size, where "micronized particles" or “micronized form” means at least about 90% of the particles have a diameter of less than about 10 ⁇ m.
  • a representative pharmaceutical composition for use in a DPI comprises dry lactose having a particle size between about 1 ⁇ m and about 100 ⁇ m and micronized particles of a compound of formula I, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • a dry powder formulation can be made, for example, by combining the lactose with the active agent and then dry blending the components.
  • the active agent can be formulated without an excipient.
  • the pharmaceutical composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
  • DPI delivery devices include Diskhaler (GlaxoSmithKline, Research Triangle Park, NC) (see, e.g., U.S. Patent No. 5,035,237 to Newell et al.); Dislcus (GlaxoSmithKline) (see, e.g., U.S. Patent No. 6,378,519 to Davies et al.); Turbuhaler (AstraZeneca, Wilmington, DE) (see, e.g., U.S. Patent No.
  • the pharmaceutical composition comprising the active agent is administered by inhalation using an MDI, which typically discharges a measured amount of the active agent or a pharmaceutically acceptable salt or solvate or stereoisomer thereof using compressed propellant gas.
  • MDI typically discharges a measured amount of the active agent or a pharmaceutically acceptable salt or solvate or stereoisomer thereof using compressed propellant gas.
  • pharmaceutical compositions administered using an MDI typically comprise a solution or suspension of the active agent in a liquefied propellant.
  • Any suitable liquefied propellant may be employed including chlorofluorocarbons, such as CCI3F, and hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and I 5 1,1,2,3,3 ,3 -heptafluoro-n-propane, (HFA 227).
  • Formulations containing HFAs are generally preferred due to concerns about chlorofluorocarbons affecting the ozone layer.
  • Additional optional components of HFA formulations include co-solvents, such as ethanol or pentane, and surfactants, such as sorbitan trioleate, oleic acid, lecithin, and glycerin. See, for example, U.S. Patent No. 5,225,183 to Purewal et al., EP 0717987 A2 (Minnesota Mining and Manufacturing Company), and WO 92/22286 (Minnesota Mining and Manufacturing Company), the disclosures of which are incorporated herein by reference in their entirety.
  • a representative pharmaceutical composition for use in an MDI comprises from about 0.01-5 % by weight of a compound of formula I, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof; from about 0-20 % by weight ethanol; and from about 0-5 % by weight surfactant; with the remainder being an HFA propellant.
  • Such compositions are typically prepared by adding chilled or pressurized hydro fluoroalkane to a suitable container containing the active agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the active agent is micronized and then combined with the propellant. The formulation is then loaded into an aerosol canister, which forms a portion of a metered-dose inhaler device.
  • a suspension formulation can be prepared by spray drying a coating of surfactant on micronized particles of the active agent. See, for example, WO 99/53901 (Glaxo Group Ltd.) and WO 00/61108 (Glaxo Group Ltd.). The disclosures of the aforementioned patents and publications are incorporated herein by reference in their entirety.
  • compositions of the invention are suitable for oral administration.
  • suitable compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • the composition When intended for oral administration in a solid dosage form, i.e., as capsules, tablets, pills and the like, the composition will typically comprise a compound of the invention as the active ingredient and one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate.
  • a pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate.
  • the solid dosage forms may also comprise one or more of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and/or sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as cetyl alcohol and/or glycerol monostearate; absorbents, such as kaolin and/or bentonite clay; lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate,
  • antioxidants can also be present in the compositions of the invention.
  • pharmaceutically acceptable antioxidants include: water- soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and metal-chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water- soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), but
  • Coating agents for tablets, capsules, pills and like include those used for enteric coatings, such as cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acid-methacrylic acid ester copolymers, cellulose acetate trimellitate (CAT), carboxymethyl ethyl cellulose (CMEC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and the like.
  • enteric coatings such as cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acid-methacrylic acid ester copolymers, cellulose acetate trimellitate (CAT), carboxymethyl ethyl cellulose (CMEC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and the like.
  • enteric coatings such as cellulose acetate phthal
  • compositions of the invention may also be formulated to provide slow or controlled release of the active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres.
  • compositions of the invention may optionally contain opacifying agents and may be formulated so that they release the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • Liquid dosage forms typically comprise the active ingredient and an inert diluent, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (e.g., cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluent such as, for example, water or other solvents, solubilizing agents and emulsifier
  • Suspensions in addition to the active ingredient, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • the compositions of the invention are preferably packaged in a unit dosage form.
  • the term "unit dosage form" means a physically discrete unit suitable for dosing a patient, i.e., each unit containing a predetermined quantity of active agent calculated to produce the desired therapeutic effect either alone or in combination with one or more additional units.
  • such unit dosage forms may be capsules
  • the compounds of the invention can also be administered transdermally using known transdermal delivery systems and excipients.
  • a compound of the invention can be admixed with permeation enhancers, such as propylene glycol, polyethylene glycol monolaurate, azacycloalkan-2-ones and the like, and incorporated into a patch or similar delivery system.
  • permeation enhancers such as propylene glycol, polyethylene glycol monolaurate, azacycloalkan-2-ones and the like
  • Additional excipients including gelling agents, emulsifiers and buffers, may be used in such transdermal compositions if desired.
  • the compounds of the invention can also be co-administered with other therapeutic agents.
  • This combination therapy involves using a compound of the invention combined with one or more of these secondary agents, either formulated together (e.g., packaged together in a single formulation) or formulated separately (e.g., packaged as separate unit dosage forms). Methods of formulating multiple agents together in the same formulation or in separate unit dosage forms, are well known in the art.
  • the secondary agents can be used in the form of pharmaceutically acceptable salts or solvates, and if appropriate, as optically pure stereoisomers.
  • the secondary therapeutic agent(s) can be selected from other bronchodilators (e.g., PDE 3 inhibitors, adenosine 2b modulators and ⁇ 2 adrenergic receptor agonists); antiinflammatory agents (e.g., steroidal anti-inflammatory agents, such as corticosteroids; nonsteroidal antiinflammatory agents (NSAIDs), and PDE 4 inhibitors); other muscarinic receptor antagonists (i.e., antichlolinergic agents); antiinfective agents (e.g., Gram positive and Gram negative antibiotics or antivirals); antihistamines; protease inhibitors; and afferent blockers (e.g., D 2 agonists and neurokinin modulators).
  • bronchodilators e.g., PDE 3 inhibitors, adenosine 2b modulators and ⁇ 2 adrenergic receptor agonists
  • antiinflammatory agents e.g., steroidal anti-inflammatory agents, such as corticosteroids; non
  • One particular embodiment of the invention is directed to a composition
  • a composition comprising (a) a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof; and (b) a pharmaceutically acceptable carrier and a therapeutically effective amount of an agent selected from a steroidal anti-inflammatory agent such as a corticosteroid; a ⁇ 2 adrenergic receptor agonist; a phosphodiesterase-4 inhibitor; or a combination thereof; wherein the compound of formula I and the agent are formulated together or separately.
  • a pharmaceutically acceptable carrier and a therapeutically effective amount of a ⁇ 2 adrenergic receptor agonist and a steroidal anti-inflammatory agent is a pharmaceutically acceptable carrier and a therapeutically effective amount of a ⁇ 2 adrenergic receptor agonist and a steroidal anti-inflammatory agent.
  • Representative ⁇ 2 adrenergic receptor agonists include, but are not limited to, salmeterol, salbutamol, formoterol, salmefamol, fenoterol, terbutaline, albuterol, isoetharine, metaproterenol, bitolterol, pirbuterol, levalbuterol and the like, or pharmaceutically acceptable salts thereof.
  • ⁇ 2 adrenergic receptor agonists that can be used in combination with the compounds of the invention include, but are not limited to, 3- (4- ⁇ [6-( ⁇ (2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)-phenyl]ethyl ⁇ amino)- hexyl]oxy ⁇ butyl)benzenesulfonamide and 3-(-3- ⁇ [7-( ⁇ (2R)-2-hydroxy-2-[4-hydroxy-3- (hydroxymethyl)phenyl] ethyl ⁇ -amino)heptyl]oxy ⁇ -propyl)benzenesulfonamide and related compounds disclosed in WO 02/066422 (Glaxo Group Ltd.); 3-[3-(4- ⁇ [6-([(2R)-2- hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl] ethyl ⁇ amino)hexyl]oxy ⁇ butyl)- phenyl]irnidazolidine
  • the ⁇ 2 - adrenoreceptor agonist is a crystalline monohydrochloride salt of N- ⁇ 2-[4-((JR)-2-hydroxy- 2-phenylethylarnino)phenyl] ethyl ⁇ -(R)-2-hydroxy-2-(3 -formamido-4-hydroxyphenyl) ethylamine.
  • the ⁇ 2 -adrenoreceptor agonist will be present in the pharmaceutical composition in a therapeutically effective amount.
  • the ⁇ 2 - adrenoreceptor agonist will be present in an amount sufficient to provide from about 0.05- 500 ⁇ g per dose.
  • steroidal anti-inflammatory agents include, but are not limited to, methyl prednisolone, prednisolone, dexamethasone, fluticasone propionate, 6 ⁇ ,9 ⁇ -difluoro- 17 ⁇ -[(2-furanylcarbonyl)oxy] - 11 ⁇ -hydroxy- 16 ⁇ -methyl-3 -oxoandrosta- 1 ,4-diene- 17 ⁇ - carbothioic acid tf-fmoromethyl ester, 6 ⁇ ,9 ⁇ -difluoro- 11 ⁇ -hydroxy- 16 ⁇ -methyl-3 -oxo- 17 ⁇ - propionyloxy-androsta-l,4-diene-17 ⁇ -carbothioic acid S-(2-oxo-tetrahydrofuran-3S-yi) ester, beclomethasone esters (e.g., the 17-propionate ester or the 17,21-dipropionate ester), budesonide, flunisolide, momet
  • An exemplary combination is a compound of formula I, or pharmaceutically acceptable salt or solvate or stereoisomer thereof, co-administered with salmeterol as the ⁇ 2 adrenergic receptor agonist, and fluticasone propionate as the steroidal anti-inflammatory agent.
  • Another exemplary combination is a compound of formula I, or pharmaceutically acceptable salt or solvate or stereoisomer thereof, co-administered with a crystalline monohydrochloride salt of N- ⁇ 2-[4-((i?)-2-hydroxy-2-phenylethylamino)phenyl]ethyl ⁇ -(R)- 2-hydroxy-2-(3-formamido-4-hydroxyphenyl)ethylamine as the ⁇ 2 -adrenoreceptor agonist, and 6 ⁇ 5 9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-ll ⁇ -hydroxy-16 ⁇ -methyl-3-oxoandrosta- l,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester as the steroidal anti-inflammatory agent.
  • NSAIDs such as sodium cromoglycate; nedocromil sodium; phosphodiesterase (PDE) inhibitors (e.g., theophylline, PDE4 inhibitors or mixed PDE3/PDE4 inhibitors); leukotriene antagonists (e.g., monteleukast); inhibitors of leukotriene synthesis; iNOS inhibitors; protease inhibitors, such as tryptase and elastase inhibitors; beta-2 integrin antagonists and adenosine receptor agonists or antagonists (e.g., adenosine 2a agonists); cytokine antagonists (e.g., chemokine antagonists such as, an interleukin antibody ( ⁇ lL antibody), specifically, an ⁇ IL-4 therapy, an ⁇ IL-13 therapy, or a combination thereof); or inhibitors of cytokine synthesis.
  • NSAIDs such as sodium cromoglycate; nedocromil sodium; phosphodie
  • Representative phosphodiesterase-4 (PDE4) inhibitors or mixed PDE3/PDE4 inhibitors include, but are not limited to cis 4-cyano-4-(3-cyclopentyloxy-4- methoxyphenyl)cyclohexan-l-carboxylic acid, 2-carbomethoxy-4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-l-one; czs-[4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-l-ol]; cz5-4-cyano-4-[3- (cyclopentyloxy)-4-methoxyphenyl]cyclohexane-l-carboxylic acid and the like, or pharmaceutically acceptable salts thereof.
  • PDE4 or mixed PDE4/PDE3 inhibitors include AWD-12-281 (elbion); NCS-613 (INSERM); D-4418 (Chiroscience and Schering-Plough); CI-1018 or PD-168787 (Pfizer); benzodioxole compounds disclosed in WO99/16766 (Kyowa Hakko); K-34 (Kyowa Hakko); V-11294A (Napp); roflumilast (Byk-Gulden); pthalazinone compounds disclosed in WO99/47505 (Byk-Gulden); Pumafentrine (Byk-Gulden, now Altana); arofylline (Almirall- Prodesfarma); VM554/UM565 (Vernalis); T-440 (Tanabe Seiyaku); and T2585 (Tanabe Seiyaku).
  • muscarinic antagonists i.e., anticholinergic agents
  • muscarinic antagonists include, but are not limited to, atropine, atropine sulfate, atropine oxide, methylatropine nitrate, homatropine hydrobromide, hyoscyamine (d, I) hydrobromide, scopolamine hydrobromide, ipratropium bromide, oxitropium bromide, tiotropium bromide, methantheline, propantheline bromide, anisotropine methyl bromide, clidinium bromide, copyrrolate (Robinul), isopropamide iodide, mepenzolate bromide, tridihexethyl chloride (Pathilone), hexocyclium methylsulfate, cyclopentolate hydrochloride, tropicamide, trihexyphenidyl hydrochloride,
  • antihistamines include, but are not limited to, ethanolamines, such as carbinoxamine maleate, clemastine fumarate, diphenylhydramine hydrochloride and dimenhydrinate; ethylenediamines, such as pyrilamine amleate, tripelennarnine hydrochloride and tripelennamine citrate; alkylamines, such as chlorpheniramine and acrivastine; piperazines, such as hydroxyzine hydrochloride, hydroxyzine pamoate, cyclizine hydrochloride, cyclizine lactate, meclizine hydrochloride and cetirizine hydrochloride; piperidines, such as astemizole, levocabastine hydrochloride, loratadine or its descarboethoxy analogue, terfenadine and fexofenadine hydrochloride; azelastine hydrochloride; and
  • exemplary suitable doses for the other therapeutic agents administered in combination with a compound of the invention are in the range of about 0.05 ⁇ g/day to about 100 mg/day.
  • the following formulations illustrate representative compositions of the invention, as well as exemplary methods of preparation.
  • One or more secondary agents can optionally be formulated with the compound of the invention (primary active agent). Alternately, the secondary agents(s) can be formulated separately and co-administered with the primary active agent, either simultaneously or sequentially.
  • a single dry powder formulation can be manufactured to include both the compound of the invention and one or more secondary agents.
  • one formulation is manufactured to contain the compound of the invention and separate formulation(s) are manufactured to contain the secondary agent(s). Such dry powder formulations can then be packaged in separate blister packs and administered with a single DPI device.
  • Exemplary Dry Powder Formulation For Administration By Inhalation 0.2 mg of a compound of the invention is micronized and then blended with 25 mg of lactose. The blended mixture is then loaded into a gelatin inhalation cartridge. The contents of the cartridge are administered using a powder inhaler.
  • Exemplary Dry Powder Formulation For Administration By A Dry Powder Inhaler 0.2 mg of a compound of the invention is micronized and then blended with 25 mg of lactose. The blended mixture is then loaded into a gelatin inhalation cartridge. The contents of the cartridge are administered using a powder inhaler.
  • a dry powder is prepared having a bulk formulation ratio of micronized compound of the invention (active agent) to lactose of 1 :200.
  • the powder is packed into a dry powder inhalation device capable of delivering between about 10 ⁇ g and 100 ⁇ g of active agent per dose.
  • a suspension containing 5 wt% of a compound of the invention (active agent) and 0.1 wt% lecithin is prepared by dispersing 1O g of the active agent as micronized particles with a mean size less than 10 ⁇ m in a solution formed from 0.2 g of lecithin dissolved in 200 niL of demineralized water.
  • the suspension is spray dried and the resulting material is micronized to particles having a mean diameter less than 1.5 ⁇ m.
  • the particles are loaded into cartridges with pressurized 1,1,1,2-tetrafluoroethane.
  • a suspension containing 5 wt% of the active agent, 0.5 wt% lecithin, and 0.5 wt% trehalose is prepared by dispersing 5 g of the active agent as micronized particles with a mean size less than 10 ⁇ m in a colloidal solution formed from 0.5 g of trehalose and 0.5 g of lecithin dissolved in 100 mL of demineralized water.
  • the suspension is spray dried and the resulting material is micronized to particles having a mean diameter less than 1.5 ⁇ m.
  • the particles are loaded into canisters with pressurized 1,1,1,2-tetrafluoroethane.
  • a pharmaceutical composition is prepared by dissolving 0.5 mg of a compound of the invention (active agent) in 1 mL of a 0.9% sodium chloride solution acidified with citric acid. The mixture is stirred and sonicated until the active agent is dissolved. The pH of the solution is adjusted to a value in the range of from 3 to 8 (typically about 5) by the slow addition of NaOH.
  • Exemplary Hard Gelatin Capsule Formulation For Oral Administration The following ingredients are thoroughly blended and then loaded into a hard gelatin capsule: 250 mg of a compound of the invention, 200 mg of lactose (spray-dried), and 10 mg of magnesium stearate, for a total of 460 mg of composition per capsule.
  • Veegum k (Vanderbilt Co.) 1.0 g
  • the compounds of the invention are expected to be useful as muscarinic receptor antagonists and therefore, are expected to be useful for treating medical conditions mediated by muscarinic receptors, i.e., medical conditions which are ameliorated by treatment with a muscarinic receptor antagonist.
  • medical conditions include, by way of example, pulmonary disorders or diseases including those associated with reversible airway obstruction, such as chronic obstructive pulmonary disease (e.g., chronic and whez bronchitis and emphysema), asthma, pulmonary fibrosis, allergic rhinitis, rhinorrhea, and the like.
  • muscarinic receptor antagonists include genitourinary tract disorders, such as overactive bladder or detrusor hyperactivity and their symptoms; gastrointestinal tract disorders, such as irritable bowel syndrome, diverticular disease, achalasia, gastrointestinal hypermotility disorders and diarrhea; cardiac arrhythmias, such as sinus bradycardia; Parkinson's disease; cognitive disorders, such as Alzheimer's disease; dismenorrhea; and the like.
  • the compounds of the invention are useful for treating smooth muscle disorders in mammals, including humans and their companion animals (e.g., dogs, cats etc.). Smooth muscle disorders include, by way of illustration, overactive bladder, chronic obstructive pulmonary disease and irritable bowel syndrome.
  • the compounds of the invention When used to treat smooth muscle disorders or other conditions mediated by muscarinic receptors, the compounds of the invention will typically be administered orally, rectally, parenterally or by inhalation in a single daily dose or in multiple doses per day.
  • the amount of active agent administered per dose or the total amount administered per day will typically be determined by the patient's physician and will depend on factors such as the nature and severity of the patients condition, the condition being treated, age and general health of the patient, tolerance of the patient to the active agent, route of administration and the like.
  • suitable doses for treating smooth muscle disorders or other disorders mediated by muscarinic receptors will range from about 0.14 ⁇ g/kg/day to about 7 mg/kg/day of active agent; including from about 0.15 ⁇ g/kg/day to about 5 mg/kg/day. For an average 70 kg human, this would amount to about 10 ⁇ g per day to about 500 mg per day of active agent.
  • the compounds of the invention are useful for treating pulmonary or respiratory disorders, such as COPD or asthma, in mammals including humans.
  • pulmonary or respiratory disorders such as COPD or asthma
  • the compounds of the invention will typically be administered by inhalation in multiple doses per day, in a single daily dose or a single weekly dose.
  • the dose for treating a pulmonary disorder will range from about 10-200 ⁇ g/day.
  • COPD includes chronic obstructive bronchitis and emphysema (see, for example, Barnes, Chronic Obstructive Pulmonary Disease, N Engl J Med 343:269-78 (2000)).
  • the compounds of the invention are optionally administered in combination with other therapeutic agents such as a ⁇ 2 -adrenoreceptor agonist; a corticosteroid, a non-steroidal anti-inflammatory agent, or combinations thereof.
  • other therapeutic agents such as a ⁇ 2 -adrenoreceptor agonist; a corticosteroid, a non-steroidal anti-inflammatory agent, or combinations thereof.
  • the compounds of the invention When administered by inhalation, the compounds of the invention typically have the effect of producing bronchodilation. Accordingly, in another embodiment, the invention is directed to a method of producing bronchodilation in a patient, the method comprising administering to a patient a bronchodilation-producing amount of a compound of the invention.
  • the therapeutically effective dose for producing bronchodilation will range from about 10-200 ⁇ g/day.
  • the compounds of the invention are used to treat overactive bladder.
  • the compounds of the invention When used to treat overactive bladder, the compounds of the invention will typically be administered orally in a single daily dose or in multiple doses per day; preferably in a single daily dose.
  • the dose for treating overactive bladder will range from about 1.0-500 mg/day.
  • the compounds of the invention are used to treat irritable bowel syndrome.
  • the compounds of the invention When used to treat irritable bowel syndrome, the compounds of the invention will typically be administered orally or rectally in a single daily dose or in multiple doses per day.
  • the dose for treating irritable bowel syndrome will range from about 1.0-500 mg/day.
  • compounds of the invention are muscarinic receptor antagonists, such compounds are also useful as research tools for investigating or studying biological systems or samples having muscarinic receptors.
  • Such biological systems or samples may comprise M 1 , M 2 , M 3 , M 4 and/or M 5 muscarinic receptors.
  • Any suitable biological system or sample having muscarinic receptors may be employed in such studies which may be conducted either in vitro or in vivo.
  • a biological assay is conducted by contacting a biological system or sample (comprising a muscarinic receptor) with a muscarinic receptor-antagonizing amount of a compound of the invention.
  • a biological system or sample comprising a muscarinic receptor
  • the effects of antagonizing the muscarinic receptor are then determined using conventional procedures and equipment, such as radioligand binding assays and functional assays.
