WO2004074246A2 - Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity - Google Patents

Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity Download PDF

Info

Publication number
WO2004074246A2
WO2004074246A2 PCT/US2004/004449 US2004004449W WO2004074246A2 WO 2004074246 A2 WO2004074246 A2 WO 2004074246A2 US 2004004449 W US2004004449 W US 2004004449W WO 2004074246 A2 WO2004074246 A2 WO 2004074246A2
Authority
WO
WIPO (PCT)
Prior art keywords
hydroxy
piperidin
ester
ethylamino
oxo
Prior art date
Application number
PCT/US2004/004449
Other languages
French (fr)
Other versions
WO2004074246A3 (en
Inventor
Mathai Mammen
Sarah Dunham
Adam Hughes
Tae Weon Lee
Craig Husfeld
Eric Stangeland
Yan Chen
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=32912274&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2004074246(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to SI200431566T priority Critical patent/SI1615889T1/en
Priority to MXPA05008528A priority patent/MXPA05008528A/en
Priority to NZ541579A priority patent/NZ541579A/en
Priority to KR1020117010078A priority patent/KR101224497B1/en
Priority to EP04711253A priority patent/EP1615889B1/en
Priority to BRPI0407508A priority patent/BRPI0407508B8/en
Priority to AT04711253T priority patent/ATE482934T1/en
Priority to CA2515777A priority patent/CA2515777C/en
Priority to DE602004029347T priority patent/DE602004029347D1/en
Application filed by Theravance Inc. filed Critical Theravance Inc.
Priority to AU2004213411A priority patent/AU2004213411B2/en
Priority to DK04711253.7T priority patent/DK1615889T3/en
Priority to KR1020057014929A priority patent/KR101223991B1/en
Priority to JP2006503604A priority patent/JP4555283B2/en
Publication of WO2004074246A2 publication Critical patent/WO2004074246A2/en
Publication of WO2004074246A3 publication Critical patent/WO2004074246A3/en
Priority to IL169922A priority patent/IL169922A/en
Priority to IS7964A priority patent/IS2847B/en
Priority to NO20054206A priority patent/NO331947B1/en
Priority to HK06106230.1A priority patent/HK1086266A1/en
Priority to AU2010219338A priority patent/AU2010219338B2/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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/452Piperidinium derivatives
    • 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
    • A61P11/06Antiasthmatics
    • 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
    • A61P11/12Mucolytics
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/40Mineralocorticosteroids, e.g. aldosterone; Drugs increasing or potentiating the activity of mineralocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C07D211/48Oxygen atoms attached in position 4 having an acyclic carbon atom attached in position 4
    • 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
    • C07D211/56Nitrogen atoms
    • C07D211/58Nitrogen atoms attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • C07D409/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D453/00Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids
    • C07D453/02Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids containing not further condensed quinuclidine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • G01N33/9406Neurotransmitters
    • G01N33/9433(Nor)adrenaline

Definitions

  • the present invention relates to novel biphenyl derivatives that are useful for treating pulmonary disorders.
  • This invention also relates to phannaceutical compositions comprising such biphenyl derivatives, processes and intermediates for preparing such biphenyl derivatives and methods of using such biphenyl derivatives to treat pulmonary disorders.
  • bronchodilators Pulmonary disorders, such as asthma and chronic obstructive pulmonary disease (COPD), are commonly treated with bronchodilators.
  • bronchodilator in widespread use consists of ⁇ 2 adrenergic receptor (adrenoceptor) agonists, such as albuterol, formoterol and salmeterol. These compounds are generally administered by inhalation.
  • adrenoceptor ⁇ 2 adrenergic receptor
  • albuterol albuterol
  • formoterol formoterol and salmeterol
  • adrenoceptor ⁇ 2 adrenergic receptor
  • adrenoceptor ⁇ 2 adrenergic receptor
  • albuterol albuterol
  • formoterol formoterol
  • salmeterol adrenoceptor
  • muscarinic receptor antagonists anticholinergic compounds
  • compositions containing both a ⁇ 2 adrenergic receptor agonist and a muscarinic receptor antagonist are also known in the art for use in treating pulmonary disorders.
  • U.S. Patent No. 6,433,027 discloses medicament compositions containing a muscarinic receptor antagonist, such as tiotropium bromide, and a ⁇ 2 adrenergic receptor agonist, such as formoterol fumarate.
  • the present invention provides novel biphenyl derivatives that are useful for treating pulmonary disorders.
  • compounds of this invention have been found to possess both ⁇ adrenergic receptor agonist and muscarinic receptor antagonist activity.
  • the present invention is directed to a compound of formula I:
  • each R 1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR la , -C(O)OR lb , -SR lc , -S(O)R ld , -S(O) 2 R le and -NR lf R lg ; each of R la , R lb , R lc , R ld , R le , R lf and R l is independently hydrogen, (l-4C)alkyl or phenyl-(l-4C)alkyl; b is 0 or an integer of from 1 to 3; each R 2 is independently selected from ( 1 -4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl,
  • W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring and represents O or NW a ;
  • W a is hydrogen or (l-4C)alkyl;
  • c is 0 or an integer of from 1 to 4;
  • each R 3 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR 3a , -C(O)OR 3b , -SR 3c , -S(O)R 3d , -S(O) 2 R 3e and -NR 3 R 3g ; or two R 3 groups are joined to form (l-3C)alkylene, (2-3C)alkenylene or oxiran- 2,3-diyl; each of R 3a , R 3b , R 3c , R 3d , R 3e , R 3f and R 3g is independently hydrogen or (1
  • R 4 is a divalent group ofthe formula:
  • d, e, f, g, h and i are each independently selected from 0 and 1 ;
  • R 4a , R 4b , R 4 ° and R 4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl-(l-4C)alkyl;
  • a 1 and A 2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, -O-(6-10C)arylene, (6-10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O) 2 -(l-4C)alkyl, -C(O)O(l-4
  • a 3 and A 4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R 4 is attached is in the range of from 4 to 16;
  • R 5 represents hydrogen or (l-4C)alkyl
  • this invention is directed to a compound of formula II:
  • R 4 is as defined herein (including any specific or preferred embodiments); W represents O or NH; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • this invention is directed to a compound of formula III:
  • R 4 is as defined herein (including any specific or preferred embodiments);
  • W represents O or NH; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • this invention is directed to a compound of formula IN:
  • R 4 is as defined herein (including any specific or preferred embodiments);
  • W represents O or NH; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising 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.
  • Such pharmaceutical compositions may optionally contain other therapeutic agents.
  • this invention is directed to such a pharmaceutical composition wherein the composition further comprises a therapeutically effective amount of a steroidal anti-inflammatory agent, such as a corticosteroid.
  • Compounds of this invention possess both ⁇ 2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity. Accordingly, the compounds of formula I are useful for treating pulmonary disorders, such as asthma and chronic obstructive pulmonary disease.
  • this invention is directed to a method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Additionally, in another of its method aspects, this invention is directed to a method of providing bronchodilation in a patient, the method comprising administering to a patient requiring bronchodilation a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • This invention is also directed to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • this invention is directed to a method 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 both ⁇ 2 adrenergic agonist activity and muscarinic receptor antagonist activity.
  • This invention is also directed to processes and novel intermediates useful for preparing compounds of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Accordingly, in another of its method aspects, this invention is directed to a process of preparing a compound of formula I, the process comprising:
  • the above process further comprises the step of forming a pharmaceutically acceptable salt of a compound of formula I.
  • this invention is directed to the other processes described herein; and to the product prepared by any ofthe processes described herein.
  • This 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, this 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.
  • this invention is directed to novel biphenyl derivatives of formula I or pharmaceutically acceptable salts or solvates or stereoisomers thereof. These compounds contain one or more chiral centers and therefore, this invention is directed to racemic mixtures; pure stereoisomers (i.e., enantiomers or diastereomers); stereoisomer-enriched mixtures and the like unless otherwise indicated.
  • pure stereoisomers i.e., enantiomers or diastereomers
  • stereoisomer-enriched mixtures unless otherwise indicated.
  • compounds of formula I contain a chiral center at the carbon atom indicated by the symbol * in the following formula:
  • the carbon atom identified by the symbol * has the (R) configuration. In this embodiment, it is preferred for compounds of formula I to have the (R) configuration at the carbon atom identified by the symbol * or to be enriched in a stereoisomeric form having the (R) configuration at this carbon atom. In another embodiment of this invention, the carbon atom identified by the symbol * has the (S) configuration. In this embodiment, it is preferred for compounds of formula I to have the (S) configuration at the carbon atom identified by the symbol * or to be enriched in a stereoisomeric form having the (S) configuration at this carbon atom. In some cases, in order to optimize the ⁇ 2 adrenergic agonist activity ofthe compounds of this invention, it is preferred that the carbon atom identified by the symbol * has the (R) configuration.
  • 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. All such salt forms are included within the scope of this invention. Furthermore, solvates of compounds of formula I or salts thereof are included within the scope of this invention.
  • each R 1 may be at the 2, 3, 4, 5 or 6-position ofthe phenyl ring to which it is attached. In one embodiment, each R 1 is independently selected from (1-
  • R 1 4C)alkyl, halo, -OR Ia and -NR lf R lg ; such as methyl, fluoro, chloro, bromo, hydroxy, methoxy, amino, methylamino, dimethylamino and the like. Particular values for R 1 are fluoro or chloro.
  • 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 (1- 4C)alkyl, halo, -OR 2a and -NR 2f R 2g ; such as methyl, fluoro, chloro, bromo, hydroxy, methoxy, amino, methylamino, dimethylamino and the like. Particular values for R 2 are fluoro or chloro.
  • Each R la , R lb , R lc , R ld , R le , R lf and R lg and R 2a , R 2b , R 2c , R 2d , R 2e , R 2f and R 2g as used in R 1 and R 2 , respectively, is independently hydrogen, (l-4C)alkyl or phenyl-(l- 4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl or benzyl.
  • these groups are independently hydrogen or (1- 3C)alkyl.
  • these groups are independently hydrogen, methyl or ethyl.
  • W is O. In another embodiment, W is NW a . Generally, it has been found that compounds in which W represents O exhibit particularly high affinity for muscarinic and ⁇ 2 adrenergic receptors. Accordingly, in a particular embodiment of this invention, W preferably represents O.
  • W When referring to W, particular mention may be made of compounds wherein W is attached to the piperidine ring at the 4-position with respect to the nitrogen atom ofthe piperidine ring.
  • W a is hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl.
  • W a is hydrogen or (l-3C)alkyl.
  • W a is hydrogen, methyl or ethyl; such as hydrogen or methyl.
  • W a is hydrogen and NW a is NH.
  • c is 0, 1 or 2; including 0 or 1. In one embodiment, c is 0. In one embodiment, each R 3 is at the 3, 4 or 5-position on the piperidine ring
  • each R 3 is independently selected from (l-4C)alkyl; such as methyl, ethyl, n-propyl, isopropyl, n- butyl, sec-butyl, isobutyl and tert-butyl. In another aspect, each R 3 is independently methyl or ethyl.
  • R 3 is at the 1 -position ofthe piperidine ring, i.e., on the nitrogen atom ofthe piperidine ring thus forming a quaternary amine salt.
  • each R 3 is independently selected from (l-4C)alkyl; such as methyl, ethyl, n-propyl, isopropyl, n-butyl, .fee-butyl, isobutyl and tert-butyl.
  • each R 3 is independently methyl or ethyl.
  • two R 3 groups are joined to form a (l-3C)alkylene or (2-3C)alkenylene group.
  • two 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- ⁇ ositions 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).
  • 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.
  • Each R 3a , R 3b , R 3c , R 3d , R 3e , R 3f and R 3g as used in R 3 is independently hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl.
  • these groups are independently hydrogen or (l-3C)alkyl.
  • these groups are independently hydrogen, methyl or ethyl.
  • R 5 is hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert- butyl.
  • each R 5 is independently hydrogen, methyl or ethyl.
  • R 5 is hydrogen.
  • R 6 is -NR 6 CR 6b (O) and R 7 is hydrogen, where each of R 6a and R 6b is independently hydrogen or (l-4C)alkyl, such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl.
  • these groups are independently hydrogen or (l-3C)alkyl.
  • these groups are independently hydrogen, methyl or ethyl.
  • a particular value for R 6 in this embodiment is -NHCHO.
  • R 7a , R 7b , R 7 °, R 7d , R 7e , R 7f , R 7g , R 7h , R 7i , R 7j , R 7k , R 71 , R 7m , R 7n , R 7 ° and R 7p is independently hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n- propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl. In one embodiment, these groups are independently hydrogen or (l-3C)alkyl.
  • these groups are independently hydrogen, methyl or ethyl.
  • R 4 is a divalent group ofthe formula:
  • R 4a , A 1 , R 4b , Q, R 4c , A 2 , R 4d , d, e, f, g h and i are as defined herein.
  • the values of each ofthe components R 4a , A 1 , R 4b , Q, R 4c , A 2 and R 4d are selected such that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R 4 is attached is in the range of from 4 to 16, (specifically, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16); including 8, 9, 10, 11, 12, 13 or 14; such as 8, 9, 10 or 11 ; or 9 or 10.
  • values for each variable in R it will be appreciated by those skilled in the art that values should be selected such that a chemically stable group is formed.
  • each contiguous atom ofthe chain is counted consecutively starting from the first atom in the R 4 group adjacent to the nitrogen ofthe piperidine ring ending with the last atom in the R 4 group adjacent to the nitrogen of the aminohydroxyethyl group.
  • the shortest chain is used to determine the number of contiguous atoms.
  • R 4 is -(CH 2 ) 2 -NHC(O)-CH 2 -(phen-l,4-ylene)-CH 2 -
  • R 4a is selected from (l-lOC)alkylene, (2-10C)alkenylene and (2-10C)alkynylene wherein the alkylene group is unsubstituted or substituted with 1 or 2 substituents independently selected from (l-4C)alkyl, hydroxy and phenyl.
  • R 4a Representative examples of particular values for R 4a are -(CH 2 ) 2 -, -(CH 2 ) 3 -, -(CH 2 ) 4 -, -(CH 2 ) 5 -, -(CH 2 ) 6 -, -(CH 2 ) 7 -, -(CH 2 ) 8 -, -(CH 2 ) 9 -, -(CH 2 ) 10 -, -(CH 2 )CH(CH 3 )-,
  • d is 1.
  • a 1 is an optionally substituted (3-7C)cycloalkylene group; including a cyclohexylene group, such as cyclohex-l,4-ylene and cyclohex-l,3-ylene; and a cyclopentylene group, such as cyclopent- 1 ,3-ylene.
  • a 1 is an optionally substituted (6-10C)arylene group, including a phenylene group, such as phen-l,4-ylene, phen-l,3-ylene and phen-l,2-ylene; and a naphthylene group, such as naphth-l,4-ylene and napth-l,5-ylene.
  • a phenylene group such as phen-l,4-ylene, phen-l,3-ylene and phen-l,2-ylene
  • a naphthylene group such as naphth-l,4-ylene and napth-l,5-ylene.
  • a 1 is an optionally substituted (2-9C)heteroarylene group, including a pyridylene group, such as pyrid-l,4-ylene; a furylene group, such as fur- 2,5-ylene and fur-2,4-ylene; a thienylene group, such as thien-2,5-ylene and thien-2,4- ylene; and a pyrrolylene, such as pyrrol-2,5-ylene and pyrrol-2,4-ylene.
  • a pyridylene group such as pyrid-l,4-ylene
  • a furylene group such as fur- 2,5-ylene and fur-2,4-ylene
  • a thienylene group such as thien-2,5-ylene and thien-2,4- ylene
  • a pyrrolylene such as pyrrol-2,5-ylene and pyrrol-2,4-ylene.
  • a 1 is an optionally substituted (3-6C)heterocyclene group, including a piperidinylene group, such as piperidin-l,4-ylene; and a pyrrolidinylene group, such as pyrrolidin-2,5-ylene.
  • a piperidinylene group such as piperidin-l,4-ylene
  • a pyrrolidinylene group such as pyrrolidin-2,5-ylene
  • a 1 is an optionally substituted phenylene, thienylene, cyclopentylene, cyclohexylene or piperidinylene.
  • e 0.
  • R 4b is (l-5C)alkylene.
  • Representative examples of particular values for R 4b are -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 3 -, -(CH 2 ) 4 -, -(CH 2 ) 5 -; including methylene, ethylene and propylene.
  • f 0.
  • Q is selected from a bond, -N(Q a )C(O)-, -C(O)N(Q b )-, -N(Q°)S(O) 2 -, -S(O) 2 N(Q d )-, -N(Q e )C(O)N(Q f )-, -OC(O)N(Q , -N(Q )' )C(O)O- or-N(Q k ); such as where Q is a bond, -N(Q a )C(O)- or -C(O)N(Q b )-.
  • Q is a bond, O, NH, -C(O)NH-, -C(O)N(CH 3 )-, -NHC(O)-, -N(CH 3 )C(O)-, -S(O) 2 NH-, -S(O) 2 N(CH 3 )-, -NHS(O) 2 -, -N(CH 3 )S(O) 2 - and -NHC(O)NH-.
  • Another example of a value for Q, together with R 4c is -C(O)(piperidin- 1,4-ylene).
  • Q a , Q b , Q c , Q d , Q e , Q f , Q g , Q h , Q Oj and Q k are each independently selected from hydrogen and (l-6C)alkyl, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy.
  • Q 1 and Q k are each independently selected from hydrogen, and (l-3C)alkyl, including hydrogen, methyl, ethyl, n-propyl and isopropyl.
  • An example of a value for each of Q a , Q b , Q°, Q d , Q Q f , Q g , Q h , O 0 s and Q k is hydrogen.
  • Q a , Q b , Q°, Q d , Q e , Q f , Q g , Q h , Q 1 , Oj and Q k together with the nitrogen atom and the group R b or R 4c to which they are attached form a 4-6 membered azacycloalkylene group.
  • Q a and Q b together with the nitrogen atom and the group R 4b or R 4 ° to which they are attached form a piperidin-4-ylene group.
  • R 4 is a group of formula:
  • R 4 is a group of formula:
  • R 4c is (l-5C)alkylene.
  • Representative examples of particular values for R 4 ° are -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 3 -, -(CH 2 ) 4 -, -(CH 2 ) 5 -; including methylene, ethylene and propylene.
  • a 2 is an optionally substituted (3-7C)cycloalkylene group; including a cyclohexylene group, such as cyclohex-l,4-ylene and cyclohex-l,3-ylene; and a cyclopentylene group, such as cyclopent- 1,3 -ylene.
  • a 2 is an optionally substituted (6-10C)arylene group, including a phenylene group, such as phen- 1 ,4-ylene, phen- 1 ,3 -ylene and phen- 1 ,2-ylene; and a naphthylene group, such as naphth-l,4-ylene and napth-l,5-ylene.
  • a phenylene group such as phen- 1 ,4-ylene, phen- 1 ,3 -ylene and phen- 1 ,2-ylene
  • a naphthylene group such as naphth-l,4-ylene and napth-l,5-ylene.
  • a 2 is an optionally substituted (2-9C)heteroarylene group, including a pyridylene group, such as ⁇ yrid-l,4-ylene; a furylene group, such as fur- 2,5-ylene and fur-2,4-ylene; a thienylene group, such as thien-2,5-ylene and thien-2,4- ylene; and a pyrrolylene, such as pyrrol-2,5-ylene and pyrrol-2,4-ylene.
  • a pyridylene group such as ⁇ yrid-l,4-ylene
  • a furylene group such as fur- 2,5-ylene and fur-2,4-ylene
  • a thienylene group such as thien-2,5-ylene and thien-2,4- ylene
  • a pyrrolylene such as pyrrol-2,5-ylene and pyrrol-2,4-ylene.
  • a 2 is an optionally substituted (3-6C)heterocyclene group, including a piperidinylene group, such as piperidin-l,4-ylene; and a pyrrolidinylene group, such as pyrrolidin-2,5-ylene.
  • a piperidinylene group such as piperidin-l,4-ylene
  • a pyrrolidinylene group such as pyrrolidin-2,5-ylene
  • a 2 is optionally substituted phenylene, thienylene, cyclopentylene, cyclohexylene or piperidinylene.
  • a 1 or A 2 or both can be phenylene, such as phen- 1,4- ylene or phen-l,3-ylene, where the phenylene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O) 2 -(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy.
  • substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O) 2 -(l-4C)alkyl, -
  • a 1 or A 2 or both can be cyclopentylene or cyclohexylene; wherein the cyclopentylene or cyclohexylene group is unsubstituted or substituted with (l-4C)alkyl.
  • Representative examples include cz's-cyclopent- 1,3 -ylene, tr ns-cyclopent-l,3-ylene, cis- cyclohex-l,4-ylene and tr ⁇ ns-cyclohex-l,4-ylene.
  • a 1 or A 2 or both can also be optionally substituted thienylene or piperidinylene, for example, thien-2,5-ylene or piperidin-1,4- ylene.
  • R 4d is selected from (l-lOC)alkylene, (2-10C)alkenylene and (2-10C)alkynylene wherein the alkylene is unsubstituted or substituted with 1 or 2 substituents independently selected from (l-4C)alkyl, hydroxy and phenyl.
  • R 4d Representative examples of particular values for R 4d are -(CH 2 )-, -(CH 2 ) 2 -, -(CH 2 ) 3 -, -(CH 2 ) 4 -, -(CH 2 ) 5 -, -(CH 2 ) 6 -, -(CH 2 ) 7 -, -(CH 2 ) 8 -, -(CH 2 ) 9 -, -(CH 2 ) 10 - and -(CH 2 )CH(CH 3 )-(CH 2 )-C(CH 3 ) 2 - (CH 2 ) 2 -.
  • R 4 is a divalent group ofthe formula: -(R 4a ) d - where R 4a is (4-10C)alkylene. In one aspect of this embodiment, R 4 is a divalent group ofthe formula: -(CH )j- where j is 8, 9 or 10.
  • R 4 in this embodiment examples include -(CH 2 ) 4 -, -(CH 2 ) 5 -, -(CH 2 ) 6 -, -(CH 2 ) 7 -, -(CH 2 ) 8 -, -(CH 2 ) 9 , and -(CH 2 ) 10 -; including -(CH 2 ) 8 -, -(CH 2 ) 9 , and -(CH 2 ) 10 -.
  • R 4 is a divalent group ofthe formula:
  • R 4a is (l-lOC)alkylene, such as -(CH 2 )-, -(CH 2 ) 2 -, -(CH 2 ) 3 -;
  • a 2 is (6- 10C)arylene, such as phen-l,4-ylene or phen- 1,3 -ylene, or (2-9C)heteroarylene, such as thien-2,5-ylene or thien-2,4-ylene; and
  • R 4d is (l-lOC)alkylene, such as -(CH 2 )-, -(CH 2 ) 2 -, -(CH 2 ) 3 -.
  • R 4 in this embodiment examples are -(CH 2 )-(phen-l,4- ylene)-(CH 2 )-; -(CH 2 )-(phen- 1 ,4-ylene)-(CH 2 ) 2 -; -(CH 2 )-(phen- 1 ,4-ylene)-(CH 2 ) 3 -; - (CH 2 ) 2 -(phen-l,4-ylene)-(CH 2 )-; -(CH 2 ) 2 -(phen-l,4-ylene)-(CH 2 ) 2 -; -(CH 2 ) 2 -(phen-l,4- ylene)-(CH 2 ) 3 -; -(CH 2 ) 3 -(phen-l,4-ylene)-(CH 2 )-; -(CH 2 ) 3 -(phen-l,4-ylene)-(CH 2 )-; -(CH 2 ) 3 -(phen-l,4-
  • Q is -O- or -N(Q k )-;
  • Q k is hydrogen or (l-3C)alkyl, such as methyl or ethyl;
  • R 4a is (l-lOC)alkylene, such as -(CH 2 )-, -(CH 2 ) 2 -, -(CH 2 ) 3 -;
  • a 2 is (6-10C)arylene, such as phen-l,4-ylene or phen-l,3-ylene, or (2-9C)heteroarylene, such as thien-2,5-ylene or thien- 2,4-ylene;
  • R 4d is (l-lOC)alkylene, such as -(CH 2 )-, -(CH 2 ) 2 -, -(CH 2 ) 3 -.
  • R 4 in this embodiment examples are -(CH 2 ) 2 -O-(phen-l,4-ylene)-(CH 2 )-; -(CH 2 ) 2 -0-(phen-l,4-ylene)-(CH 2 ) 2 -; -(CH 2 ) 2 -O-(phen-l,4-ylene)-(CH 2 ) 3 -; -(CH 2 ) 3 -O- (phen- 1 ,4-ylene)-(CH 2 )-; -(CH 2 ) 3 -O-(phen- 1 ,4-ylene)-(CH 2 ) 2 -; -(CH 2 ) 3 -O-(phen- 1 ,4- ylene)-(CH 2 ) 3 -; -(CH 2 ) 2 -NH-(phen- 1 ,4-ylene)-(CH 2 )-; -(CH 2 ) 2 -NH-(phen- 1 ,4-ylene)-(
  • R 4 in this embodiment is the formula:
  • m is an integer from 2 to 10; and n is an integer from 2 to 10; provided that m + n is an integer from 4 to 12.
  • d and g are 1 and e, f, h and i are 0; and R 4a is -(CH 2 ) m -, R 4c is -(CH 2 ) disguise- and Q is -C(O)NH-.
  • Particular values for m are 2 or 3; and for n, 4, 5 or 6.
  • R 4 Another particular value for R 4 is the formula:
  • R 4 where o is an integer from 2 to 7; and p is an integer from 1 to 6; provided that o + p is an integer from 3 to 8.
  • d, h and i are 1 and e, f and g are 0; and R 4a is -(CH 2 )o-, A 2 is phen- 1 ,4-ylene, R 4d is -(CH 2 ) P - and Q is -C(O)NH-.
  • Particular values for o are 2 or 3; and for p, 1 or 2.
  • the phen-l,4-ylene group may be optionally substituted as defined herein for A 2 .
  • Another particular value for R 4 is the formula:
  • R 4 is an integer from 2 to 6; r is an integer from 1 to 5; and s is an integer from 1 to 5; provided that q + r + s is an integer from 4 to 8.
  • R 4a is -(CH 2 ) q -
  • R 4c is -(CH 2 )
  • a 2 is 1 ,4-phenylene
  • R 4d is -(CH 2 ) S - and Q is -C(O)NH-.
  • Particular values for q are 2 or 3; for r, 1 or 2; and for s, 1 or 2.
  • the phen-l,4-ylene group may be optionally substituted as defined herein for A 2 .
  • R 4 Another particular value for R 4 is the formula:
  • t is an integer from 2 to 10; and u is an integer from 2 to 10; provided that t + u is an integer from 4 to 12.
  • d and g are 1 and e, f, h and i are 0; and R 4a is -(CH 2 ) r , R 4c is -(CH 2 ) U - and Q is -NHC(O)-.
  • Particular values for t are 2 or 3; and for u, 4, 5 or 6.
  • R 4 Another particular value for R 4 is the formula:
  • R 4 where v is an integer from 2 to 7; and w is an integer from 1 to 6; provided that v + w is an integer from 3 to 8.
  • d, h and i are 1 and e, f and g are 0; and R 4a is -(CH 2 ) V -, A 2 is 1 ,4-phenylene, R 4d is -(CH 2 ) W - and Q is -NHC(O)-.
  • Particular values for v are 2 or 3; and for w, 1 or 2.
  • the phen- 1 ,4-ylene group may be optionally substituted as defined herein for A 2 .
  • Another particular value for R 4 is the formula:
  • x is an integer from 2 to 6; y is an integer from 1 to 5; and z is an integer from 1 to 5; provided that x + y + z is an integer from 4 to 8.
  • R 4 d, g, h and i are 1 and e and f are 0; and R 4a is -(CH 2 ) X -, R 4c is -(CH 2 ) y -, A 2 is 1,4-phenylene, R 4d is -(CH 2 ) Z - and Q is -NHC(O)-.
  • Particular values for x are 2 or 3; for y, 1 or 2; and for z,
  • the phen-l,4-ylene group may be optionally substituted as defined herein for A 2 .
  • R 4 can be selected from:
  • a particular group of compounds of formula I are those disclosed in U.S. Provisional Application No. 60/447,843, filed on February 14, 2003.
  • This group includes compounds of formula I; wherein: a is 0 or an integer of from 1 to 3; each R 1 is independently selected from (1 -4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl,
  • each of R la , R lb , R ,c , R Id , R le , R lf and R lg is independently hydrogen or (1- 4C)alkyl;
  • b is 0 or an integer of from 1 to 3;
  • each R 2 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR 2a , -C(O)OR 2b , SR 2 °, -S(O)R 2d , -S(O) 2 R 2e and -NR 2f R 2g ; each of R 2a
  • W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring, and represents O or NW a ;
  • W a is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R 3 is a substituent on carbon independently selected from (l-4C)alkyl, (2-
  • R 4a , R 4b , R 4c and R 4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from ( 1 -4C)alkyl, fluoro, hydroxy, phenyl and phenyl( 1 -4C)-alkyl;
  • a 1 and A 2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, (2-9C)heteroarylene and (3-6C)heterocyclene; wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (1- 4C)alkyl and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy;
  • Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O) 2 -, -N(Q a )C(O)-, -C(O)N(Q b )-, -N(Q c )S(O) 2 -, -S(O) 2 N(Q d )-, -N(Q e )C(O)N(Q f )-, -N(Q g )S(O) 2 N(Q h )-, -OC(O)N(Q and -N( )C(O)O-; Q a , Q , Q c , Q d , Q e , Q f , Q g , Q h , Q 1 and are each independently selected from hydrogen, (l-6C)alkyl, A 3 and (l-4C)alkylene-A
  • each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R 4 is attached is in the range of from 8 to 14;
  • R 5 represents hydrogen or (l-4C)alkyl
  • Another particular group of compounds of formula I are those disclosed in U.S. Provisional Application No. 60/467,035, filed on May 1, 2003.
  • This group of compounds includes compounds of formula I; wherein: a is 0 or an integer of from 1 to 3; each R 1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR la , -C(O)OR l , SR lc , -S(O)R ld , -S(O) 2 R le , and -NR lf R l ; each of R la , R Ib , R lc , R ld , R l ⁇ , R lf and R l is independently hydrogen or (1- 4C)alkyl; b is 0 or an integer of from 1 to 3; each R 2 is independently selected from (l-4C)alkyl,
  • W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring, and represents O or NW a ;
  • W a is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R 3 is a substituent on carbon independently selected from (l-4C)alkyl, (2- 4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR 3a , -C(O)OR 3b , SR 3c , -S(O)R 3d , -S(O) 2 R 3e , and -NR 3f R 3g ; each of R 3a , R 3b , R 3 °, R 3d , R 3e , R 3f and R 3g is independently hydrogen or (1- 4C)alkyl;
  • R 4 is a divalent group ofthe formula:
  • R 4a , R 4b , R 4 ° and R 4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl(l-4C)-alkyl;
  • a and A are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, (2-9C)heteroarylene and (3-6C)heterocyclene; wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (1- 4C)alkyl and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy;
  • Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O , -S(O) 2 -, -N(Q a )C(O)-, -C(O)N(Q b )-, -N(Q°)S(O) 2 -, -S(O) 2 N(Q d )-, -N(Q e )C(O)N(Q f )-, -N(Q g )S(O) 2 N(Q h )-, -OC(O)N(Q and
  • Q a , Q b , Q c , Q d , Q e , Q f , Q g , Q h , Q ; and OJ are each independently selected from hydrogen, (l-6C)alkyl, A 3 and (l-4C)alkylene-A 4 ; wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and ( 1 -4C)alkoxy; or together with the nitrogen atom and the group R 4b or R 4c to which they are attached, form a 4-6 membered azacycloalkylene group;
  • a 3 and A 4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl; wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R is attached is in the range of from 4 to 14;
  • R 5 represents hydrogen or (l-4C)alkyl
  • Another particular group of compounds of formula I are those where: a is 0; b is 0; c is 0; W is O; W is attached at the 4-position ofthe piperidinyl ring; R 5 is hydrogen; and R 4 , R 6 and R 7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Still another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is NH; W is attached at the 4-position ofthe piperidinyl ring; R 5 is hydrogen; and R 4 , R and R 7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Yet another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is O; W is attached at the 4-position ofthe piperidinyl ring; R 4 is -(CH 2 )j- where j is 8, 9 or 10; R 5 is hydrogen; and R 6 and R 7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is NH; W is attached at the 4-position ofthe piperidinyl ring; R 4 is -(CH 2 )j- where j is 8, 9 or 10; R 5 is hydrogen; and R 6 and R 7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Yet another particular group of compounds of fonnula I are those wherein: a is 0; b is 0; c is 0; W is O; W is attached at the 4-position ofthe piperidinyl ring; R 4 is -(CH 2 ) 2 - C(O)NH-(CH ) 5 -; R 5 is hydrogen; and R 6 and R 7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is NH; W is attached at the 4-position ofthe piperidinyl ring; R 4 is -(CH 2 ) 2 - C(O)NH-(CH 2 ) 5 -; R 5 is hydrogen; and R 6 and R 7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are those of formula II as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are those of formula III as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are those of formula IN as defined herein; or a phannaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are those of formula II, III or IN as defined herein, wherein the piperidinyl ring is substitued at the 4-position with a methyl group; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • Another particular group of compounds of formula I are compounds of formula N:
  • W, R 4 , R 6 and R 7 are as defined in Table I; or a pharmaceutically acceptable salt or solvate thereof.
  • racemic means the compound is racemic at the chiral carbon bearing the hydroxyl group in formula V, VI or NIL
  • W, R 4 , R 6 and R 7 are as defined in Table III; or a pharmaceutically acceptable salt or solvate thereof.
  • 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, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, n- 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
  • 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.
  • alkoxy means a monovalent group ofthe 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, n-but-2-enyl, n-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
  • alkynyl groups include, by way of example, ethynyl, n-propynyl, n-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
  • 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.
  • azacycloalkyl means a monovalent heterocyclic ring containing one nitrogen atom, i.e., a cycloalkyl group in which one carbon atom has been replaced with a nitrogen atom. Unless otherwise defined, such azacycloalkyl groups typically contain from 2 to 9 carbon atoms. Representative examples of an azacycloalkyl group are pyrrolidinyl and piperidinyl groups.
  • azacycloalkylene means a divalent azacycloakyl group. Representative examples of an azacycloalkylene group are pyrrolidinylene and piperidinylene groups.
  • the tenn "cycloalkyl” means a monovalent saturated carbocyclic hydrocarbon group.
  • 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, by way 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, by way 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.
  • 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 phannaceutically acceptable inorganic or organic acids.
  • Salts derived from pharmaceutically acceptable inorganic bases include ammonium, calcium, copper, ferric, fenous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like. Particularly prefened 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, N,N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- 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, glucoronic, 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 prefened 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.
  • 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.
  • a phannaceutically 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.
  • terapéuticaally effective amount means an amount sufficient to effect treatment when administered to a patient in need of treatment.
  • 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:
  • treating the disease or medical condition i.e., eliminating or causing regression ofthe disease or medical condition in a patient
  • suppressing the disease or medical condition i.e., slowing or arresting the development ofthe disease or medical condition in a patient
  • alleviating the symptoms ofthe disease or medical condition in a patient 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.
  • protected derivatives thereof means a derivative ofthe specified compound in which one or more functional groups ofthe compound are protected from undesired reactions with a protecting or blocking group.
  • 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 for carboxylic acids include esters (such as aj ⁇ -methoxybenzyl ester), amides and hydrazides; for amino groups, carbamates (such as tert-butoxycarbonyl) and amides; for hydroxyl groups, ethers and esters; for thiol groups, thioethers and thioesters; for carbonyl groups, acetals and ketals; and the like.
  • Such protecting groups are well-known to those skilled in the art and are described, for example, 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 group means a protecting group suitable for preventing undesired reactions at an amino group.
  • Representative amino-protecting groups include, but are not limited to, tert-butoxycarbonyl (BOC), trityl (Tr), benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc), formyl, trimethylsilyl (TMS), tert- butyldimethylsilyl (TBS), and the like.
  • carboxy-protecting group means a protecting group suitable for preventing undesired reactions at a carboxy group.
  • Representative carboxy-protecting groups include, but are not limited to, esters, such as methyl, ethyl, tert-butyl, benzyl (Bn), / nethoxybenzyl (PMB), 9-fluroenylmethyl (Fm), trimethylsilyl (TMS), tert- butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like.
  • hydroxyl-protecting group means a protecting group suitable for preventing undesirable reactions at a hydroxyl group.
  • Representative hydroxyl-protecting groups 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) 5 j p-methoxybenzyl (PMB), 9-fluorenylmethyl (Fm), diphenylmethyl (benzhydryl, DPM) and the like.
  • 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 biphenyl derivatives of this invention can be prepared from readily available starting materials using the following general methods and procedures or by using other information readily available to those of ordinary skill in the art. Although a particular embodiment ofthe present invention may be shown or described herein, those skilled in the art will recognize that all embodiments or aspects ofthe 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 prefened 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.
  • Protecting groups other than those illustrated in the procedures described herein may be used, if desired. For example, numerous protecting groups, and 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.
  • biphenyl derivatives of this invention can be prepared by a process comprising:
  • X Qa and X Qb each independently represent functional groups that couple to form a group Q
  • P 5a represents a hydrogen atom or an amino-protecting group
  • P 5b and P 6 each independently represent a hydrogen atom or a hydroxyl-protecting group
  • P 8 and P 9 each independently represent a hydrogen atom or a hydroxyl-protecting group
  • P 10 represents a hydrogen atom or an amino-protecting group
  • R 4 represents a residue that, together with the carbon to which it is attached, affords a group R 4 upon completion ofthe reaction
  • R 4 represents a residue that, together with the carbon to which it is attached, affords a group R 4 upon completion ofthe reaction; with a compound of formula 10 in the presence of a reducing agent; and then removing any protecting group P 1 , P 2 , P 3 , P 4 , P 5a , P 5b , P 6 , P 7 , P 8 , P 9 , P 10 , P 11 , P 12 or P to provide a compound of formula I; and optionally, forming a pharmaceutically acceptable salt thereof.
  • a salt of one ofthe starting materials 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 leaving group represented by X can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
  • the groups P 1 and P 2 can be, for example, trimethylsilyl and benzyl, respectively.
  • This reaction is typically conducted in an inert diluent, such as acetonitrile, in the presence of a base.
  • this reaction can be conducted in the presence of a tertiary amine, such as diisopropylethylamine.
  • this reaction is conducted at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
  • the reaction product is then isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • P 14 represents an amino-protecting group, such as a benzyl group.
  • a benzyl group can be readily removed by reduction using, for example, hydrogen or ammonium formate and a group NIII metal catalyst, such as palladium on carbon.
  • W represents ⁇ W a
  • the hydrogenation reaction is conveniently performed using Pearlman's catalyst (i.e., Pd(OH) 2 ).
  • Compounds of formula 12 can be prepared by reacting an isocyanate compound of formula 13:
  • hydroxyl group of a compound of formula 23 can be readily converted into a leaving group using well-known reagents and procedures.
  • a hydroxyl group can be converted into a halo group using an inorganic acid halide, such as thionyl chloride, phosphorous trichloride, phosphorous tribromide, phosphorous oxychloride and the like, or a halogen acid, such a hydrogen bromide.
  • the leaving represented by X 2 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
  • the groups P 3 and P 4 can be, for example, tert-butyldimethylsilyl and benzyl, respectively.
  • This reaction is typically conducted in the presence of a base, such as sodium bicarbonate, and an alkali metal iodide, such as sodium iodide.
  • P 15 and P 16 independently represents a protecting group, such as tert-butoxycarbonyl, and any remainder represents a hydrogen atom.
  • a tert-butoxycarbonyl group can be removed by treating the protected compound with trifluoroacetic acid.
  • X represents a leaving group such as halo, such as chloro, bromo or iodo, or sulfonic ester group, such as mesylate or tosylate.
  • This reaction is typically conducted by contacting a compound of formula 1 with a compound of fonnula 16 in an inert diluent, such as acetonitrile, DMF or mixtures thereof, at a temperature ranging from about 0 °C to about 100 °C until the reaction is substantially complete.
  • an inert diluent such as acetonitrile, DMF or mixtures thereof
  • compounds of formula 3 can be obtained by reductive amination of a compound of formula 11.
  • the reductive amination can be performed by reacting the compound of formula 11 with, for example, benzylamine and hydrogen in the presence of palladium on carbon.
  • oxidizing agent such as sulfur trioxide pyridine complex and dimethyl sulfoxide.
  • This oxidation reaction is typically conducted in an inert diluent, such as dichloromethane, the presence of a tertiary amine, such as diisopropylethylamine, at a temperature ranging from about -20 °C to about 25 °C.
  • Compounds of fonnula 17 can be prepared by reacting a compound of formula 1 with a compound of fonnula 18:
  • X 4 represents a leaving group such as halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
  • compounds of formula 19 can be reduced in the presence of a chiral catalyst formed from (R)-(+)- ⁇ , ⁇ -diphenyl-2-pynolidinemethanol and trimethylboroxine; or alternatively, from (_S)-(-)- ⁇ , ⁇ -diphenyl-2-pyreolidinemethanol and trimethylboroxine.