  • Such functional assays include ligand- mediated changes in intracellular cyclic adenosine monophosphate (cAMP), ligand- mediated changes in activity of the enzyme adenylyl cyclase (which synthesizes cAMP), ligand-mediated changes in incorporation of guanosine 5'-O-( ⁇ -thio)tri ⁇ hosphate ([ S]GTPyS) into isolated membranes via receptor catalyzed exchange of [ S]GTPyS for GDP, ligand-mediated changes in free intracellular calcium ions (measured, for example, with a fluorescence-linked imaging plate reader or FLIPR ® from Molecular Devices, Inc.).
  • cAMP cyclic adenosine monophosphate
  • adenylyl cyclase which synthesizes cAMP
  • a compound of the invention will antagonize or decrease the activation of muscarinic receptors in any of the functional assays listed above, or assays of a similar nature.
  • a muscarinic receptor-antagonizing amount of a compound of the invention will typically range from about 0.1-100 nanomolar.
  • a method of evaluating a test compound in a biological assay comprises: (a) conducting a biological assay with a test compound to provide a first assay value; (b) conducting the biological assay with a compound of formula I to provide a second assay value; wherein step (a) is conducted either before, after or concurrently with step (b); and (c) comparing the first assay value from step (a) with the second assay value from step (b).
  • Exemplary biological assays include, by way of illustration and not limitation, a muscarinic receptor binding assay or a bronchoprotection assay in a mammal.
  • muscarinic receptor binding data e.g., as determined by in vitro radioligand displacement assays
  • This aspect of the invention includes, as separate embodiments, both the generation of comparison data (using the appropriate assays) and the analysis of the test data to identify test compounds of interest.
  • the compounds of the invention are used to antagonize a muscarinic receptor in a biological system, and a mammal in particular, such as mice, rats, guinea pigs, rabbits, dogs, pigs, humans and so forth.
  • a therapeutically effective amount of a compound of formula I is administered to the mammal, and the effects of antagonizing the muscarinic receptor can then determined using conventional procedures and equipment, examples of which are described above.
  • compounds of formula I have been found to be potent inhibitors of M 3 muscarinic receptor activity. Accordingly, in a specific embodiment, the invention is directed to those compounds having an inhibition dissociation constant (K;) for the M 3 receptor subtype of less than or equal to 10 nM; preferably, less than or equal to 5 nM; (as determined, for example, by an in vitro radioligand displacement assay). Additionally, compounds of formula I are expected to possess a desirable duration of action. Accordingly, in another specific embodiment, the invention is directed to those compounds having a duration of action greater than or equal to about 24 hours. Moreover, compounds of the invention are also expected to possess reduced side effects, such as dry mouth, at efficacious doses when administered by inhalation compared to other known muscarinic receptor antagonists administered by inhalation (such as tiotropium).
  • K inhibition dissociation constant
  • HPLC analysis was conducted using an Agilent (Palo Alto, CA) Series 1100 instrument equipped with a Zorbax Bonus RP 2.1 x 50 mm column (Agilent) having a 3.5 micron particle size. Detection was by UV absorbance at 214 nm.
  • the mobile phases employed were as follows (by volume): A is ACN (2%), water (98%) and TFA (0.1%); and B is ACN (90%), water (10%) and TFA (0.1%).
  • HPLC 10-70 data was obtained using a flow rate of 0.5 mL/minute of 10 to 70% B over a 6 minute gradient (with the remainder being A).
  • HPLC 5-35 data and HPLC 10-90 data were obtained using 5 to 35% B; or 10 to 90% B over a 5 minute gradient.
  • LCMS Liquid chromatography mass spectrometry
  • Biphenyl-2-isocyanate 97.5 g, 521 mmol
  • 4-hydroxy-N-benzylpiperidine 105 g, 549 mmol
  • the reaction mixture was then cooled to 50 0 C and EtOH (1 L) was added and then 6M HCl (191 mL) was added slowly.
  • the resulting mixture was then cooled to ambient temperature and ammonium formate (98.5 g, 1.56 mol) was added and then nitrogen gas was bubbled through the solution vigorously for 20 minutes.
  • Palladium on activated carbon (20 g, 10 wt% dry basis) was then added and the reaction mixture was heated at 40 °C for 12 hours, and then filtered through a pad of Celite.
  • the product was extracted using DCM washes (3 X 50 niL) and the organic layer washed with water (100 mL), NaCl (sat.) (100 mL), dried over MgSO 4 and then filtered. The solvent removed under reduced pressure. The crude material was sufficiently pure to use without further purification. The title compound was obtained in 78% yield (2.0 g, 11.2 mmol).
  • Radioligand binding assays were performed in 96-well microtiter plates in a total assay volume of 100 ⁇ L.
  • CHO cell membranes stably expressing either the hM l5 hM 2 , hM 3 , hM4 or hMs muscarinic subtype were diluted in assay buffer to the following specific target protein concentrations ( ⁇ g/well): 10 ⁇ g for hM l5 10-15 ⁇ g for hM 2 , 10-20 ⁇ g for hM 3 , 10-20 ⁇ g for ILM 4 , and 10-12 ⁇ g for hM 5 .
  • the membranes were briefly homogenized using a Polytron tissue disruptor (10 seconds) prior to assay plate addition.
  • the addition order and volumes to the assay plates were as follows: 25 ⁇ L radioligand, 25 ⁇ L diluted test compound, and 50 ⁇ L membranes. Assay plates were incubated for 60 minutes at 37 °C. Binding reactions were terminated by rapid filtration over GFfB glass fiber filter plates (PerkinElmer Inc., Wellesley, MA) pre-treated in 1% BSA. Filter plates were rinsed three times with wash buffer (10 mM HEPES) to remove unbound radioactivity. Plates were then air dried, and 50 ⁇ L Microscint-20 liquid scintillation fluid (PerkinElmer Inc., Wellesley, MA) was added to each well.
  • a lower K ; - value indicates that the test compound has a higher binding affinity for the receptor tested.
  • Muscarinic Receptor Functional Potency Assays Blockade of Agonist-Mediated Inhibition of cAMP Accumulation
  • the functional potency of a test compound is determined by measuring the ability of the test compound to block oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM 2 receptor.
  • cAMP assays are performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with 125 I-cAMP (NEN SMP004B, PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturer's instructions.
  • Cells are rinsed once with dPBS and lifted with Trypsin-EDTA solution (0.05% trypsin/0.53 mM EDTA) as described in the Cell Culture and Membrane Preparation section above.
  • the detached cells are washed twice by centrifugation at 650 x g for five minutes in 5OmLs dPBS.
  • the cell pellet is then re-suspended in 10 mL dPBS, and the cells are counted with a Coulter Zl Dual Particle Counter (Beckman Coulter, Fullerton, CA).
  • the cells are centrifuged again at 650 x g for five minutes and re-suspended in stimulation buffer to an assay concentration of 1.6 x 10 6 - 2.8 x 10 6 cells/mL.
  • the test compound is initially dissolved to a concentration of 400 ⁇ M in dilution buffer (dPBS supplemented with 1 mg/mL BSA (0.1%)), and then serially diluted with dilution buffer to final molar concentrations ranging from 100 ⁇ M to 0.1 nM.
  • Oxotremorine is diluted in a similar manner.
  • oxotremorine inhibition of adenylyl cyclase (AC) activity 25 ⁇ L forskolin (25 ⁇ M final concentration diluted in dPBS), 25 ⁇ L diluted oxotremorine, and 50 ⁇ L cells are added to agonist assay wells.
  • 25 ⁇ L forskolin and oxotremorine 25 ⁇ M and 5 ⁇ M final concentrations, respectively, diluted in dPBS
  • 25 ⁇ L diluted test compound, and 50 ⁇ L cells are added to remaining assay wells.
  • the Cheng-Prusoff equation is used to calculate the K;, using the EC 50 of the oxotremorine concentration- response curve and the oxotremorine assay concentration as the K D and [L], respectively.
  • the Ki values are converted to pKj values to determine the geometric mean and 95% confidence intervals. These summary statistics are then converted back to Ki values for data reporting, hi this assay, a lower Ki value indicates that the test compound has a higher functional activity at the receptor tested.
  • Compounds of the invention are expected to have a Kj value of less than about 10 nM for blockade of oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM 2 receptor, when tested in this or a similar assay.
  • the functional potency of test compounds can be determined by measuring the ability of the compounds to block oxotremorine-stimulated [ 35 S]GTPyS binding in CHO-Kl cells expressing the I1M 2 receptor, At the time of use, frozen membranes are thawed and then diluted in assay buffer with a final target tissue concentration of 5-10 ⁇ g protein per well. The membranes are briefly homogenized using a Polytron PT-2100 tissue disrupter and then added to the assay plates. The EC 90 value (effective concentration for 90% maximal response) for stimulation of [ 35 S]GTPyS binding by the agonist oxotremorine is determined in each experiment.
  • test compound To determine the ability of a test compound to inhibit oxotremorine-stimulated [ 35 S]GTPyS binding, the following is added to each well of 96 well plates: 25 ⁇ L of assay buffer with [ 35 S]GTPyS (0.4nM), 25 ⁇ L of oxotremorme(EC 90 ) and GDP (3 ⁇ M), 25 ⁇ L of diluted test compound and 25 ⁇ L CHO cell membranes expressing the hM 2 receptor. The assay plates are then incubated at 37 °C for 60 minutes. The assay plates are filtered over 1% BSA- pretreated GFTB filters using a PerkinElmer 96-well harvester.
  • the plates are rinsed with ice-cold wash buffer for 3 x 3 seconds and then air or vacuum dried.
  • Microscint-20 scintillation liquid 50 ⁇ L is added to each well, and each plate is sealed and radioactivity counted on a topcounter (PerkinElmer).
  • Data are analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, hie, San Diego, CA) using the non-linear regression, one-site competition equation.
  • the Cheng-Prusoff equation is used to calculate the Kj, using the IC 50 values of the concentration-response curve for the test compound and the oxotremorine concentration in the assay as the K D and [L], ligand concentration, respectively.
  • a lower Kj value indicates that the test compound has a higher functional activity at the receptor tested.
  • Compounds of the invention are expected to have a Kj value of less than about 10 nM for blockade of oxotremorine-stimulated [ 35 S]GTP ⁇ S_binding in CHO-Kl cells expressing the hM 2 receptor, when tested in this or a similar assay. Blockade of Agonist-Mediated Calcium Release via FLIPR Assays
  • Muscarinic receptor subtypes (M 1 , M 3 and M 5 receptors), which couple to G q proteins, activate the phospholipase C (PLC) pathway upon agonist binding to the receptor.
  • PLC phospholipase C
  • activated PLC hydrolyzes phosphatyl inositol diphosphate (PIP 2 ) to diacylglycerol (DAG) and phosphatidyl-l,4,5-triphosphate (IP 3 ), wliich in turn generates calcium release from intracellular stores, i.e., endoplasmic and sarcoplasmic reticulum.
  • the FLIPR Molecular Devices, Sunnyvale, CA
  • assay capitalizes on this increase in intracellular calcium by using a calcium sensitive dye (Fluo-4AM, Molecular Probes, Eugene, OR) that fluoresces when free calcium binds.
  • This fluorescence event is measured in real time by the FLIPR, which detects the change in fluorescence from a monolayer of cells cloned with human M 1 and M 3 , and chimpanzee M 5 receptors.
  • Antagonist potency can be determined by the ability of antagonists to inhibit agonist-mediated increases in intracellular calcium.
  • FLIPR calcium stimulation assays CHO cells stably expressing the hM l5 hM 3 and cM 5 receptors are seeded into 96-well FLIPR plates the night before the assay is done.
  • Seeded cells are washed twice by Cellwash (MTX Labsystems, Inc.) with FLIPR buffer (10 mM HEPES, pH 7.4, 2 mM calcium chloride, 2.5 mM probenecid in Hank's Buffered Salt Solution (HBSS) without calcium and magnesium) to remove growth media and leaving 50 ⁇ L/well of FLIPR buffer.
  • FLIPR buffer 10 mM HEPES, pH 7.4, 2 mM calcium chloride, 2.5 mM probenecid in Hank's Buffered Salt Solution (HBSS) without calcium and magnesium
  • HBSS Hank's Buffered Salt Solution
  • FLUO-4AM a 2X solution was made
  • the dose-dependent stimulation of intracellular Ca 2+ release for oxotremorine is first determined so that antagonist potency can later be measured against oxotremorine stimulation at an ECg 0 concentration.
  • Cells are first incubated with compound dilution buffer for 20 minutes, followed by agonist addition, which is performed by the FLIPR.
  • An oxotremorine concentration of 3 x ECF is prepared in stimulation plates such that an EC 90 concentration of oxotremorine is added to each well in the antagonist inhibition assay plates.
  • the parameters used for the FLIPR are: exposure length of 0.4 seconds, laser strength of 0.5 watts, excitation wavelength of 488 nm, and emission wavelength of 550 nm. Baseline is determined by measuring the change in fluorescence for 10 seconds prior to addition of agonist. Following agonist stimulation, the FLIPR continuously measured the change of fluorescence every 0.5 to 1 second for 1.5 minutes to capture the maximum fluorescence change. The change of fluorescence is expressed as maximum fluorescence minus baseline fluorescence for each well.
  • the raw data is analyzed against the logarithm of drug concentration by nonlinear regression with GraphPad Prism (GraphPad Software, Inc., San Diego, CA) using the built-in model for sigmoidal dose-response.
  • Antagonist Kj values are determined by Prism using the oxotremorine EC 50 value as the KQ and the oxotremorine EC 9 Q for the ligand concentration according to the Cheng-Prusoff equation (Cheng & Prusoff, 1973). In this assay, a lower Ki value indicates that the test compound has a higher functional activity at the receptor tested.
  • Compounds of the invention are expected to have a Kj value of less than about 10 nM for blockade of agonist-mediated calcium release in CHO cells stably expressing the hM 3 receptor, when tested in this or a similar assay.
  • ASSAY 3 ASSAY 3
  • This in vivo assay is used to assess the bronchoprotective effects of test compounds exhibiting muscarinic receptor antagonist activity.
  • Groups of six male guinea pigs (Duncan-Hartley (HsdPoc:DH) Harlan, Madison, WI) weighing between 250 and 350 g are individually identified by cage cards. Throughout the study animals are allowed access to food and water ad libitum.
  • Test compounds are administered via inhalation over 10 minutes in a whole-body exposure dosing chamber (R&S Molds, San Carlos, CA). The dosing chambers are arranged so that an aerosol was simultaneously delivered to 6 individual chambers from a central manifold. Guinea pigs are exposed to an aerosol of a test compound or vehicle (WFI).
  • WFI test compound or vehicle
  • bronchoprotective effects of test compounds administered via inhalation are evaluated using whole body plethysmography at 1.5, 24, 48 and 72 hours post-dose. Forty- five minutes prior to the start of the pulmonary evaluation, each guinea pig is anesthetized with an intramuscular injection of ketamine (43.75 mg/kg), xylazine (3.50 mg/kg) and acepromazine (1.05 mg/kg). After the surgical site is shaved and cleaned with 70% alcohol, a 2-3 cm midline incision of the ventral aspect of the neck was made.
  • the jugular vein is isolated and cannulated with a saline-filled polyethylene catheter (PE- 50, Becton Dickinson, Sparks, MD) to allow for intravenous infusions of ACh (Sigma- Aldrich, St. Louis, MO) in saline.
  • PE- 50 Becton Dickinson, Sparks, MD
  • ACh Sigma- Aldrich, St. Louis, MO
  • the trachea is then dissected free and cannulated with a 14G teflon tube (#NE- 014, Small Parts, Miami Lakes, FL).
  • anesthesia is maintained by additional intramuscular injections of the aforementioned anesthetic mixture. The depth of anesthesia is monitored and adjusted if the animal responds to pinching of its paw or if the respiration rate is greater than 100 breaths/minute. Once the cannulations are complete, the animal is placed into a plethysmo graph
  • a heating lamp is used to maintain body temperature and the guinea pig's lungs are inflated 3 times with 4 mL of air using a 10 mL calibration syringe (#5520 Series, Hans Rudolph, Kansas City, MO) to ensure that the lower airways do not collapse and that the animal does not suffer from hyperventilation.
  • the changes in volume over time that occur within the plethysmograph with each breath are measured via a Buxco pressure transducer. By integrating this signal over time, a measurement of flow is calculated for each breath.
  • This signal, together with the pulmonary driving pressure changes, which are collected using a Sensym pressure transducer (#TRD4100), is connected via a Buxco (MAX 2270) preamplifier to a data collection interface (#'s SFT3400 and SFT3813). All other pulmonary parameters are derived from these two inputs. Baseline values are collected for 5 minutes, after which time the guinea pigs are challenged with ACh.
  • ACh (0.1 mg/mL) is infused intravenously for 1 minute from a syringe pump (sp210iw, World Precision Instruments, Inc., Sarasota, FL) at the following doses and prescribed times from the start of the experiment: 1.9 ⁇ g/minute at 5 minutes, 3.8 ⁇ g/minute at 10 minutes, 7.5 ⁇ g/minute at 15 minutes, 15.0 ⁇ g/minute at 20 minutes, 30 ⁇ g/minute at 25 minutes and 60 ⁇ g/minute at 30 minutes. If resistance or compliance has not returned to baseline values at 3 minutes following each ACh dose, the guinea pig's lungs are inflated 3 times with 4 mL of air from a 10 mL calibration syringe. Recorded pulmonary parameters includes respiration frequency (breaths/minute), compliance
  • the mean ACh response in vehicle-treated animals, at each pre-treatment time, is calculated and used to compute % inhibition of ACh response, at the corresponding pre-treatment time, at each test compound dose.
  • Inhibition dose-response curves for 'R L ' are fitted with a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) to estimate bronchoprotective ID 5O (dose required to inhibit the ACh (60 ⁇ g/min) bronchoconstrictor response by 50%).
  • Y Min + (Max-Min)/(l + ⁇ 0 ( ⁇ so-xr rnu o ⁇
  • X the logarithm of dose
  • Y the response (% Inhibition of ACh induced increase in R L ).
  • the quantity PD 2 which is defined as the amount of ACh or histamine needed to cause a doubling of the baseline pulmonary resistance, is calculated using the pulmonary resistance values derived from the flow and the pressure over a range of ACh or histamine challenges using the following equation (which is derived from a equation used to calculate PC 20 values described in American Thoracic Society. Guidelines for methacholine and exercise challenge testing - 1999. Am JRespir Crit Care Med. 161: 309-329 (2000)):
  • PD 2 antilog [ log C 1 + (log C 2 - log CI)(2RQ - RQ ]
  • C 1 is the concentration of ACh or histamine preceding C 2 ;
  • C 2 is the concentration of
  • An efficacious dose is defined as a dose that limits the bronchrestriction response to a 50 ⁇ g/mL dose of ACh to a doubling of the baseline pulmonary resistance (PD 2(50) ).
  • test compounds having a PD 2 ( 5 o) less than about 200 ⁇ g/mL for ACh-induced bronchoconstriction at 1.5 hours post-dose in this assay are preferred.
  • Compounds of the invention are expected to have a PD 2(S0) of less than about 200 ⁇ g/mL for ACh-induced bronchoconstriction at 1.5 hours post-dose, when tested in this or a similar assay.
  • ASSAY 4 Inhalation Guinea Pig Salivation Assay Guinea pigs (Charles River, Wilmington, MA) weighing 200-350 g are acclimated to the in-house guinea pig colony for at least 3 days following arrival.
  • Test compound or vehicle are dosed via inhalation (IH) over a 10 minute time period in a pie shaped dosing chamber (R&S Molds, San Carlos, CA).
  • Test solutions are dissolved in sterile water and delivered using a nebulizer filled with 5.0 mL of dosing solution.
  • Guinea pigs are restrained in the inhalation chamber for 30 minutes. During this time, guinea pigs are restricted to an area of approximately 110 sq. cm.
  • guinea pigs are exposed to an aerosol generated from a LS Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA) driven by house air at a pressure of 22 psi.
  • LS Star Nebulizer Set Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA
  • guinea pigs are evaluated at 1.5, 6, 12, 24, 48, or 72 hrs after treatment.
  • Guinea pigs are anesthetized one hour before testing with an intramuscular (IM) injection of a mixture of ketamine 43.75 mg/kg, xylazine 3.5 mg/kg, and acepromazine 1.05 mg/kg at an 0.88 mL/kg volume.
  • Animals are placed ventral side up on a heated (37°C) blanket at a 20 degree incline with their head in a downward slope.
  • a 4-ply 2 x 2 inch gauze pad (Nu-Gauze General-use sponges, Johnson and Johnson, Arlington, TX) is inserted in the guinea pig's mouth.
  • the gauze pad is immediately discarded and replaced by a new pre-weighed gauze pad.
  • Saliva is collected for 10 minutes, at which point the gauze pad is weighed and the difference in weight recorded to determine the amount of accumulated saliva (in mg).
  • the mean amount of saliva collected for animals receiving the vehicle and each dose of test compound is calculated.
  • Y Min + (Max-Min)/(1 + 10 « log ID5 °- ⁇ )* WM °& )
  • X is the logarithm of dose
  • Y is the response (% inhibition of salivation).
  • Y starts at Min and approaches asymptotically to Max with a sigmoidal shape.
  • the ratio of the anti- sialagogue ID 5O to bronchoprotective ID 50 is used to compute the apparent lung-selectivity index of the test compound.
  • compounds having an apparent lung-selectivity index greater than about 5 are preferred.
  • Compounds of the invention are expected to have an apparent lung-selectivity index greater than 5, when tested in this or a similar assay.
  • Each animal is administered saline (3 mL, SC) at the end of surgery as well as buprenorphine (0.05 mg/kg, IM). Animals are allowed to recover on a heating pad before being returned to their holding rooms. Approximately 18 to 20 hours following surgery, the animals are weighed and the carotid artery catheter on each animal is connected to a transducer for recording arterial pressure. Arterial pressure and heart rate are recorded using a Biopac MP-100 Acquisition System. Animals are allowed to acclimate and stabilize for a period of 20 minutes.
  • Each animal is challenged with MCh (0.3 mg/kg, IY) administered through the jugular venous line and the cardiovascular response is monitored for 10 minutes.
  • the animals are then placed into the whole body dosing chamber, which is connected to a nebulizer containing the test compound or vehicle solution.
  • the solution is nebulized for 10 minutes using a gas mixture of breathable air and 5% carbon dioxide with a flow rate of 3 liters/minute.
  • the animals are then removed from the whole body chamber and returned to their respective cages.
  • the animals are re-challenged with MCh (0.3 mg/kg, IV) and the hemodynamic response is determined.
  • MCh produces a decrease in mean arterial pressure (MAP) and decrease in heart rate (bradycardia).
  • MAP mean arterial pressure
  • MAP heart rate
  • the peak decrease, from baseline, in MAP (depressor responses) is measured for each MCh challenge (before and after IH dosing).
  • the effects of treatment on the MCh responses are expressed as % inhibition (mean +/- SEM) of the control depressor responses.
  • Two-way ANOVA with the appropriate post-hoc test is used to test the effects of treatment and pre- treatment time.
  • the depressor responses to MCh are expected to be relatively unchanged at 1.5 and 24 hours after inhalation dosing with vehicle.
  • the ratio of the anti-depressor ID 50 to bronchoprotective ID 50 is used to compute apparent lung-selectivity of the test compound.
  • compounds having an apparent lung-selectivity index greater than 5 are preferred. It is expected that the compounds of the invention will exhibit an apparent lung-selectivity index greater than 5, as measured in this or a similar assay.