  • a chiral catalyst formed from (R)-(+)- ⁇ , ⁇ -diphenyl-2-pynolidinemethanol and trimethylboroxine; or alternatively, from (_S)-(-)- ⁇ , ⁇ -diphenyl-2-pyreolidinemethanol and trimethylboroxine.
  • the resulting hydroxyl group can then be protected with a hydroxyl-protecting group, P , by reaction with, for example, tert-butyldimethylsilyl trifluoromethanesulfonate.
  • one of X Qa and X Qb can be an amine group (i.e., -NHQ a or -NHQ b ) and the other can be a carboxyl group (i.e., -COOH) or a reactive derivative thereof (such as acyl halide, such as an acyl chloride or acyl bromide).
  • the groups P 5a , P 5b and P 6 can be, for example, benzyl, trimethylsilyl and benzyl, respectively.
  • the reaction can be performed under conventional amide coupling conditions.
  • Q is a sulfonamide, i.e., -N(Q c )S(O) 2 - or -S(O) 2 N(Q d )-
  • one of X Qa and X Qb can be an amine group, -NHQ C or -NHQ d and the other can be a sulfonyl halide group (such as sulfonyl chloride or sulfonyl bromide).
  • X 5 represents a leaving group including halo, such as chloro, bromo or iodo, and a sulfonic ester group, such as mesylate or tosylate
  • X Qa represents X Qa , such as a carboxyl group or an amino group NHQ a , or a protected derivative thereof, such as a (1- 6C)alkoxycarbonylamino group or a tert-butoxycarbonylamino group.
  • X 6 represents a leaving group including halo, such as chloro, bromo or iodo, and a sulfonic ester group, such as mesylate or tosylate
  • X Qb represents X Q , such as a carboxyl group or an amino group NHQ b , or a protected derivative thereof, such as a (1- 6C)alkoxycarbonyl group or a tert-butoxycarbonylamino group.
  • any suitable reducing agent may be used in this reaction.
  • the reducing agent can be hydrogen in the presence of a Group NIII metal catalyst, such as palladium on carbon; or a metal hydride reagent, such as sodium triacetoxyborohydride.
  • the group P 7 can be, for example, benzyl.
  • This reaction is typically conducted in an inert diluent and a protic solvent, such as a mixture of dichloroethane and methanol, at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
  • Compounds of formula 7 in the form of a hydrate can be prepared by conventional procedures, for example, by dibrominating a compound of formula 19 (where X 2 in this case can also be hydrogen), and then hydrolyzing the resulting dibromide to form a glyoxal or a hydrate thereof.
  • a compound of formula 19 can be reacted with hydrogen bromide and then hydrolyzed with water to form the corresponding glyoxal hydrate.
  • any suitable reducing agent may be used in this reaction.
  • the reducing agent may be hydrogen in the presence of a Group NIII metal catalyst, such as palladium on carbon; or a metal hydride reagent, such as sodium triacetoxyborohydride.
  • the groups P 8 , P 9 and P 10 can be, for example, trimethylsilyl, benzyl and benzyl, respectively.
  • this reduction reaction is conducted in an inert diluent and a protic solvent, such as dichloroethane and methanol, at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
  • oxidizing agent such as sulfur trioxide pyridine complex and dimethyl sulfoxide.
  • This reaction is typically conducted in the presence of a tertiary amine, such as diisopropylethylamine, at a temperature in the range of from about -20 °C to about 25 °C until the oxidation is substantially complete.
  • X 7 represents a leaving group including halo, such as chloro, bromo or iodo, and a sulfonic ester group, such as mesylate or tosylate.
  • the leaving group represented by X 3 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
  • the groups P 11 , P 12 and P 13 can be, for example, trimethylsilyl, benzyl and benzyl, respectively.
  • This reaction is typically conducted an inert diluent, such as acetonitrile, in the presence of a suitable base.
  • this reaction can be conducted in the presence of a tertiary amine, such as diisopropylethylamine. Generally, this reaction is conducted at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
  • a tertiary amine such as diisopropylethylamine.
  • this reaction is conducted at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
  • Compounds of formula 9 can be prepared by steps analogous to those of methods
  • compounds of fonnula 10 can be prepared from compounds of formula 4 by reaction with an amine of formula P I3 NH 2 .
  • any suitable reducing agent may be used in this reaction.
  • the reducing agent may be hydrogen in the presence of a Group NIII metal catalyst, such as palladium on carbon; or a metal hydride reagent, such as sodium triacetoxyborohydride.
  • the groups P 11 , P 12 and P 13 can be, for example, tert- butyldimethylsilyl, benzyl and benzyl, respectively.
  • this reduction reaction is conducted in an inert diluent and a protic solvent, such as dichloroethane and methanol, at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
  • a protic solvent such as dichloroethane and methanol
  • P 17 represents a hydrogen atom or an amino-protecting group
  • each of P 18 , P 19 and P 20 independently represent a hydrogen atom or a hydroxyl-protecting group; provided that at least one of P 17 , P 18 , P 19 or P 20 is a protecting group; (i) deprotecting a compound of formula 26:
  • P 21 represents a hydrogen atom or an amino-protecting group
  • each of P 22 and P 23 independently represent a hydrogen atom or a hydroxyl-protecting group; provided that at least one of P , P or P is a protecting group; or (j) deprotecting a compound of formula 27:
  • P 24 represents a hydrogen atom or an amino-protecting group
  • each of P 25 and P 26 independently represent a hydrogen atom or a hydroxyl-protecting group
  • benzyl protecting groups are conveniently removed by catalytic hydrogenation in the presence of a Group NIII metal catalyst, such as palladium on carbon; a tert-butyldimethylsilyl group is conveniently removed by treatment with hydrogen fluoride, such as triethylamine trihydrofluoride; and a propylidine group is conveniently removed by treatment with an acid, such as trifluoroacetic acid.
  • a Group NIII metal catalyst such as palladium on carbon
  • hydrogen fluoride such as triethylamine trihydrofluoride
  • a propylidine group is conveniently removed by treatment with an acid, such as trifluoroacetic acid.
  • R 8 represents -CH 2 OP 19 , -CHO, -COOH or -C(O)O(l-6C)alkoxy, such as carbomethoxy
  • R 9 represents -OP 18 and R 10 represents a hydrogen atom
  • Any suitable reducing agent may be used in this reaction including, by way of example, metal hydride reducing agents, such as sodium borohydride, lithium aluminum hydride and the like.
  • X represents a leaving group, such as a bromo.
  • examples of particular values for P , P and P are: for P 21 , hydrogen or benzyl; for P 22 hydrogen or tert-butyldimethylsilyl; and for P 23 hydrogen or benzyl.
  • benzyl protecting groups are conveniently removed by catalytic hydrogenation in the presence of a Group NIII metal catalyst, such as palladium on carbon; and a tert-butyldimethylsilyl group is conveniently removed by treatment with hydrogen fluoride, such as triethylamine trihydrofluoride.
  • Compounds of formula 26 can be prepared by the methods described herein, such as by processes (a) to (g).
  • examples of particular values for P , P and P are: for P , hydrogen or benzyl; for P hydrogen or tert-butyldimethylsilyl; and for P hydrogen or benzyl.
  • benzyl protecting groups are conveniently removed by catalytic hydrogenation in the presence of a Group NIII metal catalyst, such as palladium on carbon; and a tert-butyldimethylsilyl group is conveniently removed by treatment with hydrogen fluoride, such as triethylamine trihydrofluoride.
  • Compounds of formula 27 can be prepared by the methods described herein, such as by processes (a) to (g).
  • the biphenyl derivatives of this invention are typically administered to a patient in the form of a pharmaceutical composition or formulation.
  • Such pharmaceutical compositions may be administered to the patient by any acceptable route of administration including, but not limited to, inhaled, oral, nasal, topical (including transdermal) and parenteral modes of administration.
  • any form ofthe compounds of this invention, (i.e., free base, pharmaceutically acceptable salt, solvate, etc.) that is suitable for the particular mode of administration can be used in the pharmaceutical compositions discussed herein.
  • this 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 thereof.
  • such pharmaceutical compositions may contain other therapeutic and/or formulating agents if desired.
  • compositions of this invention typically contain a therapeutically effective amount of a compound ofthe present invention or a pharmaceutically acceptable salt thereof.
  • pharmaceutical compositions will contain from about 0.01 to about 95% by weight ofthe active agent; including, from about 0.01 to about 30% by weight; such as from about 0.01 to about 10% by weight ofthe active agent.
  • any conventional carrier or excipient may be used in the pharmaceutical compositions of this invention.
  • the choice of a particular carrier or excipient, or combinations of carriers or exipients, 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 pharmaceutical 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 l Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
  • compositions which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium magnesium
  • compositions of this invention are typically prepared by thoroughly and intimately mixing or blending a compound ofthe invention 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.
  • the pharmaceutical compositions of this invention are suitable for inhaled administration. Suitable pharmaceutical compositions for inhaled administration will typically be in the form of an aerosol or a powder.
  • Such compositions are generally administered using well-known delivery devices, such as a nebulizer inhaler, a metered-dose inhaler (MDI), a dry powder inhaler (DPI) or a similar delivery device.
  • the pharmaceutical composition comprising the active agent is administered by inhalation using a nebulizer inhaler.
  • a nebulizer inhaler typically produce a stream of high velocity air that causes the pharmaceutical 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 ofthe particles with a diameter of less than about 10 ⁇ m.
  • Suitable nebulizer devices are provided commercially, for example, by PARI GmbH (Stamberg, German). Other nebulizer devices include Respimat (Boehringer Ingelheim) and those disclosed, for example, in U.S. Patent No. 6,123,068 and WO 97/12687.
  • 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/mL of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the pharmaceutical composition comprising the active agent is administered by inhalation using a dry powder inhaler.
  • dry powder inhalers 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.
  • a representative pharmaceutical composition for use in a dry powder inhaler 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.
  • Such 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 phannaceutical 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 dry powder inhaler delivery devices include Diskhaler (GlaxoSmithKline, Research Triangle Park, NC) (see, e.g., U.S. Patent No. 5,035,237); Diskus (GlaxoSmithKline) (see, e.g., U.S. Patent No. 6,378,519; Turbuhaler (AstraZeneca, Wilmington, DE) (see, e.g., U.S. Patent No. 4,524,769); Rotahaler (GlaxoSmithKline) (see, e.g., U.S. Patent No. 4,353,365) and Handihaler (Boehringer Ingelheim). Further examples of suitable DPI devices are described in U.S. Patent Nos.
  • the pharmaceutical composition comprising the active agent is administered by inhalation using a metered- dose inhaler.
  • metered-dose inhalers typically discharge a measured amount ofthe active agent or a pharmaceutically acceptable salt thereof using compressed propellant gas.
  • pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension ofthe active agent in a liquefied propellant.
  • Any suitable liquefied propellant may be employed including chlorofluorocarbons, such as CC1 3 F, and hydro fluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoro-n-propane, (HFA 227).
  • chlorofluorocarbons such as CC1 3 F
  • HFAs hydro fluoroalkanes
  • HFA 134a 1,1,1,2-tetrafluoroethane
  • HFA 227 1,1,1,2,3,3,3-heptafluoro-n-propane
  • co-solvents such as ethanol or pentane
  • surfactants such as sorbitan trioleate, oleic acid, lecithin, and glycerin. See, for example, U.S.
  • a representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01 % to about 5 % by weight of a compound of formula I, or a phannaceutically acceptable salt or solvate or stereoisomer thereof; from about 0 % to about 20 % by weight ethanol; and from about 0 % to about 5 % by weight surfactant; with the remainder being an HFA propellant.
  • 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).
  • 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 metered-dose inhaler devices developed specifically for use with HFA propellants are provided in U.S. Patent Nos. 6,006,745 and 6,143,277.
  • a suspension formulation can be prepared by spray drying a coating of surfactant on micronized particles ofthe active agent. See, for example, WO 99/53901 and WO 00/61108.
  • compositions of this invention are suitable for oral administration.
  • suitable pharmaceutical 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 ofthe present invention as an active ingredient.
  • the pharmaceutical compositions of this invention When intended for oral administration in a solid dosage form (i.e., as capsules, tablets, pills and the like), the pharmaceutical compositions of this invention will typically comprise a compound ofthe present invention as the active ingredient and one or more pharmaceutically acceptable earners, such as sodium citrate or dicalcium phosphate.
  • a pharmaceutically acceptable earners such as sodium citrate or dicalcium phosphate.
  • such solid dosage forms may also comprise: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar- agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and/or sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as cetyl alcohol and/or glycerol monostearate; (8) absorbents, such as kaolin and/or bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid poly
  • antioxidants include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) 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), butylated hydroxytoluene (BHT), lecithin
  • Coating agents for tablets, capsules, pills and like include those used for enteric coatings, such as cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PNAP), 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 (PNAP), 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.
  • the pharmaceutical compositions ofthe present invention may also be formulated to provide slow or controlled release ofthe active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres.
  • the pharmaceutical compositions of the present invention may optionally contain opacifying agents and may be formulated so that they release the active ingredient only, or preferentially, in a certain portion ofthe 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 ofthe above-described excipients.
  • Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • Such 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-butyl ene glycol, oils (esp., cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • an inert diluent such as, for example, water or other solvents, solubilizing agents and emul
  • 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.
  • the pharmaceutical compositions of this 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, tablets, pills, and the like.
  • the compounds of this invention can also be administered transdermally using known transdermal delivery systems and excipents.
  • a compound of this invention can be admixed with permeation enhancers, such as propylene glycol, polyethylene glycolm 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 pharmaceutical compositions of this invention may also contain other therapeutic agents that are co-administered with a compound of formula I, or pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the phannaceutical compositions of this invention may further comprise one or more therapeutic agents selected from other bronchodilators (e.g., PDE 3 inhibitors,adenosine 2b modulators and ⁇ 2 adrenergic receptor agonists); anti-inflammatory agents (e.g. steroidal anti-inflammatory agents, such as corticosteroids; non-steroidal anti-inflammatory agents (NSAIDs), and PDE 4 inhibitors); other muscarinic receptor antagonists (i.e., antichlolinergic agents); antiinfective agents (e.g.
  • other bronchodilators e.g., PDE 3 inhibitors,adenosine 2b modulators and ⁇ 2 adrenergic receptor agonists
  • anti-inflammatory agents e.g. steroidal anti-inflammatory agents, such as corticosteroids; non-steroidal anti-inflammatory agents (NSAIDs), and PDE 4 inhibitors
  • other muscarinic receptor antagonists i.e., antichloline
  • the other therapeutic agents can be used in the form of pharmaceutically acceptable salts or solvates. Additionally, if appropriate, the other therapeutic agents can be used as optically pure stereoisomers.
  • Representative ⁇ adrenergic receptor agonists that can be used in combination with, and in addition to, the compounds of this invention 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 this 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, published on August 29, 2002; 3-[3-(4- ⁇ [6- ([(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]ethyl ⁇ amino)hexyl]oxy ⁇ butyl)- phenyl]imidazolidine-2,4-d
  • the ⁇ 2 -adrenoreceptor agonist When employed, the ⁇ 2 -adrenoreceptor agonist will be present in the pharmaceutical composition in a therapeutically effective amount. Typically, the ⁇ 2 -adrenoreceptor agonist will be present in an amount sufficient to provide from about 0.05 ⁇ g to about 500 ⁇ g per dose.
  • steroidal anti-inflammatory agents that can be used in combination with the compounds of this invention 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 S-fluoromethyl ester, 6,9-difluoro- 11 -hydroxy- 16 -methyl-3-oxo-17 -propionyloxy- androsta-l,4-diene-l 7-carbothioic acid 5 , -(2-oxo-tetrahydrofuran-3S-yl) ester, beclomethasone esters (e.g.
  • the steroidal anti- inflammatory 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 ⁇ g to about 500 ⁇ g per dose.
  • 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 ( IL antibody), specifically, an IL-4 therapy, an IL-13 therapy, or a combination thereof); or inhibitors of cytokine synthesis.
  • IL antibody interleukin antibody
  • IL-4 therapy an interleukin antibody
  • IL-13 therapy an IL-13 therapy
  • representative phosphodiesterase-4 (PDE4) inhibitors or mixed PDE3/PDE4 inhibitors that can be used in combination with the compounds of this invention include, but are not limited to cis 4-cyano-4-(3-cyclopentyloxy-4- methoxyphenyl)cyclohexan- 1 -carboxylic acid, 2-carbomethoxy-4-eyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan- 1 -one; c/s-[4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan- 1 -ol] ; czs-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 (LNSERM); 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-l 1294A (Napp); roflumilast (Byk-Gulden); pthalazinone compounds disclosed in WO99/47505 (Byk-Gulden); Pumafentrine (Byk-Gulden, now Altana); arofylline (Almirall- Prodesfarma); NM554/UM565 (Nernalis); 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, l) hydrobromide, scopolamine hydrobromide, ipratropium bromide, oxitropium bromide, tiotropium bromide, methantheline, propantheline bromide, anisotropine methyl bromide, clidinium bromide, copynolate (Robinul), isopropamide iodide, mepenzolate bromide, tridihexethyl chloride (Pathilone), hexocyclium methylsulfate, cyclopentolate hydrochloride, tropicamide, trihexyphenidyl hydrochloride
  • antihistamines i.e., Hi-receptor antagonists
  • ethanolamines such as carbinoxamine maleate, clemastine fumarate, diphenylhydramine hydrochloride and dimenhydrinate
  • ethylenediamines such as pyrilamine amleate, tripelennamine 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
  • ethanolamines such as carbinoxamine maleate, clemastine fumarate
  • Formulation Example A A dry powder for administration by inhalation is prepared as follows: Ingredients Amount
  • the compound ofthe invention is micronized and then blended with lactose. This blended mixture is then loaded into a gelatin inhalation cartridge. The contents ofthe cartridge are administered using a powder inhaler.
  • Formulation Example B A dry powder formulation for use in a dry powder inhalation device is prepared as follows:
  • a pharmaceutical composition is prepared having a bulk formulation ratio of micronized compound ofthe invention to lactose of 1 :200.
  • the composition is packed into a dry powder inhalation device capable of delivering between about 10 ⁇ g and about 100 ⁇ g ofthe compound ofthe invention per dose.
  • Formulation Example C A dry powder for administration by inhalation in a metered dose inhaler is prepared as follows:
  • a suspension containing 5 wt. % of a compound ofthe invention and 0.1 wt. % lecithin is prepared by dispersing 10 g ofthe compound ofthe invention as micronized particles with mean size less than 10 ⁇ m in a solution formed from 0.2 g of lecithin dissolved in 200 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 cartridges with pressurized 1,1,1,2-tetrafluoroethane.
  • a pharmaceutical composition for use in a metered dose inhaler is prepared as follows: Representative Procedure: A suspension containing 5 % compound ofthe invention, 0.5 % lecithin, and 0.5 % trehalose is prepared by dispersing 5 g of active ingredient as micronized particles with 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 for use in a nebulizer inhaler is prepared as follows: Representative Procedure: An aqueous aerosol formulation for use in a nebulizer is prepared by dissolving 0.1 mg ofthe compound ofthe invention in 1 mL of a 0.9 % sodium chloride solution acidified with citric acid. The mixture is stined and sonicated until the active ingredient is dissolved. The pH ofthe solution is adjusted to a value in the range of from 3 to 8 by the slow addition of NaOH.
  • Hard gelatin capsules for oral administration are prepared as follows:
  • Neegum k (Nanderbilt Co.) 1.0 g
  • An injectable formulation is prepared as follows: Ingredients Amount
  • the biphenyl derivatives of this invention possess both ⁇ 2 adrenergic receptor agonist and muscarinic receptor antagonist activity and therefore, such compounds are useful for treating medical conditions mediated by ⁇ 2 adrenergic receptors or muscarinic receptors, i.e., medical conditions that are ameliorated by treatment with a ⁇ 2 adrenergic receptor agonist or a muscarinic receptor antagonist.
  • medical conditions include, by way of example, pulmonary disorders or diseases associated with reversible airway obstruction, such as chronic obstructive pulmonary disease (e.g., chronic and whez bronchitis and emphysema), asthma, pulmonary fibrosis and the like.
  • Other conditions which may be treated include premature labor, depression, congestive heart failure, skin diseases (e.g., inflammatory, allergic, psoriatic and proliferative skin diseases, conditions where lowering peptic acidity is desirable (e.g., peptic and gastric ulceration) and muscle wasting disease.