Abstract

The invention provides compounds of formula (I) wherein a, b, c, m, s, t, W, Z, Ar, R1, R2, R3, R6, and R7 are as defined in the specification. The compounds of formula (I) are muscarinic receptor antagonists. The invention also provides pharmaceutical compositions containing such compounds, processes and intermediates for preparing such compounds and methods of using such compounds to treat pulmonary disorders.

Description

BIPHENYL COMPOUNDS
USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS BACKGROUND OF THE INVENTION
Field of the Invention The present invention relates to novel biphenyl compounds having muscarinic receptor antagonist or anticholinergic activity. The invention also relates to pharmaceutical compositions comprising these compounds, processes and intermediates for preparing these compounds and methods of using these compounds to treat pulmonary disorders.
State of the Art Pulmonary or respiratory disorders, such as chronic obstructive pulmonary disease
(COPD) and asthma, afflict many millions of people worldwide and are a leading cause of morbidity and mortality. Muscarinic receptor antagonists are known to provide bronchoprotective effects and therefore are useful for treating respiratory disorders, such as COPD and asthma. Muscarinic receptor antagonists are typically administered by inhalation to treat these disorders. However, even when administered by inhalation, a significant amount of the antagonist is often absorbed into the systemic circulation resulting in systemic side effects, such as dry mouth, mydriasis and cardiovascular side effects. In addition, many inhaled muscarinic receptor antagonists have a relatively short duration of action requiring that they be administered several times per day. This multiple- daily dosing regime is inconvenient and creates a significant risk of inadequate treatment due to patient non-compliance with the required frequent dosing schedule.
Accordingly, a need exists for new muscarinic receptor antagonists, in particular, those having high potency and reduced systemic side effects when administered by inhalation. Additionally, a need exists for inhaled muscarinic receptor antagonists having a long duration of action thereby allowing for once-daily or even once-weekly dosing. Such compounds are expected to be particularly effective for treating pulmonary disorders, such as COPD and asthma, while reducing or eliminating side effects, such as dry-mouth and constipation.
SUMMARY OF THE INVENTION The present invention provides novel biphenyl compounds having muscarinic receptor antagonist or anticholinergic activity. Among other properties, compounds of the invention are expected to possess high potency and reduced systemic side effects when administered by inhalation and are also expected to possess a long duration of action. One aspect of the invention relates to a compound of formula I:
Figure imgf000003_0001
I wherein: a is 0 or an integer of from 1 to 5; each R1 is independently selected from -C1-5alkyl, -C2-5alkenyl, -C2-5alkynyl, -C3-6cycloalkyl, -cyano, -halo, -ORla, -C(O)ORlb, -SRlc, -S(O)Rld, -S(O)2R16, -NRlfRlg 5 -NRlhS(O)2Rlj, and -NRljC(O)Rlk; where each of Rla'lk is independently -H, -C1-5alkyl or -phenyl-C1-5alkyl; b is 0 or an integer of from 1 to 4; each R2 is independently selected from -C1-5alkyl, -C2-5alkenyl, -C2-5alkynyl, -Cs-ecycloalkyl, -cyano, -halo, -OR2a, -C(O)OR2b, -SR2c, -S(O)R2d, -S(O)2R26, -NR2fR2g, -NR211S(O)2R21, and -NR2jC(O)R2k; where each of R2a'2k is independently -H, -C1-5alkyl or -phenyl-C \ -salkyl; W is -O- or -NWa-, where Wa is -H or -C1-5alkyl; c is O or an integer from 1 to 5; each R3 independently is -C1-5alkyl or two R3 groups are joined to form C1-3alkylene, C2-3alkenylene or oxiran-2,3-diyl; m is O or 1; Z is selected from -C(O)N(R4)- and -N(R4)C(O)-, where R4 is selected from -H,
-C1-5alkyl, and -C3-5CyClOaUCyI; s is 0, 1 or 2;
Ar is a phenylene group or a C3-5heteroarylene group containing 1 or 2 heteroatoms independently selected from oxygen, nitrogen or sulfur; wherein the phenylene or C3-5heteroarylene group is substituted with (R5)q where q is 0 or an integer from 1 to 4 and each R5 is independently selected from -halo, -OH, -Q.salkyl or -C^salkoxy; t is 0, 1 or 2;
R6 is selected from -H, -C1-5alkyl, and -X6R6a; where X6 is -C1-5alkylene and R6a is selected from -OH, -C1-5alkoxy and -heteroaryl; and R7 is selected from -H, -C1-5alkyl, -C3-6cycloalkyl, and -X7R7a; where X7 is selected from -C1-5alkylene, -C(O)-, -C1-5alkylene-C(O)-, -S(O2)-, -C1-5alkylene-S(O2)-, and -S(O2)-C1-5alkylene; and R7a is selected from -OH, -Q^alkyl, -Cs-βcycloalkyl, -C^alkoxy, heteroaryl, -NR7bR7c, where R7b and R7c are independently -H or -C1-5alkyl, and aryl optionally substituted with 1-2 -Q.salkyl or halo groups; or R6 and R7 are taken together to form:
Figure imgf000004_0001
or
— N O
optionally substituted with one to three -C^alkyl groups, or
Figure imgf000004_0002
where: d is 1 or 2; e is 0, 1 or 2; f is 0, 1, 2 or 3; R8 is selected from -C1-5alkyl and =0; R9 is selected from -H, -C1-5alkyl, hydroxyphenyl, heteroaryl, and -X9R9a; where X9 is selected from -C1-5alkylene, -C(O)-, -C1-5alkylene-C(O)-, -C(O)-C 1-5alkylene, -S(O2)-, -C1-5alkylene-S(O2)-, and -S(O2)-C1-5alkylene; and R9a is selected from -H, -OH, -C1-5alkyl, -C1-5alkoxy, aryl, heteroaryl, heterocyclyl, and -NR9bR9°, where R9b and R9c are independently -H or -C1-5alkyl; wherein each alkyl and alkoxy group in R1, Rla"lk, R2, R2a-2k, R3, R5, R6, R6a, R7, R7a"c, R8, R9, and R9a"c is optionally substituted with 1 to 5 fluoro substituents; or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Another aspect of the invention pertains to pharmaceutical compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof. Yet another aspect of the invention pertains to compositions comprising a compound of formula I in combination with one or more other therapeutic agents. Accordingly, in one embodiment, the invention is directed to compositions comprising (a) a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof; and (b) a therapeutically effective amount of an agent selected from a steroidal anti-inflammatory agent such as a corticosteroid; a β2 adrenergic receptor agonist; a phosρhodiesterase-4 inhibitor; or a combination thereof; wherein the compound of formula I and the agent are formulated together or separately. When the agent is formulated separately, a pharmaceutically acceptable carrier may be included.
Compounds of the invention possess muscarinic receptor antagonist activity. Accordingly, compounds of formula I are expected to be useful for treating pulmonary disorders, such as chronic obstructive pulmonary disease and asthma.
Yet another aspect of the invention is directed to methods for treating a pulmonary disorders comprising administering to a patient a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Still another aspect of the invention pertains to methods of producing bronchodilation in a patient comprising administering to a patient a bronchodilation- producing amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. The invention is also directed to methods of treating chronic obstructive pulmonary disease or asthma comprising administering to a patient a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. In another aspect, the invention is directed to methods for antagonizing a muscarinic receptor in a mammal comprising administering to the mammal a therapeutically effective amount of the compound of formula I. Since compounds of the invention possess muscarinic receptor antagonist activity, such compounds are also useful as research tools. Accordingly, in yet another embodiment, the invention is directed to methods for using a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof as a research tool for studying a biological system or sample, or for discovering new chemical compounds having muscarinic receptor antagonist activity.
The invention is also directed to processes and intermediates useful for preparing compounds of formula I and pharmaceutically acceptable salts or solvates or stereoisomers thereof. Accordingly, in another embodiment, the invention is directed to a process of preparing a compound of formula I, the process comprising: (a) reacting a compound of formula II with a compound of formula III; or (b) coupling a compound of formula IVa with a compound of formula Va or coupling a compound of formula IVb with a compound of formula Vb; or (c) reacting a compound of formula VI with a compound of formula VII; or (d) reacting a compound of formula II with a compound of formula VIII in the presence of a reducing agent; or (e) reacting a compound of formula IX with a compound of formula VII in the presence of a reducing agent; and then removing any protecting groups that may be present to provide a compound of formula I, and optionally, forming a pharmaceutically acceptable salt thereof, wherein compounds of formula I-IX, are as defined herein. In one embodiment, the above process further comprises the step of forming a pharmaceutically acceptable salt of a compound of formula I. In other embodiments, the invention is directed to the other processes described herein; and to the products prepared by any of the processes described herein.
The invention is also directed to a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof, for use in therapy or as a medicament. Additionally, the invention is directed to the use of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof, for the manufacture of a medicament; especially for the manufacture of a medicament for the treatment of a pulmonary disorder or for antagonizing a muscarinic receptor in a mammal. DETAILED DESCRIPTION OF THE INVENTION
In one embodiment, the invention is directed to novel biphenyl compounds of formula I, and pharmaceutically acceptable salts or solvates or stereoisomers thereof. The compounds may contain one or more chiral centers and therefore, the invention is also directed to racemic mixtures, pure stereoisomers (i.e., enantiomers or diastereomers), stereoisomer-enriched mixtures, and the like unless otherwise indicated. When a particular stereoisomer is shown or named herein, it will be understood by those skilled in the art that minor amounts of other stereoisomers may be present in the compositions of the invention unless otherwise indicated, provided that the desired utility of the composition as a whole is not eliminated by the presence of such other isomers. The compounds of formula I also contain several basic groups (e.g., amino groups) and therefore, the compounds of formula I can exist as the free base or in various salt forms. AU such salt forms are included within the scope of the invention. Furthermore, solvates of compounds of formula I or salts thereof are included within the scope of the invention. Additionally, where applicable, all cis-trans or E/Z isomers (geometric isomers), tautomeric forms and topoisomeric forms of the compounds of formula I are included within the scope of the invention unless otherwise specified.
The compounds of formula I, as well as those compounds used in its synthesis, may also include isotopically-labeled compounds, i.e., where one or more atoms have been enriched with atoms having an atomic mass different from the atomic mass predominately found in nature. Examples of isotopes that may be incorporated into the compounds of formula I include, but are not limited to, 2H, 3H5 13C, 14C, 15N, 18O and 17O.
The nomenclature used herein to name the compounds of the invention is illustrated in the Examples. This nomenclature has been derived using the commercially-available AutoNom software (MDL, San Leandro, California). For example, compounds of formula I wherein W is O have typically been named as ester derivatives of biphenyl-2-ylcarbamic acid.
Representative Embodiments The following substituents and values are intended to provide representative examples of various aspects and embodiments of the invention. These representative values are intended to further define and illustrate such aspects and embodiments and are not intended to exclude other embodiments or to limit the scope of the invention, hi this regard, the representation that a particular value or substituent is preferred is not intended in any way to exclude other values or substituents from the invention unless specifically indicated.
The values for a and b are independently 0, 1, 2, 3, 4 or 5; particularly independently 0, 1 or 2, and even more particularly 0 or 1. In one embodiment, both a and b are 0. When present, each R1 may be at the 2, 3, 4, 5 or 6-position of the phenyl ring to which it is attached. Each R1 is independently selected from -Ci-salkyl, -C2-5alkenyl,
-C2-5alkynyl, -C3-6cycloalkyl, -cyano, -halo, -ORla, -C(O)ORlb, -SRlc, -S(O)Rld, -S(O)2R16, -NRlfRlg, -NR111S(O)2R11, and -NRljC(O)Rlk, examples of which include methyl, fluoro, chloro, bromo, hydroxy, methoxy, amino, methylamino, dimethylamino and the like. Particular values for R1 are fluoro or chloro. When present, each R2 may be at the 3, 4, 5 or 6-position on the phenylene ring to which it is attached (where the carbon atom on the phenylene ring attached to the nitrogen atom is position 1). Each R2 is independently selected from -C1-5alkyl, -C2-salkenyl, -C2-5alkynyl, -C3-6CyClOaIkVl, -cyano, -halo, -OR2a, -C(O)OR2b, -SR2c, -S(O)R2d, -S(O)2R26, -NR2fR2g, -NR2hS(O)2R2i, and -NR2jC(O)R2k, examples of which include methyl, fluoro, chloro, bromo, hydroxy, methoxy, amino, methylamino, dimethylamino and the like. Particular values for R2 are fluoro or chloro. Each Rla, Rlb, Rlc, Rld, Rle, Rlf, Rlg, Rlh, R11, Rlj, and Rlk and R2a, R2b, R2c, R2d, R2e, R2f, R2g, R2h, R21, R2j, and R2k as used in R1 and R2, respectively, is independently -H, -Chalky! or -phenyl-Cμsalkyl, examples of which include hydrogen, methyl, ethyl, n-propyl, isopropyl, «-butyl, sec-butyl, isobutyl, tert-butyl or benzyl. In one embodiment, these groups are independently hydrogen or -C1-3alkyl. In another embodiment, these groups are independently hydrogen, methyl or ethyl. In addition, each alkyl and alkoxy group in R1, Rla'lk, R2, and R2a"2k is optionally substituted with 1 to 5 fluoro substituents. In one embodiment of the invention, W is -O-. Generally, it has been found that compounds in which W represents -O- exhibit particularly high affinity for muscarinic receptors, hi another embodiment W is -NWa-, where Wa is -H or -C1-4alkyl, examples of which include hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl. hi one embodiment, Wa is hydrogen or -C1-3alkyl. hi another embodiment, Wa is hydrogen, methyl or ethyl, particularly hydrogen or methyl, hi yet another embodiment, Wa is hydrogen and -NWa- is -NH-.
The value for c is 0, 1, 2, 3, 4, or 5; particularly 0, 1, or 2; and more particularly 0 or 1. hi one particular embodiment, c is 0. Each R3 independently represents -d^alkyl or two R3 groups that are joined to form C1-3alkylene, C2.3alkenylene or oxiran-2,3-diyl. hi one embodiment, each R3 is independently -Chalky!, such as methyl, ethyl, «-propyl, isopropyl, o-butyl, sec-butyl, isobutyl and tert-butyl. In addition, each alkyl group in R3 is optionally substituted with 1 to 5 fluoro substituents. hi one embodiment, each R3 is independently -C1-3alkyl, and in another embodiment, each R3 is independently methyl or ethyl. hi one embodiment, each R3 is at the 3, 4 or 5-position on the piperidine ring
(where the nitrogen atom of the piperidine ring is position 1). hi a particular embodiment, R3 is at the 4-position on the piperidine ring, hi another embodiment, R3 is at the 1-position of the piperidine ring, i.e., on the nitrogen atom of the piperidine ring thus forming a quaternary amine salt. hi yet another embodiment, two R3 groups are joined to form a C1-3alkylene or
C2-3alkenylene group. For example, two R3 groups at the 2 and 6-positions on the piperidine ring can be joined to form an ethylene bridge (i.e., the piperidine ring and the R3 groups form an 8-azabicyclo[3.2.1]octane ring); or two R3 groups at the 1 and 4-positions on the piperidine ring can be joined to form an ethylene bridge (i.e., the piperidine ring and the R3 groups form an l-azabicyclo[2.2.2]octane ring), hi this embodiment, other R3 groups as defined herein may also be present.
In still another embodiment, two R3 groups are joined to form a oxiran-2,3-diyl group. For example, two R3 groups at the 2 and 6-positions on the piperidine ring can be joined to form a 3-oxatricyclo[3.3.1.02>4]nonane ring). In this embodiment, other R3 groups as defined herein may also be present.
The value for m is 0 or 1. In one embodiment, m is 0. Z is selected from -C(O)N(R4)- and -N(R4)C(O)-. R4 is -H, -C1-5alkyl, or -C3-5CyClOaIkVl. Examples of -C1-5alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, -fee-butyl, isobutyl, tert-butyl, and pentyl. Examples of -C3-5cycloalkyl groups include cyclopropyl, cyclobutyl, and cyclopentyl. In one embodiment R4 is hydrogen or -C1-3alkyl, in particular hydrogen or methyl. Exemplary embodiments of Z include -NHC(O)-, -N(CH3)C(O)-, and -C(O)NH-. The value for s is 0, 1 or 2. One particular value for s is 0; in another embodiment, s is 1; and in yet another embodiment, the value for s is 2.
Ar is a phenylene group or a C3-5heteroarylene group containing 1 or 2 heteroatoms independently selected from oxygen, nitrogen or sulfur. The phenylene or heteroarylene group may be unsubstituted (q is 0) or substituted with 1, 2, 3, or 4 (q is 1, 2, 3, or 4) R5 substituents, which are independently selected from -halo, -OH, -C1-5alkyl or -C1-5alkoxy. In addition, each alkyl and alkoxy group in R5 is optionally substituted with 1 to 5 fluoro substituents. In one embodiment, q is 0, 1, 2 or 3; in another embodiment, q is 0, 1 or 2. In another embodiment, q is 1 and R5 is methoxy. The point of attachment for Ar is at any available carbon or heteroatom ring atom. In certain embodiments, Ar is a phenylene group attached at the meta or para position.
In one embodiment Ar is phen-l,3-ylene or phen-l,4-ylene wherein the phenylene group is unsubstituted or substituted with 1, 2 or 3 R5 substituents. Representative R5 substituents include fluoro, chloro, bromo, methyl, ethyl, «-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, isopropoxy, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl and trifluoromethoxy. Particular examples of Ar1 groups in this embodiment include 2-fluorophen-l,4-ylene, 3-fluorophen-l,4-ylene, 2- chlorophen-l,4-ylene, 3-chlorophen-l,4-ylene, 2-methylphen-l,4-ylene, 3-methylphen-l,4- ylene, 2-methoxyphen-l,4-ylene, 3-methoxyphen-l,4-ylene, 2-trifluoromethoxyphen-l,4- ylene, 3-trifluoromethoxyphen-l,4-ylene, 2,3-difluorophen-l,4-ylene, 2,5-difluorophen- 1,4-ylene, 2,6-difluorophen-l,4-ylene, 2,3-dichlorophen-l,4-ylene, 2,5-dichlorophen-l,4- ylene, 2,6-dichlorophen- 1,4-ylene, 2-chloro-5-methoxyphen- 1,4-ylene, 2-chloro-6- methoxyphen- 1 ,4-ylene, 2-chloro-5-trifluoromethoxyphen- 1 ,4-ylene, 2-chloro-6- trifluoromethoxyphen- 1,4-ylene, and 2,5-dibromophen-l,4-ylene. In another embodiment, Ar is a C3-5heteroarylene group containing 1 or 2 heteroatoms, and is unsubstituted or substituted with 1 or 2 R5 substituents. Representative C3-5heteroarylene groups include divalent species of pyrrole, imidazole, thiazole, oxazole, furan, thiophene, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine and pyrimidine, where the point of attachment is at any available carbon or nitrogen ring atom. More specific examples of such Ar groups include: pyridylene such as 2,5-pyridylene and 2,6-pyridylene; thienylene such as 2,4-thienylene and 2,5-thienylene; pyrrolylene such as 2,4-pyrrolylene and 2,5-pyrrolylene; and furylene such as 2,5-furylene. Representative R5 substituents include fluoro, chloro, methyl, ethyl, w-propyl, isopropyl, π-butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, isopropoxy, difluoromethyl, trifluoromethyl, 2,2,2- trifluoroethyl and trifluoromethoxy. Particular examples of substituted Ar groups include 3-fluoro-2,5-thienylene, 3-chloro-2,5-thienylene, 3-methyl-2,5-thienylene, 3-methoxy-2,5- thienylene, and 3-methoxy-6-chloro-2,5-pyridylene.
In one particular embodiment, Ar represents phen-l,3-ylene, phen-l,4-ylene, 2-methoxyphen-l,4-ylene, 2,5-pyridylene, 2,5-thienylene, 2,4-pyrrolylene, 2,5-pyrrolylene, or 2,5-furylene.
The value for t is 0, 1 or 2. A particular value for t is 1.
R6 is selected from -H, -C1-5alkyl, and -X6R6a, where X6 is -C1-5alkylene and R6a is selected from -OH, -C1-5alkoxy and -heteroaryl. In one embodiment, R6 is -H. hi another embodiment, R6 is -C1-5alkyl such as methyl or ethyl. In another embodiment, R6 is -X6R6a, where X6 is -C1-5alkylene such as -(CBb)2-, and R6a is -OH. hi yet another embodiment, R6 is -X6R6a, where X6 is -C1-5alkylene such as -(CH2)2-, and R6a is -Q-salkoxy such as -OCH3 or -OCH2CH3. In addition, each alkyl and alkoxy group in R6 and R a is optionally substituted with 1 to 5 fluoro substituents. R7 is selected from -H, -C1-5alkyl, -C3-6cycloalkyl, and -X7R7a. X7 is selected from
-C1-5alkylene, -C(O)-, -C1-5alkylene-C(O)-, -S(O2)-, -C1-5alkylene-S(O2)-, and -S(O2)-C1-5alkylene. R7a is selected from -OH, -C1-5alkyl, -C3-6CyClOaIkVl, -C1-5alkoxy, heteroaryl, -NR7bR7c (R7b and R7c are independently -H or -C1-5alkyl), and aryl (which may be substituted with 1 or 2 -C1-5alkyl or halo groups). In addition, each alkyl and alkoxy group in R7 and R7a'c is optionally substituted with 1 to 5 fluoro substituents. For example, R can be -CH2CH2CF3 or -S(O2)CF3. In one embodiment, R is -H. In one embodiment, R7 is -C1-5alkyl such as methyl, ethyl, isopropyl, isobutyl, and tert-butyl. hi another embodiment, R7 is -C3-6cycloalkyl such as cyclopropyl, cyclobutyl, or cyclopentyl. In yet another embodiment, R7 is -X7R7a, where X7 is -C1-5alkylene such as -CH2-, and R7a is: -C3-6cycloalkyl such as cyclopropyl; heteroaryl such as 2-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 5-benzo[l,3]dioxole, or 7-lH-indole. X7 can also be -C1-5alkylene such as -(CH2)2-, and R7a can be -OH, or -Ci^alkoxy such as -OCH3 and -OCH2CH3. R7 can also be -X7R7a, where X7 is -C(O)-, and R7a is -C1-5alkyl such as methyl, -C3-6cycloalkyl such as cyclopropyl or cyclobutyl, heteroaryl such as 2-furyl, 2-thienyl, or 4-pyridyl. In another embodiment, R7 is -X7R7a, where X7 is -C1-5alkylene-C(O)- such as -CH2C(O)-, and R7a is: -OH; or -NR7bR7c such as -NH2. R7 can also be -X7R7a, where X7 is -S(O2)-, and R7a is: -Ci-salkyl such as methyl, ethyl, w-propyl, isopropyl, and 7j-butyl; heteroaryl such as 2-thienyl and 8-quinolinyl; or aryl such as 4-trifluoromethylphenyl,
2-fluorophenyl, 2,6-dichlorophenyl, 4-methylphenyl, and phenyl. In another embodiment, R7 is -X7R7a, where X7 is -C1-5alkylene-S(O2)- such as -(CH2)2S(O2)-, and R7a is -C1-5alkyl such as methyl. In yet another embodiment, R7 is -X7R7a, where X7 is -S(O2)-C1-5alkylene such as -S(O2)(CH2)2-, and R7a is -NR7bR7c such as -NHCH3. One particular R6/R7 combination is where R6 is -H and R7 is -H, -C1-5alkyl (e.g., methyl, ethyl, isopropyl, isobutyl, or tert-butyϊ), or -C3-6CyClOaUCyI (e.g., cyclopropyl, cyclobutyl, or cyclopentyl). In another combination, R6 is -H and R7 is -X7R7a, where X7 is -C1-5alkylene (e.g., -CH2-), and R7a is -C3-6cycloalkyl (e.g., cyclopropyl) or heteroaryl (e.g., 2-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 5-benzo[l,3]dioxole, or 7-lH-indole). X7 may also be -Q.salkylene such as -(CH2)2-, and R7a is -OH or -C1-5alkoxy such as
-OCH3. Another particular R6/R7 combination is where R6 is -H, and R7 is -X7R7a, where X7 is -C(O)-, and R7a is: -Q-salkyl such as methyl; -Cs-όCycloalkyl such as cyclopropyl or cyclobutyl; or heteroaryl such as 2-furyl, 2-thienyl, or 4-pyridyl.
Another particular R /R combination is where R is -H, and R is -X R , where X is -C1-5alkylene-C(O)- such as -CH2C(O)-, and R7a is -NR7bR7c such as -NH2. Another particular R6/R7 combination is where R6 is -H, and R7 is -X7R7a, where X7 is -S(O2)-, and R7a is -Q.salkyl (e.g., methyl, ethyl, ra-propyl, isopropyl, or n-butyl), heteroaryl (e.g., thiophen-2-yl or 8-quinolinyl), or aryl (e.g., 4-trifluoromethylphenyl, 2-fluorophenyl, 2,6- dichlorophenyl, 4-methylphenyl, or phenyl). Another particular R6/R7 combination is where R6 is -H, and R7 is -X7R7a, where X7 is -S(O2)-C1-5alkylene such as -S(O2)(CH2)2-, and R7a is -NR7bR7c such as -NHCH3.
Another particular R6ZR7 combination is where R6 is -C1-5alkyl such as methyl, and R7 is: -X7R7a, where X7 is -C1-5alkylene such as -(CH2)2-, and R7a is -OH; -X7R7a, where X7 is -C^alkylene such as -CH2-, and R7a is heteroaryl such as 3-pyridyl, 4-pyridyl, or 2-furyl; X7R7a, where X7 is -C1-5alkylene-C(O)- such as -CH2C(O)-, and R7a is -OH; or X7R7a, where X7 is -C1-5alkylene-S(O2)- such as -(CH2)2S(O2)-, and R7a is -C1-5alkyl such as methyl. Another particular R6/R7 combination is where R6 and R7 are different -C1-5alkyl groups. For example, R6 can be ethyl, and R7 can be methyl or isopropyl.
Another particular R6ZR7 combination is where R6 and R7 are the same. R6 and R7 can both be a -C1-5alkyl group such as methyl and ethyl. R6 and R7 can also both be a -C^salkylene-d-salkoxy group (X6 and X7 are -Q.salkylene, R6a and R7a are -C1-5alkoxy), examples of which include -(CH2)2OCH3 and -(CH2)2OCH2CH3. R6 and R7 can also both be a -C1-5alkylene-OH group (X6 and X7 are -C1-5alkylene, R6a and R7a are -OH), an example of which includes -(CH2)2OH.
Alternately, R6 and R7 may be taken together to form a thiomorpholine 1,1 -dioxide ring:
Figure imgf000012_0001
R6 and R7 may also be taken together to form morpholin-4-yl:
— N O
which may be substituted with one, two or three -C1-5alkyl groups. In one embodiment, the morpholine ring is substituted with two -C1-5alkyl groups such as methyl, for example 2,6-dimethylmorpholin-4-yl. In addition, R6 and R7 may also be taken together to form:
Figure imgf000012_0002
The value for d is 1 or 2. In one embodiment, d is 1. The value for e is 0, 1 or 2. In one embodiment, e is 0 and in another embodiment e is 1. In one embodiment, d is 1 and e is 0 or 1. The value for f is 0, 1, 2 or 3. In one embodiment, f is 0. R8 is selected from -C1-5alkyl and =0. In one embodiment, R8 is =0. In addition, each alkyl group in R8 is optionally substituted with 1 to 5 fluoro substituents.
R9 is selected from -H, -C1-5alkyl, hydroxyphenyl, heteroaryl, and -X9R9a. X9 is selected from -C1-5alkylene, -C(O)-, -C1-5alkylene-C(O)-, -C(O)-C 1-5alkylene, -S(O2)-, -C1-5alkylene-S(O2)-, and -S(O2)-C1-5alkylene. R9a is selected from -H, -OH, -C1-5alkyl, -C1-5alkoxy, aryl, heteroaryl, heterocyclyl, and -NR9bR9c (R9b and R9c are independently -H or -C^aUcyl). In addition, each alkyl and alkoxy group in R9 and R9a"c is optionally substituted with 1 to 5 fluoro substituents.
In one embodiment, R9 is -H or hydroxyphenyl. R9 can also be -C1-5alkyl such as ethyl and isopropyl. In another embodiment, R9 is heteroaryl such as 4-pyridyl. In another embodiment, R9 is -X9R9a, where X9 is -C1-5alkylene such as -CH2-, and R9a is heteroaryl such as 4-pyridyl, or where X9 is -Q-salkylene such as -(CH2)2-, and R9a is -C1-SaIkOXy such as -OCH3. R9 can also be -X9R9a, where X9 is -C(O)-, and R9a is: -H; -C1-5alkyl such as methyl; -C1-5alkoxy such as -OCH3; heterocyclyl such as 1-pyrrolidinyl, 1-piperidyl, and 4-morpholinyl; or -NR9bR9c such as -N(CH3)2. hi another embodiment, R9 is -X9R9a, where X9 is -C1-5alkylene-C(O)- such as -CH2C(O)-, and R9a is heterocyclyl such as 1- pyrrolidinyl, or -NR9bR9° such as -N(CH3)2. R9 can also be -X9R9a, where X9 is -S(O2)-, and R9a is -C1-5alkyl such as methyl and ethyl. hi one embodiment, d is 1, e is O, and f is 1, i.e., R6 and R7 are taken together to form a piperidine ring. Particular embodiments of the piperidine ring include those where R is =0 and R is -H, for example, a piperazin-2-one ring.
In one embodiment, d is 1, and e and f are O, i.e., R6 and R7 are taken together to form a piperidine ring. Particular embodiments of the piperidine ring include those where R9 is: -Q-salkyl such as ethyl and isopropyl; hydroxyphenyl; heteroaryl such as 4-pyridyl; -X9R9a, where X9 is -C1-5alkylene such as -CH2-, and R9a is heteroaryl such as 4-pyridyl; -X9R9a, where X9 is -C1-5alkylene such as -(CH2)2-, and R9a is -C1-5alkoxy such as -OCH3; -X9R9a, where X9 is -C(O)-, and R9a is -H, -C1-5alkyl such as methyl, -C1-5alkoxy such as -OCH3, heterocyclyl such as 1-pyrrolidinyl, 1-piperidyl, and 4-morpholinyl, or -NR9bR9c such as -N(CH3)2; -X9R9a, where X9 is -C1-5alkylene-C(O)- such as -CH2C(O)-, and R9a is heterocyclyl such as 1-ρyrrolidinyl, or -NR9bR9c such as -N(CH3)2; and -X9R9a, where X9 is -S(O2)-, and R9a is -C1-5alkyl such as methyl and ethyl. hi one embodiment, d is 2, and e and f are O, i.e., R6 and R7 are taken together to form a [l,4]diazepane ring. Particular embodiments of the [l,4]diazepane ring include those where R9 is -X9R9a, where X9 is -C(O)-, and R9a is -C1-5alkyl such as methyl, hi another embodiment, d and e are 1, and f is O, i.e., R6 and R7 are taken together to form a 2,5-diaza-bicyclo[2.2.1]heptane ring. Particular embodiments ofthe 2,5-diaza- bicyclo[2.2.1]heptane ring include those where R9 is: -H; or -X9R9a, where X9 is -C1-5alkylene such as -CH2-, and R9a is aryl such as phenyl.
A particular group of compounds of interest are compounds of formula I wherein a, b, and c are 0. Another group of compounds of interest are compounds of formula I wherein W is -O-. A particular group of compounds of interest are compounds of formula I where m is 0 and t is 1. A particular group of compounds of interest are compounds of formula I where Z is -NHC(O)-, -N(CH3)C(O)-, or -C(O)NH-. Other compounds of interest include those where Ar is phenylene, phenylene substituted with methoxy, pyridylene, thienylene, pyrrolylene, or furylene, and in particular where Ar is phen-1,3- ylene, phen-l,4-ylene, 2-methoxyphen-l,4-ylene, 2,5-pyridylene, 2,5-thienylene, 2,4-pyrrolylene, 2,5-pyrrolylene, or 2,5-furylene.
Combinations of the foregoing are also of interest. For example, one group of compounds of interest are compounds of formula I wherein m is 0, t is 1, and W is -O-. These compounds can also be depicted by formula Ia:
Figure imgf000014_0001
Ia where a, b, c, s, R1"3, R6"7, Z, and Ar3 are as defined for formula I. Another example of a group of compounds of interest are compounds of formula Ia wherein a, b, and c are 0; Z is selected from -NHC(O)-, -N(CH3)C(O)-, and -C(O)NH-; and Ar is selected from phenylene, phenylene substituted with methoxy, pyridylene, thienylene, pyrrolylene, and furylene.
In addition, particular compounds of formula I that are of interest include: biphenyl-2-yl-carbamic acid l-(2-{[4-(4-acetylpiperazin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(4-aminomethylbenzoyl)methylamino]ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4- {[(furan-2-carbonyl)amino]methyl}benzoyl) methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[4-(acetylaminomethyl)benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl-(4- { [(pyridine-4-carbonyl)amino]methyl} benzoyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(cyclopropanecarbonylamino)methyl] benzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl-(4-{[(thiophene-2-carbonyl)amino] methyl}benzoyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(cyclobutanecarbonylamino)methyl]benzoyl} methylamino)ethyl]piperidm-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(isopropylaminomethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(cyclopropylmethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl-(4-morpholin-4-ybnethylbenzoyl)amino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- {[(2-methanesulfonylethyl)methylamino] methyl}benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-ethylpiperazin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-isopropylpiperazin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(ethylisopropylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(2,6-dimethylmorpholin-4-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4- {[bis-(2-hydroxyethyl)amino]methyl} benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {[4-(tert-butylaminomethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(ethyhnethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- {[bis-(2-ethoxyethyl)amino]methyl}benzoyl) methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl-{4-[(methylpyridin-3-ylmethylamino) methyl]benzoyl} ammo)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4- {[bis(2-methoxyethyl)amino]methyl} benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4- {[(2-hydroxyethyl)methylamino]methyl} benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl- {4-[(methyl-pyridin-4-ylmetliylamino) methyljbenzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[4-(2-methoxyethyl)piperazin-l-ylmethyl] benzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-ethanesulfonylpiperazin-l-ylmethyl) benzoyljmethylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(2,5-diaza-bicyclo[2.2.1]hept-2-ylmethyl) benzoyljmethylamino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(2-hydroxyethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {methyl- [4-(4-pyridin-4-ylpiperazin-l-ylmethyl) benzoyl]amino}ethyl)piperidm-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-dimethylcarbamoylmethylpiperazin-l- ylmethyl)benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {methyl-[4-(4-pyridin-4-ylmethylpiperazin- 1 - ylmethyl)benzoyl]amino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl- {4-[4-(2-oxo-2-pyrrolidin-l-ylethyl) piperazin- 1 -ylmethyl]benzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(2-hydroxyethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4-ethylaminomethylbenzoyl)methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(4-cyclopropylaminomethylbenzoyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(2-methoxyethylamino)methyl] benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {methyl-[4-(3-oxopiperazin- 1 -ylmethyl)benzoyl] amino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {[4-(4-acetyl-[ 1 ,4]diazepan- 1 -ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[4-(4-hydroxyphenyl)piperazin-l-ylmethyl] benzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(carbamoylmethylamino)inethyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-dimethylcarbamoylρiperazin- 1 -ylmethyl) benzoyl]methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(methyl- {4-[4-(pyrrolidine- 1 -carbonyl)piperazin- 1 -ylmethyl]benzoyl} amino)ethyl]ρiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(methyl- {4-[4-(piperidine- 1 -carbonyl)piperazin- 1 - ylmethyl]benzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl-{4-[4-(moφholine-4-carbonyl)piperazin-
1 -ylmethyl]benzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4-ammomethylbenzoylamino)ethyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 - {2-[3-((lR,4R)-5-benzyl-2,5-diaza-bicyclo[2.2.1 ] hept-2-ylmethyl)benzylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-aminomethylbenzylcarbamoyl)ethyl]piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{[(pyridm-4-ylmethyl)amino]methyl} benzylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[(ruran-2-ylmethyhnethylamino)methyl] benzylcarbamoyl} ethyl)piperidin-4-yl ester;
{[3-({3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionylamino}methyl) benzyl]methylamino} acetic acid; biphenyl-2-ylcarbamic acid l-[2-(3-cyclobutylaminomethylbenzylcarbamoyl) ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3-cyclopropylaminomethylbenzylcarbamoyl) ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3-morpholin-4-ylmethylbenzylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-methylaminomethylbenzylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3-dimethylaminomethylbenzylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {methyl-[3-(4-methylaminomethylphenyl) propionyl] amino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[3-(4-ethylaminomethylphenyl)propionyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-aminomethylphenylcarbamoyl)ethyl]piperidin-
4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(4-trifluoromethylbenzenesulfonylamino) methyljphenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(butane-l-sulfonylamino)methyl] phenylcarbamoyl}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(2-fluorobenzenesulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {4-[(quinoline-8-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[4-(ethanesulfonylaminomethyl) phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(2,6-dichlorobenzenesulfonylamino)methyl] phenylcarbamoyl} ethyl)piρeridin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(toluene-4-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {4-[(thiophene-2-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[4-(benzenesulfonylaminomethyl) phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[4-(methanesulfonylaminomethyl) phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[4-(trifluoromethanesulfonylaminomethyl) phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(propane-l-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(propane-2-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(pyridin-2-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(pyridin-3-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(pyridin-4-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(benzo[ 1 ,3]dioxol-5-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4- {[(lH-indol-7-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[4-(isobutylaminomethyl)phenylcarbamoyl] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4- {[(thiophen-2-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl]ρiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(3,3,3-trifluoropropylamino)methyl] phenylcarbamoyl}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(2-methylaminoethanesulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[3-(4-methylaminomethylphenyl)propionylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(4-ethylaminomethylphenyl)propionylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {3-[4-(isopropylaminomethyl)phenyl] propionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(4-cyclopropylaminomethylphenyl) propionylamino] ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3- {4-[(cyclopropylmethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(4-cyclopentylaminomethylphenyl) propionylamino] ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{4-[(2-hydroxyethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {4-[(2-methoxyethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-acetyl-[l,4]diazepan-l-ylmethyl)phenyl] propionylamino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-acetylpiperazin-l-ylmethyl)phenyl] propionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{4-[(carbamoylmethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-dimethylcarbamoylmethylpiρerazin-l- ylmethyl)phenyl]propionylamino} ethyl)piperidin-4-yl ester;
4-[4-(2-{2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethylcarbamoyl}ethyl) benzyljpiperazine- 1 -carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(l,l-dioxo-lλ6-thiomorpholin-4-ylmethyl) phenyl]propionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[3-(4- {[bis-(2-hydroxyethyl)amino]methyl} phenyl)propionylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-dimethylcarbamoylpiperazin-l-ylmethyl) phenyljpropionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{4-[4-(pyrrolidine-l-carbonyl)piperazin-l- ylmethyl]phenyl}propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{4-[4-(piperidine-l-carbonyl)piperazin-l- ylmethyl]phenyl}propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - [2-(3 - {4- [4-(morpholine-4-carbonyl)piperazin- 1 - ylmethyl]phenyl}propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[2-(4-methylaminomethylphenyl)acetylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[2-(4-ethylaminomethylphenyl)acetylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2- {4-[(2-hydroxyethylamino)methyl]phenyl} acetylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2- {4-[(2-methoxyethylamino)methyl]phenyl} acetylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2- {4-[(carbamoylmethylamino)methyl]phenyl} acetylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{2-[4-(4-dimethylcarbamoylpiperazin-l- ylmethyl)phenyl]acetylamino}etliyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(2-methoxy-4-methylaminomethylbenzoyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[2-methoxy-4-(3-oxopiperazin-l-ylmethyl) benzoyl]methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(2-hydroxyethylamino)methyl]-2- methoxybenzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2-methoxy-4-methylaminomethylbenzoylamino) ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[2-methoxy-4-(3-oxopiperazin-l-ylmethyl) benzoylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(2-hydroxyethylamino)methyl]-2- methoxybenzoylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl(5-methylaminomethylpyridine-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5-diethylaminomethylpyridine-2-carbonyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {5-[(2-hydroxyethylammo)methyl]pyridine-2- carbonyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl(5-morpholm-4-ylmethylpyridine-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- {[(furan-2-ylmethyl)amino]methyl}pyridine- 2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[5-(4-formylpiperazin- 1 -ylmethyl)pyridine-2- carbonyl]methylammo}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(5-{[bis-(2-hydroxyethyl)amino]methyl} pyridine-2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{methyl-[5-(3-oxo-piperazin-l-ylmethyl)pyridine- 2-carbonyl]amino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetyl-[l,4]diazepan-l-ylmethyl)pyridine-
2-carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {[5-(4-acetylpiperazin-l-ylmethyl)pyridine-2- carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbaniic acid 1 -(2- { [5-(4-methanesulfonylpiperazin- 1 -ylmethyl) pyridine-2-carbonyl]methylamino} ethyl)piperidin-4-yl ester;
4-[6-({2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}methylcarbamoyl) pyridin-3-ylmethyl]piperazine-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl-(5-memylaminomethylthiophene-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5-diethylaminomethylthiophene-2-carbonyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {5-[(2-hydroxyethylamino)methyl]thiophene-2- carbonyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl-(5-morpholin-4-ylmethylthiophene-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(5- {[(furan-2-ylmethyl)amino]methyl}thiophene- 2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [5-(4-formylpiperazin- 1 -ylmethyl)thiophene-2- carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(5-{[bis-(2-hydroxy-ethyl)amino]methyl} thiophene-2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-yl-carbamic acid l-(2-{methyl-[5-(3-oxopiperazin-l-ylmethyl) thiophene-2-carbonyl]amino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetyl-[l,4]diazepan-l-ylmethyl)thiophene- 2-carbonyl]methylamino} ethyl)piperidin-4-yl ester;
4-[5-({2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}methylcarbamoyl) thiophen-2-ylmethyl]piperazine-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(l,l-dioxo-lλ6-thiomorpholin-4-ylmethyl) thiophene-2-carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[5-(4-acetylpiperazin-l -ylmethyl)thiophene-2- carbonyljmethylamino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [5-(4-methanesulfonylpiperazin- 1 -ylmethyl) thiophene-2-carbonyl]methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5-methylaminomethylthiophene-2-carbonyl) amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]thiophene-2- carbonyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl(4-memylaminomethyl-lH-pyrrole-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl-(5-methylaminomethyl-lH-pyrrole-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetylpiperazin-l-ylmethyl)-lH-pyrrole-2- carbonyl]methylamino} ethyl) piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]-lH-pyrrole-2- carbonyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl(5-methylaminomethylfuran-2-carbonyl) amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(5-diethylaminomethylfuran-2-carbonyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]furan-2- carbonyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[methyl(5-morpholin-4-ylmethylfuran-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- { [(furan-2-ymiethyl)amino]methyl} furan-2- carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-formylpiperazin-l-ylmethyl)furan-2- carbonyljmethylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- {[bis-(2-hydroxyethyl)amino]methyl} furan-2- carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-yl-carbamic acid l-(2-{methyl-[5-(3-oxopiperazin-l-ylmethyl)furan-2- carbonyl]amino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetyl[l,4]diazepan-l-ylmethyl)furan-2- carbonyl]ethylamino} ethyl)piperidin-4-yl ester;
4-[5-({2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}methylcarbamoyl) furan-2-yhnethyl]piperazine-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(l,l-dioxo-lλδ-thiomorpholin-4-yhnethyl) furan-2-carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {[5-(4-acetylpiperazin-l-ylmethyl)furan-2- carbonyl]methylamino} ethyl) piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-methanesulfonylpiperazin-l-ylmethyl) furan-2-carbonyl]niethylamino} ethyl)piperidin-4-yl ester; biphenyl-2-yl-carbamic acid l-{2-[(5-aminomethylfuran-2-carbonyl)amino]ethyl} piperidin-4-yl ester; biphenyl-2-yl-carbamic acid l-{2-[(5-methylaminomethylruran-2-carbonyl)amino] ethyl}piperidin-4-yl ester; and biphenyl-2-ylcarbamic acid 1 -[2-( {5-[(2-hydroxyethylamino)methyl]ruran-2- carbonyl} amino)ethyl]piperidin-4-yl ester; or a pharmaceutically acceptable salt or solvate thereof. Definitions
When describing the compounds, compositions, methods and processes of the invention, the following terms have the following meanings unless otherwise indicated.
The term "alkyl" means a monovalent saturated hydrocarbon group which may be linear or branched. Unless otherwise defined, such alkyl groups typically contain from 1 to 10 carbon atoms. Representative alkyl groups include, by way of example, methyl, ethyl, rø-propyl, isopropyl, π-butyl, sec-butyl, isobutyl, tert-bntyl, «-pentyl, rø-hexyl, n-heptyl, Tz-octyl, n-nonyl, n-decyl and the like.
The term "alkylene" means a divalent saturated hydrocarbon group which may be linear or branched. Unless otherwise defined, such alkylene groups typically contain from 1 to 10 carbon atoms. Representative alkylene groups include, by way of example, methylene, ethane- 1,2-diyl ("ethylene"), propane- 1,2-diyl, propane- 1, 3 -diyl, butane- 1,4- diyl, pentane-l,5-diyl and the like.
The term "alkoxy" means a monovalent group of the formula (alkyl)-O-, where alkyl is as defined herein. Representative alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, isobutoxy, tert-butoxy and the like.
The term "alkenyl" means a monovalent unsaturated hydrocarbon group which may be linear or branched and which has at least one, and typically 1, 2 or 3, carbon-carbon double bonds. Unless otherwise defined, such alkenyl groups typically contain from 2 to 10 carbon atoms. Representative alkenyl groups include, by way of example, ethenyl, n-propenyl, isopropenyl, «-but-2-enyl, «-hex-3-enyl and the like. The term "alkenylene" means a divalent alkenyl group.
The term "alkynyl" means a monovalent unsaturated hydrocarbon group which may be linear or branched and which has at least one, and typically 1, 2 or 3, carbon- carbon triple bonds. Unless otherwise defined, such alkynyl groups typically contain from 2 to 10 carbon atoms. Representative alkynyl groups include, by way of example, ethynyl, 7?-propynyl, w-but-2-ynyl, n-hex-3-ynyl and the like. The term "alkynylene" means a divalent alkynyl group.
The term "aryl" means a monovalent aromatic hydrocarbon having a single ring (i.e., phenyl) or fused rings (i.e., naphthalene). Unless otherwise defined, such aryl groups typically contain from 6 to 10 carbon ring atoms. Representative aryl groups include, by way of example, phenyl and naphthalene- 1-yl, naphthalene-2-yl, and the like. The term "arylene" means a divalent aryl group.
The term "cycloalkyl" means a monovalent saturated carbocyclic hydrocarbon group. Unless otherwise defined, such cycloalkyl groups typically contain from 3 to 10 carbon atoms. Representative cycloalkyl groups include, by way of example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. The term "cycloalkylene" means a divalent cycloalkyl group.
The term "halo" means fluoro, chloro, bromo and iodo.
The term "heteroaryl" means a monovalent aromatic group having a single ring or two fused rings and containing in the ring at least one heteroatom (typically 1 to 3 heteroatoms) selected from nitrogen, oxygen or sulfur. Unless otherwise defined, such heteroaryl groups typically contain from 5 to 10 total ring atoms. Representative heteroaryl groups include, byway of example, monovalent species of pyrrole, imidazole, thiazole, oxazole, furan, thiophene, triazole, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine, pyrimidine, triazine, indole, benzofuran, benzothiophene, benzimidazole, benzthiazole, quinoline, isoquinoline, quinazoline, quinoxaline and the like, where the point of attachment is at any available carbon or nitrogen ring atom. The term "heteroarylene" means a divalent heteroaryl group.
The term "heterocyclyl" or "heterocyclic" means a monovalent saturated or unsaturated (non-aromatic) group having a single ring or multiple condensed rings and containing in the ring at least one heteroatom (typically 1 to 3 heteroatoms) selected from nitrogen, oxygen or sulfur. Unless otherwise defined, such heterocyclic groups typically contain from 2 to 9 total ring carbon atoms. Representative heterocyclic groups include, byway of example, monovalent species of pyrrolidine, imidazolidine, pyrazolidine, piperidine, 1,4-dioxane, morpholine, thiomorpholine, piperazine, 3-pyrroline and the like, where the point of attachment is at any available carbon or nitrogen ring atom. The term "heterocyclene" means a divalent heterocyclyl or heterocyclic group.
When a specific number of carbon atoms is intended for a particular term used herein, the number of carbon atoms is shown preceding the term. For example, the term "(l-5C)alkyl" or "C^alkyl" means an alkyl group having from 1 to 5 carbon atoms.
The term "pharmaceutically acceptable salt" means a salt which is acceptable for administration to a patient, such as a mammal (e.g., salts having acceptable mammalian safety for a given dosage regime). Such salts can be derived from pharmaceutically acceptable inorganic or organic bases and from pharmaceutically acceptable inorganic or organic acids. Salts derived from pharmaceutically acceptable inorganic bases include ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like. Particularly preferred are ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from pharmaceutically acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, iV.N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, 7V-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperadine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like. Salts derived from pharmaceutically acceptable acids include acetic, ascorbic, benzenesulfonic, benzoic, camphosulfonic, citric, ethanesulfonic, edisylic, fumaric, gentisic, gluconic, glucuronic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, lactobionic, maleic, malic, mandelic, methanesulfonic, mucic, naphthalenesulfonic, naphthalene- 1, 5 -disulfonic, naphthalene-2,6-disulfonic, nicotinic, nitric, orotic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, xinafoic and the like. Particularly preferred are citric, hydrobromic, hydrochloric, isethionic, maleic, naphthalene- 1,5-disulfonic, phosphoric, sulfuric and tartaric acids.
The term "salt thereof means a compound formed when the hydrogen of an acid is replaced by a cation, such as a metal cation or an organic cation and the like. Preferably, the salt is a pharmaceutically acceptable salt, although this is not required for salts of intermediate compounds that are not intended for administration to a patient.
The term "solvate" means a complex or aggregate formed by one or more molecules of a solute, i.e. a compound of formula I or a pharmaceutically acceptable salt thereof, and one or more molecules of a solvent. Such solvates are typically crystalline solids having a substantially fixed molar ratio of solute and solvent. Representative solvents include, by way of example, water, methanol, ethanol, isopropanol, acetic acid and the like. When the solvent is water, the solvate formed is a hydrate.
It will be appreciated that the term "or a pharmaceutically acceptable salt or solvate or stereoisomer thereof is intended to include all permutations of salts, solvates and stereoisomers, such as a solvate of a pharmaceutically acceptable salt of a stereoisomer of a compound of formula I. The term "therapeutically effective amount" means an amount sufficient to effect treatment when administered to a patient in need of treatment. For example, a therapeutically effective amount for antagonizing a muscarinic receptor is that amount which will achieve the desired antagonizing effect. Similarly, a therapeutically effective amount for treating a pulmonary disorder is the amount that will achieve the desired therapeutic result, which may be disease prevention, amelioration, suppression or alleviation, as described below.
The term "treating" or "treatment" as used herein means the treating or treatment of a disease or medical condition (such as COPD) in a patient, such as a mammal (particularly a human) that includes: (a) preventing the disease or medical condition from occurring, i.e., prophylactic treatment of a patient; (b) ameliorating the disease or medical condition, i.e., eliminating or causing regression of the disease or medical condition in a patient; (c) suppressing the disease or medical condition, i.e., slowing or arresting the development of the disease or medical condition in a patient; or (d) alleviating the symptoms of the disease or medical condition in a patient. The term "leaving group" means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction. By way of example, representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; and acyloxy groups, such as acetoxy, trifluoroacetoxy and the like. General Synthetic Procedures
The biphenyl compounds of the invention can be prepared from readily available starting materials using the following general methods, the procedures set forth in the Examples, or by using other methods, reagents, and starting materials that are readily available to those of ordinary skill in the art. Although a particular embodiment of the present invention may be shown or described herein, those skilled in the art will recognize that all embodiments or aspects of the present invention can be prepared using the methods described herein or by using other methods, reagents and starting materials known to those skilled in the art. It will also be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. While the optimum reaction conditions may vary depending on the particular reactants or solvent used, such conditions can be readily determined by one skilled in the art by routine optimization procedures.
Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary or desired to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection of such functional groups are well-known in the art. Functional groups which may be protected include, by way of example, carboxylic acid groups, amino groups, hydroxyl groups, thiol groups, carbonyl groups and the like. Protecting groups other than those illustrated in the procedures described herein may be used, if desired. Representative protecting groups for carboxylic acids include esters, amides and hydrazides; for amino groups, carbamates and amides; for hydroxyl groups, ethers and esters; for thiol groups, thioethers and thioesters; for carbonyl groups, acetals and ketals; and the like. Exemplary protecting groups are listed below, and other groups as well as details of their introduction and removal, are described in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein. Amino-protecting groups are suitable for preventing undesired reactions at an amino group, and include, but are not limited to, tert-butoxycarbonyl (BOC), trityl (Tr), benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc), formyl, trimethylsilyl (TMS), fert-butyldimethylsilyl (TBS), and the like. Carboxy-protecting groups are suitable for preventing undesired reactions at a carboxy group, and include, but are not limited to, esters, such as methyl, ethyl, tert-butyl, benzyl (Bn),p-methoxybenzyl (PMB), 9- fluroenylmethyl (Fm), trimethylsilyl (TMS), tert-butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like. Hydroxyl-protecting groups are suitable for preventing undesirable reactions at a hydroxyl group, and include, but are not limited to, silyl groups including tri(l-6C)alkylsilyl groups, such as trimethylsilyl (TMS), triethylsilyl (TES), tert- butyldimethylsilyl (TBS) and the like; esters (acyl groups) including (l-6C)alkanoyl groups, such as formyl, acetyl and the like; arylmethyl groups, such as benzyl (Bn), p- methoxybenzyl (PMB), 9-fluorenylmethyl (Fm), diphenylmethyl (benzhydryl, DPM) and the like. Additionally, two hydroxyl groups can also be protected as an alkylidene group, such as prop-2-ylidine, formed, for example, by reaction with a ketone, such as acetone.
By way of illustration, the compounds of formula I can be prepared by a process comprising:
(a) reacting a compound of formula II:
Figure imgf000029_0001
II or a salt thereof, with a compound of formula III:
Figure imgf000029_0002
III wherein L represents a leaving group; or
(b) coupling a compound of formula IVa:
Figure imgf000029_0003
IVa or a reactive derivative thereof, with a compound of formula Va:
Figure imgf000029_0004
Va or coupling a compound of formula IVb:
Figure imgf000030_0001
IVb with a compound of formula Vb:
Figure imgf000030_0002
Vb or a reactive derivative thereof; or
(c) reacting a compound of formula VI:
Figure imgf000030_0003
VI wherein L2 represents a leaving group, with a compound of formula VII:
.R°
HN
V
VII or
(d) reacting a compound of formula II with a compound of formula VIII:
Figure imgf000030_0004
VIII in the presence of a reducing agent; or
(e) reacting a compound of formula IX:
Figure imgf000030_0005
IX with a compound of formula VII in the presence of a reducing agent; and then
(f) removing any protecting groups that may be present to provide a compound of formula I; and optionally, forming a pharmaceutically acceptable salt thereof.
Generally, if a salt of one of the starting materials is used in the processes described above, such as an acid addition salt, the salt is typically neutralized before or during the reaction process. This neutralization reaction is typically accomplished by contacting the salt with one molar equivalent of a base for each molar equivalent of acid addition salt.
In process (a), the reaction between the compounds of formula II and III, the leaving represented by L1 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate. The reaction is conveniently performed in the presence of a base, for example, a tertiary amine such as diisopropylethylamine. Convenient solvents include nitriles, such as acetonitrile. The reaction is conveniently conducted at a temperature in the range of from 0 0C to 100 °C.
Compounds of formula II are generally known in the art, or can be prepared by deprotecting a compound of formula X:
Figure imgf000031_0001
X wherein P1 represents an amino-protecting group, such as a benzyl group. Benzyl groups are conveniently removed by reduction, using a hydrogen or ammonium formate and a Group VIII metal catalyst, such as palladium. When W represents NWa, the hydrogenation is conveniently performed using Pearlman's catalyst (Pd(OH)2).
Compounds of formula X can be prepared by reacting an isocyanate compound of formula XI with a compound of formula XII:
Figure imgf000031_0002
XI XII
Compounds of formula III can be prepared starting from a corresponding compound in which L1 represents a hydroxyl group, for example, by reaction of a halogenating agent, such as thionyl chloride, to afford a compound of formula III in which L1 represents halo, such as chloro. Compounds in which L1 represents a hydroxyl group may be prepared, for example, by reacting a compound of formula Vb with an appropriate amino-substituted alcohol, such as 2-aminoethanol or 3-aminopropan-l-ol. In process (b), the term "reactive derivative" of compound IVa or Vb is intended to mean that the carboxylic acid is activated, for example, by forming an anhydride or carboxylic acid halide, such as a carboxylic acid chloride. Alternatively, the carboxylic acid can be activated using conventional carboxylic acid/amine coupling reagents, such carbodiimides, O-(7-azabenzotriazol-l-yl-iV,ΛζN',N' tetramethyluronium hexafluorophosphate (HATU) and the like. This reaction is conveniently performed under conventional amide bond-forming conditions. The process is conveniently conducted at a temperature in the range of from -10 °C to 100 °C.
Compounds of formula IVa can be prepared by reacting a compound of formula II with a compound of formula XIII: L3-CH2(CH2)mCH2COOP2
XIII wherein L3 represents a leaving group including, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate; and P2 represents a hydrogen atom or a carboxyl-protecting group, such as a (l-4C)alkyl group. If necessary, the carboxyl-protecting group P2, is then removed, for example, by hydrolysis under conventional conditions, such as by using lithium hydroxide.
Compounds of formula Va can be prepared by reacting a compound of formula VII with a compound of formula XIV:
Figure imgf000032_0001
XIV wherein P represents hydrogen or an amino-protecting group, such as fert-butoxycarbonyl, and L4 represents a leaving group including, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate; followed if necessary, by removing an amino-protecting group P3. Alternatively, such compounds can be prepared by reductive amination of a compound of formula XV:
Figure imgf000033_0001
XV using a compound of formula VII. The reducing agent may be, for example, hydrogen in the presence of a Group VDI metal catalyst, such as palladium, or a metal hydride reducing agent, such as a borohydride, including sodium triacetoxyborohydride. Convenient solvents include alcohols, such as methanol. The reaction is conveniently performed at a temperature in the range of from 0 0C to 100 °C.
Compounds of formula IVb can be prepared by reacting a compound of formula II with a compound of formula XVI: C(O)H(CH2)mCH2NR4P4
XVI wherein P4 represents hydrogen or an amino-protecting group, such as benzyl, in the presence of a reducing agent, such as sodium triacetoxyborohydride, followed if necessary by removing the amino-protecting group P4 by, for example, hydrogenation in the presence of palladium.
Compounds of formula Vb can be prepared by reacting a compound of formula VII with a compound of formula XVII:
Figure imgf000033_0002
XVII wherein P5 represents hydrogen or a carboxyl-protecting group, such as methyl or ethyl, and L represents a leaving group, followed if necessary by removing the carboxyl protecting group P5. Alternatively, such compounds can be prepared by reductive animation of a compound of formula XVIII with a compound of formula VII:
Figure imgf000033_0003
xvπi
The reducing agent may be, for example, hydrogen in the presence of a Group VIII metal catalyst, such as palladium, or a metal hydride reducing agent, such as a borohydride, including sodium triacetoxyborohydride. Convenient solvents include alcohols, such as methanol. The reaction is conveniently performed at a temperature in the range of from 0 °C to lOO °C. Referring to process (c), the leaving group represented by L2 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate. This reaction is conveniently performed in the presence of a base, for example, a tertiary amine such as dϋsopropylethylamine. Convenient solvents include nitriles, such as acetonitrile. The reaction is conveniently conducted at a temperature in the range of from 0 0C to 100 0C. The compounds of formula VI can be prepared by reacting a compound of formula IVa with a compound of formula XIX:
Figure imgf000034_0001
XIX or by reacting a compound of formula IVb with a compound of formula XX:
Figure imgf000034_0002
XX
The reaction is conveniently performed following, for example, the method of process (b) described herein. Compounds of formula VII are generally known or can be prepared from readily available starting materials using well-known synthetic methods.
In process (d), the reducing agent may be, for example, hydrogen in the presence of a Group VIII metal catalyst, such as palladium, or a metal hydride reducing agent, such as a borohydride, including sodium triacetoxyborohydride, optionally used in combination with a titanium tetraalkoxide, such as titanium tetraisopropoxide. Convenient solvents include alcohols, such as methanol and halogenated hydrocarbons, such as dichloromethane. The reaction is conveniently performed at a temperature in the range of from 0 0C to 100 0C.
The compound of formula VIII may be prepared by oxidizing a compound corresponding to formula III in which L1 represents a hydroxyl group. Such oxidation reactions can be conducted, for example, using sulfur dioxide pyridine complex in dimethylsulfoxide in the presence of a tertiary amine, such as diisopropylethylamine. In process (e), the reduction is preformed as described for process (d). Compounds of formula IX may be prepared by reacting a compound of formula rVb with a compound of formula XXI:
Figure imgf000035_0001
XXI in the presence of a carboxylic acid/amine coupling agent, such as l-(3- dimethylaminopropyl)-3-ethylcarbodiimide (EDC) and 1-hydroxybenzotriazole hydrate (HOBT) and the like.
As will be apparent to those skilled in the art, compounds of formula I prepared by any of steps (a) to (f) herein may be further derivatized to form other compounds of formula I using methods and reagents well-known in the art. By way of illustration, a compound of formula I may be reacted with bromine to afford a corresponding compound of formula I in which R2, for example, represents a bromo group. Additionally, a compound of formula I in which R4 represents a hydrogen atom may be alkylated to afford a corresponding compound of formula I in which R4 represents a (1-4C) alkyl group. Further details regarding specific reaction conditions and other procedures for preparing representative compounds of the invention or intermediates thereof are described in the Examples set forth below.
Pharmaceutical Compositions and Formulations
The compounds of the invention are typically administered to a patient in the form of a pharmaceutical composition or formulation. Such compositions may be administered by any acceptable route of administration including, but not limited to, inhaled, oral, nasal, topical (including transdermal) and parenteral modes of administration. It will be understood that any form of the compound, i.e., free base, pharmaceutically acceptable salt, solvate, etc., that is suitable for the particular mode of administration can be used in the compositions described herein.
Accordingly, in one embodiment, the invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. The pharmaceutical composition may contain other therapeutic and/or formulating agents if desired.
The compositions of the invention typically contain a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof, as the active agent. Typically, the composition will contain from about 0.01-95% by weight of the active agent; including, from about 0.01-30% by weight; such as from about 0.01-10% by weight of the active agent.
Any conventional carrier or excipient may be used in the compositions of the invention. The choice of a particular carrier or excipient, or combination of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease state. In this regard, the preparation of a suitable composition for a particular mode of administration is well within the scope of those skilled in the pharmaceutical arts. Additionally, the ingredients for such compositions are commercially available from, for example, Sigma, P.O. Box 14508, St. Louis, MO 63178. Byway of further illustration, conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar;) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; compressed propellant gases, such as chlorofluorocarbons and hydrofluorocarbons; and other non-toxic compatible substances employed in pharmaceutical compositions.
The compositions of the invention are typically prepared by thoroughly and intimately mixing or blending a compound of formula I with a pharmaceutically acceptable carrier and one or more optional ingredients. If necessary or desired, the resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills, canisters, cartridges, dispensers and the like using conventional procedures and equipment.
In one embodiment, the pharmaceutical compositions of the invention are suitable for inhaled administration. Suitable compositions for inhaled administration will typically be in the form of an aerosol or a powder, and are generally administered using well-known delivery devices, such as a nebulizer inhaler, a dry powder inhaler (DPI), a metered-dose inhaler (MDI) or similar delivery device.
In a specific embodiment of the invention, a pharmaceutical composition comprising the active agent is administered by inhalation using a nebulizer inhaler. Such nebulizer devices typically produce a stream of high velocity air that causes the composition comprising the active agent to spray as a mist that is carried into the patient's respiratory tract. Accordingly, when formulated for use in a nebulizer inhaler, the active agent is typically dissolved in a suitable carrier to form a solution. Alternatively, the active agent can be micronized and combined with a suitable carrier to form a suspension of micronized particles of respirable size, where micronized is typically defined as having about 90% or more of the particles with a diameter of less than about 10 μm. Suitable nebulizer devices are commercially available, for example, by PARI GmbH (Sternberg, German). Other nebulizer devices include Respimat (Boehringer Ingelheim) and those described, for example, in U.S. Patent No. 6,123,068 to Lloyd et al. and WO 97/12687 (Eicher et al.), the disclosures of which are incorporated herein by reference in their entirety. A representative pharmaceutical composition for use in a nebulizer inhaler comprises an isotonic aqueous solution comprising from about 0.05 μg/mL to about 10 mg/niL of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
In another specific embodiment of the invention, a pharmaceutical composition comprising the active agent is administered by inhalation using a DPI. Such DPIs typically administer the active agent as a free-flowing powder that is dispersed in a patient's air- stream during inspiration. In order to achieve a free flowing powder, the active agent is typically formulated with a suitable excipient such as lactose or starch. Micronization is a common method of reducing crystal size to that suitable for pulmonary delivery. Typically, the active agent is micronized and combined with a suitable carrier to form a suspension of micronized particles of respirable size, where "micronized particles" or "micronized form" means at least about 90% of the particles have a diameter of less than about 10 μm. Other methods of reducing particle size may also be used such as fine milling, chopping, crushing, grinding, milling, screening, trituration, pulverization, and so forth, as long as the desired particle size can be obtained. A representative pharmaceutical composition for use in a DPI comprises dry lactose having a particle size between about 1 μm and about 100 μm and micronized particles of a compound of formula I, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Such a dry powder formulation can be made, for example, by combining the lactose with the active agent and then dry blending the components. Alternatively, if desired, the active agent can be formulated without an excipient. The pharmaceutical composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device. Examples of DPI delivery devices include Diskhaler (GlaxoSmithKline, Research Triangle Park, NC) (see, e.g., U.S. Patent No. 5,035,237 to Newell et al.); Dislcus (GlaxoSmithKline) (see, e.g., U.S. Patent No. 6,378,519 to Davies et al.); Turbuhaler (AstraZeneca, Wilmington, DE) (see, e.g., U.S. Patent No. 4,524,769 to Wetterlin); Rotahaler (GlaxoSmithKline) (see, e.g., U.S. Patent No. 4,353,365 to Hallworth et al.); Handihaler (Boehringer Ingelheim); and those described in U.S. Patent Nos. 5,415,162 to Casper et al., 5,239,993 to Evans, and 5,715,810 to Armstrong et al., and references cited therein; the disclosures of which are incorporated herein by reference in their entirety.
In yet another specific embodiment of the invention, the pharmaceutical composition comprising the active agent is administered by inhalation using an MDI, which typically discharges a measured amount of the active agent or a pharmaceutically acceptable salt or solvate or stereoisomer thereof using compressed propellant gas. Accordingly, pharmaceutical compositions administered using an MDI typically comprise a solution or suspension of the active agent in a liquefied propellant. Any suitable liquefied propellant may be employed including chlorofluorocarbons, such as CCI3F, and hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and I51,1,2,3,3 ,3 -heptafluoro-n-propane, (HFA 227). Formulations containing HFAs are generally preferred due to concerns about chlorofluorocarbons affecting the ozone layer. Additional optional components of HFA formulations include co-solvents, such as ethanol or pentane, and surfactants, such as sorbitan trioleate, oleic acid, lecithin, and glycerin. See, for example, U.S. Patent No. 5,225,183 to Purewal et al., EP 0717987 A2 (Minnesota Mining and Manufacturing Company), and WO 92/22286 (Minnesota Mining and Manufacturing Company), the disclosures of which are incorporated herein by reference in their entirety. A representative pharmaceutical composition for use in an MDI comprises from about 0.01-5 % by weight of a compound of formula I, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof; from about 0-20 % by weight ethanol; and from about 0-5 % by weight surfactant; with the remainder being an HFA propellant. Such compositions are typically prepared by adding chilled or pressurized hydro fluoroalkane to a suitable container containing the active agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the active agent is micronized and then combined with the propellant. The formulation is then loaded into an aerosol canister, which forms a portion of a metered-dose inhaler device. Examples of MDI devices developed specifically for use with HFA propellants are described in U.S. Patent Nos. 6,006,745 to Marecki and 6,143,277 to Ashurst et al. Alternatively, a suspension formulation can be prepared by spray drying a coating of surfactant on micronized particles of the active agent. See, for example, WO 99/53901 (Glaxo Group Ltd.) and WO 00/61108 (Glaxo Group Ltd.). The disclosures of the aforementioned patents and publications are incorporated herein by reference in their entirety.
For additional examples of processes of preparing respirable particles, and formulations and devices suitable for inhalation dosing see U.S. Patent Nos. 6,268,533 to Gao et al., 5,983,956 to Trofast, 5,874,063 to Briggner et al., and 6,221,398 to Jakupovic et al.; and WO 99/55319 (Glaxo Group Ltd.) and WO 00/30614 (AstraZeneca AB); the disclosures of which are incorporated herein by reference in their entirety.
In another embodiment, the pharmaceutical compositions of the invention are suitable for oral administration. Suitable compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present invention as an active ingredient. When intended for oral administration in a solid dosage form, i.e., as capsules, tablets, pills and the like, the composition will typically comprise a compound of the invention as the active ingredient and one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate. The solid dosage forms may also comprise one or more of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and/or sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as cetyl alcohol and/or glycerol monostearate; absorbents, such as kaolin and/or bentonite clay; lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and/or mixtures thereof; coloring agents; and buffering agents. Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions of the invention. Examples of pharmaceutically acceptable antioxidants include: water- soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and metal-chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Coating agents for tablets, capsules, pills and like, include those used for enteric coatings, such as cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acid-methacrylic acid ester copolymers, cellulose acetate trimellitate (CAT), carboxymethyl ethyl cellulose (CMEC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and the like.
If desired, the compositions of the invention may also be formulated to provide slow or controlled release of the active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres.
In addition, the compositions of the invention may optionally contain opacifying agents and may be formulated so that they release the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. Liquid dosage forms typically comprise the active ingredient and an inert diluent, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (e.g., cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Suspensions, in addition to the active ingredient, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. When intended for oral administration, the compositions of the invention are preferably packaged in a unit dosage form. The term "unit dosage form" means a physically discrete unit suitable for dosing a patient, i.e., each unit containing a predetermined quantity of active agent calculated to produce the desired therapeutic effect either alone or in combination with one or more additional units. For example, such unit dosage forms may be capsules, tablets, pills, and the like.
The compounds of the invention can also be administered transdermally using known transdermal delivery systems and excipients. For example, a compound of the invention can be admixed with permeation enhancers, such as propylene glycol, polyethylene glycol monolaurate, azacycloalkan-2-ones and the like, and incorporated into a patch or similar delivery system. Additional excipients including gelling agents, emulsifiers and buffers, may be used in such transdermal compositions if desired.
The compounds of the invention can also be co-administered with other therapeutic agents. This combination therapy involves using a compound of the invention combined with one or more of these secondary agents, either formulated together (e.g., packaged together in a single formulation) or formulated separately (e.g., packaged as separate unit dosage forms). Methods of formulating multiple agents together in the same formulation or in separate unit dosage forms, are well known in the art. The secondary agents can be used in the form of pharmaceutically acceptable salts or solvates, and if appropriate, as optically pure stereoisomers.
The secondary therapeutic agent(s) can be selected from other bronchodilators (e.g., PDE3 inhibitors, adenosine 2b modulators and β2 adrenergic receptor agonists); antiinflammatory agents (e.g., steroidal anti-inflammatory agents, such as corticosteroids; nonsteroidal antiinflammatory agents (NSAIDs), and PDE4 inhibitors); other muscarinic receptor antagonists (i.e., antichlolinergic agents); antiinfective agents (e.g., Gram positive and Gram negative antibiotics or antivirals); antihistamines; protease inhibitors; and afferent blockers (e.g., D2 agonists and neurokinin modulators). One particular embodiment of the invention is directed to a composition comprising (a) a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof; and (b) a pharmaceutically acceptable carrier and a therapeutically effective amount of an agent selected from a steroidal anti-inflammatory agent such as a corticosteroid; a β2 adrenergic receptor agonist; a phosphodiesterase-4 inhibitor; or a combination thereof; wherein the compound of formula I and the agent are formulated together or separately. In another embodiment, (b) is a pharmaceutically acceptable carrier and a therapeutically effective amount of a β2 adrenergic receptor agonist and a steroidal anti-inflammatory agent. Representative β2 adrenergic receptor agonists include, but are not limited to, salmeterol, salbutamol, formoterol, salmefamol, fenoterol, terbutaline, albuterol, isoetharine, metaproterenol, bitolterol, pirbuterol, levalbuterol and the like, or pharmaceutically acceptable salts thereof. Other β2 adrenergic receptor agonists that can be used in combination with the compounds of the invention include, but are not limited to, 3- (4-{[6-({(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)-phenyl]ethyl}amino)- hexyl]oxy}butyl)benzenesulfonamide and 3-(-3-{[7-({(2R)-2-hydroxy-2-[4-hydroxy-3- (hydroxymethyl)phenyl] ethyl} -amino)heptyl]oxy} -propyl)benzenesulfonamide and related compounds disclosed in WO 02/066422 (Glaxo Group Ltd.); 3-[3-(4-{[6-([(2R)-2- hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl] ethyl} amino)hexyl]oxy} butyl)- phenyl]irnidazolidine-2,4-dione and related compounds disclosed in WO 02/070490 (Glaxo Group Ltd.); 3-(4-{[6-({(2R)-2-[3-(formylamino)-4-hydroxyρhenyl]-2- hydroxyethyl} amino)hexyl]oxy}butyl)-benzenesulfonamide, 3-(4- {[6-({(2S)-2-[3- (formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]oxy}butyl)- benzenesulfonamide, 3-(4-{[6-({(2R/S)-2-[3~(formylamino)-4-hydroxyphenyl]-2- hydroxyethyl} amino)hexyl]oxy}butyl)-benzenesulfonamide, JV-(teπN-butyl)-3-(4- {[6- ( {(2R)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl} amino)hexyl]-oxy} butyl) benzenesulfonamide, iV-(tert-butyl)-3-(4-{[6-({(2S)-2-[3-(formylamino)-4- hydroxyphenyl]-2-hydroxyethyl}amino)-hexyl]oxy}butyl)-benzenesulfonamide, iV-(tert- butyl)-3-(4-{[6-({(2R/S)-2-[3-(formylamino)-4-hydroxyρhenyl]-2-hydroxyethyl} amino) hexyl]-oxy}butyl)benzenesulfonamide and related compounds disclosed in WO 02/076933 (Glaxo Group Ltd.); 4-{(lR)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-l- hydroxyethyl}-2-(hydroxymethyl)phenol and related compounds disclosed in WO 03/024439 (Glaxo Group Ltd.); JV-{2-[4-((i?)-2-hydroxy-2-phenylethylamino)phenyl] ethyl} -(i?)-2-hydroxy-2-(3-formamido-4-hydroxyphenyl)ethylamme and related compounds disclosed in U.S. Patent No. 6,576,793 to Moran et al.; iV-{2-[4-(3-phenyl-4- methoxyph.enyl)aminophenyl]ethyl}-(i?)-2-hydroxy-2-(8-hydroxy-2(lH)-quinolinon-5- yl)ethylamine and related compounds disclosed in U.S. Patent No. 6,653,323 to Moran et al.; and pharmaceutically acceptable salts thereof, hi a particular embodiment, the β2- adrenoreceptor agonist is a crystalline monohydrochloride salt of N- {2-[4-((JR)-2-hydroxy- 2-phenylethylarnino)phenyl] ethyl} -(R)-2-hydroxy-2-(3 -formamido-4-hydroxyphenyl) ethylamine. When employed, the β2-adrenoreceptor agonist will be present in the pharmaceutical composition in a therapeutically effective amount. Typically, the β2- adrenoreceptor agonist will be present in an amount sufficient to provide from about 0.05- 500 μg per dose. The disclosures of the aforementioned patents and publications are incorporated herein by reference in their entirety.
Representative steroidal anti-inflammatory agents include, but are not limited to, methyl prednisolone, prednisolone, dexamethasone, fluticasone propionate, 6α,9α-difluoro- 17α-[(2-furanylcarbonyl)oxy] - 11 β-hydroxy- 16α-methyl-3 -oxoandrosta- 1 ,4-diene- 17 β- carbothioic acid tf-fmoromethyl ester, 6α,9α-difluoro- 11 β-hydroxy- 16α-methyl-3 -oxo- 17α- propionyloxy-androsta-l,4-diene-17β-carbothioic acid S-(2-oxo-tetrahydrofuran-3S-yi) ester, beclomethasone esters (e.g., the 17-propionate ester or the 17,21-dipropionate ester), budesonide, flunisolide, mometasone esters (e.g., the furoate ester), triamcinolone acetonide, rofleponide, ciclesonide, butixocort propionate, RPR-106541, ST-126 and the like, or pharmaceutically-acceptable salts thereof. When employed, the steroidal antiinflammatory agent will be present in the pharmaceutical composition in a therapeutically effective amount. Typically, the steroidal anti-inflammatory agent will be present in an amount sufficient to provide from about 0.05-500 μg per dose.
An exemplary combination is a compound of formula I, or pharmaceutically acceptable salt or solvate or stereoisomer thereof, co-administered with salmeterol as the β2 adrenergic receptor agonist, and fluticasone propionate as the steroidal anti-inflammatory agent. Another exemplary combination is a compound of formula I, or pharmaceutically acceptable salt or solvate or stereoisomer thereof, co-administered with a crystalline monohydrochloride salt of N- {2-[4-((i?)-2-hydroxy-2-phenylethylamino)phenyl]ethyl} -(R)- 2-hydroxy-2-(3-formamido-4-hydroxyphenyl)ethylamine as the β2-adrenoreceptor agonist, and 6α59α-difluoro-17α-[(2-furanylcarbonyl)oxy]-llβ-hydroxy-16α-methyl-3-oxoandrosta- l,4-diene-17β-carbothioic acid S-fluoromethyl ester as the steroidal anti-inflammatory agent. Other suitable combinations include, for example, other anti-inflammatory agents, e.g., NSAIDs (such as sodium cromoglycate; nedocromil sodium; phosphodiesterase (PDE) inhibitors (e.g., theophylline, PDE4 inhibitors or mixed PDE3/PDE4 inhibitors); leukotriene antagonists (e.g., monteleukast); inhibitors of leukotriene synthesis; iNOS inhibitors; protease inhibitors, such as tryptase and elastase inhibitors; beta-2 integrin antagonists and adenosine receptor agonists or antagonists (e.g., adenosine 2a agonists); cytokine antagonists (e.g., chemokine antagonists such as, an interleukin antibody (αlL antibody), specifically, an αIL-4 therapy, an αIL-13 therapy, or a combination thereof); or inhibitors of cytokine synthesis. Representative phosphodiesterase-4 (PDE4) inhibitors or mixed PDE3/PDE4 inhibitors include, but are not limited to cis 4-cyano-4-(3-cyclopentyloxy-4- methoxyphenyl)cyclohexan-l-carboxylic acid, 2-carbomethoxy-4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-l-one; czs-[4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-l-ol]; cz5-4-cyano-4-[3- (cyclopentyloxy)-4-methoxyphenyl]cyclohexane-l-carboxylic acid and the like, or pharmaceutically acceptable salts thereof. Other representative PDE4 or mixed PDE4/PDE3 inhibitors include AWD-12-281 (elbion); NCS-613 (INSERM); D-4418 (Chiroscience and Schering-Plough); CI-1018 or PD-168787 (Pfizer); benzodioxole compounds disclosed in WO99/16766 (Kyowa Hakko); K-34 (Kyowa Hakko); V-11294A (Napp); roflumilast (Byk-Gulden); pthalazinone compounds disclosed in WO99/47505 (Byk-Gulden); Pumafentrine (Byk-Gulden, now Altana); arofylline (Almirall- Prodesfarma); VM554/UM565 (Vernalis); T-440 (Tanabe Seiyaku); and T2585 (Tanabe Seiyaku).
Representative muscarinic antagonists (i.e., anticholinergic agents) that can be used in combination with, and in addition to, the compounds of the invention include, but are not limited to, atropine, atropine sulfate, atropine oxide, methylatropine nitrate, homatropine hydrobromide, hyoscyamine (d, I) hydrobromide, scopolamine hydrobromide, ipratropium bromide, oxitropium bromide, tiotropium bromide, methantheline, propantheline bromide, anisotropine methyl bromide, clidinium bromide, copyrrolate (Robinul), isopropamide iodide, mepenzolate bromide, tridihexethyl chloride (Pathilone), hexocyclium methylsulfate, cyclopentolate hydrochloride, tropicamide, trihexyphenidyl hydrochloride, pirenzepine, telenzepine, AF-DX 116 and methoctramine and the like, or a pharmaceutically acceptable salt thereof; or, for those compounds listed as a salt, alternate pharmaceutically acceptable salt thereof.
Representative antihistamines (i.e., Hi-receptor antagonists) include, but are not limited to, ethanolamines, such as carbinoxamine maleate, clemastine fumarate, diphenylhydramine hydrochloride and dimenhydrinate; ethylenediamines, such as pyrilamine amleate, tripelennarnine hydrochloride and tripelennamine citrate; alkylamines, such as chlorpheniramine and acrivastine; piperazines, such as hydroxyzine hydrochloride, hydroxyzine pamoate, cyclizine hydrochloride, cyclizine lactate, meclizine hydrochloride and cetirizine hydrochloride; piperidines, such as astemizole, levocabastine hydrochloride, loratadine or its descarboethoxy analogue, terfenadine and fexofenadine hydrochloride; azelastine hydrochloride; and the like, or a pharmaceutically acceptable salt thereof; or, for those compounds listed as a salt, alternate pharmaceutically acceptable salt thereof.
Unless otherwise indicated, exemplary suitable doses for the other therapeutic agents administered in combination with a compound of the invention are in the range of about 0.05 μg/day to about 100 mg/day. The following formulations illustrate representative compositions of the invention, as well as exemplary methods of preparation. One or more secondary agents can optionally be formulated with the compound of the invention (primary active agent). Alternately, the secondary agents(s) can be formulated separately and co-administered with the primary active agent, either simultaneously or sequentially. For example, in one embodiment, a single dry powder formulation can be manufactured to include both the compound of the invention and one or more secondary agents. In another embodiment, one formulation is manufactured to contain the compound of the invention and separate formulation(s) are manufactured to contain the secondary agent(s). Such dry powder formulations can then be packaged in separate blister packs and administered with a single DPI device.
Exemplary Dry Powder Formulation For Administration By Inhalation 0.2 mg of a compound of the invention is micronized and then blended with 25 mg of lactose. The blended mixture is then loaded into a gelatin inhalation cartridge. The contents of the cartridge are administered using a powder inhaler. Exemplary Dry Powder Formulation For Administration By A Dry Powder Inhaler
A dry powder is prepared having a bulk formulation ratio of micronized compound of the invention (active agent) to lactose of 1 :200. The powder is packed into a dry powder inhalation device capable of delivering between about 10 μg and 100 μg of active agent per dose.
Exemplary Formulations For Administration By A Metered Dose Inhaler A suspension containing 5 wt% of a compound of the invention (active agent) and 0.1 wt% lecithin is prepared by dispersing 1O g of the active agent as micronized particles with a mean size less than 10 μm in a solution formed from 0.2 g of lecithin dissolved in 200 niL of demineralized water. The suspension is spray dried and the resulting material is micronized to particles having a mean diameter less than 1.5 μm. The particles are loaded into cartridges with pressurized 1,1,1,2-tetrafluoroethane.
Alternately, a suspension containing 5 wt% of the active agent, 0.5 wt% lecithin, and 0.5 wt% trehalose is prepared by dispersing 5 g of the active agent as micronized particles with a mean size less than 10 μm in a colloidal solution formed from 0.5 g of trehalose and 0.5 g of lecithin dissolved in 100 mL of demineralized water. The suspension is spray dried and the resulting material is micronized to particles having a mean diameter less than 1.5 μm. The particles are loaded into canisters with pressurized 1,1,1,2-tetrafluoroethane.
Exemplary Aqueous Aerosol Formulation For Administration By Nebulizer A pharmaceutical composition is prepared by dissolving 0.5 mg of a compound of the invention (active agent) in 1 mL of a 0.9% sodium chloride solution acidified with citric acid. The mixture is stirred and sonicated until the active agent is dissolved. The pH of the solution is adjusted to a value in the range of from 3 to 8 (typically about 5) by the slow addition of NaOH.
Exemplary Hard Gelatin Capsule Formulation For Oral Administration The following ingredients are thoroughly blended and then loaded into a hard gelatin capsule: 250 mg of a compound of the invention, 200 mg of lactose (spray-dried), and 10 mg of magnesium stearate, for a total of 460 mg of composition per capsule.
Exemplary Suspension Formulation For Oral Administration The following ingredients are mixed to form a suspension containing 100 mg of active ingredient per 10 mL of suspension.
Ingredients Amount
Compound of the invention 1.0 g
Fumaric acid 0.5 g
Sodium chloride 2.O g
Methyl paraben 0.15 g
Propyl paraben 0.05 g Ingredients Amount
Granulated sugar 25.5 g
Sorbitol (70% solution) 12.85 g
Veegum k (Vanderbilt Co.) 1.0 g
Flavoring 0.035 mL
Colorings 0.5 mg
Distilled water q.s. to 100 mL
Exemplary Injectable Formulation
The following ingredients are blended and the pH is adjusted to 4 ± 0.5 using 0.5 N HCl or 0.5 N NaOH.
Ingredients Amount
Compound of the invention 0.2 g
Sodium acetate buffer solution (0.4 M) 2.0 mL HCl (0.5 N) or NaOH (0.5 N) q.s. to pH 4
Water (distilled, sterile) q.s. to 20 mL
UTILITY
The compounds of the invention are expected to be useful as muscarinic receptor antagonists and therefore, are expected to be useful for treating medical conditions mediated by muscarinic receptors, i.e., medical conditions which are ameliorated by treatment with a muscarinic receptor antagonist. These medical conditions include, by way of example, pulmonary disorders or diseases including those associated with reversible airway obstruction, such as chronic obstructive pulmonary disease (e.g., chronic and wheezy bronchitis and emphysema), asthma, pulmonary fibrosis, allergic rhinitis, rhinorrhea, and the like. Other medical conditions that can be treated with muscarinic receptor antagonists are genitourinary tract disorders, such as overactive bladder or detrusor hyperactivity and their symptoms; gastrointestinal tract disorders, such as irritable bowel syndrome, diverticular disease, achalasia, gastrointestinal hypermotility disorders and diarrhea; cardiac arrhythmias, such as sinus bradycardia; Parkinson's disease; cognitive disorders, such as Alzheimer's disease; dismenorrhea; and the like. In one embodiment, the compounds of the invention are useful for treating smooth muscle disorders in mammals, including humans and their companion animals (e.g., dogs, cats etc.). Smooth muscle disorders include, by way of illustration, overactive bladder, chronic obstructive pulmonary disease and irritable bowel syndrome. When used to treat smooth muscle disorders or other conditions mediated by muscarinic receptors, the compounds of the invention will typically be administered orally, rectally, parenterally or by inhalation in a single daily dose or in multiple doses per day. The amount of active agent administered per dose or the total amount administered per day will typically be determined by the patient's physician and will depend on factors such as the nature and severity of the patients condition, the condition being treated, age and general health of the patient, tolerance of the patient to the active agent, route of administration and the like. Typically, suitable doses for treating smooth muscle disorders or other disorders mediated by muscarinic receptors will range from about 0.14 μg/kg/day to about 7 mg/kg/day of active agent; including from about 0.15 μg/kg/day to about 5 mg/kg/day. For an average 70 kg human, this would amount to about 10 μg per day to about 500 mg per day of active agent.