  • skin diseases e.g., inflammatory, allergic, psoriatic and proliferative skin diseases, conditions where lowering peptic acidity is desirable (e.g., peptic and gastric ulceration) and muscle wasting disease.
  • this invention is directed to a method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the compounds of this invention When used to treat a pulmonary disorder, the compounds of this 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 ⁇ g/day to about 200 ⁇ g/day.
  • this invention is directed to a method of providing bronchodilation in a patient, the method comprising administering to a patient requiring bronchodilation a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the dose for providing bronchodilation will range from about 10 ⁇ g/day to about 200 ⁇ g/day.
  • this invention is directed to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
  • the compounds of this invention When used to treat a COPD or asthma, the compounds of this invention will typically be administered by inhalation in multiple doses per day or in a single daily dose. Generally, the dose for treating COPD or asthma will range from about 10 ⁇ g/day to about 200 ⁇ g/day.
  • COPD includes chronic obstructive bronchitis and emphysema (see, for example, Barnes, Chronic Obstructive Pulmonary Disease, NEngl JMed 2000: 343:269-78).
  • the compounds of this invention are optionally administered in combination with other therapeutic agents.
  • a steroidal anti-inflammatory agent e.g. a corticosteroid
  • the pharmaceutical compositions of this invention can provide triple therapy, i.e., ⁇ 2 adrenergic receptor agonist, muscarinic receptor antagonist and anti- inflammatory activity, using only two active components. Since pharmaceutical compositions containing two active components are typically easier to formulate compared to compositions containing three active components, such two component compositions provide a significant advantage over compositions containing three active components. Accordingly, in a particular embodiment, the pharmaceutical compositions and methods of this invention further comprise a therapeutically effective amount of a steroidal anti- inflammatory agent.
  • compounds of this invention exhibit both muscarinic receptor antagonist and ⁇ 2 adrenergic receptor agonist activity. Accordingly, among other properties, compounds of particular interest are those that demonstrate an inhibitory constant K; value for binding at the M 3 muscarinic receptor and an EC 50 value for ⁇ 2 adrenergic receptor agonist activity of less than about 100 nM; particularly less than 10 nM.
  • compounds of special interest include those having muscarinic activity, expressed in terms ofthe inhibitory constant Kj for binding at the M 3 muscarinic receptor, that is about equal to the compound's ⁇ 2 adrenergic agonist activity, expressed in terms ofthe half maximal effective concentration EC 50 , as determined in the in vitro assays described herein, or in similar assays.
  • compounds of particular interest are those having a ratio of the inhibitory constant Kj for the M 3 muscarinic receptor to the EC 50 for the ⁇ 2 adrenergic receptor ranging from about 30:1 to about 1:30; including about 20:1 to about 1:20; such as about 10:1 to about 1:10.
  • the present invention also provides a method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound having both muscarinic receptor antagonist and ⁇ 2 adrenergic receptor agonist activity.
  • the compound administered has an inhibitory constant K; for the M 3 muscarinic receptor that is less than about 100 nM and a half maximal effective concentration EC 50 for agonism at the ⁇ 2 adrenergic receptor that is less than about 100 nM.
  • the method for treating a pulmonary disorder comprises administering a therapeutically effective amount of a compound for which the ratio ofthe inhibitory constant K; for the M 3 muscarinic receptor to the EC 50 for agonism ofthe ⁇ 2 adrenergic receptor is between about 30: 1 and about 1 :30.
  • compounds of this invention possess both ⁇ 2 adrenergic agonist activity and muscarinic receptor antagonist activity, such compounds are also useful as research tools for investigating or studying biological systems or samples having ⁇ 2 adrenergic receptors or muscarinic receptors, or for discovering new compounds having both ⁇ 2 adrenergic agonist activity and muscarinic receptor antagonist activity.
  • biological systems or samples may comprise ⁇ 2 adrenergic receptors and/or muscarinic receptors. Any suitable biological system or sample having ⁇ 2 adrenergic and/or 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.
  • a biological system or sample comprising a ⁇ 2 adrenergic receptor or a muscarinic receptor is contacted with a ⁇ 2 adrenergic receptor-agonizing or muscarinic receptor-antagonizing amount of a compound of this invention.
  • the effects 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 ofthe enzyme adenylyl cyclase (which synthesizes cAMP), ligand- mediated changes in incorporation of guanosine 5'-O-( -thio)triphosphate ([ 35 S]GTP S) into isolated membranes via receptor catalyzed exchange of [ 35 S]GTP S 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
  • ligand- mediated changes in incorporation of guanosine 5'-O-( -thio)triphosphate [ 35 S]GTP S
  • a compound of this invention will agonize or cause activation of a ⁇ 2 adrenergic receptor and antagonize or decrease the activation of muscarinic receptors in any ofthe functional assays listed above, or assays of a similar nature.
  • the amount of compound used in these studies will typically range from about 0.1 nanomolar to about 100 nanomolar.
  • the compounds of this invention can be used as research tools for discovering new compounds that have both a ⁇ 2 adrenergic receptor agonist and muscarinic receptor antagonist activity.
  • a ⁇ 2 adrenergic receptor and muscarinic receptor binding data for example, as determined by in vitro radioligand displacement assays
  • a compound of this invention is compared to the ⁇ 2 adrenergic receptor and muscarinic receptor binding data for a compound of this invention to identify those test compounds that have about equal or superior ⁇ 2 adrenergic receptor and/or 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 ofthe test data to identify test compounds of interest.
  • compounds of this invention may possess either weak muscarinic receptor antagonist activity or weak ⁇ 2 adrenergic receptor agonist activity. In these cases, those of ordinary skill in the art will recognize that such compounds still have utility as primarily either a ⁇ 2 adrenergic receptor agonist or a muscarinic receptor antagonist, respectively.
  • HPLC 10-70 data was obtained with a flow rate of 0.5 mL/minute of 10%-70% B over 6 minutes.
  • Mobile phase A was 2 % - 98 % - 0.1% AC ⁇ -H 2 O-TFA; and mobile phase B was 90 % - 10 % - 0.1 % ACN-H 2 O-TFA.
  • HPLC 5-35 data and HPLC 10-90 data were obtained with a 5 minute gradient.
  • LCMS Liquid chromatography mass spectrometry
  • N-1,1 '-biphenyl-2-yl-iV-4-piperidinylurea was synthesized by heating together biphenyl-2-isocyanate (50 g, 256 mmol) and 4-amino-N-benzylpiperidine (51.1 g, 269 mmol) at 70 °C for 12 h (the reaction was monitored by LCMS). The reaction mixture was cooled to 50 °C and ethanol (500 mL) added, followed by slow addition of 6M hydrochloric acid (95 mL). The reaction mixture was cooled to room temperature.
  • step (b) 5-Acetyl-8-hydroxy-lH-quinolin-2-one
  • a sluny of aluminum chloride (85.7 g, 640 mmol) in 1 ,2-dichloroethane (280 mL) was cooled in ice, and the product of step (a) (56.8 g, 280 mmol) was added.
  • the mixture was warmed to room temperature and then heated at 85°C. After 30 min, acetyl chloride (1.5 mL, 21 mmol) was added and the mixture was heated an additional 60 min.
  • the reaction mixture was then cooled and added to IN hydrochloric acid (3 L) at 0 °C with good stirring.
  • step (b) To the product of step (b) (37.7 g, 186 mmol) was added N,N-dimethylformamide (200 mL) and potassium carbonate (34.5 g, 250 mmol) followed by benzyl bromide (31.8 g, 186 mmol). The mixture was stined at room temperature for 2.25 hour and then poured into saturated sodium chloride (3.5 L) at 0 °C and stined for 1 hour. The product was collected and dried on a Buchner funnel for 1 hour, and the resulting solids were dissolved in dichloromethane (2 L) and this mixture was dried over sodium sulfate.
  • N,N-dimethylformamide 200 mL
  • potassium carbonate 34.5 g, 250 mmol
  • benzyl bromide 31.8 g, 186 mmol
  • step (d) 8-Benzyloxy-5-(2,2-dihydroxyacetyl)-lH-quinolin-2-one
  • DMSO 60 mL
  • a 48% w/w hydrobromic acid solution 11.8 mL, 102.3 mmol
  • the mixture was warmed to 60 °C for 16 h then allowed to cool to room temperature.
  • Water (100 mL) was added and the resulting slurry stirred at room temperature for 0.5 h before being cooled to 0°C.
  • the product was collected on a Buchner funnel then dried under reduced pressure to give 8-benzyloxy-5-(2,2-dihydroxyacetyl)-lH-quinolin-2-one (12.2 g) as a solid.
  • Biphenyl-2-isocyanate 97.5 g, 521 mmol
  • 4-hydroxy-l-benzylpiperidine 105 g, 549 mmol
  • the reaction mixture was then cooled to 50 °C and ethanol (1 L) was added, and then 6M hydrochloric acid (191 mL) was added slowly.
  • reaction mixture was then cooled to ambient temperature and ammonium formate (98.5 g, 1.56 mol) was added and nitrogen gas was bubbled through the solution vigorously for 20 min. Palladium (10 wt. % (dry basis) on activated carbon) (20 g) was then added. The reaction mixture was heated at 40 °C for 12 h and then filtered through a pad of Celite. The solvent was then removed under reduced pressure and IM hydrochloric acid (40 mL) was added to the crude residue. Sodium hydroxide (ION) was then added to adjust the pH to 12.
  • Trifluoroacetic acid (11 mL) was added to a solution ofthe product of Preparation 10 (7.2 g, 11.3 mmol) in dichloromethane (56 mL). After 2 h, LCMS analysis showed that the reaction was completed. The reaction mixture was then concentrated to dryness and diluted with ethyl acetate (75 mL). Sodium hydroxide (IN) was then added until the pH of the mixture reached 14. The organic phase was then collected and washed with saturated sodium bicarbonate (2 x 50 mL) and brine (50 mL). The organic phase was then dried over magnesium sulfate and concentrated to provide the title compound (5.5 g). MS m/z: [M + H + ] calcd for C 27 H 39 N 3 O 2 438.30; found 439.
  • step (b) To the product of step (b) (70.2 g, 189 mmol) was added NN-dimethylformamide (260 mL) and this mixture was cooled in an ice bath under nitrogen. 2,6-Lutidine (40.3 g, 376 mmol) was added over 5 min and then tert-butyldimethylsilyl trifluoromethanesulfonate (99.8 g, 378 mmol) was added slowly while maintaining the temperature below 20°C. The mixture was allowed to Trust to room temperature for 45 min.
  • the product of Preparation 13 (3.9 g, 8.17 mmol) was added to a solution ofthe product of Preparation 11 (5.0 g, 11.4 mmol) in THF (20 mL), followed by sodium bicarbonate (2.0 g, 24.5 mmol) and sodium iodide (1.8 g, 12.2 mmol).
  • the reaction mixture was heated to 80 °C for 72 h.
  • the reaction mixture was then cooled, diluted with dichloromethane (20 mL) and the organic phase was washed with saturated sodium bicarbonate (2 x 50 mL) and brine (50 mL).
  • the organic phase was then dried (magnesium sulfate) and concentrated to give 6.5 g of a crude product.
  • the crude product was purified by chromatography on silica gel eluting with 3% methanol in dichloromethane to provide the title compound (1.4 g, 21% yield).
  • Triethylamine hydrogen fluoride (376 ⁇ L, 2.3 mmol) was added to a solution ofthe product of Preparation 14 (1.3 g, 1.5 mmol) in THF (8 mL) and the reaction mixture was stirred at ambient temperature. After 5 h, the reaction was complete as determined by LCMS analysis. The reaction mixture was then quenched with IN NaOH until the pH was 14 and then diluted with ethyl acetate (20 mL) and washed with IN NaOH (20 mL) and brine (20 mL). The organic phase was then separated, dried over magnesium sulfate, and concentrated to yield the title compound (1.1 g).
  • TEMPO 2,2,6,6-Tetramethyl-l-piperidinyloxy free radical
  • the residue was purified by silica gel chromatography (15g silica/1 g crude) using 5% MeOH in DCM/0.5% NH 4 OH (10 x 150 mL), 8% MeOH in DCM/0.5% NH 4 OH (10 x 150 mL) and 10% MeOH in DCM/0.5% NH 4 OH (10 x 150 mL). The appropriate fractions were combined and the solvent was removed under reduced pressure while maintaining the temperature ⁇ 35 °C to give the title intermediate (4.05 g , 97% purity).
  • the reaction mixture was then diluted with a mixture of isopropyl acetate (53 mL) and hexanes (27 mL) and transfened to a separatory funnel.
  • the organic layer was washed twice with a mixture of water (27 mL) and saturated aqueous sodium chloride (27 mL) followed by a final wash with saturated aqueous sodium chloride (27 mL).
  • the organic layer was dried over sodium sulfate.
  • Silica gel (23.6 g) and hexanes (27 mL) were added and the suspension was stirred for 10 min. The solids were removed by filtration and the filtrate concentrated under vacuum.
  • Methyl 3-bromopropionate (553 ⁇ L, 5.07 mmol) was added to a stined solution of the product of Preparation 8 (1.00 g, 3.38 mmol) and DIPEA (1.76 mL, 10.1 mmol) in acetonitrile (34 mL) at 50 °C and the reaction mixture was heated at 50 °C overnight. The solvent was then removed under reduced pressure and the residue was dissolved in dichloromethane (30 mL). The resulting solution was washed with saturated aqueous sodium bicarbonate solution (10 mL), dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (5-10% MeOH/DCM) to give the title compound (905 mg, 70%).
  • palladium (10 wt. % (dry basis) on activated carbon) 81 mg
  • the title compound can be prepared as follows: (a) 5-ChloropentanaI
  • TEMPO 2,2,6,6-Tetramethyl-l-piperidinyloxy free radical
  • the layers ofthe mixture were then separated, and the aqueous layer was extracted with dichloromethane (1 x 50 mL). The combined dichloromethane layers were then washed with water (1 x 50 mL), dried (MgSO 4 ), filtered and concentrated under reduced pressure to afford the title compound (53 g). The product was distilled at 65°C/8 ton to afford the title compound (31.16 g) as an orange oil (GC purity was 70 to 80 %). The product was further purified by adding the crude material (4 g) to a mixture of ethanol (920 mL), ethyl acetate (12 mL) and water (4 mL).
  • Aqueous saturated sodium bicarbonate solution (200 mL) was then added slowly (gas evolution) and stirring was continued for 15 min. The pH of the solution was then adjusted with solid sodium carbonate to a pH of about 9 and the layers were separated. The organic layer was washed with aqueous 5% sodium chloride solution (200 mL), dried (MgSO 4 ), filtered and concentrated under reduced pressure to afford the title compound (53 g).
  • step (c) 5-[(R)-2-[(5-N ⁇ V-Diformylaminopentyl)benzylamino]-l-(tert- butyldimethyl-silanyloxy)ethyl]-8-benzyloxy-lH-quinolin-2-one
  • sodium diformylamide 6.1 g, 64.2 mmol
  • sodium iodide 2.13 g, 14.3 mmol
  • step (e) A mixture ofthe product of step (e) (3.68 g, 10 mmol) and N,N-dimethylformamide (50 mL) was heated at 60 °C until the solid completely dissolved and then cooled to room temperature.
  • the product of step (d) (6 g, 10 mmol) and diisopropylethylamine (3.5 mL) was added and the reaction mixture was cooled to 0°C.
  • PyBOP (6.25 g, 12 mmol) was added in one portion and the reaction mixture was stined at 0 °C to room temperature for 2 hours.
  • the reaction mixture was then poured into cold water (500 mL) with stirring and the pH ofthe resulting mixture was adjusted to about 2 using aqueous 1 M hydrochloric acid. This mixture was stined for 15 min and then filtered to collect the solid, which was washed with water (100 mL) and dried to afford the title compound (8.7 g, HPLC purity >95%) as an
  • step (f) can be deprotected using essentially the same procedures as those described in Preparation 24 and Example 6 to afford the title compound.
  • DMSO 71 mL,l mol
  • DIPEA 87.1 mL, 0.5 mol
  • the reaction mixture was stined at -10 °C for 15 min and then sulfur trioxide pyridine complex (79.6 g, 0.5 mol) was added and the resulting mixture was stined for 1 hour.
  • the reaction mixture was quenched with addition of IM hydrochloric acid (200 mL).
  • the organic layer was separated and washed with saturated aqueous sodium bicarbonate (100 mL), brine (100 mL), dried (potassium carbonate) and solvent removed under reduced pressure to give the title compound (20.7 g, ⁇ 100% yield).
  • N-tert-butoxycarbonyl-l,5-diaminopentane (1.04 g, 5.12 mmol) was added to a solution ofthe product of Preparation 13 (1.00 g, 2.05 mmol) in dimethyl sulfoxide (2 mL).
  • the solution was stined at 75 °C for 12 hours, at which time LCMS analysis showed that the reaction was complete.
  • the reaction mixture was then concentrated under vacuum to dryness. To the residue was added dichloromethane (2 mL) and trifluoroacetic acid (1 mL) was then added. The solution was stined at room temperature for about 3 hours, at which time MS analysis showed that the reaction was complete.
  • Triethylamine trihydrofluoride (0.16 mL, 1.02 mmol) was added to a solution of the product of Preparation 41 (597 mg, 0.68 mmol) in tetrahydrofuran (3.4 mL) and this mixture was stined at room temperature for about 12 hours, at which time the reaction was determined to be completed by MS analysis.
  • pH 7.0
  • the diethyl ether phase was washed once with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a light orange oil.
  • the oil was dissolved in a minimum volume of ethyl acetate, diluted with hexanes, and to give the title compound as a crystalline solid.
  • step (b) To the product of step (b) (23.4 g, 0.113 mol) in 600 mL of THF at -78 °C was added 135 mL of 1.0 M sodium hexamethyldisilazane in THF (Sigma- Aldrich). After 1 hour, trimethylsilyl chloride (15.8 mL, 0.124 mol) was added. After another 30 minutes, bromine (5.82 mL, 0.113 mol) was added. After 10 minutes, the reaction was quenched by diluting the reaction mixture with diethyl ether and pouring it onto 500 mL of 5% aqueous Na 2 SO 3 premixed with 500 mL of 5% aqueous NaHCO 3 . The phases were separated and the organic phase was washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give the title compound as an oil that solidified upon storage in the freezer.
  • step (d) (R)-2-Bromo-l-(2,2-dimethyl-4H-benzo[l,3]dioxin-6-yI)ethanol
  • step (c)(1) (0.97 g, 3.5 mmol).
  • the solution was cooled to between -20 °C and -10 °C and BH 3 -THF (35 mL, 35 mmol) diluted with 50 mL THF was added dropwise via a dropping funnel. After the addition was complete, the reaction mixture was allowed to warm to ambient temperature.
  • reaction mixture was quenched by slow addition of 50 mL of methanol and then concentrated to a thick oil.
  • the oil was purified by silica gel chromatography eluted with 1 :2 ethyl acetate/hexanes. The fractions were combined and concentrated to give the title compound as an off-white solid.
  • step (d) To the product of step (d) (10 g, 34.8 mmol) and imidazole (4.7 g, 69.7 mmol) dissolved in 100 mL DMF was added tert-butyldimethylsilyl chloride (5.78 g, 38.3 mmol). The reaction mixture was stined for 18 hours. The reaction mixture was then partitioned between 200 mL of saturated sodium chloride and 200 mL of diethyl ether. The aqueous layer was extracted with 200 mL of diethyl ether. The organic layers were then combined, washed with saturated sodium chloride (3 x 100 mL), dried over MgSO and concentrated. The product was purified by silica gel chromatography, eluting with hexanes followed by 5% ethyl acetate in hexanes. The desired fractions were combined and concentrated to give the title compound as an oil.
  • Triethylamine trihydrofluoride (342 ⁇ L, 2.10 mmol) was added to a stirred solution ofthe Product of Preparation 44 (798 mg, 1.05 mmol) in dichloromethane (10.5 mL) at ambient temperature. The reaction mixture was stined for 24 h and it was then diluted with dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate (15 mL). The organic layer was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude title compound was isolated as an oil (659 mg, 1.02 mmol), which was used in the next step without further purification. MS m/z: [M + H + ] calcd for C 39 H 53 N 3 O 5 644.4; found 644.8.
  • step (a) The product of step (a) (10.0 g, 35.2 mmol) was dissolved in chloroform (250 mL) in a 500 mL flask under a nitrogen atmosphere. Bromine (1.63 mL, 31.7 mmol) dissolved in chloroform (50 mL) was added using a dropping funnel over 30 min. The reaction mixture was stined for 2.5 h and then concentrated to give a solid. The solid was dissolved in toluene (150 mL) with some gentle heat, followed by the addition of ethyl ether (150 mL) to yield the title compound as a crystalline solid (55% yield).