In a specific embodiment, the compounds of the invention are useful for treating pulmonary or respiratory disorders, such as COPD or asthma, in mammals including humans. When used to treat such disorders, the compounds of the invention will typically be administered by inhalation in multiple doses per day, in a single daily dose or a single weekly dose. Generally, the dose for treating a pulmonary disorder will range from about 10-200 μg/day. As used herein, COPD includes chronic obstructive bronchitis and emphysema (see, for example, Barnes, Chronic Obstructive Pulmonary Disease, N Engl J Med 343:269-78 (2000)). When used to treat a pulmonary disorder, the compounds of the invention are optionally administered in combination with other therapeutic agents such as a β2-adrenoreceptor agonist; a corticosteroid, a non-steroidal anti-inflammatory agent, or combinations thereof.
When administered by inhalation, the compounds of the invention typically have the effect of producing bronchodilation. Accordingly, in another embodiment, the invention is directed to a method of producing bronchodilation in a patient, the method comprising administering to a patient a bronchodilation-producing amount of a compound of the invention. Generally, the therapeutically effective dose for producing bronchodilation will range from about 10-200 μg/day.
In another embodiment, the compounds of the invention are used to treat overactive bladder. When used to treat overactive bladder, the compounds of the invention will typically be administered orally in a single daily dose or in multiple doses per day; preferably in a single daily dose. Preferably, the dose for treating overactive bladder will range from about 1.0-500 mg/day. In yet another embodiment, the compounds of the invention are used to treat irritable bowel syndrome. When used to treat irritable bowel syndrome, the compounds of the invention will typically be administered orally or rectally in a single daily dose or in multiple doses per day. Preferably, the dose for treating irritable bowel syndrome will range from about 1.0-500 mg/day.
Since compounds of the invention are muscarinic receptor antagonists, such compounds are also useful as research tools for investigating or studying biological systems or samples having muscarinic receptors. Such biological systems or samples may comprise M1, M2, M3, M4 and/or M5 muscarinic receptors. Any suitable biological system or sample having muscarinic receptors may be employed in such studies which may be conducted either in vitro or in vivo. Representative biological systems or samples suitable for such studies include, but are not limited to, cells, cellular extracts, plasma membranes, tissue samples, mammals (such as mice, rats, guinea pigs, rabbits, dogs, pigs, etc.), and the like, m one embodiment, a biological assay is conducted by contacting a biological system or sample (comprising a muscarinic receptor) with a muscarinic receptor-antagonizing amount of a compound of the invention. The effects of antagonizing the muscarinic receptor are then determined using conventional procedures and equipment, such as radioligand binding assays and functional assays. Such functional assays include ligand- mediated changes in intracellular cyclic adenosine monophosphate (cAMP), ligand- mediated changes in activity of the enzyme adenylyl cyclase (which synthesizes cAMP), ligand-mediated changes in incorporation of guanosine 5'-O-(γ-thio)triρhosphate ([ S]GTPyS) into isolated membranes via receptor catalyzed exchange of [ S]GTPyS for GDP, ligand-mediated changes in free intracellular calcium ions (measured, for example, with a fluorescence-linked imaging plate reader or FLIPR® from Molecular Devices, Inc.). A compound of the invention will antagonize or decrease the activation of muscarinic receptors in any of the functional assays listed above, or assays of a similar nature. A muscarinic receptor-antagonizing amount of a compound of the invention will typically range from about 0.1-100 nanomolar.
Additionally, the compounds of the invention can be used as research tools for discovering and evaluating new compounds that have muscarinic receptor antagonist activity. In one embodiment, a method of evaluating a test compound in a biological assay, comprises: (a) conducting a biological assay with a test compound to provide a first assay value; (b) conducting the biological assay with a compound of formula I to provide a second assay value; wherein step (a) is conducted either before, after or concurrently with step (b); and (c) comparing the first assay value from step (a) with the second assay value from step (b). Exemplary biological assays include, by way of illustration and not limitation, a muscarinic receptor binding assay or a bronchoprotection assay in a mammal. For example, muscarinic receptor binding data (e.g., as determined by in vitro radioligand displacement assays) for a test compound or a group of test compounds can be compared to the muscarinic receptor binding data for a compound of the invention to identify those test compounds that have about equal or superior muscarinic receptor binding, if any. This aspect of the invention includes, as separate embodiments, both the generation of comparison data (using the appropriate assays) and the analysis of the test data to identify test compounds of interest.
In another embodiment, the compounds of the invention are used to antagonize a muscarinic receptor in a biological system, and a mammal in particular, such as mice, rats, guinea pigs, rabbits, dogs, pigs, humans and so forth. A therapeutically effective amount of a compound of formula I is administered to the mammal, and the effects of antagonizing the muscarinic receptor can then determined using conventional procedures and equipment, examples of which are described above.
Among other properties, compounds of formula I have been found to be potent inhibitors of M3 muscarinic receptor activity. Accordingly, in a specific embodiment, the invention is directed to those compounds having an inhibition dissociation constant (K;) for the M3 receptor subtype of less than or equal to 10 nM; preferably, less than or equal to 5 nM; (as determined, for example, by an in vitro radioligand displacement assay). Additionally, compounds of formula I are expected to possess a desirable duration of action. Accordingly, in another specific embodiment, the invention is directed to those compounds having a duration of action greater than or equal to about 24 hours. Moreover, compounds of the invention are also expected to possess reduced side effects, such as dry mouth, at efficacious doses when administered by inhalation compared to other known muscarinic receptor antagonists administered by inhalation (such as tiotropium).
These properties, as well as the utility of the compounds of the invention, can be demonstrated using various in vitro and in vivo assays well-known to those skilled in the art. For example, representative assays are described in further detail in the following Examples. EXAMPLES
The following Preparations and Examples illustrate specific embodiments of the invention. In these examples, the following abbreviations have the following meanings:
AC adenylyl cyclase
ACh acetylcholine
ACN acetonitrile
BOC tert-butoxycarbonyl
BSA bovine serum albumin cAMP 3 '-5' cyclic adenosine monophosphate
CHO Chinese hamster ovary cM5 cloned chimpanzee M5 receptor
DCM dichloromethane (i.e., methylene chloride)
DIPEA N,iV-diisopropylethylamine
DMSO dimethyl sulfoxide dPBS Dulbecco's phosphate buffered saline
DMAP 4-dimethylaminopyridine
DMF dimethylformamide
DMSO dimethyl sulfoxide
EDC l-ethyl-3-(3-dimethylaminopropyl)carbodiimide
EDCI l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride
EtOAc ethyl acetate
EtOH ethanol
FBS fetal bovine serum
FLIPR fluorometric imaging plate reader
HATU O-(7-azabenzotriazol-l-yl-N,N,N',iV'-tetramethyluronium hexafluorophosphate
HBSS Hank's buffered salt solution
HEPES 4-(2-hydroxyethyl)- 1 -piperazineethanesulfonic acid
HOAt 1 -hydroxy-7-azabenzotriazole hMi cloned human M1 receptor hM2 cloned human M2 receptor hM3 cloned human M3 receptor KM4 cloned human M4 receptor hM5 cloned human M5 receptor
HOAc acetic acid
HOAt l-hydroxy-7-azabenzotriazole HOBT 1-hydroxybenzotriazole hydrate
HPLC high-performance liquid chromatography
IPA isopropanol
MCh niethylcholine
MeCN methylcyanide MeOH methanol
NaBH(OAc)3 sodium triacetoxyborohydride TFA trifhioroacetic acid
THF tetrahydrofuran
Any other abbreviations used herein but not defined have their standard, generally accepted meaning. Unless noted otherwise, all materials, such as reagents, starting materials and solvents, were purchased from commercial suppliers (such as Sigma- Aldrich, Fluka, and the like) and were used without further purification.
Unless otherwise indicated, HPLC analysis was conducted using an Agilent (Palo Alto, CA) Series 1100 instrument equipped with a Zorbax Bonus RP 2.1 x 50 mm column (Agilent) having a 3.5 micron particle size. Detection was by UV absorbance at 214 nm. The mobile phases employed were as follows (by volume): A is ACN (2%), water (98%) and TFA (0.1%); and B is ACN (90%), water (10%) and TFA (0.1%). HPLC 10-70 data was obtained using a flow rate of 0.5 mL/minute of 10 to 70% B over a 6 minute gradient (with the remainder being A). Similarly, HPLC 5-35 data and HPLC 10-90 data were obtained using 5 to 35% B; or 10 to 90% B over a 5 minute gradient.
Liquid chromatography mass spectrometry (LCMS) data were obtained with an Applied Biosystems (Foster City, CA) Model API-150EX instrument. LCMS 10-90 data was obtained using 10 to 90% Mobile Phase B over a 5 minute gradient. Small-scale purification was conducted using an API- 15 OEX Prep Workstation system from Applied Biosystems. The mobile phases employed were as follows (by volume): A is water and 0.05% TFA; and B is ACN and 0.05% TFA. For arrays (typically about 3 to 50 mg recovered sample size) the following conditions were used: 20 mL/min flow rate; 15 minute gradients and a 20 mm x 50 mm Prism RP column with 5 micron particles (Thermo Hypersil-Keystone, Bellefonte, PA). For larger scale purifications (typically greater than 100 mg crude sample), the following conditions were used: 60 niL/min flow rate; 30 minute gradients and a 41.4 mm x 250 mm Microsorb BDS column with 10 micron particles (Varian, Palo Alto, CA).
PREPARATION 1 Biρhenyl-2-ylcarbamic Acid Piperidin-4-yl Ester
Figure imgf000053_0001
Biphenyl-2-isocyanate (97.5 g, 521 mmol) and 4-hydroxy-N-benzylpiperidine (105 g, 549 mmol) were heated together at 70 0C for 12 hours. The reaction mixture was then cooled to 50 0C and EtOH (1 L) was added and then 6M HCl (191 mL) was added slowly. The resulting mixture was then cooled to ambient temperature and ammonium formate (98.5 g, 1.56 mol) was added and then nitrogen gas was bubbled through the solution vigorously for 20 minutes. Palladium on activated carbon (20 g, 10 wt% dry basis) was then added and the reaction mixture was heated at 40 °C for 12 hours, and then filtered through a pad of Celite. The solvent was then removed under reduced pressure and IM HCl (40 mL) was added to the crude residue. The pH of the mixture was then adjusted with 10 N NaOH to pH 12. The aqueous layer was extracted with EtOAc (2 x 150 mL) and the organic layer was dried over MgSO4, filtered and the solvent removed under reduced pressure to give 155 g of the title intermediate (100% yield). HPLC (10-70) Rt = 2.52; m/z: [M + H+] calcd for C18H20N2O2, 297.15; found, 297.3.
PREPARATION 2
JV-Benzyl-JV-methylaminoacetaldehyde
Figure imgf000053_0002
To a 3-necked 2-L flask was added 7V-benzyl-7V-methylethanolamine (30.5 g, 0.182 mol), DCM (0.5 L), DIPEA (95 mL, 0.546 mol) and DMSO (41 mL, 0.728 mol). Using an ice bath, the mixture was cooled to about -10 0C and sulfur trioxide pyridine- complex (87 g, 0.546 mol) was added in 4 portions over 5 minute intervals. The reaction was stirred at -10 °C for 2 hours. Before removing the ice-bath, the reaction was quenched by adding water (0.5 L). The aqueous layer was separated and the organic layer was washed with water (0.5 L) and brine (0.5 L) and then dried over MgSO4 and filtered to provide the title compound which was used without further purification.
PREPARATION 3 Biphenyl-2-ylcarbamic Acid l-[2-(Benzylmethylammo)ethyl]piperidin-4-yl Ester
Figure imgf000054_0001
To a 2-L flask, containing N-benzyl-iV-methylaminoacetaldehyde (19.8g, 0.121 mol; prepared as described in Preparation 2) in DCM (0.5 L) was added biphenyl-2- ylcarbamic acid piperidin-4-yl ester (30 g, 0.101 mol; prepared as described in Preparation 1) followed by NaBH(OAc)3 (45 g, 0.202 mol). The reaction mixture was stirred overnight and then quenched by the addition of 1 N HCl (0.5 L) with vigorous stirring. Three layers were observed and the aqueous layer was removed. After washing with IN NaOH (0.5 L), a homogenous organic layer was obtained which was then washed with a saturated solution of aqueous NaCl (0.5 L), dried over MgSO4, filtered and the solvent removed under reduced pressure. The residue was purified by dissolving it in a minimal amount of IPA and cooling this solution to 0 °C to form a solid which was collected and washed with cool IPA to provide 42.6 g of the title intermediate (95% yield). MS m/z: [M + H+] calcd f for C28H33N3O2, 444.3; found, 444.6. Rf = 3.51 min (10-70 ACN: H2O, reverse phase HPLC).
PREPARATION 3A Biρhenyl-2-ylcarbarnic Acid l-[2-(Benzyhnethylammo)ethyl]piperidin-4-yl Ester The title compound was prepared by mesylation of ΛMDenzyl-iV-methyl ethanolamine, which was then reacted with biphenyl-2-ylcarbamic acid piρeridin-4-yl ester in an alkylation reaction.
A 500 mL flask (reactor flask) was charged with N-benzyl-iV-methylethanolamine (24.5 mL), DCM (120 mL), NaOH (80 mL; 30wt%) and tetrabutylammonium chloride. Mixing at low speed throughout the reaction, the mixture was cooled to -10 °C (cooling bath), and the addition funnel charged with DCM (30 mL) and mesyl chloride (15.85 mL), which was added drop wise at a constant rate over 30 minutes. The addition was exothermic, and stirring was continued for 15 minutes while the temperature equilibrated back to -10 °C. The reaction was held for at least 10 minutes to ensure full hydrolysis of the excess mesyl chloride.
A 250 mL flask was charged with biphenyl-2-ylcarbamic acid piperidin-4-yl ester (26 g; prepared as described in Preparation 1) and DCM (125 mL), stirred for 15 minutes at room temperature, and the mixture chilled briefly to 10 °C to form a slurry. The slurry was then charged into the reactor flask via the addition funnel. The cooling bath was removed and the reaction mixture was warmed to 5 0C. The mixture was transferred to a separatory funnel, the layers allowed to settle, and the aqueous layer removed. The organic layer was transferred back to the reactor flask, stirring resumed, the mixture held to room temperature, and the reaction monitored by HPLC for a total of 3.5 hours. The reactor flask was charged with NaOH (IM solution; 100 mL), stirred, and the layers allowed to settle. The organic layer was separated, washed (NaCl satd. solution), its volume partially reduced under vacuum, and subjected to repeated IPA washings. The solids were collected and allowed to air-dry (25.85 g, 98% purity). Additional solids were obtained from further processing of the mother liquor (volume reduction, IPA, cooling). PREPARATION 4
Biphenyl-2-ylcarbamic Acid l-(2-Methylaminoethyr)piperidin-4-yl Ester
Figure imgf000055_0001
To a Parr hydrogenation flask was added biphenyl-2-ylcarbamic acid l-[2- (benzylmethylamino)ethyl]piperidin-4-yl ester (40 g, 0.09 mol; prepared as described in Preparation 3) and EtOH (0.5 L). The flask was flushed with nitrogen gas and palladium on activated carbon (15g, 10 wt. % (dry basis), 37% wt/wt) was added along with HOAc (20 mL). The mixture was kept on the Parr hydrogenator under a hydrogen atmosphere (~50 psi) for 3 hours. The mixture was then filtered and washed with EtOH. The filtrate was condensed and the residue was dissolved in a minimal amount of DCM. Isopropyl acetate (10 volumes) was added slowly to form a solid which was collected to provide 22.0 g of the title intermediate (70% yield). MS m/z: [M + H+] calcd for C21H27N3O2, 354.2; found, 354.3. Rf = 2.96 min (10-70 ACN: H2O, reverse phase HPLC). PREPARATION 5
Biphenyl-2-ylcarbamic Acid l-{2-r(4-Formylbenzoyl)methylamino] ethvl}piperidin-4-yl Ester
Figure imgf000056_0001
To a three-necked 1-L flask was added 4-carboxybenzaldehyde (4.77 g,
31.8 mmol), EDC (6.64 g, 34.7 mmol), HOBT (1.91 g, 31.8 mmol), and DCM (200 niL). When the mixture was homogenous, a solution of biphenyl-2-ylcarbamic acid l-(2- methylaminoethyl)piperidin-4-yl ester (10 g, 31.8 mmol; prepared as described in Preparation 4) in DCM (100 mL) was added slowly. The reaction mixture was stirred at room temperature for 16 hours and then washed with water (1 x 100 mL), IN HCl (5 x 60 mL), IN NaOH (1 x 100 mL), brine (1 x 5OmL), dried over sodium sulfate, filtered and concentrated to afford 12.6 g of the title intermediate (92% yield; 85% purity based on HPLC). MS m/z: [M + H+] calcd for C29H31N3O4, 486.2; found, 486.4. 22,3.12 min (10- 70 ACN: H2O, reverse phase HPLC). EXAMPLE 1
Figure imgf000056_0002
To a stirred solution of biphenyl-2-ylcarbamic acid l-{2-[(4-formylbenzoyl) methylamino]ethyl}piperidin-4-yl ester (50mg, O.lmmol; prepared as described in Preparation 5) in MeOH (1 mL) was added 1-acetylpiperazine (26mg, 0.2mmol). The mixture was shake for an hour and NaBH(OAc)3 ( 67mg, 0.3 mmol) was added. This was allowed to stir for 14 hours. The solvent was then removed under reduced pressure. To the mixture was added 1 : 1 HOAdH2O (1.0 mL) and then purified by reverse phase HPLC (gradient elution, ACN/H2O) to provide 37.8 mg of Compound 1-1 with 99% purity.
Compounds 1-2 to 1-44 were made in a similar manner by substituting the appropriate amine in the reductive amination step.
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
PREPARATION 6
Figure imgf000061_0001
To a stirred solution of biphenyl-2-ylcarbamic acid piperidin-4-yl ester (2.00 g, 6.76 mmol; prepared as described in Preparation 1) and DIPEA (3.54 mL, 20.3 mmol) in MeCN (67.6 mL) at 50 °C, was added 2-BOC-amino ethyl bromide (1.82 g, 8.11 mmol) and the reaction mixture was heated at 50 °C overnight. The solvent was removed under reduced pressure and dissolved in DCM (60 mL) and washed with saturated aqueous sodium bicarbonate solution (30 mL), organics dried (MgSO4) and solvent removed under reduced pressure. The crude residue was purified by column chromatography (5 % MeOH/DCM) to give the title intermediate as a white solid (2.32 g, 78%).
PREPARATION 7 Biphenyl-2-ylcarbamic acid l-(2-ammoethyl)piperidm-4-vl ester
Figure imgf000061_0002
The product of Preparation 6 was dissolved in TFA/DCM (25%, 52 mL) and stirred at room temperature for 2 hours. The solvent was removed under reduced pressure and the crude residue dissolved in DCM (30 mL) and washed with IN NaOH (15 mL). The organics were separated, dried (MgSO4) and solvent removed under reduced pressure to give the title intermediate (1.61 g, 90%).
PREPARATION 8
Figure imgf000061_0003
To a stirred solution of the product of Preparation 7 (339 mg, 1 mmol), BOC-(4- aminomethyl)benzoic acid (301 mg, 1.2 mmol) and HATU (456 mg, 1.2 mmol) in DMF (2 mL), was added DIPEA (226 μL, 1.3 mmol). The reaction mixture was stirred at room temperature overnight before removing the solvent under reduced pressure. The resulting residue was dissolved in DCM (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL), organics dried (MgSO4) and solvent removed under reduced pressure to yield the crude title intermediate, which was used directly in the next step.
PREPARATION 9 Biphenyl-2-ylcarbamic Acid l-r2-(4-Formylbenzoylammo)ethyl1piperidm-4-yl Ester
Figure imgf000062_0001
A mixture of 4-carboxybenzaldehyde (0.95 g, 6.35 mmol) and HATU (3.02 g, 7.94 mmol) in 55 mL of DCM was stirred at room temperature for 1 hour and then biphenyl-2- ylcarbamic acid l-(2-aminoethyl)piperidin-4-yl ester (3g, 5.29 mmol; prepared as described in Preparation 7) and DIPEA (4.6 mL, 26.45 mmol) were added. The resulting mixture was stirred at room temperature for 2 hours and then diluted with 100 mL of DCM and washed with saturated sodium bicarbonate (150 mL) and brine (150 mL). The organic layer was dried over MgSO4, filtered and concentrated to provide 2.3 g of the title intermediate (92% yield), which was suitable for use without further purification.
EXAMPLE 2
Figure imgf000062_0002
The crude product of Preparation 8 was dissolved in TFA/DCM (25%, 10 mL) and stirred at room temperature for 2 hours. The solvent was removed under reduced pressure and crude residue dissolved in DCM (15 mL) and washed with IN NaOH (5 mL). The organics were separated, dried (MgSO4) and solvent removed under reduced pressure to give Compound 2-1 (R6 and R7 = H) (472 mg, -100% over 2 steps): biphenyl-2-ylcarbamic acid l-[2-(4-aminomethylbenzoyl amino)ethyl]piperidin-4-yl ester. MS m/z: [M + H+] calcd for C28H32N4O3, 473.25; found, 473.2. Using the procedure of Example 1, but substituting the product of Preparation 9 in place of the product of Preparation 5, and substituting the appropriate amine in the reductive animation step, compounds having other R6 and R7 groups can also be made.
PREPARATION 10 3-r4-(Biphenyl-2-ylcarbamoyloxy1piperidin-l-yl1propionic Acid Methyl Ester
Figure imgf000063_0001
Methyl 3-bromopropionate (553 μL, 5.07 mmol) was added to a stirred solution of biphenyl-2-ylcarbamic acid piperidin-4-yl ester (1.00 g, 3.38 mmol; prepared as described in Preparation 1) and DIPEA (1.76 mL, 10.1 mmol) in ACN (34 mL) at 50 0C and the reaction mixture was heated at 50 0C overnight. The solvent was then removed under reduced pressure, and the residue was dissolved in DCM (30 mL). The resulting solution was washed with saturated aqueous sodium bicarbonate solution (10 mL), dried over MgSO4, filtered and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (5-10% MeOH/DCM) to give 905 mg of the title intermediate (70% yield). PREPARATION 11
3-r4-(Biphenyl-2-ylcarbamoyloxy)pir)eridin-l-vllpropionic Acid
Figure imgf000063_0002
A stirred solution of 3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic acid methyl ester (902 mg, 2.37 mmol; prepared as described in Preparation 10) and lithium hydroxide (171 mg, 7.11 mmol) in 50% THFiH2O (24 mL) was heated at 30 °C overnight, and then acidified with concentrated HCl and lyophilized to give the title intermediate (~ 100% yield, also contains LiCl salts). PREPARATION 12
Biρhenyl-2-ylcarbamic acid 1 -r2-(3-hydroxymethylbenzylcarbamoyl)ethyl] piperidin-4-yl Ester
Figure imgf000064_0001
To a stirred solution of 3-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic acid (0.5 g, 1.36 mmol; prepared as described in Preparation 11) in DCM (200 niL) was added DIPEA (10.7 mL, 6.12 mmol), 3-aminomethylbenzyl alcohol (0.33 g, 1.9 mmol), DMAP (0.14 g, 0.14 mmol) and EDCI (0.364 g, 1.9 mmol). The reaction mixture was stirred at room temperature for 2 hours. The mixture was concentrated, taken up in EtOAc (20 mL) and then washed with K2CO3 (3 x 10 mL), H2O (1 x 10 mL), brine (1 x 1OmL), dried over magnesium sulfate, filtered and concentrated. The mixture was purified on silica gel (7% MeOH/CHCl3) to afford 0.38 g of the title intermediate (57% yield).
PREPARATION 13
Biphenyl-2-ylcarbamic Acid l-[2-(3-formylbenzylcarbamoyl)ethyr| piperidin-4-yl Ester
Figure imgf000064_0002
To a flask was added biphenyl-2-ylcarbamic acid l-[2-(3-hydroxymethylbenzyl carbamoyl)ethyl]piperidin-4-yl ester (0.33 g, 0.68 mmol; prepared as described in Preparation 12), DCM (5 mL), DIPEA (0.47 mL, 2.7 mol) and DMSO (0.2 mL, 2.7 mol). Using an ice bath, the mixture was cooled to about -10 °C and sulfur trioxide pyridine- complex (0.32 g, 2.0 mol) was added. The reaction was stirred at -10 °C for 0.75 hour. Before removing the ice-bath, the reaction was quenched by adding water (5 mL). The aqueous layer was separated and the organic layer was washed with water (2 x 5 mL), 1.0N NaHSO4(aq) (2 x 5 mL), 1.0N NaHCO3(aq) (1 x 5 mL) and brine (0.5 L) and then dried over MgSO4 and filtered to provide the title compound which was used without further purification. EXAMPLE 3
Figure imgf000065_0001
To a stirred solution of biphenyl-2-ylcarbarnic acid l-[2-(3-formylbenzyl carbamoyl)ethyl]piperidin-4-yl ester (39 mg, 0.08 mmol; prepared as described in Preparation 13) in DCM (1 niL) was added 2-benzyl-2,5-diaza-bicyclo[2.2.1]heptane (18.8 mg, 0.1 mmol), and NaBH(OAc)3 (275 mg, 1.24 mmol). The mixture was stirred for 2 days and then the solvent was removed under reduced pressure. A 1 : 1 solution of HOAc and water (8.0 mL) was added to the reaction mixture. The mixture was chromatographed on reverse-phase HPLC (gradient elution, 10-50% ACN/H2O) to afford 4.0 mg of the title compound (5.6 % yield) as a bis(trifluoroacetate) salt.
Compounds 3-2 to 3-10 were made in a similar manner by substituting the appropriate amine in the reductive animation step.
Figure imgf000065_0002
Figure imgf000066_0002
EXAMPLE 4
Figure imgf000066_0001
Using the procedure of Example 1, but substituting 3-(4-formylphenyl)propionic acid in place of 4-carboxybenzaldehyde in Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared. The preparation of 3-(4-formylphenyl)propionic acid was done according to Tetrahedron 52{20}:6913-6930 (2001).
Figure imgf000066_0003
PREPARATION 14
Biphenyl-2-ylcarbamic Acid l-r2-f4-Aminomethylphenylcarbamoyr)ethyl1 piperidin-4-yl Ester
Figure imgf000067_0001
To a stirred solution of 4-(N-tørt-butoxycarbonylaminomethyl)aniline (756 nig,
3.4 mmol), 3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic acid (1.5 g, 4.08 mmol; prepared as described in Preparation 11) and HATU (1.55 g, 4.08 mmol) in DMF (6.8 mL), was added DIPEA (770 μL, 4.42 mmol). The reaction mixture was stirred at 50 °C overnight, and then the solvent was removed under reduced pressure. The resulting residue was dissolved in DCM (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic phase was then dried over MgSO4 and the solvent was removed under reduced pressure. The crude product was purified by flash chromatography (5-10% MeOH/DCM) to give a solid, which was dissolved in TFA/DCM (25%, 30 mL) and stirred at room temperature for 2 hours. The solvent was then removed under reduced pressure and the crude residue was dissolved in DCM (30 mL) and washed with IN NaOH (15 mL). The organic phase was separated, dried over MgSO4, filtered and the solvent was removed under reduced pressure to give 1.5 g of the title intermediate (94% yield over 2 steps).
PREPARATION 15 Biphenyl-2-ylcarbamic Acid l-f2-(4-Formylphenylcarbamoyl)ethyll piperidin-4-yl Ester
Figure imgf000067_0002
A mixture of biphenyl-2-ylcarbamic acid l-[2-(4-aminomethylphenylcarbamoyl) ethyl]piperidin-4-yl ester (1 g, 2.7 mmol; prepared as described in Preparation 14), 4- aminobenzyl alcohol (498 mg, 4.05 mmol), HATU (1.54 g, 4.05 mmol) and DIPEA (1.41 mL, 8.1 mmol) in DCM (14 mL) was stirred at room temperature for 2 hours. The reaction mixture was diluted with 100 mL of DCM and washed first with a 1 :3 mixture of IN HCl/water (100 mL), then brine (100 niL). The organic layer was dried over MgSO4, filtered and concentrated. The residue was dissolved in DCM (100 mL) and the solution was cooled to -5 0C. DIPEA (1.1 mL, 6.33 mmol) and DMSO (1.5 mL, 21.1 mmol) were added to the solution, followed by sulfur trioxide pyridine complex (1 g, 6.33 mmol). The reaction mixture was stirred at 0 °C for 1 hour then washed with a saturated solution of sodium bicarbonate (100 mL) and brine (100 mL). The organic layer was dried over MgSO4, filtered and concentrated to give 0.84 g of the title intermediate (66% yield) as an oil.
EXAMPLE 5
Figure imgf000068_0001
Using the procedure of Example 1, but substituting biphenyl-2-ylcarbamic acid 1- [2-(4-formylphenylcarbamoyl)ethyl]piperidin-4-yl ester (prepared as described in Preparation 15) in place of the product of Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000068_0002
Figure imgf000069_0001
Figure imgf000070_0001
EXAMPLE 6
Figure imgf000070_0002
Using the procedure of Example 1, substituting 3-(4-formylphenyl)propionic acid in place of 4-carboxybenzaldehyde in Preparation 9 and using that product in place of the product of Preparation 5, and substituting the appropriate amine in the reductive animation step, the following compounds were prepared.
Figure imgf000071_0001
Figure imgf000072_0002
PREPARATION 16 4-(Formylphenyl')acetic Acid
Figure imgf000072_0001
4-(formylphenyl)acetic acid methyl ester was prepared as described in Chem.
Commun. 7:669-670 (2001). To a solution of (4-formylphenyl)acetic acid methyl ester (2.76 g, 14.4 mmol) in 40 mL of MeOH was added IN NaOH (30 ml). The solution was stirred at room temperature for 16 hours and then the MeOH was removed in vacuo. The aqueous layer was then washed with EtOAc (40 mL), followed by wash with DCM (40 niL). The aqueous layer was then acidified to pH=l using IN HCl. The product was extracted using DCM washes (3 X 50 niL) and the organic layer washed with water (100 mL), NaCl (sat.) (100 mL), dried over MgSO4 and then filtered. The solvent removed under reduced pressure. The crude material was sufficiently pure to use without further purification. The title compound was obtained in 78% yield (2.0 g, 11.2 mmol).
EXAMPLE 7
Figure imgf000073_0001
Using the procedure of Example 1, substituting 4-(formylphenyl)acetic acid (prepared as described in Preparation 16) in place of 4-carboxybenzaldehyde in Preparation 9 and using that product in place of the product of Preparation 5, and substituting the appropriate amine in the reductive animation step, the following compounds were prepared.
Figure imgf000073_0002
PREPARATION 17 4-Formyl-3-methoxybenzoic Acid. Methyl Ester
Figure imgf000074_0001
To a stirred solution of 4-bromo-3-methoxybenzoic acid (15.0 g, 58 mmol) in DMSO (150 mL) was added NaHCO3 (20.0 g, 230 mmol). This was heated to 80 °C for 18 hours. The reaction was then cooled to room temperature and the solvent removed under reduced pressure. The crude reaction mixture was then dissolved in DCM (200 mL) and washed with IN HCl (100 mL), water (100 mL), NaCl (sat.) (100 mL), dried over MgSO4 and then filtered. The solvent was removed under reduced pressure. The crude material was sufficiently pure to use without further purification. The title intermediate was obtained in 79% yield (8.9 g, 45.8 mmol).
PREPARATION 18 2-Methoxyterephthalic Acid 4-Methvl Ester
Figure imgf000074_0002
To a stirred solution of 4-formyl-3-methoxybenzoic acid methyl ester (5.0 g, 26 mmol; prepared as described in Preparation 17) in tert-butyl alcohol (200 mL) was added NaH2PO4-2H2O (3.6 g, 26 mmol), water (50 mL), 2-methyl-2-butene (11 mL, 104 mmol), and finally NaClO2 (7.02 g, 78 mmol). The reaction was allowed to stir at room temperature for 4 hours. The solvent was then removed under reduced pressure. The crude reaction mixture was then dissolved in DCM (200 mL) and the product was extracted with IN NaOH (200 mL). The aqueous layer was washed with DCM (200 mL) and then neutralized with 6N HCl (-40 mL) and the product extracted with DCM (200 mL). The organic layer was then washed with water (100 mL), NaCl (sat.) (100 mL), dried over MgSO4 and then filtered. The solvent was removed under reduced pressure. The crude material was sufficiently pure to use without further purification. The title intermediate was obtained in 47% yield (2.4 g, 12.3 mmol). PREPARATION 19
JV-(2-[4-(Biphenyl-2-ylcarbamoyloxy')piperidin-l-yllethyl}-3-methoxy-iV- methylterephthalamic Acid Methyl Ester
Figure imgf000075_0001
To a stirred solution of 2-methoxyterephthalic acid 4-methyl ester (450 mg,
2.1 mmol; prepared as described in Preparation 18) in DMF (10 mL) was added EDC (630 mg, 3.3 mmol), HOAt (2.4 mL, 1.18 mmol, 0.5M in DMF) and DIPEA (1.3 mL, 7.05 mmol). When the mixture was homogenous, a solution of biplienyl-2-ylcarbamic acid 1- (2-methylaminoethyl)piperidin-4-yl ester (830 mg, 2.4 mmol; prepared as described in Preparation 4) was added slowly. The reaction mixture was stirred at room temperature for 16 hours and then washed with water (100 mL), IN HCl (100 mL), IN NaOH (100 mL), brine (100 mL), dried over MgSO4, filtered and concentrated to afford the title intermediate in 89% yield (1.04 g, 1.9 mmol). MS m/z: [M + H+] calcd for C31H35N3O6, 545.6; found, 546.6. PREPARATION 20