  • the product of Preparation 47 (371 mg, 1.00 mmol) was added to a solution ofthe product of Preparation 46 (448 mg, 0.85 mmol) in dimethyl sulfoxide (4.5 mL) followed by the addition of potassium carbonate (234 mg, 1.7 mmol).
  • the reaction mixture was stined at 40 °C for 6 h, at which time the product of Preparation 46 was no longer observed by HPLC analysis.
  • the reaction mixture was cooled to ambient temperature and filtered, and then diluted with ethanol (4 mL). Sodium borohydride (63 mg, 1.7 mmol) was added to the reaction mixture and the reaction was stined at ambient temperature for 24 h.
  • reaction mixture was quenched with 0.5 M ammonium chloride (5 mL) and extracted into ethyl acetate (2 x 10 mL). The combined organic layers were washed with saturated sodium bicarbonate (10 mL) and then with brine (5 mL). The organic layer was dried over magnesium sulfate and the solvent was removed under reduced pressure. The crude residue was purified by chromatography on silica gel (3% methanol in chloroform) to give the title compound.
  • the crude product was purified by flash chromatography (5-10% MeOH/DCM) and then dissolved in a trifluoroacetic acid/DCM mixture (1 mL/5 mL) and stirred at room temperature for lh. The solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (20 mL) and washed with IM sodium hydroxide (10 mL), dried (magnesium sulfate) and the solvent reduced to yield the title compound (167 mg, 39% yield).
  • step (a) To the product of step (a) (1.71 g, 7.2 mmol) was added a solution of 4 M hydrochloric acid in dioxane (9 mL, 36 mmol) and the resulting mixture was stined at room temperature for 1 h. The reaction mixture was then concentrated and the residue was diluted with diethyl ether (50 mL) and filtered to provide the title compound as a white solid (1.09 g).
  • step (a) To the product from step (a) was added acetonitrile (20 mL), diisopropylethylamine (0.5 mL, 2.8 mmol) and the product of Preparation 8 (626 mg, 2.11 mmol). The reaction mixture was heated to 50 °C for 20 h and then cooled to room temperature and the solvent was removed under reduced pressure. The residue was purified by silica gel chromatography (5% MeOH/DCM with 0.6% NH 3 (aq)) to afford the title compound (450 mg, 44% yield). MS m/z (MH + ) 479.6; R 4.15 min (10-70% ACN: H 2 O, reverse phase HPLC).

Abstract

This invention provides biphenyl derivatives of formula I: wherein R1, R2, R3, R4, R5, R6, R7, W, a, b and c are as defined in the specification, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. The biphenyl derivatives of this invention possess both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity and therefore, such biphenyl derivatives are useful for treating pulmonary disorders, such as chronic obstructive pulmonary disease and asthma.

Description

BIPHENYL DERIVATIVES
BACKGROUND OF THE INVENTION
Field ofthe Invention
The present invention relates to novel biphenyl derivatives that are useful for treating pulmonary disorders. This invention also relates to phannaceutical compositions comprising such biphenyl derivatives, processes and intermediates for preparing such biphenyl derivatives and methods of using such biphenyl derivatives to treat pulmonary disorders.
State ofthe Art
Pulmonary disorders, such as asthma and chronic obstructive pulmonary disease (COPD), are commonly treated with bronchodilators. One class of bronchodilator in widespread use consists of β2 adrenergic receptor (adrenoceptor) agonists, such as albuterol, formoterol and salmeterol. These compounds are generally administered by inhalation. Another class of bronchodilator consists of muscarinic receptor antagonists (anticholinergic compounds), such as ipratropium and tiotropium. These compounds are also typically administered by inhalation.
Pharmaceutical compositions containing both a β2 adrenergic receptor agonist and a muscarinic receptor antagonist are also known in the art for use in treating pulmonary disorders. For example, U.S. Patent No. 6,433,027 discloses medicament compositions containing a muscarinic receptor antagonist, such as tiotropium bromide, and a β2 adrenergic receptor agonist, such as formoterol fumarate.
Although compounds having either β2 adrenergic receptor agonist or muscarinic receptor antagonist activity are known, no compound having both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity has been previously disclosed. Compounds possessing both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity are highly desirable since such bifunctional compounds would provide bronchodilation through two independent modes of action while having single molecule pharmacokinetics.
SUMMARY OF THE INVENTION
The present invention provides novel biphenyl derivatives that are useful for treating pulmonary disorders. Among other properties, compounds of this invention have been found to possess both β adrenergic receptor agonist and muscarinic receptor antagonist activity.
Accordingly, in one of its composition aspects, the present invention is directed to a compound of formula I:
Figure imgf000003_0001
I wherein: a is 0 or an integer of from 1 to 3; each R1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -ORla, -C(O)ORlb, -SRlc, -S(O)Rld, -S(O)2Rle and -NRlfRlg; each of Rla, Rlb, Rlc, Rld, Rle, Rlf and Rl is independently hydrogen, (l-4C)alkyl or phenyl-(l-4C)alkyl; b is 0 or an integer of from 1 to 3; each R2 is independently selected from ( 1 -4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl,
(3-6C)cycloalkyl, cyano, halo, -OR2a, -C(O)OR2b, -SR2c, -S(O)R2d, -S(O)2R2e and -NR2fR2g; each of R2a, R2b, R2°, R2d, R2e, R2f and R2g is independently hydrogen, (l-4C)alkyl or ρhenyl-(l-4C)alkyl;
W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring and represents O or NWa; Wa is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R3 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR3a, -C(O)OR3b, -SR3c, -S(O)R3d, -S(O)2R3e and -NR3 R3g; or two R3 groups are joined to form (l-3C)alkylene, (2-3C)alkenylene or oxiran- 2,3-diyl; each of R3a, R3b, R3c, R3d, R3e, R3f and R3g is independently hydrogen or (1- 4C)alkyl;
R4 is a divalent group ofthe formula:
-(R4a)d-(A1)e-(R4b)f-Q-(R4o)g-(A2)h-(R4d)i-
wherein d, e, f, g, h and i are each independently selected from 0 and 1 ;
R4a, R4b, R4° and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl-(l-4C)alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, -O-(6-10C)arylene, (6-10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O , -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Qc)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q;)-, ^( ^)0(0)0- and -N(Qk); Qa, θΛ Q°, Qd, Qe, Qf, Qg, θΛ QΛ J and Qk are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 16;
R5 represents hydrogen or (l-4C)alkyl;
R6 is -NR6aCR6b(O) or -CR6oR6dOR6e and R7 is hydrogen; or R6 and R7 together form -NR7aC(O)-CR7b=CR7c- , -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7i-CR7jR7k- or - CR71R7m-CR7nR70- C(O) -NR7p-; each of R6a, R6 , R6°, R6d and R6e is independently hydrogen or (l-4C)alkyl; and each of R7a, R7b, R7°, R7d, R7e, R7f, R7g, R7h, R7i, R7j, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or (l-4C)alkyl; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
In another of its composition aspects, this invention is directed to a compound of formula II:
Figure imgf000005_0001
II wherein
R4 is as defined herein (including any specific or preferred embodiments); W represents O or NH; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. In yet another of its composition aspects, this invention is directed to a compound of formula III:
Figure imgf000006_0001
III wherein
R4 is as defined herein (including any specific or preferred embodiments);
W represents O or NH; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
In still another of its composition aspects, this invention is directed to a compound of formula IN:
Figure imgf000006_0002
IN wherein R4 is as defined herein (including any specific or preferred embodiments);
W represents O or NH; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
In another of its composition aspects, this invention is directed to a pharmaceutical composition comprising 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. Such pharmaceutical compositions may optionally contain other therapeutic agents. Accordingly, in one embodiment, this invention is directed to such a pharmaceutical composition wherein the composition further comprises a therapeutically effective amount of a steroidal anti-inflammatory agent, such as a corticosteroid.
Compounds of this invention possess both β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity. Accordingly, the compounds of formula I are useful for treating pulmonary disorders, such as asthma and chronic obstructive pulmonary disease.
Accordingly, in one of its method aspects, this invention is directed to a method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Additionally, in another of its method aspects, this invention is directed to a method of providing bronchodilation in a patient, the method comprising administering to a patient requiring bronchodilation a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
This invention is also directed to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Since compounds of this invention possess both β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity, such compounds are also useful as research tools. Accordingly, in yet another of its method aspects, this invention is directed to a method 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 both β2 adrenergic agonist activity and muscarinic receptor antagonist activity.
This invention is also directed to processes and novel intermediates useful for preparing compounds of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Accordingly, in another of its method aspects, this invention is directed to a process of preparing a compound of formula I, the process comprising:
(a) reacting a compound of formula 1 or a salt thereof, with a compound of formula 2;
(b) reacting a compound of formula 3 or a salt thereof, with a compound of formula 4;
(c) coupling a compound of fonnula 5 with a compound of formula 6;
(d) for a compound of formula I wherein R5 represents a hydrogen atom, reacting a compound of formula 3 with a compound of formula 7 or a hydrate thereof, in the presence of a reducing agent; (e) reacting a compound of formula 1 with a compound of formula 8 or a hydrate thereof, in the presence of a reducing agent;
(f) reacting a compound of formula 9, with a compound of formula 10; or
(g) reacting a compound of formula 11 or a hydrate thereof, with a compound of formula 10, in the presence of a reducing agent; and then removing any protecting groups to form a compound of formula I; wherein the compounds of formula 1-11 are as defined therein.
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, this invention is directed to the other processes described herein; and to the product prepared by any ofthe processes described herein.
This 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, this 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. DETAILED DESCRIPTION OF THE INVENTION
In one of its composition aspects, this invention is directed to novel biphenyl derivatives of formula I or pharmaceutically acceptable salts or solvates or stereoisomers thereof. These compounds contain one or more chiral centers and therefore, this invention is 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 this invention unless otherwise indicated, provided that the utility ofthe composition as a whole is not eliminated by the presence of such other isomers.
In particular, compounds of formula I contain a chiral center at the carbon atom indicated by the symbol * in the following formula:
Figure imgf000009_0001
In one embodiment of this invention, the carbon atom identified by the symbol * has the (R) configuration. In this embodiment, it is preferred for compounds of formula I to have the (R) configuration at the carbon atom identified by the symbol * or to be enriched in a stereoisomeric form having the (R) configuration at this carbon atom. In another embodiment of this invention, the carbon atom identified by the symbol * has the (S) configuration. In this embodiment, it is preferred for compounds of formula I to have the (S) configuration at the carbon atom identified by the symbol * or to be enriched in a stereoisomeric form having the (S) configuration at this carbon atom. In some cases, in order to optimize the β2 adrenergic agonist activity ofthe compounds of this invention, it is preferred that the carbon atom identified by the symbol * has the (R) configuration.
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. All such salt forms are included within the scope of this invention. Furthermore, solvates of compounds of formula I or salts thereof are included within the scope of this invention.
Additionally, where applicable, all cis-trans or E/Z isomers (geometric isomers), tautomeric forms and topoisomeric forms ofthe compounds of formula I are included within the scope of this invention unless otherwise specified.
The nomenclature used herein to name the compounds of this invention and intennediates thereof has generally been derived using the commercially-available AutoNom software (MDL, San Leandro, California). Typically, compounds of formula I wherein W is O have been named as ester derivatives of biphenyl-2-ylcarbamic acid; and compounds of formula I wherein W is NWa have been named as urea derivatives.
Representative Embodiments
The following substituents and values are intended to provide representative examples of various aspects and embodiments of this 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 this invention. In 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 this invention unless specifically indicated. In particular embodiments ofthe compounds of fonnula I, a and b are independently 0, 1 or 2; including 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 ofthe phenyl ring to which it is attached. In one embodiment, each R1 is independently selected from (1-
4C)alkyl, halo, -ORIa and -NRlfRlg; such as 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). In one embodiment, each R2 is independently selected from (1- 4C)alkyl, halo, -OR2a and -NR2fR2g; such as 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 and Rlg and R2a, R2b, R2c, R2d, R2e, R2f and R2g as used in R1 and R2, respectively, is independently hydrogen, (l-4C)alkyl or phenyl-(l- 4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl or benzyl. In one embodiment, these groups are independently hydrogen or (1- 3C)alkyl. In another embodiment, these groups are independently hydrogen, methyl or ethyl.
In one embodiment of this invention, W is O. In another embodiment, W is NWa. Generally, it has been found that compounds in which W represents O exhibit particularly high affinity for muscarinic and β2 adrenergic receptors. Accordingly, in a particular embodiment of this invention, W preferably represents O.
When referring to W, particular mention may be made of compounds wherein W is attached to the piperidine ring at the 4-position with respect to the nitrogen atom ofthe piperidine ring.
When W is NWa, Wa is hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl. In one embodiment, Wa is hydrogen or (l-3C)alkyl. In another embodiment, Wa is hydrogen, methyl or ethyl; such as hydrogen or methyl. In yet another embodiment, Wa is hydrogen and NWa is NH.
In a particular embodiment ofthe compounds of fonnula I, c is 0, 1 or 2; including 0 or 1. In one embodiment, c is 0. In one embodiment, each R3 is at the 3, 4 or 5-position on the piperidine ring
(where the nitrogen atom ofthe piperidine ring is position 1). In another embodiment, R3 is at 4-position on the piperidine ring. In a particular aspect of these embodiments, each R3 is independently selected from (l-4C)alkyl; such as methyl, ethyl, n-propyl, isopropyl, n- butyl, sec-butyl, isobutyl and tert-butyl. In another aspect, each R3 is independently methyl or ethyl.
In another embodiment, R3 is at the 1 -position ofthe piperidine ring, i.e., on the nitrogen atom ofthe piperidine ring thus forming a quaternary amine salt. In a particular aspect of this embodiment, each R3 is independently selected from (l-4C)alkyl; such as methyl, ethyl, n-propyl, isopropyl, n-butyl, .fee-butyl, isobutyl and tert-butyl. In another aspect, each R3 is independently methyl or ethyl.
In yet another embodiment, two R3 groups are joined to form a (l-3C)alkylene or (2-3C)alkenylene 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-ρositions 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). In 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.
Each R3a, R3b, R3c, R3d, R3e, R3f and R3g as used in R3 is independently hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl. In one embodiment, these groups are independently hydrogen or (l-3C)alkyl. In another embodiment, these groups are independently hydrogen, methyl or ethyl.
In one embodiment ofthe compounds of formula I, R5 is hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert- butyl. In another embodiment, each R5 is independently hydrogen, methyl or ethyl. In a particular embodiment, R5 is hydrogen.
In one embodiment of this invention, R6 is -NR6 CR6b(O) and R7 is hydrogen, where each of R6a and R6b is independently hydrogen or (l-4C)alkyl, such as hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl. In one embodiment, these groups are independently hydrogen or (l-3C)alkyl. In another embodiment, these groups are independently hydrogen, methyl or ethyl. A particular value for R6 in this embodiment is -NHCHO.
In another embodiment, R6 and R7 together form -NR7aC(O)-CR7b=CR7c- , -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7i-CR7jR7k- or - CR71R7m-CR7nR7°- C(O)
-NR7p-; where each of R7a, R7b, R7°, R7d, R7e, R7f, R7g, R7h, R7i, R7j, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or (l-4C)alkyl; such as hydrogen, methyl, ethyl, n- propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl. In one embodiment, these groups are independently hydrogen or (l-3C)alkyl. In another embodiment, these groups are independently hydrogen, methyl or ethyl. Particular values for R6 and R7 in this embodiment are R6 and R7 together fonn -NHC(O)-CH=CH-, -CH=CH-C(O)-NH-, -CH2- CH2-C(O)NH- or-NHC(O)-CH2-CH2-; including where R6 and R7 together form -NHC(O)-CH=CH- or -CH=CH-C(O)-NH-; and in particular, where R6 and R7 together
--I I- form -NHC(O)-CH=CH- (i.e., the nitrogen atom is attached at R6 and the carbon atom is attached at R7 to form, together with the hydroxyphenyl ring to which R6 and R7 are attached, a 8-hydroxy-2-oxo-l,2-dihydroquinolin-5-yl group).
In the compounds of fonnula I, R4 is a divalent group ofthe formula:
Figure imgf000013_0001
wherein R4a, A1, R4b, Q, R4c, A2, R4d, d, e, f, g h and i are as defined herein. In the compound of this invention, the values of each ofthe components R4a, A1, R4b, Q, R4c, A2 and R4d are selected such that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 16, (specifically, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16); including 8, 9, 10, 11, 12, 13 or 14; such as 8, 9, 10 or 11 ; or 9 or 10. When selecting values for each variable in R , it will be appreciated by those skilled in the art that values should be selected such that a chemically stable group is formed.
When determining the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached, each contiguous atom ofthe chain is counted consecutively starting from the first atom in the R4 group adjacent to the nitrogen ofthe piperidine ring ending with the last atom in the R4 group adjacent to the nitrogen of the aminohydroxyethyl group. Where two or more chains are possible, the shortest chain is used to determine the number of contiguous atoms. As shown below, for example, when R4 is -(CH2)2-NHC(O)-CH2-(phen-l,4-ylene)-CH2-, there are 10 contiguous atoms in the shortest chain counted consecutively starting from the first atom in the R group adjacent to the nitrogen ofthe piperidine ring ending with the last atom in the R4 group adjacent to the nitrogen ofthe aminohydroxyethyl group as shown below:
Figure imgf000013_0002
In one embodiment of R4, R4a is selected from (l-lOC)alkylene, (2-10C)alkenylene and (2-10C)alkynylene wherein the alkylene group is unsubstituted or substituted with 1 or 2 substituents independently selected from (l-4C)alkyl, hydroxy and phenyl. Representative examples of particular values for R4a are -(CH2)2-, -(CH2)3-, -(CH2)4-, -(CH2)5-, -(CH2)6-, -(CH2)7-, -(CH2)8-, -(CH2)9-, -(CH2)10-, -(CH2)CH(CH3)-,
-(CH2)C(CH3)2-, and -(CH2)2C(phenyl)2-. In another aspect, R4a is -(CH )C(=CH2)-.
In one embodiment, d is 1.
In one embodiment, A1 is an optionally substituted (3-7C)cycloalkylene group; including a cyclohexylene group, such as cyclohex-l,4-ylene and cyclohex-l,3-ylene; and a cyclopentylene group, such as cyclopent- 1 ,3-ylene.
In another embodiment, A1 is an optionally substituted (6-10C)arylene group, including a phenylene group, such as phen-l,4-ylene, phen-l,3-ylene and phen-l,2-ylene; and a naphthylene group, such as naphth-l,4-ylene and napth-l,5-ylene.
In yet another embodiment, A1 is an optionally substituted (2-9C)heteroarylene group, including a pyridylene group, such as pyrid-l,4-ylene; a furylene group, such as fur- 2,5-ylene and fur-2,4-ylene; a thienylene group, such as thien-2,5-ylene and thien-2,4- ylene; and a pyrrolylene, such as pyrrol-2,5-ylene and pyrrol-2,4-ylene.
In still another embodiment, A1 is an optionally substituted (3-6C)heterocyclene group, including a piperidinylene group, such as piperidin-l,4-ylene; and a pyrrolidinylene group, such as pyrrolidin-2,5-ylene.
In a particular embodiment, A1 is an optionally substituted phenylene, thienylene, cyclopentylene, cyclohexylene or piperidinylene.
In one embodiment, e is 0.
In a particular embodiment, R4b is (l-5C)alkylene. Representative examples of particular values for R4b are -CH2-, -(CH2)2-, -(CH2)3-, -(CH2)4-, -(CH2)5-; including methylene, ethylene and propylene.
In one embodiment, f is 0.
In a particular embodiment, Q is selected from a bond, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Q°)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -OC(O)N(Q , -N(Q)')C(O)O- or-N(Qk); such as where Q is a bond, -N(Qa)C(O)- or -C(O)N(Qb)-. Representative examples of particular values for Q are a bond, O, NH, -C(O)NH-, -C(O)N(CH3)-, -NHC(O)-, -N(CH3)C(O)-, -S(O)2NH-, -S(O)2N(CH3)-, -NHS(O)2-, -N(CH3)S(O)2- and -NHC(O)NH-. Another example of a value for Q, together with R4c, is -C(O)(piperidin- 1,4-ylene).