Biphenyl-2-ylcarbamic Acid l-{2-[(4-hydroxymethyl-2- methoxybenzoyDmethylaminolethvUpiperidm-4-vl Ester
Figure imgf000075_0002
To a stirred solution of iV-{2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}- 3-methoxy-n-methylterephthalamic acid methyl ester (1.0 g, 1.8 mmol; prepared as described in Preparation 19) in THF (100 mL) at 0 0C, was added MeOH (57 μL, l.δmmol), followed by LiAlH4 (1.8 mL, 1.8 mmol, 1.0M in THF) was added. The ice bath was removed, and the reaction mixture was stirred at room temperature for 1 hour. The reaction was quenched with IN HCl (aq) at 0 °C until no more bubbling, and stirring was continued for 10 minutes. The solvent was removed under reduced pressure. The crude reaction mixture was taken up in DCM (100 mL) and washed with water (100 mL), NaCl (sat.) (100 mL), dried over MgSO4 and then filtered. The solvent was removed under reduced pressure. The crude material was sufficiently pure to use without further purification. The title intermediate was obtained in 89% yield (831 mg, 1.6 mmol). MS m/z: [M + H+] calcd for C30H35N3O5, 517.6; found, 518.6.
PREPARATION 21
Biphenyl-2-ylcarbamic acid l-{2-[Y4-formyl-2- methoxybenzovDmethylaminol ethyl}piperidin-4- yl Ester
Figure imgf000076_0001
To a stirred solution of biphenyl-2-ylcarbamic acid l-{2-[(4-hydroxymethyl-2- methoxybenzoyl)methylamino]ethyl}piperidin-4-yl ester (78 mg, 1.5 mmol; prepared as described in Preparation 20) in DCM (2.5 mL) at -15 0C was added DMSO (130 μL, 22.5 mmol), and DIPEA (130 μL, 7.5 mmol). To the solution was added sulfur trioxide- pyridine complex (240 mg, 15 mmol). After 30 minutes, the reaction mixture was quenched with water (~3 mL). Two layers were separated, the organic layer was dried over MgSO4, filtered and the title intermediate was used directly in the next reaction.
EXAMPLE 8
Figure imgf000076_0002
Using the procedure of Example 1, but substituting biphenyl-2-ylcarbamic acid 1- {2-[(4-formyl-2-methoxybenzoyl)methylamino]ethyl}piperidin-4-yl ester (prepared as described in Preparation 21) in place of the product of Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000077_0002
EXAMPLE 9
Figure imgf000077_0001
Using the procedure of Example 8, but substituting biphenyl-2-ylcarbamic acid 1- (2-aminoethyl)piperidin-4-yl ester (prepared as described in Preparation 7) in place of the product of Preparation 4 in Preparation 19 and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000077_0003
EXAMPLE 10
Figure imgf000078_0001
Using the procedure of Example 1, but substituting 5-formylpyridine-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 5, and substituting the appropriate amine in the reductive animation step, the following compounds were prepared.
Figure imgf000078_0002
Figure imgf000079_0002
EXAMPLE 11
Figure imgf000079_0001
Using the procedure of Example 1, but substituting 5-formylthiophene-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000079_0003
Figure imgf000080_0001
EXAMPLE 12
Figure imgf000080_0002
Using the procedure of Example 1, substituting 5-formylthiophene-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 9 and using that product in place of the product of Preparation 5, and substituting the appropriate amine in the reductive animation step, the following compounds were prepared.
Figure imgf000081_0003
EXAMPLE 13
Figure imgf000081_0001
Using the procedure of Example 1, substituting 4-formylpyrrole-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 9 and using that product in place of the product of Preparation 5, and substituting the appropriate amine in the reductive amination step, Compound 13-1 (R6 = H, R7 = -CH3) was prepared: biphenyl-2-ylcarbamic acid l-{2- [methyl(4-methylamino methyl-lH-pyrrole-2-carbonyl)amino]ethyl}piperidin-4-yl ester. MS m/z: [M + H+] calcd for C28H35N5O3, 490.27; found, 490.2.
EXAMPLE 14
Figure imgf000081_0002
Using the procedure of Example 1, but substituting 5-formylpyrrole-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000081_0004
Figure imgf000082_0002
EXAMPLE 15
Figure imgf000082_0001
Using the procedure of Example 1, but substituting 5-formylfuran-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000082_0003
Figure imgf000083_0002
EXAMPLE 16
Figure imgf000083_0001
Using the procedure of Example 1, substituting 5-formylfuran-2-carboxylic acid in place of 4-carboxybenzaldehyde in Preparation 9 and using that product in place of the product of Preparation 5, and substituting the appropriate amine in the reductive amination step, the following compounds were prepared.
Figure imgf000083_0003
Figure imgf000084_0001
ASSAY 1
Radioligand Binding Assay Membrane Preparation from Cells Expressing hMi, hM.2, hMs and hM.4 Muscarinic Receptor Subtypes
CHO cell lines stably expressing cloned human !1M1 , hM2, hM3 and hM4 muscarinic receptor subtypes, respectively, were grown to near confluency in medium consisting of HAM's F-12 supplemented with 10% FBS and 250 μg/mL Geneticin. The cells were grown in a 5% CO2, 37 0C incubator and lifted with 2 niM EDTA in dPBS. Cells were collected by 5 minute centrifugation at 650 x g, and cell pellets were either stored frozen at -80 °C or membranes were prepared immediately. For membrane preparation, cell pellets were resuspended in lysis buffer and homogenized with a Polytron PT-2100 tissue disrupter (Kinematica AG; 20 seconds x 2 bursts). Crude membranes were centrifuged at 40,000 x g for 15 minutes at 4 0C. The membrane pellet was then resuspended with resuspension buffer and homogenized again with the Polytron tissue disrupter. The protein concentration of the membrane suspension was determined by the method described in Lowry, O. et al, Journal of Biochemistry 193:265 (1951). All membranes were stored frozen in aliquots at -80 °C or used immediately. Aliquots of prepared hM5 receptor membranes were purchased directly from Perkin Elmer and stored at -80 0C until use. Radioligand Binding Assay on Muscarinic Receptor
Subtypes hMi, I1M2, hMs, I1M4 and hMs
Radioligand binding assays were performed in 96-well microtiter plates in a total assay volume of 100 μL. CHO cell membranes stably expressing either the hMl5 hM2, hM3, hM4 or hMs muscarinic subtype were diluted in assay buffer to the following specific target protein concentrations (μg/well): 10 μg for hMl5 10-15 μg for hM2, 10-20 μg for hM3, 10-20 μg for ILM4, and 10-12 μg for hM5. The membranes were briefly homogenized using a Polytron tissue disruptor (10 seconds) prior to assay plate addition. Saturation binding studies for determining K# values of the radioligand were performed using L-[N- methyl-3H] scopolamine methyl chloride ([3H]-NMS) (TRK666, 84.0 Ci/mmol, Amersham Pharmacia Biotech, Buckinghamshire, England) at concentrations ranging from 0.001 nM to 20 nM. Displacement assays for determination of K; values of test compounds were performed with [3H]-NMS at 1 nM and eleven different test compound concentrations. The test compounds were initially dissolved to a concentration of 400 μM in dilution buffer and then serially diluted 5x with dilution buffer to final concentrations ranging from 10 pM to 100 μM. The addition order and volumes to the assay plates were as follows: 25 μL radioligand, 25 μL diluted test compound, and 50 μL membranes. Assay plates were incubated for 60 minutes at 37 °C. Binding reactions were terminated by rapid filtration over GFfB glass fiber filter plates (PerkinElmer Inc., Wellesley, MA) pre-treated in 1% BSA. Filter plates were rinsed three times with wash buffer (10 mM HEPES) to remove unbound radioactivity. Plates were then air dried, and 50 μL Microscint-20 liquid scintillation fluid (PerkinElmer Inc., Wellesley, MA) was added to each well. The plates were then counted in a PerkinElmer Topcount liquid scintillation counter (PerkinElmer Inc., Wellesley, MA). Binding data were analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the one-site competition model. K/ values for test compounds were calculated from observed IC50 values and the K# value of the radioligand using the Cheng-Prusoff equation (Cheng Y; Prusoff W. H. Biochemical Pharmacology 22(23):3099-108 (1973)). Ki values were converted to pKi values to determine the geometric mean and 95% confidence intervals. These summary statistics were then converted back to Ki values for data reporting. In this assay, a lower K;- value indicates that the test compound has a higher binding affinity for the receptor tested. Exemplary compounds of the invention that were tested in this or a similar assay, typically were found to have a K,- value of less than about 10 nM for the M3 muscarinic receptor subtype.
ASSAY 2
Muscarinic Receptor Functional Potency Assays Blockade of Agonist-Mediated Inhibition of cAMP Accumulation In this assay, the functional potency of a test compound is determined by measuring the ability of the test compound to block oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM2 receptor. cAMP assays are performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with 125I-cAMP (NEN SMP004B, PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturer's instructions.
Cells are rinsed once with dPBS and lifted with Trypsin-EDTA solution (0.05% trypsin/0.53 mM EDTA) as described in the Cell Culture and Membrane Preparation section above. The detached cells are washed twice by centrifugation at 650 x g for five minutes in 5OmLs dPBS. The cell pellet is then re-suspended in 10 mL dPBS, and the cells are counted with a Coulter Zl Dual Particle Counter (Beckman Coulter, Fullerton, CA). The cells are centrifuged again at 650 x g for five minutes and re-suspended in stimulation buffer to an assay concentration of 1.6 x 106 - 2.8 x 106 cells/mL. The test compound is initially dissolved to a concentration of 400 μM in dilution buffer (dPBS supplemented with 1 mg/mL BSA (0.1%)), and then serially diluted with dilution buffer to final molar concentrations ranging from 100 μM to 0.1 nM. Oxotremorine is diluted in a similar manner. To measure oxotremorine inhibition of adenylyl cyclase (AC) activity, 25 μL forskolin (25 μM final concentration diluted in dPBS), 25 μL diluted oxotremorine, and 50 μL cells are added to agonist assay wells. To measure the ability of a test compound to block oxotremorine-inhibited AC activity, 25 μL forskolin and oxotremorine (25 μM and 5 μM final concentrations, respectively, diluted in dPBS) 25 μL diluted test compound, and 50 μL cells are added to remaining assay wells. Reactions are incubated for 10 minutes at 37 °C and stopped by addition of 100 μL ice-cold detection buffer. Plates are sealed, incubated overnight at room temperature and counted the next morning on a PerkinElmer TopCount liquid scintillation counter (PerkinElmer hie, Wellesley, MA). The amount of c AMP produced (pmol/well) is calculated based on the counts observed for the samples and cAMP standards, as described in the manufacturer's user manual. Data are analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the non-linear regression, one-site competition equation. The Cheng-Prusoff equation is used to calculate the K;, using the EC50 of the oxotremorine concentration- response curve and the oxotremorine assay concentration as the KD and [L], respectively. The Ki values are converted to pKj values to determine the geometric mean and 95% confidence intervals. These summary statistics are then converted back to Ki values for data reporting, hi this assay, a lower Ki value indicates that the test compound has a higher functional activity at the receptor tested. Compounds of the invention are expected to have a Kj value of less than about 10 nM for blockade of oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM2 receptor, when tested in this or a similar assay.
Blockade of Agonist-Mediated [35S] GTPyS Binding
In a second functional assay, the functional potency of test compounds can be determined by measuring the ability of the compounds to block oxotremorine-stimulated [35S]GTPyS binding in CHO-Kl cells expressing the I1M2 receptor, At the time of use, frozen membranes are thawed and then diluted in assay buffer with a final target tissue concentration of 5-10 μg protein per well. The membranes are briefly homogenized using a Polytron PT-2100 tissue disrupter and then added to the assay plates. The EC90 value (effective concentration for 90% maximal response) for stimulation of [35S]GTPyS binding by the agonist oxotremorine is determined in each experiment. To determine the ability of a test compound to inhibit oxotremorine-stimulated [35S]GTPyS binding, the following is added to each well of 96 well plates: 25 μL of assay buffer with [35S]GTPyS (0.4nM), 25 μL of oxotremorme(EC90) and GDP (3 μM), 25 μL of diluted test compound and 25 μL CHO cell membranes expressing the hM2 receptor. The assay plates are then incubated at 37 °C for 60 minutes. The assay plates are filtered over 1% BSA- pretreated GFTB filters using a PerkinElmer 96-well harvester. The plates are rinsed with ice-cold wash buffer for 3 x 3 seconds and then air or vacuum dried. Microscint-20 scintillation liquid (50 μL) is added to each well, and each plate is sealed and radioactivity counted on a topcounter (PerkinElmer). Data are analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, hie, San Diego, CA) using the non-linear regression, one-site competition equation. The Cheng-Prusoff equation is used to calculate the Kj, using the IC50 values of the concentration-response curve for the test compound and the oxotremorine concentration in the assay as the KD and [L], ligand concentration, respectively. In this assay, a lower Kj value indicates that the test compound has a higher functional activity at the receptor tested. Compounds of the invention are expected to have a Kj value of less than about 10 nM for blockade of oxotremorine-stimulated [35S]GTPγS_binding in CHO-Kl cells expressing the hM2 receptor, when tested in this or a similar assay. Blockade of Agonist-Mediated Calcium Release via FLIPR Assays
Muscarinic receptor subtypes (M1, M3 and M5 receptors), which couple to Gq proteins, activate the phospholipase C (PLC) pathway upon agonist binding to the receptor.
As a result, activated PLC hydrolyzes phosphatyl inositol diphosphate (PIP2) to diacylglycerol (DAG) and phosphatidyl-l,4,5-triphosphate (IP3), wliich in turn generates calcium release from intracellular stores, i.e., endoplasmic and sarcoplasmic reticulum. The FLIPR (Molecular Devices, Sunnyvale, CA) assay capitalizes on this increase in intracellular calcium by using a calcium sensitive dye (Fluo-4AM, Molecular Probes, Eugene, OR) that fluoresces when free calcium binds. This fluorescence event is measured in real time by the FLIPR, which detects the change in fluorescence from a monolayer of cells cloned with human M1 and M3, and chimpanzee M5 receptors. Antagonist potency can be determined by the ability of antagonists to inhibit agonist-mediated increases in intracellular calcium. For FLIPR calcium stimulation assays, CHO cells stably expressing the hMl5 hM3 and cM5 receptors are seeded into 96-well FLIPR plates the night before the assay is done. Seeded cells are washed twice by Cellwash (MTX Labsystems, Inc.) with FLIPR buffer (10 mM HEPES, pH 7.4, 2 mM calcium chloride, 2.5 mM probenecid in Hank's Buffered Salt Solution (HBSS) without calcium and magnesium) to remove growth media and leaving 50 μL/well of FLIPR buffer. The cells are then incubated with 50 μL/well of 4 μM FLUO-4AM (a 2X solution was made) for 40 minutes at 37 °C, 5% carbon dioxide. Following the dye incubation period, cells are washed two times with FLIPR buffer, leaving a final volume of 50 μL/well.
To determine antagonist potency, the dose-dependent stimulation of intracellular Ca2+ release for oxotremorine is first determined so that antagonist potency can later be measured against oxotremorine stimulation at an ECg0 concentration. Cells are first incubated with compound dilution buffer for 20 minutes, followed by agonist addition, which is performed by the FLIPR. An EC90 value for oxotremorine is generated according to the method detailed in the FLIPR measurement and data reduction section below, in conjunction with the formula ECF = ((F/100-F)Λl/H) * EC50. An oxotremorine concentration of 3 x ECF is prepared in stimulation plates such that an EC90 concentration of oxotremorine is added to each well in the antagonist inhibition assay plates. The parameters used for the FLIPR are: exposure length of 0.4 seconds, laser strength of 0.5 watts, excitation wavelength of 488 nm, and emission wavelength of 550 nm. Baseline is determined by measuring the change in fluorescence for 10 seconds prior to addition of agonist. Following agonist stimulation, the FLIPR continuously measured the change of fluorescence every 0.5 to 1 second for 1.5 minutes to capture the maximum fluorescence change. The change of fluorescence is expressed as maximum fluorescence minus baseline fluorescence for each well. The raw data is analyzed against the logarithm of drug concentration by nonlinear regression with GraphPad Prism (GraphPad Software, Inc., San Diego, CA) using the built-in model for sigmoidal dose-response. Antagonist Kj values are determined by Prism using the oxotremorine EC50 value as the KQ and the oxotremorine EC9Q for the ligand concentration according to the Cheng-Prusoff equation (Cheng & Prusoff, 1973). In this assay, a lower Ki value indicates that the test compound has a higher functional activity at the receptor tested. Compounds of the invention are expected to have a Kj value of less than about 10 nM for blockade of agonist-mediated calcium release in CHO cells stably expressing the hM3 receptor, when tested in this or a similar assay. ASSAY 3
Determination of Duration of Bronchoprotection in Guinea Pig Model of Acetylcholine-Induced Bronchoconstriction
This in vivo assay is used to assess the bronchoprotective effects of test compounds exhibiting muscarinic receptor antagonist activity. Groups of six male guinea pigs (Duncan-Hartley (HsdPoc:DH) Harlan, Madison, WI) weighing between 250 and 350 g are individually identified by cage cards. Throughout the study animals are allowed access to food and water ad libitum. Test compounds are administered via inhalation over 10 minutes in a whole-body exposure dosing chamber (R&S Molds, San Carlos, CA). The dosing chambers are arranged so that an aerosol was simultaneously delivered to 6 individual chambers from a central manifold. Guinea pigs are exposed to an aerosol of a test compound or vehicle (WFI). These aerosols are generated from aqueous solutions using an LC Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA) driven by a mixture of gases (CO2 = 5%, O2 = 21% and N2 = 74%) at a pressure of 22 psi. The gas flow through the nebulizer at this operating pressure is approximately 3 L/minute. The generated aerosols are driven into the chambers by positive pressure. No dilution air is used during the delivery of aerosolized solutions. During the 10 minute nebulization, approximately 1.8 mL of solution is nebulized. This is measured gravimetrically by comparing pre-and post-nebulization weights of the filled nebulizer. The bronchoprotective effects of test compounds administered via inhalation are evaluated using whole body plethysmography at 1.5, 24, 48 and 72 hours post-dose. Forty- five minutes prior to the start of the pulmonary evaluation, each guinea pig is anesthetized with an intramuscular injection of ketamine (43.75 mg/kg), xylazine (3.50 mg/kg) and acepromazine (1.05 mg/kg). After the surgical site is shaved and cleaned with 70% alcohol, a 2-3 cm midline incision of the ventral aspect of the neck was made. Then, the jugular vein is isolated and cannulated with a saline-filled polyethylene catheter (PE- 50, Becton Dickinson, Sparks, MD) to allow for intravenous infusions of ACh (Sigma- Aldrich, St. Louis, MO) in saline. The trachea is then dissected free and cannulated with a 14G teflon tube (#NE- 014, Small Parts, Miami Lakes, FL). If required, anesthesia is maintained by additional intramuscular injections of the aforementioned anesthetic mixture. The depth of anesthesia is monitored and adjusted if the animal responds to pinching of its paw or if the respiration rate is greater than 100 breaths/minute. Once the cannulations are complete, the animal is placed into a plethysmo graph
(#PLY3114, Buxco Electronics, Inc., Sharon, CT) and an esophageal pressure cannula (PE-160, Becton Dickinson, Sparks, MD) is inserted to measure pulmonary driving pressure (pressure). The teflon tracheal tube is attached to the opening of the plethysmograph to allow the guinea pig to breathe room air from outside the chamber. The chamber is then sealed. A heating lamp is used to maintain body temperature and the guinea pig's lungs are inflated 3 times with 4 mL of air using a 10 mL calibration syringe (#5520 Series, Hans Rudolph, Kansas City, MO) to ensure that the lower airways do not collapse and that the animal does not suffer from hyperventilation. Once it is determined that baseline values are within the range 0.3 - 0.9 mL/cm H2O for compliance and within the range 0.1 - 0.199 cm H20/mL per second for resistance, the pulmonary evaluation is initiated. A Buxco pulmonary measurement computer progam enables the collection and derivation of pulmonary values. Starting this program initiates the experimental protocol and data collection. The changes in volume over time that occur within the plethysmograph with each breath are measured via a Buxco pressure transducer. By integrating this signal over time, a measurement of flow is calculated for each breath. This signal, together with the pulmonary driving pressure changes, which are collected using a Sensym pressure transducer (#TRD4100), is connected via a Buxco (MAX 2270) preamplifier to a data collection interface (#'s SFT3400 and SFT3813). All other pulmonary parameters are derived from these two inputs. Baseline values are collected for 5 minutes, after which time the guinea pigs are challenged with ACh. ACh (0.1 mg/mL) is infused intravenously for 1 minute from a syringe pump (sp210iw, World Precision Instruments, Inc., Sarasota, FL) at the following doses and prescribed times from the start of the experiment: 1.9 μg/minute at 5 minutes, 3.8 μg/minute at 10 minutes, 7.5 μg/minute at 15 minutes, 15.0 μg/minute at 20 minutes, 30 μg/minute at 25 minutes and 60 μg/minute at 30 minutes. If resistance or compliance has not returned to baseline values at 3 minutes following each ACh dose, the guinea pig's lungs are inflated 3 times with 4 mL of air from a 10 mL calibration syringe. Recorded pulmonary parameters includes respiration frequency (breaths/minute), compliance
(mL/cm H2O) and pulmonary resistance (cm H2O/ mL per second). Once the pulmonary function measurements are completed at minute 35 of this protocol, the guinea pig is removed from the plethysmograph and euthanized by carbon dioxide asphyxiation. The data are evaluated in one or both of the following ways: (a) Pulmonary resistance (RL, cm H2OZmL per second) is calculated from the ratio of "change in pressure" to "the change in flow." The RL response to ACh (60 μg/min, IH) is computed for the vehicle and the test compound groups. The mean ACh response in vehicle-treated animals, at each pre-treatment time, is calculated and used to compute % inhibition of ACh response, at the corresponding pre-treatment time, at each test compound dose. Inhibition dose-response curves for 'RL' are fitted with a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) to estimate bronchoprotective ID5O (dose required to inhibit the ACh (60 μg/min) bronchoconstrictor response by 50%). The equation used is as follows: Y = Min + (Max-Min)/(l + ι0 (^so-xr rnuo^ where X is the logarithm of dose, Y is the response (% Inhibition of ACh induced increase in RL). Y starts at Min and approaches asymptotically to Max with a sigmoidal shape.
(b) The quantity PD2, which is defined as the amount of ACh or histamine needed to cause a doubling of the baseline pulmonary resistance, is calculated using the pulmonary resistance values derived from the flow and the pressure over a range of ACh or histamine challenges using the following equation (which is derived from a equation used to calculate PC20 values described in American Thoracic Society. Guidelines for methacholine and exercise challenge testing - 1999. Am JRespir Crit Care Med. 161: 309-329 (2000)):
PD2 = antilog [ log C1 + (log C2 - log CI)(2RQ - RQ ]
R2 - R1 where: C1 is the concentration of ACh or histamine preceding C2; C2 is the concentration of
ACh or histamine resulting in at least a 2-fold increase in pulmonary resistance (RL); R0 is the baseline RL value; R1 is the RL value after C1; and R2 is the RL value after C2. An efficacious dose is defined as a dose that limits the bronchrestriction response to a 50 μg/mL dose of ACh to a doubling of the baseline pulmonary resistance (PD2(50)).
Statistical analysis of the data is performed using a two-tailed Students t-test. A P-value <0.05 is considered significant. Generally, test compounds having a PD2(5o) less than about 200 μg/mL for ACh-induced bronchoconstriction at 1.5 hours post-dose in this assay are preferred. Compounds of the invention are expected to have a PD2(S0) of less than about 200 μg/mL for ACh-induced bronchoconstriction at 1.5 hours post-dose, when tested in this or a similar assay.
ASSAY 4 Inhalation Guinea Pig Salivation Assay Guinea pigs (Charles River, Wilmington, MA) weighing 200-350 g are acclimated to the in-house guinea pig colony for at least 3 days following arrival. Test compound or vehicle are dosed via inhalation (IH) over a 10 minute time period in a pie shaped dosing chamber (R&S Molds, San Carlos, CA). Test solutions are dissolved in sterile water and delivered using a nebulizer filled with 5.0 mL of dosing solution. Guinea pigs are restrained in the inhalation chamber for 30 minutes. During this time, guinea pigs are restricted to an area of approximately 110 sq. cm. This space is adequate for the animals to turn freely, reposition themselves, and allow for grooming. Following 20 minutes of acclimation, guinea pigs are exposed to an aerosol generated from a LS Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA) driven by house air at a pressure of 22 psi. Upon completion of nebulization, guinea pigs are evaluated at 1.5, 6, 12, 24, 48, or 72 hrs after treatment.
Guinea pigs are anesthetized one hour before testing with an intramuscular (IM) injection of a mixture of ketamine 43.75 mg/kg, xylazine 3.5 mg/kg, and acepromazine 1.05 mg/kg at an 0.88 mL/kg volume. Animals are placed ventral side up on a heated (37°C) blanket at a 20 degree incline with their head in a downward slope. A 4-ply 2 x 2 inch gauze pad (Nu-Gauze General-use sponges, Johnson and Johnson, Arlington, TX) is inserted in the guinea pig's mouth. Five minutes later, the muscarinic agonist pilocarpine (3.0 mg/kg, SC) is administered and the gauze pad is immediately discarded and replaced by a new pre-weighed gauze pad. Saliva is collected for 10 minutes, at which point the gauze pad is weighed and the difference in weight recorded to determine the amount of accumulated saliva (in mg). The mean amount of saliva collected for animals receiving the vehicle and each dose of test compound is calculated. The vehicle group mean is considered to be 100% salivation. Results are calculated using result means (n = 3 or greater). Confidence intervals (95%) are calculated for each dose at each time point using two-way ANOVA. This model is a modified version of the procedure described in Rechter, "Estimation of anticholinergic drug effects in mice by antagonism against pilocarpine-induced salivation" Ata Pharmacol Toxicol 24:243-254 (1996). The mean weight of saliva in vehicle-treated animals, at each pre-treatment time, is calculated and used to compute % inhibition of salivation, at the corresponding pre- treatment time, at each dose. The inhibition dose-response data are fitted to a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) to estimate anti-sialagogue DD50 (dose required to inhibit 50% of pilocarpine-evoked salivation). The following equation is used:
Y = Min + (Max-Min)/(1 + 10 «log ID5°-χ)* WM°& ) where X is the logarithm of dose, Y is the response (% inhibition of salivation). Y starts at Min and approaches asymptotically to Max with a sigmoidal shape. The ratio of the anti- sialagogue ID5O to bronchoprotective ID50 is used to compute the apparent lung-selectivity index of the test compound. Generally, compounds having an apparent lung-selectivity index greater than about 5 are preferred. Compounds of the invention are expected to have an apparent lung-selectivity index greater than 5, when tested in this or a similar assay.
ASSAY 5 Methacholine-Induced Depressor Responses in Conscious Guinea Pigs Healthy, adult, male Sprague-Dawley guinea pigs (Harlan, Indianapolis, IN), weighing between 200 and 300 g are used in these studies. Under isoflurane anesthesia (to effect), animals are instrumented with common carotid artery and jugular vein catheters (PE-50 tubing). The catheters are exteriorized utilizing a subcutaneous tunnel to the subscapular area. All surgical incisions are sutured with 4-0 Ethicon Silk and the catheters locked with heparin (1000 units/mL). Each animal is administered saline (3 mL, SC) at the end of surgery as well as buprenorphine (0.05 mg/kg, IM). Animals are allowed to recover on a heating pad before being returned to their holding rooms. Approximately 18 to 20 hours following surgery, the animals are weighed and the carotid artery catheter on each animal is connected to a transducer for recording arterial pressure. Arterial pressure and heart rate are recorded using a Biopac MP-100 Acquisition System. Animals are allowed to acclimate and stabilize for a period of 20 minutes.
Each animal is challenged with MCh (0.3 mg/kg, IY) administered through the jugular venous line and the cardiovascular response is monitored for 10 minutes. The animals are then placed into the whole body dosing chamber, which is connected to a nebulizer containing the test compound or vehicle solution. The solution is nebulized for 10 minutes using a gas mixture of breathable air and 5% carbon dioxide with a flow rate of 3 liters/minute. The animals are then removed from the whole body chamber and returned to their respective cages. At 1.5 and 24 hours post-dosing, the animals are re-challenged with MCh (0.3 mg/kg, IV) and the hemodynamic response is determined. Thereafter, the animals are euthanized with sodium pentobarbital (150 mg/kg, IV). MCh produces a decrease in mean arterial pressure (MAP) and decrease in heart rate (bradycardia). The peak decrease, from baseline, in MAP (depressor responses) is measured for each MCh challenge (before and after IH dosing). The effects of treatment on the MCh responses are expressed as % inhibition (mean +/- SEM) of the control depressor responses. Two-way ANOVA with the appropriate post-hoc test is used to test the effects of treatment and pre- treatment time. The depressor responses to MCh are expected to be relatively unchanged at 1.5 and 24 hours after inhalation dosing with vehicle. The ratio of the anti-depressor ID50 to bronchoprotective ID50 is used to compute apparent lung-selectivity of the test compound. Generally, compounds having an apparent lung-selectivity index greater than 5 are preferred. It is expected that the compounds of the invention will exhibit an apparent lung-selectivity index greater than 5, as measured in this or a similar assay.
While the present invention has been described with reference to specific aspects or embodiments thereof, it will be understood by those of ordinary skilled in the art that various changes can be made or equivalents can be substituted without departing from the true spirit and scope of the invention. Additionally, to the extent permitted by applicable patent statues and regulations, all publications, patents and patent applications cited herein are hereby incorporated by reference in their entirety to the same extent as if each document had been individually incorporated by reference herein.