In one embodiment, Qa, Qb, Qc, Qd, Qe, Qf, Qg, Qh, Q Oj and Qk are each independently selected from hydrogen and (l-6C)alkyl, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy. For example, Qa, Qb, Q°, Qd, Qe, Qf, Qg, Qh, Q!, Q1 and Qk are each independently selected from hydrogen, and (l-3C)alkyl, including hydrogen, methyl, ethyl, n-propyl and isopropyl. An example of a value for each of Qa, Qb, Q°, Qd, Q Qf, Qg, Qh, O 0s and Qk is hydrogen. In another embodiment, Qa, Qb, Q°, Qd, Qe, Qf, Qg, Qh, Q1, Oj and Qk together with the nitrogen atom and the group R b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group. For example, Qa and Qb together with the nitrogen atom and the group R4b or R4° to which they are attached, form a piperidin-4-ylene group. By way of illustration, when Q represents -N(Qa)C(O)- and Qa together with the nitrogen atom and the group R4b to which it is attached, forms a piperidin-4-ylene group, R4 is a group of formula:
Figure imgf000015_0001
Similarly, when Q represents -C(O)N(Q )- and Q together with the nitrogen atom and the group R4c to which it is attached, forms a piperidin-4-ylene group, R4 is a group of formula:
Figure imgf000015_0002
In a particular embodiment, R4c is (l-5C)alkylene. Representative examples of particular values for R4° are -CH2-, -(CH2)2-, -(CH2)3-, -(CH2)4-, -(CH2)5-; including methylene, ethylene and propylene. In one embodiment, A2 is an optionally substituted (3-7C)cycloalkylene group; including a cyclohexylene group, such as cyclohex-l,4-ylene and cyclohex-l,3-ylene; and a cyclopentylene group, such as cyclopent- 1,3 -ylene.
In another embodiment, A2 is an optionally substituted (6-10C)arylene group, including a phenylene group, such as phen- 1 ,4-ylene, phen- 1 ,3 -ylene and phen- 1 ,2-ylene; and a naphthylene group, such as naphth-l,4-ylene and napth-l,5-ylene.
In yet another embodiment, A2 is an optionally substituted (2-9C)heteroarylene group, including a pyridylene group, such as ρyrid-l,4-ylene; a furylene group, such as fur- 2,5-ylene and fur-2,4-ylene; a thienylene group, such as thien-2,5-ylene and thien-2,4- ylene; and a pyrrolylene, such as pyrrol-2,5-ylene and pyrrol-2,4-ylene.
In still another embodiment, A2 is an optionally substituted (3-6C)heterocyclene group, including a piperidinylene group, such as piperidin-l,4-ylene; and a pyrrolidinylene group, such as pyrrolidin-2,5-ylene.
In a particular embodiment, A2 is optionally substituted phenylene, thienylene, cyclopentylene, cyclohexylene or piperidinylene.
By way of illustration, either A1 or A2 or both can be phenylene, such as phen- 1,4- ylene or phen-l,3-ylene, where the phenylene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy. Representative examples include phen- 1,3 -ylene, phen-l,4-ylene, 4-chlorophen- 1,3 -ylene, 6-chlorophen-l,3-ylene, 4-methylphen- 1,3 -ylene, 2-fluorophen-l,4-ylene, 2-chlorophen-l,4-ylene, 2-bromophen- 1,4-ylene, 2-iodoρhen-l,4-ylene, 2-methylphen-l,4-ylene, 2-methoxyphen-l,4-ylene, 2- trifluoromethoxyphen-l,4-ylene, 3-nitrophen-l,4-ylene, 3-chlorophen-l,4-ylene, 2,5- difluorophen- 1 ,4-ylene, 2,6-dichlorophen- 1 ,4-ylene, 2,6-diiodophen- 1 ,4-ylene, 2-chloro-6- methylphen- 1 ,4-ylene, 2-chloro-5-methoxyphen- 1 ,4-ylene, 2,3,5,6-tetrafluorophen- 1 ,4- ylene.
Alternatively, A1 or A2 or both can be cyclopentylene or cyclohexylene; wherein the cyclopentylene or cyclohexylene group is unsubstituted or substituted with (l-4C)alkyl. Representative examples include cz's-cyclopent- 1,3 -ylene, tr ns-cyclopent-l,3-ylene, cis- cyclohex-l,4-ylene and trαns-cyclohex-l,4-ylene. A1 or A2 or both can also be optionally substituted thienylene or piperidinylene, for example, thien-2,5-ylene or piperidin-1,4- ylene. In one embodiment, R4d is selected from (l-lOC)alkylene, (2-10C)alkenylene and (2-10C)alkynylene wherein the alkylene is unsubstituted or substituted with 1 or 2 substituents independently selected from (l-4C)alkyl, hydroxy and phenyl. Representative examples of particular values for R4d are -(CH2)-, -(CH2)2-, -(CH2)3-, -(CH2)4-, -(CH2)5-, -(CH2)6-, -(CH2)7-, -(CH2)8-, -(CH2)9-, -(CH2)10- and -(CH2)CH(CH3)-(CH2)-C(CH3)2- (CH2)2-.
In a particular embodiment, R4 is a divalent group ofthe formula: -(R4a)d- where R4a is (4-10C)alkylene. In one aspect of this embodiment, R4 is a divalent group ofthe formula: -(CH )j- where j is 8, 9 or 10. Examples of particular values for R4 in this embodiment are -(CH2)4-, -(CH2)5-, -(CH2)6-, -(CH2)7-, -(CH2)8-, -(CH2)9, and -(CH2)10-; including -(CH2)8-, -(CH2)9, and -(CH2)10-.
In another particular embodiment, R4 is a divalent group ofthe formula:
-(R4a)d-(A2)h-(R4d);-
where R4a is (l-lOC)alkylene, such as -(CH2)-, -(CH2)2-, -(CH2)3-; A2 is (6- 10C)arylene, such as phen-l,4-ylene or phen- 1,3 -ylene, or (2-9C)heteroarylene, such as thien-2,5-ylene or thien-2,4-ylene; and R4d is (l-lOC)alkylene, such as -(CH2)-, -(CH2)2-, -(CH2)3-. Examples of particular values for R4 in this embodiment are -(CH2)-(phen-l,4- ylene)-(CH2)-; -(CH2)-(phen- 1 ,4-ylene)-(CH2)2-; -(CH2)-(phen- 1 ,4-ylene)-(CH2)3-; - (CH2)2-(phen-l,4-ylene)-(CH2)-; -(CH2)2-(phen-l,4-ylene)-(CH2)2-; -(CH2)2-(phen-l,4- ylene)-(CH2)3-; -(CH2)3-(phen-l,4-ylene)-(CH2)-; -(CH2)3-(phen-l,4-ylene)-(CH2)2-, - (CH2)3-(phen-l,4-ylene)-(CH2)3-, -(CH2)4-(phen-l,4-ylene)-(CH2)-; -(CH2)4-(phen-l,4- ylene)-(CH2)2- and -(CH2)4-(phen-l,4-ylene)-(CH2)3-. In yet another particular embodiment, R4 is a divalent group ofthe formula:
-(R4a)d-Q-(A2)h-(R4d);-
where Q is -O- or -N(Qk)-; Qk is hydrogen or (l-3C)alkyl, such as methyl or ethyl; R4a is (l-lOC)alkylene, such as -(CH2)-, -(CH2)2-, -(CH2)3-; A2 is (6-10C)arylene, such as phen-l,4-ylene or phen-l,3-ylene, or (2-9C)heteroarylene, such as thien-2,5-ylene or thien- 2,4-ylene; and R4d is (l-lOC)alkylene, such as -(CH2)-, -(CH2)2-, -(CH2)3-. Examples of particular values for R4 in this embodiment are -(CH2)2-O-(phen-l,4-ylene)-(CH2)-; -(CH2)2-0-(phen-l,4-ylene)-(CH2)2-; -(CH2)2-O-(phen-l,4-ylene)-(CH2)3-; -(CH2)3-O- (phen- 1 ,4-ylene)-(CH2)-; -(CH2)3-O-(phen- 1 ,4-ylene)-(CH2)2-; -(CH2)3-O-(phen- 1 ,4- ylene)-(CH2)3-; -(CH2)2-NH-(phen- 1 ,4-ylene)-(CH2)-; -(CH2)2-NH-(phen- 1 ,4-ylene)- (CH2)2-; -(CH2)2-NH-(phen-l,4-ylene)-(CH2)3-; -(CH2)3-NH-(phen-l,4-ylene)-(CH2)-; -(CH2)3-NH-(phen- 1 ,4-ylene)-(CH2)2- and -(CH2)3-NH-(phen- 1 ,4-ylene)-(CH2)3-. In yet another particular embodiment, R4 is a divalent group ofthe formula:
Figure imgf000018_0001
where Q is -N(Qa)C(O or -C(O)N(Qb)-. A particular value for R4 in this embodiment is the formula:
O
II
— (CH2)m-C-N— (CH2)n
where m is an integer from 2 to 10; and n is an integer from 2 to 10; provided that m + n is an integer from 4 to 12. In this formula for R4, d and g are 1 and e, f, h and i are 0; and R4a is -(CH2)m-, R4c is -(CH2)„- and Q is -C(O)NH-. Particular values for m are 2 or 3; and for n, 4, 5 or 6.
Another particular value for R4 is the formula:
Figure imgf000018_0002
where o is an integer from 2 to 7; and p is an integer from 1 to 6; provided that o + p is an integer from 3 to 8. In this formula for R4, d, h and i are 1 and e, f and g are 0; and R4a is -(CH2)o-, A2 is phen- 1 ,4-ylene, R4d is -(CH2)P- and Q is -C(O)NH-. Particular values for o are 2 or 3; and for p, 1 or 2. In this embodiment, the phen-l,4-ylene group may be optionally substituted as defined herein for A2. Another particular value for R4 is the formula:
Figure imgf000019_0001
where q is an integer from 2 to 6; r is an integer from 1 to 5; and s is an integer from 1 to 5; provided that q + r + s is an integer from 4 to 8. In this formula for R4, d, g, h and i are 1 and e and fare 0; and R4a is -(CH2)q-, R4c is -(CH2) , A2 is 1 ,4-phenylene, R4d is -(CH2)S- and Q is -C(O)NH-. Particular values for q are 2 or 3; for r, 1 or 2; and for s, 1 or 2. In this embodiment, the phen-l,4-ylene group may be optionally substituted as defined herein for A2.
Another particular value for R4 is the formula:
o
II
(CH2 2)Λ, -N-C- "(CH2)U- H
where t is an integer from 2 to 10; and u is an integer from 2 to 10; provided that t + u is an integer from 4 to 12. In this formula for R4, d and g are 1 and e, f, h and i are 0; and R4a is -(CH2)r, R4c is -(CH2)U- and Q is -NHC(O)-. Particular values for t are 2 or 3; and for u, 4, 5 or 6.
Another particular value for R4 is the formula:
Figure imgf000019_0002
where v is an integer from 2 to 7; and w is an integer from 1 to 6; provided that v + w is an integer from 3 to 8. In this formula for R4, d, h and i are 1 and e, f and g are 0; and R4a is -(CH2)V-, A2 is 1 ,4-phenylene, R4d is -(CH2)W- and Q is -NHC(O)-. Particular values for v are 2 or 3; and for w, 1 or 2. In this embodiment, the phen- 1 ,4-ylene group may be optionally substituted as defined herein for A2. Another particular value for R4 is the formula:
Figure imgf000020_0001
where x is an integer from 2 to 6; y is an integer from 1 to 5; and z is an integer from 1 to 5; provided that x + y + z is an integer from 4 to 8. In this formula for R4, d, g, h and i are 1 and e and f are 0; and R4a is -(CH2)X-, R4c is -(CH2)y-, A2 is 1,4-phenylene, R4d is -(CH2)Z- and Q is -NHC(O)-. Particular values for x are 2 or 3; for y, 1 or 2; and for z,
1 or 2. In this embodiment, the phen-l,4-ylene group may be optionally substituted as defined herein for A2.
By way of further illustration, R4 can be selected from:
-(CH2)7-;
-(CH2)8-;
-(CH2) -;
-(CH2)U-;
-(CH2)2C(O)NH(CH2)5-;
-(CH2)2N(CH3)C(O)(CH2)5-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)CH2-; -(CH2)2NHC(O)(phen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)NH(CH2)5-;
-(CH2)3NHC(O)NH(CH2)5-;
-(CH2)2C(O)NHCH2(cyclohex- 1 ,3-ylene)CH2-;
-(CH2)2NHC(O)(cyclopent- 1 ,3-ylene)-; -(CH2)2NHC(O)NH(phen- 1 ,4-ylene)(CH2)2-; l-[-(CH2)2C(O)](piperidin-4-yl)(CH2)2-;
-(CH2)2NHC(O)(trαns-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(cts-cyclopent- 1 ,3-ylene)-;
-(CH2)2NH(phen-l,4-ylene)(CH2)2-; l-[-(CH2)2NHC(O)](piperidin-4-yl)(CH2)2-;
-CH2(phen-l,4-ylene)NH(phen-l,4-ylene)CH2-;
-(CH2)2C(O)NHCH2(phen- 1 ,3-ylene)CH2-; -(CH2)2C(O)NHCH2(pyrid-2,6-ylene)CH2-;
-(CH2)2C(O)NH(cts-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(trαns-cyclohex-l,4-ylene)CH2-;
-(CH2)2NHC(O)(c s-cyclopent- 1 ,3-ylene)CH2-; -(CH2)2N(CH3)C(O)(phen- 1 ,3-ylene)CH2-;
-(CH2)2N(CH3)C(O)(trαns-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)C*H(CH3)- ((S)-isomer);
-(CH2)2C(O)NH(phen- 1 ,4-ylene)C*H(CH3)- ((i?)-isomer); 2-[(S)-(-CH2-](pyrrolidin-l-yl)C(O)(CH2)4-;
2-[(S)-(-CH2-](pyrrolidin- 1 -yl)C(O)(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(4-chlorophen- 1 ,3-ylene)CH2-;
-CH2(2-fluorophen-l,3-ylene)CH2-;
-(CH2)2C(O)NH(4-methylphen- 1 ,3-ylene)CH2-; -(CH2)2C(O)NH(6-chlorophen- 1 ,3-ylene)CH2-;
-(CH2)2C(O)NH(2-chlorophen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2,6-dichlorophen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)NHCH2(phen- 1 ,3-ylene)CH2-;
4-[-CH2-](piperidin- 1 -yl)C(O)(phen- 1 ,4-ylene)CH2-; -(CH2)2C(O)N(CH2CH3)(phen- 1 ,4-ylene)CH2-; l-[-(CH2)2NHC(O)](piperidin-4-yl)-;
-(CH2)2C(O)NH(phen-l,4-ylene)(CH2)2-;
-(CH2)2NHC(O)(thien-2,5-ylene)CH2-;
-(CH2)2N(CH3)C(O)(3-nitrophen- 1 ,4-ylene)CH2-; -(CH2)2N(CH3)C(O)(trans-cyclohex- 1 ,4-ylene)-;
1 -[-CH2(2-fluoroρhen- 1 ,3-ylene)CH2](piperidin-4-yl)-;
5-[-(CH2)2NHC(O)](pyrid-2-yl)CH2-;
-(CH2)2(phen-l,4-ylene)(CH2)2-;
-(CH2)3(thien-2,5-ylene)(CH2)3-; -(CH2)2(phen- 1 ,4-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-CH2(phen- 1 ,2-ylene)NH(phen- 1 ,4-ylene)(CH2)2-; l-[-CH2(2-fluorophen-l,3-ylene)CH2](piperidin-4-yl)(CH2)2-;
1 -[-CH2(2-fluorophen- 1 ,3-ylene)CH2](piperidin-4-yl)CH2-; -(CH2)2C(O)NH(3-chlorophen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2-(CF3O-)phen-l,4-ylene)CH2-;
-(CH2)3(phen- 1 ,3-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2S(O)2NH(CH2)5-; -CH2(phen- 1 ,3-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(2-iodophen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2-chloro-5-methoxyphen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2-chloro-6-methylphen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(CH2)5-; -(CH2)2N(CH3)S(O)2(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2-bromophen- 1 ,4-ylene)CH2-;
-(CH2)3(phen- 1 ,4-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)3(phen- 1 ,2-ylene)NH(phen- 1 ,4-ylene)(CH2)2-; l-[-CH2(2-fluorophen-l,3-ylene)CH2](piperidin-4-yl)(CH2)3-; -(CH2)2C(O)NH(2-methoxyphen- 1 ,4-ylene)CH2-;
-(CH2)5NH(phen- 1 ,4-ylene)(CH2)2-;
4-[-(CH2)2-](piperidin- 1 -yl)(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)CH(CH3)CH2-;
-(CH2)2-(trαns-cyclohex- 1 ,4-ylene)NH(phen- 1 ,4-ylene)(CH2)2-; -(CH2)2C(O)NH(2-fluorophen- 1 ,4-ylene)CH2-;
-(CH2)2(phen- 1 ,3-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(2,5-difluorophen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(phen- 1 ,4-ylene)(CH2)2-; l-[-CH2(pyrid-2,6-ylene)CH2](piperidin-4-yl)CH2-; -(CH2)3NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2NH(naphth- 1 ,4-ylene)(CH2)2-;
-(CH2)3O(phen- 1 ,4-ylene)CH2-; l-[-(CH2)3](piperidin-4-yl)CH2-;
4-[-(CH2)2](piperidin- 1 -yl)C(O)(phen- 1 ,4-ylene)CH2-; -(CH2)3(phen-l,4-ylene)NHC(O)(CH2)2-;
-(CH2)3O(phen-l,4-ylene)(CH2)2-;
2-[-(CH2)2](benzimidazol-5-yl)CH2-;
-(CH2)2-(tr ns-cyclohex- 1 ,4-ylene)NHC(O)(CH2)2-; -(CH2)2-(tr ns-cyclohex- 1 ,4-ylene)NHC(O)(CH2)4-;
-(CH2)2-(trσns-cyclohex- 1 ,4-ylene)NHC(O)(CH2)5-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)2-;
-(CH2)2NHC(O)NH(phen- 1 ,4-ylene)CH2-; -(CH2)2N(CH3)(CH2)2(c/s-cyclohex- 1 ,4-ylene)-;
-(CH2)2C(O)NH(2,3,5,6-terrafluorophen-l,4-ylene)CH2-;
-(CH2)2C(O)NH(2,6-diiodophen- 1 ,4-ylene)CH2-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)3-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)4-; 4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)5-;
-(CH2)2C(O)NHCH2(phen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)NHCH2(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2-methylphen- 1 ,4-ylene)CH2-; l-[-(CH2)3O(phen-l,4-ylene)(CH2)2](piperidin-4-yl)CH2-; -(CH2)2C(O)NHCH2(phen- 1 ,3-ylene)(CH2)2-;
-(CH2)2O(phen- 1 ,3-ylene)CH2-;
-(CH2)2N(CH3)C(O)CH2O(phen- 1 ,4-ylene)CH2-;
-(CH2)2N(CH3)C(O)CH2O(phen-l,3-ylene)CH2-;
-(CH2)2N(CH3)C(O)(fur-2,5-ylene)CH2-; -(CH2)2N(CH3)C(O)(thien-2,5-ylene)CH2-;
-(CH2)2O(phen- 1 ,4-ylene)O(CH2)2-;
-(CH2)2(trαns-cyclohex- 1 ,4-ylene)NHC(O)(phen- 1 ,4-ylene)CH2-;
-(CH2)2(tr ns-cyclohex- 1 ,4-ylene)NHC(O)CH2O(phen- 1 ,2-ylene)CH2-;
-(CH2)2(tr ns-cyclohex- 1 ,4-ylene)NHC(O)CH2O(phen- 1 ,3-ylene)CH2-; -(CH2)2(trαns-cyclohex- 1 ,4-ylene)NHC(O)CH2O(phen- 1 ,4-ylene)CH2-;
-(CH2)2(tr ns-cyclohex- 1 ,4-ylene)NHC(O)(fur-2,5-ylene)CH2-;
-(CH2)2(tr ns-cyclohex- 1 ,4-ylene)NHC(O)(thien-2,5-ylene)CH2-;
4-[-(CH2)2](piperidin- 1 -yl)C(O)CH2O(phen- 1 ,2-ylene)CH2-;
4-[-(CH2)2](piperidin-l-yl)C(O)CH2O(phen-l,3-ylene)CH2-; 4-[-(CH2)2](piperidin- 1 -yl)C(O)CH2O(phen- 1 ,4-ylene)CH2-;
4-[-(CH2)2](piperidin- 1 -yl)C(O)(fur-2,5-ylene)CH2-;
4-[-(CH2)2](piperidin- 1 -yl)C(O)(thien-2,5-ylene)CH2-;
-(CH2)2(phen- 1 ,4-ylene)NHC(O)(phen- 1 ,3-ylene)CH2-; -(CH2)2(phen- 1 ,4-ylene)NHC(O)(phen- 1 ,4-ylene)CH2-;
-(CH2)2(phen- 1 ,4-ylene)NHC(O)CH2O(phen- 1 ,2-ylene)CH2-;
-(CH2)2(ρhen-l,4-ylene)NHC(O)CH2O(phen-l,3-ylene)CH2-;
-(CH2)2(phen- 1 ,4-ylene)NHC(O)CH2O(phen- 1 ,4-ylene)CH2-; -(CH2)2(phen- 1 ,4-ylene)NHC(O)(fur-2,5-ylene)CH2-;
-(CH2)2(ρhen- 1 ,4-ylene)NHC(O)(thien-2,5-ylene)CH2-;
-(CH2)2(trαns-cyclohex- 1 ,4-ylene)NHC(O)(phen- 1 ,3 -ylene)CH2-;
-(CH2)3O(phen- 1 ,3-ylene)CH2-;
-CH2CH(OH)CH2NH(phen- 1 ,4-ylene)(CH2)2-; -(CH2)4NH(phen-l,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)CH2NHC(O)CH2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)(CH2)2NHC(O)CH2-;
-(CH2)2C(O)NHCH2(tr ns-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(CH2)5-; -(CH2)2O(phen- 1 ,3-ylene)O(CH2)2-;
-(CH2)2O(phen- 1 ,2-ylene)O(CH2)2-;
-CH2(ρhen- 1 ,2-ylene)O(ρhen- 1 ,2-ylene)CH2-;
-(CH2)2C(O)NH(CH2)6-;
-(CH2)3(phen- 1 ,4-ylene)(CH2)3-; -(CH2)3(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)4(phen-l,4-ylene)(CH2)2-;
-(CH2)3(furan-2,5-ylene)(CH2)3-;
-(CH2)2N(CH3)C(O)NH(phen-l,4-ylene)(CH2)2-;
4-[-(CH2)2](piρeridin-l-yl)C(O)NH(phen-l,4-ylene)(CH2)2-; -(CH2)3(phen-l,3-ylene)(CH2)3-;
-(CH2)3(tetrahydrofuran-2,5-ylene)(CH2)3-; and
-(CH2)2O(phen- 1 ,4-ylene)C(O)(CH2)2-. Representative Subgeneric Groupings
The following subgeneric formulae and groupings are intended to provide representative examples of various aspects and embodiments of this invention and as such, they are not intended to exclude other embodiments or to limit the scope of this invention unless otherwise indicated.
A particular group of compounds of formula I are those disclosed in U.S. Provisional Application No. 60/447,843, filed on February 14, 2003. This group includes compounds of formula I; wherein: a is 0 or an integer of from 1 to 3; each R1 is independently selected from (1 -4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl,
(3-6C)cycloalkyl, cyano, halo, -ORla, -C(O)ORIb, SRl0, -S(O)Rld, -S(O)2RIe and -NRlfRlg; each of Rla, Rlb, R,c, RId, Rle, Rlf and Rlg is independently hydrogen or (1- 4C)alkyl; b is 0 or an integer of from 1 to 3; each R2 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR2a, -C(O)OR2b, SR2°, -S(O)R2d, -S(O)2R2e and -NR2fR2g; each of R2a, R2b, R2°, R2d, R2e, R2f and R2g is independently hydrogen or (1- 4C)alkyl;
W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring, and represents O or NWa;
Wa is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R3 is a substituent on carbon independently selected from (l-4C)alkyl, (2-
4C)alkenyl, (2-4C)alkynyl, (3 -6C) cycloalkyl, cyano, halo, -OR3a, -C(O)OR3b, SR3c, -S(O)R3d, -S(O)2R3e and -NR3fR3g; each of R3a, R3b, R3°, R3d, R3e, R3f and R3g is independently hydrogen or (1- 4C)alkyl; R4 is a divalent group ofthe formula:
-(R4V(A1)e-(R4b)f-Q-(R4c)g-(A2)h-(R4d)i- wherein d, e, f, g, h and i are each independently selected from 0 and 1 ;
R4a, R4b, R4c and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from ( 1 -4C)alkyl, fluoro, hydroxy, phenyl and phenyl( 1 -4C)-alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, (2-9C)heteroarylene and (3-6C)heterocyclene; wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (1- 4C)alkyl and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy;
Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Qc)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q and -N( )C(O)O-; Qa, Q , Qc, Qd, Qe, Qf, Qg, Qh, Q1 and are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4; wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4 or R4c to which they are attached, form a 4-6 membered azacycloalkylene group; A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl,
(2-9C)heteroaryl and (3-6C)heterocyclyl; wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 8 to 14;
R5 represents hydrogen or (l-4C)alkyl;
R6 is -NR6aCR6b(O) and R7 is hydrogen, or R6 and R7 together form -NR7aC(O)-CR7b=CR70-, -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7i-CR7jR7k- or - CR71R7m-CR7nR7o-C(O)-NR7p-; each of R6 and R6b is independently hydrogen or (l-4C)alkyl; and each of R7a, R7b, R7c, R7d, R7e, R7f, R7g, R7h, R7i, R7J, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or (l-4C)alkyl; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those disclosed in U.S. Provisional Application No. 60/467,035, filed on May 1, 2003. This group of compounds includes compounds of formula I; wherein: a is 0 or an integer of from 1 to 3; each R1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -ORla, -C(O)ORl , SRlc, -S(O)Rld, -S(O)2Rle, and -NRlfRl ; each of Rla, RIb, Rlc, Rld, R, Rlf and Rl is independently hydrogen or (1- 4C)alkyl; b is 0 or an integer of from 1 to 3; each R2 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR2a, -C(O)OR2b, SR2c, -S(O)R2d, -S(O)2R, and -NR2fR2g; each of R2a, R2b, R2°, R2d, R2e, R2f and R2g is independently hydrogen or ( 1 -
4C)alkyl;
W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring, and represents O or NWa;
Wa is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R3 is a substituent on carbon independently selected from (l-4C)alkyl, (2- 4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR3a, -C(O)OR3b, SR3c, -S(O)R3d, -S(O)2R3e, and -NR3fR3g; each of R3a, R3b, R3°, R3d, R3e, R3f and R3g is independently hydrogen or (1- 4C)alkyl;
R4 is a divalent group ofthe formula:
-(R4a)d-(A])e-(R4 )f-Q-(R4o)g-(A2)h-(R4d)i- wherein d, e, f, g, h and i are each independently selected from 0 and 1 ;
R4a, R4b, R4° and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl(l-4C)-alkyl;
A and A are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, (2-9C)heteroarylene and (3-6C)heterocyclene; wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (1- 4C)alkyl and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy;
Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O , -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Q°)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q and
Figure imgf000028_0001
Qa, Qb, Qc, Qd, Qe, Qf, Qg, Qh, Q; and OJ are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4; wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and ( 1 -4C)alkoxy; or together with the nitrogen atom and the group R4b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl; wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R is attached is in the range of from 4 to 14;
R5 represents hydrogen or (l-4C)alkyl; R6 is -NR6aCR6b(O) or CR6cR6dOR6e and R7 is hydrogen, or R6 and R7 together form -NR7aC(O)-CR7b=CR7c-, -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7i-CR7jR7k- or - CR71R7m-CR7nR7o-C(O)-NR7p-; each of R6a, R6b, R6°, R6d and R6e is independently hydrogen or (l-4C)alkyl; and each of R7a, R7b, R7c, R7d, R7e, R7f, R7g, R7h, R7i, R7j, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or ( 1 -4C)alkyl; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those where: a is 0; b is 0; c is 0; W is O; W is attached at the 4-position ofthe piperidinyl ring; R5 is hydrogen; and R4, R6 and R7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Still another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is NH; W is attached at the 4-position ofthe piperidinyl ring; R5 is hydrogen; and R4, R and R7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Yet another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is O; W is attached at the 4-position ofthe piperidinyl ring; R4 is -(CH2)j- where j is 8, 9 or 10; R5 is hydrogen; and R6 and R7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is NH; W is attached at the 4-position ofthe piperidinyl ring; R4 is -(CH2)j- where j is 8, 9 or 10; R5 is hydrogen; and R6 and R7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Yet another particular group of compounds of fonnula I are those wherein: a is 0; b is 0; c is 0; W is O; W is attached at the 4-position ofthe piperidinyl ring; R4 is -(CH2)2- C(O)NH-(CH )5-; R5 is hydrogen; and R6 and R7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those wherein: a is 0; b is 0; c is 0; W is NH; W is attached at the 4-position ofthe piperidinyl ring; R4 is -(CH2)2- C(O)NH-(CH2)5-; R5 is hydrogen; and R6 and R7 are as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those of formula II as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Another particular group of compounds of formula I are those of formula III as defined herein; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those of formula IN as defined herein; or a phannaceutically acceptable salt or solvate or stereoisomer thereof.
Another particular group of compounds of formula I are those of formula II, III or IN as defined herein, wherein the piperidinyl ring is substitued at the 4-position with a methyl group; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Another particular group of compounds of formula I are compounds of formula N:
Figure imgf000030_0001
N
wherein W, R4, R6 and R7 are as defined in Table I; or a pharmaceutically acceptable salt or solvate thereof.
Table I
Figure imgf000030_0002
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
1 In Tables I-III, "(racemic)" means the compound is racemic at the chiral carbon bearing the hydroxyl group in formula V, VI or NIL
For this group, the nitrogen atom is attached at R6 and carbon atom is attached at R7. Another particular group of compounds of formula I are compounds of formula NI:
Figure imgf000037_0001
VI
wherein W, R1A, R1B, R1C, R2A, R2B, R4, R6 and R7 are as defined in Table II; or a pharmaceutically acceptable salt or solvate thereof.
Table II
Figure imgf000037_0002
Another particular group of compounds of formula I are compounds of formula
Nil:
Figure imgf000038_0001
Nil
wherein W, R4, R6 and R7 are as defined in Table III; or a pharmaceutically acceptable salt or solvate thereof.
Table III
Figure imgf000038_0002
Definitions
When describing the compounds, compositions, methods and processes of this invention, the following tenns 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, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, n- 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 ofthe 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, n-but-2-enyl, n-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, n-propynyl, n-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 "azacycloalkyl" means a monovalent heterocyclic ring containing one nitrogen atom, i.e., a cycloalkyl group in which one carbon atom has been replaced with a nitrogen atom. Unless otherwise defined, such azacycloalkyl groups typically contain from 2 to 9 carbon atoms. Representative examples of an azacycloalkyl group are pyrrolidinyl and piperidinyl groups. The term "azacycloalkylene" means a divalent azacycloakyl group. Representative examples of an azacycloalkylene group are pyrrolidinylene and piperidinylene groups. The tenn "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 tenn "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, by way 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 tenn "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, by way 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 in parentheses preceding the term. For example, the term "(l-4C)alkyl" means an alkyl group having from 1 to 4 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 phannaceutically acceptable inorganic or organic acids. Salts derived from pharmaceutically acceptable inorganic bases include ammonium, calcium, copper, ferric, fenous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like. Particularly prefened 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, N,N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- 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, glucoronic, 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 prefened 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 phannaceutically 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.
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 ofthe disease or medical condition in a patient; (c) suppressing the disease or medical condition, i.e., slowing or arresting the development ofthe disease or medical condition in a patient; or (d) alleviating the symptoms ofthe 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.
The term "protected derivatives thereof means a derivative ofthe specified compound in which one or more functional groups ofthe compound are protected from undesired reactions with a protecting or blocking group. 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. Representative protecting groups for carboxylic acids include esters (such as ajσ-methoxybenzyl ester), amides and hydrazides; for amino groups, carbamates (such as tert-butoxycarbonyl) and amides; for hydroxyl groups, ethers and esters; for thiol groups, thioethers and thioesters; for carbonyl groups, acetals and ketals; and the like. Such protecting groups are well-known to those skilled in the art and are described, for example, in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein.
The term "amino-protecting group" means a protecting group suitable for preventing undesired reactions at an amino group. Representative amino-protecting groups include, but are not limited to, tert-butoxycarbonyl (BOC), trityl (Tr), benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc), formyl, trimethylsilyl (TMS), tert- butyldimethylsilyl (TBS), and the like.
The term "carboxy-protecting group" means a protecting group suitable for preventing undesired reactions at a carboxy group. Representative carboxy-protecting groups include, but are not limited to, esters, such as methyl, ethyl, tert-butyl, benzyl (Bn), / nethoxybenzyl (PMB), 9-fluroenylmethyl (Fm), trimethylsilyl (TMS), tert- butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like.
The tenn "hydroxyl-protecting group" means a protecting group suitable for preventing undesirable reactions at a hydroxyl group. Representative hydroxyl-protecting groups 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)5 jp-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.
General Synthetic Procedures
The biphenyl derivatives of this invention can be prepared from readily available starting materials using the following general methods and procedures or by using other information readily available to those of ordinary skill in the art. Although a particular embodiment ofthe present invention may be shown or described herein, those skilled in the art will recognize that all embodiments or aspects ofthe 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 prefened 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. Protecting groups other than those illustrated in the procedures described herein may be used, if desired. For example, numerous protecting groups, and 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.
By way of illustration, the biphenyl derivatives of this invention can be prepared by a process comprising:
(a) reacting a compound of formula 1 :
Figure imgf000044_0001
or a salt thereof; with a compound of formula 2:
Figure imgf000045_0001
1 1 wherein X represents a leaving group, and P and P each independently represent a hydrogen atom or a hydroxyl-protecting group; (b) reacting a compound of formula 3:
Figure imgf000045_0002
or salt thereof; with a compound of formula 4:
Figure imgf000045_0003
wherein X2 represents a leaving group, and P3 and P4 each independently represent a hydrogen atom or a hydroxyl-protecting group; (c) coupling a compound of formula 5:
Figure imgf000046_0001
with a compound of fonnula 6:
Figure imgf000046_0002
wherein XQa and XQb each independently represent functional groups that couple to form a group Q, P5a represents a hydrogen atom or an amino-protecting group; and P5b and P6 each independently represent a hydrogen atom or a hydroxyl-protecting group;
(d) for a compound of formula I wherein R5 represents a hydrogen atom, reacting a compound of formula 3 with a compound of fonnula 7:
Figure imgf000046_0003
7 or a hydrate thereof (e.g., a glyoxal), in the presence of a reducing agent, wherein P represents a hydrogen atom or a hydroxyl-protecting group; (e) reacting a compound of formula 1 with a compound of formula 8:
Figure imgf000047_0001
8
or a hydrate thereof, in the presence of a reducing agent, wherein P8 and P9 each independently represent a hydrogen atom or a hydroxyl-protecting group, P10 represents a hydrogen atom or an amino-protecting group, and R4 represents a residue that, together with the carbon to which it is attached, affords a group R4 upon completion ofthe reaction;
(f) reacting a compound of formula 9:
Figure imgf000047_0002
wherein X represents a leaving group, with a compound of formula 10:
Figure imgf000047_0003
10 wherein P and P each independently represent a hydrogen atom or a hydroxyl- protecting group, and P13 represents a hydrogen atom or an amino-protecting group; or (g) reacting a compound of formula 11:
Figure imgf000048_0001
11
or a hydrate thereof; wherein R4 represents a residue that, together with the carbon to which it is attached, affords a group R4 upon completion ofthe reaction; with a compound of formula 10 in the presence of a reducing agent; and then removing any protecting group P1, P2, P3, P4, P5a, P5b, P6, P7, P8, P9, P10, P11, P12 or P to provide a compound of formula I; and optionally, forming a pharmaceutically acceptable salt thereof.
Generally, if a salt of one ofthe 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), i.e., the reaction between the compounds of formula 1 and 2, the leaving group represented by X can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate. The groups P1 and P2 can be, for example, trimethylsilyl and benzyl, respectively. This reaction is typically conducted in an inert diluent, such as acetonitrile, in the presence of a base. For example, this reaction can be conducted in the presence of a tertiary amine, such as diisopropylethylamine. Generally, this reaction is conducted at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete. The reaction product is then isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Compounds of formula 1 are generally known in the art or can be prepared from commercially available starting materials and reagents using well-known procedures. For example, compounds of formula 1 can be prepared by deprotecting a compound of formula 12:
Figure imgf000049_0001
12
wherein P14 represents an amino-protecting group, such as a benzyl group. By way of illustration, a benzyl group can be readily removed by reduction using, for example, hydrogen or ammonium formate and a group NIII metal catalyst, such as palladium on carbon. When W represents ΝWa, the hydrogenation reaction is conveniently performed using Pearlman's catalyst (i.e., Pd(OH)2).
Compounds of formula 12 can be prepared by reacting an isocyanate compound of formula 13:
Figure imgf000049_0002
with a compound of formula 14:
Figure imgf000049_0003
14
Compounds of fonnula 2 can be prepared by various procedures described herein or by procedures that are well-known to those skilled in the art. For example, the hydroxyl group of a compound of formula 23 below, can be readily converted into a leaving group using well-known reagents and procedures. By way of illustration, a hydroxyl group can be converted into a halo group using an inorganic acid halide, such as thionyl chloride, phosphorous trichloride, phosphorous tribromide, phosphorous oxychloride and the like, or a halogen acid, such a hydrogen bromide.
In process (b), i.e., the reaction of a compound of formula 3 with a compound of fonnula 4, the leaving represented by X2 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate. The groups P3 and P4 can be, for example, tert-butyldimethylsilyl and benzyl, respectively. This reaction is typically conducted in the presence of a base, such as sodium bicarbonate, and an alkali metal iodide, such as sodium iodide. Generally, this reaction is conducted in an inert diluent, such as tetrahydrofuran, at a temperature ranging from 25 °C to 100 °C until the reaction is substantially complete. The reaction product is then isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like. Compounds of formula 3 can be prepared by deprotecting a compound of formula
15:
Figure imgf000050_0001
15
wherein one or both of P15 and P16 independently represents a protecting group, such as tert-butoxycarbonyl, and any remainder represents a hydrogen atom. For example, a tert-butoxycarbonyl group can be removed by treating the protected compound with trifluoroacetic acid.
Compounds of formula 15 can be prepared by reacting a compound of formula 1 with a compound of fonnula 16:
X3-R4-NP15P16
16 wherein X represents a leaving group such as halo, such as chloro, bromo or iodo, or sulfonic ester group, such as mesylate or tosylate. This reaction is typically conducted by contacting a compound of formula 1 with a compound of fonnula 16 in an inert diluent, such as acetonitrile, DMF or mixtures thereof, at a temperature ranging from about 0 °C to about 100 °C until the reaction is substantially complete.
Alternatively, compounds of formula 3 can be obtained by reductive amination of a compound of formula 11. The reductive amination can be performed by reacting the compound of formula 11 with, for example, benzylamine and hydrogen in the presence of palladium on carbon.
Compounds of formula 11 may be prepared by oxidizing the conesponding alcohol of formula 17:
Figure imgf000051_0001
17
using a suitable oxidizing agent, such as sulfur trioxide pyridine complex and dimethyl sulfoxide. This oxidation reaction is typically conducted in an inert diluent, such as dichloromethane, the presence of a tertiary amine, such as diisopropylethylamine, at a temperature ranging from about -20 °C to about 25 °C.
Compounds of fonnula 17 can be prepared by reacting a compound of formula 1 with a compound of fonnula 18:
X4-R4-OH
18
wherein X4 represents a leaving group such as halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate.
Compounds of formula 4 can be prepared by reacting a compound of formula 19:
Figure imgf000052_0001
with a reducing agent, such as borane. If desired, such a reduction can be performed in the presence of a chiral catalyst to provide compounds of formula 4 in chiral form. For example, compounds of formula 19 can be reduced in the presence of a chiral catalyst formed from (R)-(+)-α,α-diphenyl-2-pynolidinemethanol and trimethylboroxine; or alternatively, from (_S)-(-)-α,α-diphenyl-2-pyreolidinemethanol and trimethylboroxine. The resulting hydroxyl group can then be protected with a hydroxyl-protecting group, P , by reaction with, for example, tert-butyldimethylsilyl trifluoromethanesulfonate.
Compounds of formula 19 in which X2 represents a bromine atom can be prepared by reacting a compound of formula 20:
Figure imgf000052_0002
20
with bromine in the presence of a Lewis acid, such as boron trifluoride diethyl etherate. Compounds of formula 20 are well-known in the art or can be prepared by well-known procedures using commercially available starting materials and reagents. Referring to process (c), i.e., the reaction of a compound of formula 5 with a compound of fonnula 6, it will be appreciated that the groups XQa and XQb should be selected so as to afford the desired group Q upon completion ofthe reaction. For example, when the desired group Q is an amide group, i.e., -N(Qa)C(O)- or -C(O)N(Qb), one of XQa and XQb can be an amine group (i.e., -NHQa or -NHQb) and the other can be a carboxyl group (i.e., -COOH) or a reactive derivative thereof (such as acyl halide, such as an acyl chloride or acyl bromide). The groups P5a, P5b and P6 can be, for example, benzyl, trimethylsilyl and benzyl, respectively. When Q is an amide group, the reaction can be performed under conventional amide coupling conditions. Similarly, when the desired group Q is a sulfonamide, i.e., -N(Qc)S(O)2- or -S(O)2N(Qd)-, one of XQa and XQb can be an amine group, -NHQC or -NHQd and the other can be a sulfonyl halide group (such as sulfonyl chloride or sulfonyl bromide).