Claims

CLAIMSWHAT IS CLAIMED IS:
1. A compound of formula I:
Figure imgf000095_0001
I wherein: a is 0 or an integer of from 1 to 5; each R1 is independently selected from -C1-5alkyl, -C2-5alkenyl, -C2-5alkynyl, -C3-6cycloalkyl, -cyano, -halo, -ORla, -C(O)ORlb, -SRlc, -S(O)Rld, -S(O)2R16, -NRlfRlg, -NRlhS(O)2RH, and -NRljC(0)Rlk; where each of Rla"lk is independently -H, -C1-5alkyl or phenyl-C^alkyl; b is 0 or an integer of from 1 to 4; each R2 is independently selected from -C1-5alkyl, -C2-5alkenyl, -C2-5alkynyl, -C3-6cycloalkyl, -cyano, -halo, -OR2a, -C(O)OR2b, -SR2c, -S(O)R2d, -S(O)2R2e, -NR2fR2g, -NR2hS(O)2R2i, and -NR2jC(O)R2k; where each of R2a"2k is independently -H5 -C1-5alkyl or -phenyl-C \ -5alkyl;
W is -O- or -NWa-, where Wa is -H or -C1-5alkyl; c is O or an integer from 1 to 5; each R3 independently is -C^salkyl or two R3 groups are joined to form Ci^alkylene, C2-3alkenylene or oxiran-2,3-diyl; m is O or 1;
Z is selected from -C(O)N(R4)- and -N(R4)C(0)-, where R4 is selected from -H, -Q-salkyl, and -C^scycloalkyl; s is 0, 1 or 2; Ar is a phenylene or C3-5heteroarylene group containing 1 or 2 heteroatoms independently selected from oxygen, nitrogen or sulfur; wherein the phenylene or C3-5heteroarylene group is substituted with (R5)q where q is 0 or an integer from 1 to 4 and each R5 is independently selected from -halo, -OH, -C1-5alkyl or -C^alkoxy; t is O, 1 or 2; R6 is selected from -H, -C1-5alkyl, and -X6R6a; where X6 is -C1-5alkylene and R6a is selected from -OH, -C1-5alkoxy and -heteroaryl; and
R7 is selected from -H, -Ci-salkyl, -Cs-βcycloalkyl, and -X7R7a; where X7 is selected from -C1-5alkylene, -C(O)-, -C1-5alkylene-C(O)-, -S(O2)-, -C1-5alkylene-S(O2)-, and -S(O2)-C1-5alkylene; and R7a is selected from -OH, -Q-salkyl, -C^ecycloalkyl, -C1-SaIkOXy, heteroaryl, -NR7bR7c, where R7b and R7c are independently -H or -C1-5alkyl, and aryl optionally substituted with 1-2 -Q.salkyl or halo groups; or R6 and R7 are taken together to form:
Figure imgf000096_0001
or
N/ optionally substituted with one to three -C1-5alkyl groups, or
Figure imgf000096_0002
where: d is 1 or 2; e is O, 1 or 2; f is 0, 1, 2 or 3; R8 is selected from -Q.salkyl and =0; R9 is selected from -H, -C1-5alkyl, hydroxyphenyl, heteroaryl, and -X9R9a; where X9 is selected from -C1-5alkylene, -C(O)-, -C1-5alkylene-C(O)-, -C(O)-C 1-5alkylene, -S(O2)-,
-C1-5alkylene-S(O2)-, and -S(O2)-C1-5alkylene; and R9a is selected from -H, -OH, -C1-5alkyl, -C1-5alkoxy, aryl, heteroaryl, heterocyclyl, and -NR9bR9c, where R9b and R9c are independently -H or -C1-5alkyl; wherein each alkyl and alkoxy group in R1, Rla4k, R2, R2a"2k, R3, R5, R6, R6a, R7, R7a"c, R8, R9, and R9a"c is optionally substituted with 1 to 5 fluoro substituents; or a pharmaceutically acceptable salt, solvate, or stereoisomer thereof.
2. The compound of Claim 1, wherein a, b and c each represent O.
3. The compound of Claim 1 , wherein W is -O-.
4. The compound of Claim 1 , wherein m is 0 and t is 1.
5. The compound of Claim 1 , wherein Z is selected from -NHC(O)-, -N(CH3)C(O)-, and -C(O)NH-.
6. The compound of Claim 1 , wherein Ar is selected from phenylene, phenylene substituted with methoxy, pyridylene, thienylene, pyrrolylene, and furylene.
7. The compound of Claim 6, wherein Ar is selected from phen-l,3-ylene, phen-1,4- ylene, 2-methoxyphen-l,4-ylene, 2,5-pyridylene, 2,5-thienylene, 2,4-pyrrolylene, 2,5- pyrrolylene, and 2,5-furylene.
8. The compound of Claim 3 , wherein m is 0 and t is 1.
9. The compound of Claim 8, wherein a, b, and c are 0; Z is selected from -NHC(O)-, -N(CH3)C(O)-, and -C(O)NH-; and Ar is selected from phenylene, phenylene substituted with methoxy, pyridylene, thienylene, pyrrolylene, and furylene.
10. The compound of any one of Claims 1 to 9, wherein R6 is -H and R7 is selected from -H, -Q.salkyl, -C3-6cycloalkyl, -C^salkylene-OH, -C1-5alkylene-C3-6cycloalkyl, -C^salkylene-C^salkoxy, -Ci-salkyleneheteroaryl, -C(O)-C1-SaIlCyI, -C(O)-C3-6cycloalkyl, -C(O)-heteroaryl, -C1-5alkylene-C(O)-NR7bR7c, -S(O2)-C1-5alkyl, -S(O2)-heteroaryl, -S(O2)- aryl, and -S(O2)-C1-5alkylene-NR7bR7c; where the aryl is optionally substituted with 1-2 -C1-5alkyl or halo groups, and wherein each alkyl and alkoxy group in R7 and R7b"c is optionally substituted with 1 to 5 fluoro substituents.
11. The compound of any one of Claims 1 to 9, wherein R6 is -C1-5alkyl and R7 is selected from -C1-5alkylene-OH,
Figure imgf000097_0001
-C1-5alkylene-C(O)OH, and
-C1-5alkylene-S(O2)-C1-5alkyl; and wherein each alkyl group in R7 is optionally substituted with 1 to 5 fluoro substituents.
12. The compound of any one of Claims 1 to 9, wherein R6 and R7 are different -Cμsalkyl groups.
13. The compound of any one of Claims 1 to 9, wherein R6 and R7 are the same, and are selected from: -C^alkyl, -C1-5alkylene-OH, and -d-salkylene-Q.salkoxy.
14. The compound of any one of Claims 1 to 9, wherein R6 and R7 are taken together to form:
\ /> O
-N
O
15. The compound of any one of Claims 1 to 9, wherein R6 and R7 are taken together to form:
— N O
optionally substituted with two -C1-5alkyl groups.
16. The compound of any one of Claims 1 to 9, wherein R6 and R7 are taken together to form:
Figure imgf000098_0001
where: d is 1; e is 0; f is 1; R8 is =0 and R9 is -H.
17. The compound of any one of Claims 1 to 9, wherein R6 and R7 are taken together to form:
Figure imgf000098_0002
where: d is 1; e and fare 0; and R9 is selected from -C1-5alkyl, hydroxyphenyl, heteroaryl, -C1-5alkylene-C1-5alkoxy, -C1-5alkyleneheteroaryl, -C(O)H, -C(O)-C1-5alkyl,
-C(O)-C1-5alkoxy, -C(O)-heterocyclyl, -C(O)-NR9bR9c, -C1-5alkylene-C(O)-heterocyclyl, -C1-5alkylene-C(O)-NR9bR9c, and -S(O2)-C1-5alkyl; and wherein each alkyl and alkoxy group in R9 and R9b"c is optionally substituted with 1 to 5 fluoro substituents.
18. The compound of any one of Claims 1 to 9, wherein R6 and R7 are taken together to form:
Figure imgf000098_0003
where: d is 2; e and f are 0; and R9 is -C(O)-C1_5alkyl such as methyl; and wherein the alkyl group in R9 is optionally substituted with 1 to 5 fluoro substituents.
19. The compound of any one of Claims 1 to 9, wherein R6 and R7 are taken together to form:
Figure imgf000099_0001
where: d and e are 1; f is 0; and R9 is selected from -H and -C1-5alkylenearyl; and wherein the alkyl group in R9 is optionally substituted with 1 to 5 fluoro substituents.
20. The compound of Claim 1, selected from: biphenyl-2-yl-carbamic acid l-(2-{[4-(4-acetylpiperazin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4-aminomethylbenzoyl)methylamino]ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(4-{[(furan-2-carbonyl)amino]methyl}benzoyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[4-(acetylaminomethyl)benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl-(4- {[(pyridine-4-carbonyl)amino]methyl} benzoyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(cyclopropanecarbonylamino)methyl] benzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[methyl-(4- {[(thiophene-2-carbonyl)amino] methyl}benzoyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(cyclobutanecarbonylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[4-(isopropylaminomethyl)benzoyl] methylamino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(cyclopropylmethylamino)methyl]benzoyl} methylammo)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl-(4-morpholin-4-ylmethylbenzoyl)amino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[(4- {[(2-methanesulfonylethyl)metliylamino] methyl}benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {[4-(4-ethylpiperazin-l-ylmethyl)benzoyl] methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-isopropylpiperazin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(ethylisopropylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(2,6-dimethylmorpholin-4-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(4-{[bis-(2-hydroxyethyl)amino]methyl} benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(tert-butylaminomethyl)benzoyl] methylamino }ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(ethylmethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(4-{[bis-(2-ethoxyethyl)amino]methyl}benzoyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl-{4-[(methylpyridin-3-ylmethylamino) methyljbenzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(4-{[bis(2-methoxyethyl)amino]methyl} benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- { [(2-hydroxyethyl)methylamino]methyl} benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl- {4-[(methyl-pyridin-4-yhnethylamino) methyl]benzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[4-(2-methoxyethyl)piperazin-l-ylmethyl] benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-ethanesulfonylpiperazin-l-ylmethyl) benzoyljmethylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {[4-(2,5-diaza-bicyclo[2.2. l]hept-2-yhnethyl) benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(2-hydroxyethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{methyl-[4-(4-pyridin-4-ylpiperazin-l-ylmethyl) benzoyl]amino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-dimethylcarbamoylmethylpiperazin-l- ylmethyl)benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{methyl-[4-(4-pyridin-4-ylmethylpiperazin-l- ylmethyl)benzoyl] amino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl-{4-[4-(2-oxo-2-pyrrolidin-l-ylethyl) piperazin-l-ylmethyl]benzoyl}amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(2-hydroxyethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4-ethylaminomethylbenzoyl)methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4-cyclopropylaminomethylbenzoyl) methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(2-methoxyethylamino)methyl] benzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{methyl-[4-(3-oxopiperazin-l-ylmethyl)benzoyl] amino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[4-(4-acetyl-[l,4]diazepan-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[4-(4-hydroxyphenyl)piperazin-l-ylmethyl] benzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(carbamoylmethylamino)methyl]benzoyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[4-(4-dimethylcarbamoylpiperazin-l -ylmethyl) benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl-{4-[4-(pyrrolidine-l-carbonyl)piperazin- l-ylmethyl]benzoyl}amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl- {4-[4-(piperidine-l-carbonyl)piperazin-l- ylmethyl]benzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(methyl- {4-[4-(morpholine-4-carbonyl)piperazin- 1 -ylmethyl]benzoyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4-aminomethylbenzoylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-((lR,4R)-5-benzyl-2,5-diaza-bicyclo[2.2.1] hept-2-ylmethyl)benzylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3-aminomethylbenzylcarbamoyl)ethyl]piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {[(pyridin-4-ylmethyl)amino]methyl} benzylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[(furan-2-ylmethyhnethylamino)methyl] benzylcarbamoyl} ethyl)piperidm-4-yl ester;
{[3-({3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionylamino}methyl) benzyl]methylamino} acetic acid; biphenyl-2-ylcarbamic acid 1 -[2-(3-cyclobutylaminomethylbenzylcarbamoyl) ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3-cyclopropylaminomethylbenzylcarbamoyl) ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-morpholin-4-ylmethylbenzylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-methylaminomethylbenzylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3-dimethylaminomethylbenzylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {methyl-[3-(4-methylaminomethylphenyl) propionyl] amino }ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[3-(4-ethylaminomethylphenyl)propionyl] methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4-aminomethylphenylcarbamoyl)ethyl]piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(4-trifluoromethylbenzenesulfonylamino) methyl]phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(butane-l-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; US2006/022829
biphenyl-2-ylcarbamic acid 1 -(2- {4-[(2-fluorobenzenesulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {4-[(qumoline-8-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[4-(ethanesulfonylaminomethyl) phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(2,6-dichlorobenzenesulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(toluene-4-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidm-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(thiophene-2-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[4-(benzenesulfonylaminomethyl) phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[4-(methanesulfonylaminomethyl) phenylcarb amoyl] ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[4-(trifluoromethanesulfonylaminomethyl) phenylcarb amoyl] ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(propane-l-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(propane-2-sulfonylamino)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(pyridin-2-ylmethyl)amino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(pyridin-3-ylmethyl)amino]methyl} phenylcarb amoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(pyridin-4-ylmethyl)amino]methyl} phenylcarb amoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(benzo[l,3]dioxol-5-yhnethyl)amino]methyl} ρhenylcarbamoyl)ethyl] piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(lH-indol-7-yhnethyl)amino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[4-(isobutylaminomethyl)phenylcarbamoyl] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4- {[(thiophen-2-ylmethyl)amino]methyl} ρhenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(3,3,3-trifluoropropylamino)methyl] phenylcarbamoyl}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(2-methylaminoethanesulfonylammo)methyl] phenylcarbamoyl} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[3-(4-methylaminomethylphenyl)propionylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(4-ethylaminomethylphenyl)propionylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2- {3-[4-(isopropylaminomethyl)phenyl] propionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[3-(4-cyclopropylaminomethylphenyl) propionylamino] ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {4-[(cycloproρylmethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[3-(4-cyclopentylaminomethylphenyl) propionylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{4-[(2-hydroxyethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3- {4-[(2-methoxyethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-acetyl-[l,4]diazepan-l-ylmethyl)phenyl] propionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-acetylρiperazin-l-ylmethyl)phenyl] propionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {4-[(carbamoylmethylamino)methyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-dimethylcarbamoylmethylpiperazin-l- yhnethyl)phenyl]propionylamino} ethyl)piperidin-4-yl ester;
4-[4-(2-{2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethylcarbamoyl}ethyl) benzyl]piperazine-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid 1 -(2- {3-[4-(l , 1 -dioxo-lλ6-tbiomorpholin-4-ylniethyl) phenyl]propionylamino} ethyl)ρiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[3-(4- {[bis-(2-hydroxyethyl)amino]methyl} phenyl)propionylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{3-[4-(4-dimethylcarbamoylρiperazin-l-ylmethyl) phenyljpropionylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {4-[4-(pyrrolidine- 1 -carbonyl)piperazin- 1 - ylmethyl]phenyl}propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - [2-(3 - {4- [4-(piperidine- 1 -carbonyl)piperazin- 1 - ylmethyl]phenyl}propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{4-[4-(morpholine-4-carbonyl)piperazin-l- ylmethyl]phenyl} propionylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[2-(4-methylaminomethylphenyl)acetylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[2-(4-ethylaminomethylphenyl)acetylamino] ethyl}piρeridin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2- {4-[(2-hydroxyethylamino)methyl]phenyl} acetylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2- {4-[(2-methoxyethylamino)methyl]phenyl} acetylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2- {4-[(carbamoylmethylamino)methyl]phenyl} acetylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{2-[4-(4-dimethylcarbamoylpiperazin-l- ylmethyl)phenyl]acetylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(2-methoxy-4-methylaminomethylbenzoyl) methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [2-methoxy-4-(3-oxopiperazin- 1 -ylmethyl) benzoyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {4-[(2-hydroxyethylamino)methyl]-2- methoxybenzoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2-methoxy-4-methylaminomethylbenzoylamino) ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[2-methoxy-4-(3-oxopiperazin-l-ylmethyl) benzoylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(2~hydroxyethylamino)methyl]-2- methoxybenzoylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl(5-methylammomethylpyridme-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5-diethylaminomethylpyridine-2-carbonyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]pyridine-2- carbonyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[niethyl(5-morpholin-4-ylmethylpyridine-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- {[(ruran-2-ylmethyl)amino]methyl}pyridine- 2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [5-(4-formylpiperazin- 1 -ylmethyl)pyridine-2- carbonyl]methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(5-{[bis-(2-hydroxyethyl)amino]methyl} pyridine-2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {methyl-[5-(3-oxo-piperazin- 1 -ylmethyl)pyridine- 2-carbonyl] amino} ethyl)piperidm-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetyl-[l,4]diazepan-l-ylmethyl)pyridine-
2-carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetylpiperazin-l-ylmethyl)pyridine-2- carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-methanesulfonylpiperazin-l- ylmethyl)pyridine-2-carbonyl]methylamino} ethyl)piperidin-4-yl ester;
4-[6-({2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}methylcarbamoyl) pyridin-3-yhnethyl]piperazine-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl-(5-methylaminomethylthiophene-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(5-diethylaminomethylthiophene-2-carbonyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]thiophene-2- carbonyl} methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[methyl-(5-morpholin-4-ylmethylthiophene-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- { [(furan-2-ylmethyl)amino]methyl} thiophene- 2-carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-formylpiperazin-l-ylmethyl)thiophene-2- carbonyljmethylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- {[bis-(2-hydroxy-ethyl)amino]methyl} thiophene-2-carbonyl)methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-yl-carbamic acid l-(2-{methyl-[5-(3-oxopiperazin-l-ylmethyl) thiophene-2-carbonyl] amino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetyl-[l,4]diazepan-l-ylmethyl)thiophene- 2-carbonyl]methylamino } ethyl)piperidin-4-yl ester;
4-[5-({2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}methylcarbamoyl) thiophen-2-ylmethyl]piperazine-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(l,l-dioxo-lλ6-miomorpholin-4-ylmethyl) thiophene-2-carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [5-(4-acetylpiperazin- 1 -ylmethyl)thiophene-2- carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [5-(4-methanesulfonylpiperazin- 1 -ylmethyl) thiophene-2-carbonyl]methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5-methylaminomethylthiophene-2-carbonyl) amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( {5-[(2-hydroxyethylamino)methyl]thiophene-2- carbonyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[methyl(4-methylaminomethyl-lH-pyrrole-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1- {2-[methyl-(5-methylaminomethyl-lH-pyrrole-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-acetylpiperazin-l-ylmethyl)-lH-pyrrole-2- carbonyljmethylamino} ethyl) piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]-lH-pyrrole-2- carbonyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl(5-methylaminomethylruran-2-carbonyl) amino] ethyl} piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5-diemylaminomethylfuran-2-carbonyl) methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]furan-2- carbonyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[methyl(5-morpholin-4-ylmethylfuran-2- carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- {[(furan-2-ylmethyl)amino]methyl} furan-2- carbonyl)methylamino] ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- { [5-(4-formylpiperazin- 1 -ylmethyl)furan-2- carbonyljmethylarnino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- {[bis-(2-hydroxyethyl)amino]methyl} furan-2- carbonyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-yl-carbamic acid l-(2-{methyl-[5-(3-oxopiperazin-l-ylmethyl)furan-2- carbonyl] amino } ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[5-(4-acetyl[ 1 ,4]diazepan-l -ylmethyl)furan-2- carbonyljethylamino} ethyl)piperidin-4-yl ester;
4-[5-({2-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]ethyl}methylcarbamoyl) furan-2-ylmethyl]piperazme-l-carboxylic acid methyl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(l,l-dioxo-lλ6-thiomorpholin-4-yhnethyl) furan-2-carbonyl]methylamino} ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {[5-(4-acetylpiperazin- 1 -ylmethyl)furan-2- carbonyljmethylamino} ethyl) piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{[5-(4-methanesulfonylpiperazin-l-ylmethyl) ruran-2-carbonyl]methylamino}ethyl)piperidin-4-yl ester; biphenyl-2-yl-carbamic acid 1 - {2-[(5-aminomethylruran-2-carbonyl)amino]ethyl} piperidin-4-yl ester; biphenyl-2-yl-carbamic acid 1 - {2-[(5-methylaminomethylfuran-2-carbonyl)amino] ethyl}piperidin-4-yl ester; and biphenyl-2-ylcarbamic acid l-[2-({5-[(2-hydroxyethylamino)methyl]furan-2- carbonyl} amino)ethyl]piperidin-4-yl ester; or a pharmaceutically acceptable salt or solvate thereof.
21. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of Claim 1 or 20.
22. The composition of Claim 21 , wherein the composition further comprises a therapeutically effective amount of an agent selected from β2 adrenergic receptor agonists, steroidal anti-inflammatory agents, phosphodiesterase-4 inhibitors, and combinations thereof.
23. The composition of Claim 22, wherein the composition comprises a therapeutically effective amount of a β2 adrenergic receptor agonist and a steroidal anti-inflammatory agent.
24. A process for preparing the compound of Claim 1 , comprising: (a) reacting a compound of formula II:
Figure imgf000109_0001
II or a salt thereof, with a compound of formula III:
Figure imgf000109_0002
III wherein L1 represents a leaving group; or
(b) coupling a compound of formula IVa:
Figure imgf000109_0003
IVa or a reactive derivative thereof, with a compound of formula Va: R4 t!ι-( >.*
H - sAArr Λ V K ~ NN
"R7
Va or coupling a compound of formula IVb:
Figure imgf000110_0001
IVb with a compound of formula Vb:
Figure imgf000110_0002
Vb or a reactive derivative thereof; or (c) reacting a compound of formula VI:
Figure imgf000110_0003
VI wherein L2 represents a leaving group, with a compound of formula VII:
Figure imgf000110_0004
VII or
(d) reacting a compound of formula II with a compound of formula VIII:
Figure imgf000110_0005
VIΠ in the presence of a reducing agent; or
(e) reacting a compound of formula IX:
Figure imgf000111_0001
IX with a compound of formula VII in the presence of a reducing agent; and then
(f) removing any protecting groups that may be present to provide a compound of formula I.
25. The process of Claim 24, which further comprises forming a pharmaceutically acceptable salt of the compound of formula I.
26. The product prepared by the process of Claim 24 or 25.
27. Use of a compound of Claim 1 or 20 for the manufacture of a medicament.
28. The use of Claim 27, wherein the medicament is for treatment of a pulmonary disorder.
29. The use of Claim 28, wherein the disorder is chronic obstructive pulmonary disease or asthma.
30. The use of Claim 27, wherein the medicament is for producing bronchodilation.
31. The use of Claim 27, wherein the medicament is for antagonizing a muscarinic receptor in a mammal.
32. Use of a compound of Claim 1 or 20 for studying a biological system or sample comprising a muscarinic receptor, comprising contacting the biological system or sample with the compound and determining the effects caused by the compound on the biological system or sample.
PCT/US2006/022829 2005-06-13 2006-06-12 Biphenyl compounds useful as muscarinic receptor antagonists WO2006138218A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP06772935A EP1893577A1 (en) 2005-06-13 2006-06-12 Biphenyl compounds useful as muscarinic receptor antagonists
JP2008516980A JP2008546695A (en) 2005-06-13 2006-06-12 Biphenyl compounds useful as muscarinic receptor antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US69000805P 2005-06-13 2005-06-13
US60/690,008 2005-06-13

Publications (1)

Publication Number Publication Date
WO2006138218A1 true WO2006138218A1 (en) 2006-12-28

Family

ID=37102351

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/022829 WO2006138218A1 (en) 2005-06-13 2006-06-12 Biphenyl compounds useful as muscarinic receptor antagonists

Country Status (4)

Country Link
US (4) US7728144B2 (en)
EP (1) EP1893577A1 (en)
JP (1) JP2008546695A (en)
WO (1) WO2006138218A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005087736A1 (en) * 2004-03-11 2005-09-22 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
TWI341836B (en) 2004-03-11 2011-05-11 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087737A1 (en) 2004-03-11 2005-09-22 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
EP1856112A1 (en) 2005-03-10 2007-11-21 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
US7659403B2 (en) * 2005-03-10 2010-02-09 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
TW200714587A (en) * 2005-03-10 2007-04-16 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
EP1866284A1 (en) 2005-03-10 2007-12-19 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
SG174389A1 (en) * 2009-03-17 2011-10-28 Daiichi Sankyo Co Ltd Amide derivative
WO2010119064A1 (en) * 2009-04-14 2010-10-21 Glaxo Group Limited Process for the preparation of a biphenyl-2-ylcarbamic acid ester
WO2011138916A1 (en) * 2010-05-07 2011-11-10 第一三共株式会社 Polycyclic compound
NZ604035A (en) 2010-06-04 2015-02-27 Albany Molecular Res Inc Glycine transporter-1 inhibitors, methods of making them, and uses thereof
CN107849047B (en) * 2015-09-28 2021-01-15 四川海思科制药有限公司 Biphenyl derivative, preparation method and medical application thereof
WO2022049604A1 (en) * 2020-09-05 2022-03-10 Cipla Limited Process for preparing revefenacin and intermediates therof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001042212A1 (en) 1999-12-07 2001-06-14 Theravance, Inc. Carbamate derivatives having muscarinic receptor antagonist activity
WO2005087737A1 (en) * 2004-03-11 2005-09-22 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995006635A1 (en) 1993-09-02 1995-03-09 Yamanouchi Pharmaceutical Co., Ltd. Carbamate derivative and medicine containing the same
WO1995021820A1 (en) 1994-02-10 1995-08-17 Yamanouchi Pharmaceutical Co., Ltd. Novel carbamate derivative and medicinal composition containing the same
SG80041A1 (en) 1998-06-08 2001-04-17 Advanced Medicine Inc Muscarinic receptor antagonists
US6693202B1 (en) * 1999-02-16 2004-02-17 Theravance, Inc. Muscarinic receptor antagonists
DE19933926A1 (en) * 1999-07-20 2001-01-25 Boehringer Ingelheim Pharma Biphenyl derivatives, their preparation and their use as medicines
UA73543C2 (en) * 1999-12-07 2005-08-15 Тераванс, Інк. Urea derivatives, a pharmaceutical composition and use of derivative in the preparation of medicament for the treatment of disease being mediated by muscarine receptor
WO2002051841A1 (en) 2000-12-22 2002-07-04 Almirall Prodesfarma Ag Quinuclidine carbamate derivatives and their use as m3 antagonists
US6656694B2 (en) * 2001-01-11 2003-12-02 Theravance, Inc. Method for identifying a ligand for a biological substrate
US20030018019A1 (en) * 2001-06-23 2003-01-23 Boehringer Ingelheim Pharma Kg Pharmaceutical compositions based on anticholinergics, corticosteroids and betamimetics
UY27927A1 (en) 2002-08-06 2003-12-31 Glaxo Group Ltd ACETILCOLINE M3 MUSCARINIC RECEIVER ANTAGONISTS
PE20040950A1 (en) * 2003-02-14 2005-01-01 Theravance Inc BIPHENYL DERIVATIVES AS AGONISTS OF ß2-ADRENERGIC RECEPTORS AND AS ANTAGONISTS OF MUSCARINAL RECEPTORS
CA2543858C (en) 2003-11-21 2014-04-15 Theravance, Inc. Compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
US7524962B2 (en) * 2004-03-11 2009-04-28 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
JP2007528416A (en) * 2004-03-11 2007-10-11 セラヴァンス, インコーポレーテッド Useful biphenyl compounds as muscarinic receptor antagonists
TWI341836B (en) * 2004-03-11 2011-05-11 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087736A1 (en) * 2004-03-11 2005-09-22 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
JP2007528412A (en) * 2004-03-11 2007-10-11 セラヴァンス, インコーポレーテッド Biphenyl compounds useful as muscarinic receptor antagonists
TW200538095A (en) * 2004-03-11 2005-12-01 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
US7501442B2 (en) * 2004-03-11 2009-03-10 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
EP1856112A1 (en) * 2005-03-10 2007-11-21 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
TW200714587A (en) * 2005-03-10 2007-04-16 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
EP1866284A1 (en) * 2005-03-10 2007-12-19 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
US7659403B2 (en) 2005-03-10 2010-02-09 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
WO2006099031A1 (en) * 2005-03-10 2006-09-21 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001042212A1 (en) 1999-12-07 2001-06-14 Theravance, Inc. Carbamate derivatives having muscarinic receptor antagonist activity
WO2005087737A1 (en) * 2004-03-11 2005-09-22 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists

Also Published As

Publication number Publication date
US8013152B2 (en) 2011-09-06
US20110281851A1 (en) 2011-11-17
US7728144B2 (en) 2010-06-01
JP2008546695A (en) 2008-12-25
US20120122850A1 (en) 2012-05-17
US20060281740A1 (en) 2006-12-14
US20100197667A1 (en) 2010-08-05
US8119796B2 (en) 2012-02-21
EP1893577A1 (en) 2008-03-05
US8399488B2 (en) 2013-03-19

Similar Documents

Publication Publication Date Title
EP1723114B1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
US8329911B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
US8399488B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
US8124627B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
US8501776B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087736A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723108A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
US7659403B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723110A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087732A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723115A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087734A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723113A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087763A1 (en) Diphenylmethyl compounds useful as muscarinic receptor antagonists

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2008516980

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006772935

Country of ref document: EP