Compounds of formula 5 can be prepared by reacting a compound of formula 1 with a compound of formula 21:
X5-(R4a)d-(AI)e-(R4b)rXQa' 21
wherein X5 represents a leaving group including halo, such as chloro, bromo or iodo, and a sulfonic ester group, such as mesylate or tosylate; and XQa represents XQa , such as a carboxyl group or an amino group NHQa, or a protected derivative thereof, such as a (1- 6C)alkoxycarbonylamino group or a tert-butoxycarbonylamino group. This reaction is typically conducted by a method analogous to that used to prepare compounds of formula 3, followed by removing any protecting group in XQ .
Compounds of formula 6 can be prepared by reacting a compound of formula 4 with a compound of formula 22:
XQb'-(R4c)g-(A2)h-(R4d)i-X6 22
wherein X6 represents a leaving group including halo, such as chloro, bromo or iodo, and a sulfonic ester group, such as mesylate or tosylate; and XQb represents XQ , such as a carboxyl group or an amino group NHQb, or a protected derivative thereof, such as a (1- 6C)alkoxycarbonyl group or a tert-butoxycarbonylamino group. This reaction is typically conducted by a method analogous to that used to prepare compounds of fonnula 3, followed by removing any protecting group in XQb . Referring to process (d), i.e., the reaction of a compound of fonnula 3 with a compound of fonnula 7, any suitable reducing agent may be used in this reaction. For example, the reducing agent can be hydrogen in the presence of a Group NIII metal catalyst, such as palladium on carbon; or a metal hydride reagent, such as sodium triacetoxyborohydride. The group P7 can be, for example, benzyl. This reaction is typically conducted in an inert diluent and a protic solvent, such as a mixture of dichloroethane and methanol, at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete. Compounds of formula 7 in the form of a hydrate can be prepared by conventional procedures, for example, by dibrominating a compound of formula 19 (where X2 in this case can also be hydrogen), and then hydrolyzing the resulting dibromide to form a glyoxal or a hydrate thereof. For example, a compound of formula 19 can be reacted with hydrogen bromide and then hydrolyzed with water to form the corresponding glyoxal hydrate.
Referring to process (e), i.e., the reaction of a compound of formula 1 with a compound of formula 8, any suitable reducing agent may be used in this reaction. For example, the reducing agent may be hydrogen in the presence of a Group NIII metal catalyst, such as palladium on carbon; or a metal hydride reagent, such as sodium triacetoxyborohydride. The groups P8, P9 and P10 can be, for example, trimethylsilyl, benzyl and benzyl, respectively. Typically, this reduction reaction is conducted in an inert diluent and a protic solvent, such as dichloroethane and methanol, at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
Compounds of formula 8 may be prepared by oxidizing a compound of formula 23:
Figure imgf000054_0001
23
using any suitable oxidizing agent, such as sulfur trioxide pyridine complex and dimethyl sulfoxide. This reaction is typically conducted in the presence of a tertiary amine, such as diisopropylethylamine, at a temperature in the range of from about -20 °C to about 25 °C until the oxidation is substantially complete.
Compounds of formula 23 can be prepared by reacting a compound of formula 10 with a compound of formula 24: HO-R4-X7 24
wherein X7 represents a leaving group including halo, such as chloro, bromo or iodo, and a sulfonic ester group, such as mesylate or tosylate.
Referring to process (f), i.e., the reaction of a compound of formula 9 with a compound of fonnula 10, the leaving group represented by X3 can be, for example, halo, such as chloro, bromo or iodo, or a sulfonic ester group, such as mesylate or tosylate. The groups P11, P12 and P13 can be, for example, trimethylsilyl, benzyl and benzyl, respectively. This reaction is typically conducted an inert diluent, such as acetonitrile, in the presence of a suitable base. For example, this reaction can be conducted in the presence of a tertiary amine, such as diisopropylethylamine. Generally, this reaction is conducted at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete. Compounds of formula 9 can be prepared by steps analogous to those of methods
(a) to (e) herein, starting from a compound of fonnula 1. Additionally, compounds of fonnula 10 can be prepared from compounds of formula 4 by reaction with an amine of formula PI3NH2.
Referring to process (g), i.e., the reaction of a compound of formula 11 with a compound of fonnula 10, any suitable reducing agent may be used in this reaction. For example, the reducing agent may be hydrogen in the presence of a Group NIII metal catalyst, such as palladium on carbon; or a metal hydride reagent, such as sodium triacetoxyborohydride. The groups P11, P12 and P13 can be, for example, tert- butyldimethylsilyl, benzyl and benzyl, respectively. Typically, this reduction reaction is conducted in an inert diluent and a protic solvent, such as dichloroethane and methanol, at a temperature in the range of from 0 °C to 100 °C until the reaction is substantially complete.
Compounds of formula 11 are readily prepared by oxidation ofthe corresponding alcohol or by hydrolysis ofthe conesponding acetal. Any suitable oxidizing agent may be employed in this reaction to provide the aldehyde, such as sulfur trioxide pyridine complex and dimethyl sulfoxide. The acetal may be hydrolyzed under conventional conditions using aqueous acid to provide the aldehyde. In a particular embodiment, certain compounds of formula I are prepared by a process comprising:
(h) deprotecting a compound of formula 25:
25
wherein P17 represents a hydrogen atom or an amino-protecting group; and each of P18, P19 and P20 independently represent a hydrogen atom or a hydroxyl-protecting group; provided that at least one of P17, P18, P19 or P20 is a protecting group; (i) deprotecting a compound of formula 26:
Figure imgf000056_0002
26
wherein P21 represents a hydrogen atom or an amino-protecting group; and each of P22 and P23 independently represent a hydrogen atom or a hydroxyl-protecting group; provided that at least one of P , P or P is a protecting group; or (j) deprotecting a compound of formula 27:
Figure imgf000057_0001
27
wherein P24 represents a hydrogen atom or an amino-protecting group; and each of P25 and P26 independently represent a hydrogen atom or a hydroxyl-protecting group;
94. 9*ϊ ή provided that at least one of P , P or P is a protecting group. to provide a compound of formula I and, optionally, forming a pharmaceutically acceptable salt of the compound of fonnula I.
17 I S 1 Q 90
Referring to process (h), examples of particular values for P , P , P and P are:
1 7 1 Ji 1 Q 90 for P , hydrogen or benzyl; for P hydrogen or tert-butyldimethylsilyl; and for P and P hydrogen or benzyl, or together propylidine. In this process, benzyl protecting groups are conveniently removed by catalytic hydrogenation in the presence of a Group NIII metal catalyst, such as palladium on carbon; a tert-butyldimethylsilyl group is conveniently removed by treatment with hydrogen fluoride, such as triethylamine trihydrofluoride; and a propylidine group is conveniently removed by treatment with an acid, such as trifluoroacetic acid.
Compounds of formula 25 can be prepared by the methods described herein, such as by processes (a) to (g). Alternatively, compounds of formula 25 can be prepared by reacting a compound of formula 28:
Figure imgf000058_0001
28
wherein R8 represents -CH2OP19, -CHO, -COOH or -C(O)O(l-6C)alkoxy, such as carbomethoxy, R9 represents -OP18 and R10 represents a hydrogen atom, or R9 and R10 together represent =O, with a reducing agent. Any suitable reducing agent may be used in this reaction including, by way of example, metal hydride reducing agents, such as sodium borohydride, lithium aluminum hydride and the like.
Compounds of formula 28 in which R9 and R10 together represent a =O group can be readily prepared by reacting a compound of formula 29:
Figure imgf000058_0002
or a salt thereof, with a compound of formula 30:
Figure imgf000058_0003
30 wherein X represents a leaving group, such as a bromo.
91 99 9^
Referring to process (i), examples of particular values for P , P and P are: for P21, hydrogen or benzyl; for P22 hydrogen or tert-butyldimethylsilyl; and for P23 hydrogen or benzyl. In this process, benzyl protecting groups are conveniently removed by catalytic hydrogenation in the presence of a Group NIII metal catalyst, such as palladium on carbon; and a tert-butyldimethylsilyl group is conveniently removed by treatment with hydrogen fluoride, such as triethylamine trihydrofluoride. Compounds of formula 26 can be prepared by the methods described herein, such as by processes (a) to (g).
Referring to process (j), examples of particular values for P , P and P are: for P , hydrogen or benzyl; for P hydrogen or tert-butyldimethylsilyl; and for P hydrogen or benzyl. In this process, benzyl protecting groups are conveniently removed by catalytic hydrogenation in the presence of a Group NIII metal catalyst, such as palladium on carbon; and a tert-butyldimethylsilyl group is conveniently removed by treatment with hydrogen fluoride, such as triethylamine trihydrofluoride. Compounds of formula 27 can be prepared by the methods described herein, such as by processes (a) to (g).
Additionally, compounds of formula I in which R6 and R7 together form -ΝR7gC(O)-CR7hR7i-CR7jR7k- or -CR7IR7m-CR7nR7o-C(O) -NR7p- may be prepared by
( 7 reducing a conesponding compound of fonnula I in which R and R together form -NR7aC(O)-CR7 =CR7c- or -CR7d=CR - C(O)-NR7f-, for example by catalytic hydrogenation as described in Example 6 hereinafter.
Further details regarding specific reaction conditions and other procedures for preparing representative compounds of this invention or intermediates thereof are described in the Examples set forth below.
Pharmaceutical Compositions and Formulations
The biphenyl derivatives of this invention are typically administered to a patient in the form of a pharmaceutical composition or formulation. Such pharmaceutical compositions may be administered to the patient 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 ofthe compounds of this invention, (i.e., free base, pharmaceutically acceptable salt, solvate, etc.) that is suitable for the particular mode of administration can be used in the pharmaceutical compositions discussed herein. Accordingly, in one of its compositions aspects, this 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 thereof. Optionally, such pharmaceutical compositions may contain other therapeutic and/or formulating agents if desired.
The pharmaceutical compositions of this invention typically contain a therapeutically effective amount of a compound ofthe present invention or a pharmaceutically acceptable salt thereof. Typically, such pharmaceutical compositions will contain from about 0.01 to about 95% by weight ofthe active agent; including, from about 0.01 to about 30% by weight; such as from about 0.01 to about 10% by weight ofthe active agent.
Any conventional carrier or excipient may be used in the pharmaceutical compositions of this invention. The choice of a particular carrier or excipient, or combinations of carriers or exipients, 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 pharmaceutical 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. By way 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, 7l 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: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; (21) compressed propellant gases, such as chlorofluorocarbons and hydrofluorocarbons; and (22) other non-toxic compatible substances employed in pharmaceutical compositions. The pharmaceutical compositions of this invention are typically prepared by thoroughly and intimately mixing or blending a compound ofthe invention 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 this invention are suitable for inhaled administration. Suitable pharmaceutical compositions for inhaled administration will typically be in the form of an aerosol or a powder. Such compositions are generally administered using well-known delivery devices, such as a nebulizer inhaler, a metered-dose inhaler (MDI), a dry powder inhaler (DPI) or a similar delivery device. In a specific embodiment of this invention, the 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 pharmaceutical 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 ofthe particles with a diameter of less than about 10 μm. Suitable nebulizer devices are provided commercially, for example, by PARI GmbH (Stamberg, German). Other nebulizer devices include Respimat (Boehringer Ingelheim) and those disclosed, for example, in U.S. Patent No. 6,123,068 and WO 97/12687. 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/mL of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
In another specific embodiment of this invention, the pharmaceutical composition comprising the active agent is administered by inhalation using a dry powder inhaler. Such dry powder inhalers 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.
A representative pharmaceutical composition for use in a dry powder inhaler 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 phannaceutical 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 dry powder inhaler delivery devices include Diskhaler (GlaxoSmithKline, Research Triangle Park, NC) (see, e.g., U.S. Patent No. 5,035,237); Diskus (GlaxoSmithKline) (see, e.g., U.S. Patent No. 6,378,519; Turbuhaler (AstraZeneca, Wilmington, DE) (see, e.g., U.S. Patent No. 4,524,769); Rotahaler (GlaxoSmithKline) (see, e.g., U.S. Patent No. 4,353,365) and Handihaler (Boehringer Ingelheim). Further examples of suitable DPI devices are described in U.S. Patent Nos. 5,415,162, 5,239,993, and 5,715,810 and references cited therein. In yet another specific embodiment of this invention, the pharmaceutical composition comprising the active agent is administered by inhalation using a metered- dose inhaler. Such metered-dose inhalers typically discharge a measured amount ofthe active agent or a pharmaceutically acceptable salt thereof using compressed propellant gas. Accordingly, pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension ofthe active agent in a liquefied propellant. Any suitable liquefied propellant may be employed including chlorofluorocarbons, such as CC13F, and hydro fluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoro-n-propane, (HFA 227). Due to concerns about chlorofluorocarbons affecting the ozone layer, formulations containing HFAs are generally prefened. 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, EP 0717987 A2, and WO 92/22286. A representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01 % to about 5 % by weight of a compound of formula I, or a phannaceutically acceptable salt or solvate or stereoisomer thereof; from about 0 % to about 20 % by weight ethanol; and from about 0 % to about 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 metered-dose inhaler devices developed specifically for use with HFA propellants are provided in U.S. Patent Nos. 6,006,745 and 6,143,277. Alternatively, a suspension formulation can be prepared by spray drying a coating of surfactant on micronized particles ofthe active agent. See, for example, WO 99/53901 and WO 00/61108. 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, 5,983,956, 5,874,063, and 6,221,398, and WO 99/55319 and WO 00/30614.
In another embodiment, the pharmaceutical compositions of this invention are suitable for oral administration. Suitable pharmaceutical 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 ofthe 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 pharmaceutical compositions of this invention will typically comprise a compound ofthe present invention as the active ingredient and one or more pharmaceutically acceptable earners, such as sodium citrate or dicalcium phosphate. Optionally or alternatively, such solid dosage forms may also comprise: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar- agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and/or sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as cetyl alcohol and/or glycerol monostearate; (8) absorbents, such as kaolin and/or bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and/or mixtures thereof; (10) coloring agents; and (11) buffering agents.
Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the pharmaceutical compositions of this invention. Examples of pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) 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 (PNAP), 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 pharmaceutical compositions ofthe present invention may also be formulated to provide slow or controlled release ofthe active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres. In addition, the pharmaceutical compositions of the present invention may optionally contain opacifying agents and may be formulated so that they release the active ingredient only, or preferentially, in a certain portion ofthe 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 ofthe above-described excipients.
Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. Such 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-butyl ene glycol, oils (esp., 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 pharmaceutical compositions of this 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 this invention can also be administered transdermally using known transdermal delivery systems and excipents. For example, a compound of this invention can be admixed with permeation enhancers, such as propylene glycol, polyethylene glycolm 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 pharmaceutical compositions of this invention may also contain other therapeutic agents that are co-administered with a compound of formula I, or pharmaceutically acceptable salt or solvate or stereoisomer thereof. For example, the phannaceutical compositions of this invention may further comprise one or more therapeutic agents selected from other bronchodilators (e.g., PDE3 inhibitors,adenosine 2b modulators and β2 adrenergic receptor agonists); anti-inflammatory agents (e.g. steroidal anti-inflammatory agents, such as corticosteroids; non-steroidal anti-inflammatory 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). The other therapeutic agents can be used in the form of pharmaceutically acceptable salts or solvates. Additionally, if appropriate, the other therapeutic agents can be used as optically pure stereoisomers.
Representative β adrenergic receptor agonists that can be used in combination with, and in addition to, the compounds of this invention 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 this 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, published on August 29, 2002; 3-[3-(4-{[6- ([(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]ethyl}amino)hexyl]oxy}butyl)- phenyl]imidazolidine-2,4-dione and related compounds disclosed in WO 02/070490, published September 12, 2002; 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-(fonnylamino)-4-hydroxyphenyl]-2- hydroxyethyl} amino)hexyl]oxy}butyl)-benzenesulfonamide, N-(tert-butyl)-3-(4- { [6- ({(2R)-2-[3-(formylamino)-4-hydroxyphenyl]-2-hydroxyethyl}amino)hexyl]- oxy}butyl)benzenesulfonamide, N-(tert-butyl)-3-(4-{[6-({(2S)-2-[3-(fonnylamino)-4- hydroxyρhenyl]-2-hydroxyethyl}amino)-hexyl]oxy}butyl)-benzenesulfonamide, N-(tert- butyl)-3-(4- {[6-( {(2R/S)-2-[3-(fonnylamino)-4-hydroxyphenyl]-2- hydroxyethyl}amino)hexyl]-oxy}butyl)benzenesulfonamide and related compounds disclosed in WO 02/076933, published on October 3, 2002; 4-{(lR)-2-[(6-{2-[(2,6- dichlorobenzyl)oxy] ethoxy} hexyl)amino] - 1 -hydroxyethyl} -2-(hydroxymethyl)phenol and related compounds disclosed in WO 03/024439, published on March 27, 2003; and pharmaceutically acceptable salts thereof. 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 μg to about 500 μg per dose.
Representative steroidal anti-inflammatory agents that can be used in combination with the compounds of this invention 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 S-fluoromethyl ester, 6,9-difluoro- 11 -hydroxy- 16 -methyl-3-oxo-17 -propionyloxy- androsta-l,4-diene-l 7-carbothioic acid 5,-(2-oxo-tetrahydrofuran-3S-yl) 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 anti- inflammatory 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 μg to about 500 μg per dose.
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 ( IL antibody), specifically, an IL-4 therapy, an IL-13 therapy, or a combination thereof); or inhibitors of cytokine synthesis.
For example, representative phosphodiesterase-4 (PDE4) inhibitors or mixed PDE3/PDE4 inhibitors that can be used in combination with the compounds of this invention include, but are not limited to cis 4-cyano-4-(3-cyclopentyloxy-4- methoxyphenyl)cyclohexan- 1 -carboxylic acid, 2-carbomethoxy-4-eyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan- 1 -one; c/s-[4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan- 1 -ol] ; czs-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 (LNSERM); 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-l 1294A (Napp); roflumilast (Byk-Gulden); pthalazinone compounds disclosed in WO99/47505 (Byk-Gulden); Pumafentrine (Byk-Gulden, now Altana); arofylline (Almirall- Prodesfarma); NM554/UM565 (Nernalis); 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 this invention include, but are not limited to, atropine, atropine sulfate, atropine oxide, methylatropine nitrate, homatropine hydrobromide, hyoscyamine (d, l) hydrobromide, scopolamine hydrobromide, ipratropium bromide, oxitropium bromide, tiotropium bromide, methantheline, propantheline bromide, anisotropine methyl bromide, clidinium bromide, copynolate (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) that can be used in combination with the compounds of this invention include, but are not limited to, ethanolamines, such as carbinoxamine maleate, clemastine fumarate, diphenylhydramine hydrochloride and dimenhydrinate; ethylenediamines, such as pyrilamine amleate, tripelennamine 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. Suitable doses for the other therapeutic agents administered in combination with a compound ofthe invention are in the range of about 0.05 g/day to about 100 mg/day.
The following formulations illustrate representative pharmaceutical compositions ofthe present invention: Formulation Example A A dry powder for administration by inhalation is prepared as follows: Ingredients Amount
Compound of the invention 0.2 mg
Lactose 25 mg
Representative Procedure: The compound ofthe invention is micronized and then blended with lactose. This blended mixture is then loaded into a gelatin inhalation cartridge. The contents ofthe cartridge are administered using a powder inhaler.
Formulation Example B A dry powder formulation for use in a dry powder inhalation device is prepared as follows:
Representative Procedure: A pharmaceutical composition is prepared having a bulk formulation ratio of micronized compound ofthe invention to lactose of 1 :200. The composition is packed into a dry powder inhalation device capable of delivering between about 10 μg and about 100 μg ofthe compound ofthe invention per dose.
Formulation Example C A dry powder for administration by inhalation in a metered dose inhaler is prepared as follows:
Representative Procedure: A suspension containing 5 wt. % of a compound ofthe invention and 0.1 wt. % lecithin is prepared by dispersing 10 g ofthe compound ofthe invention as micronized particles with mean size less than 10 μm in a solution formed from 0.2 g of lecithin dissolved in 200 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 cartridges with pressurized 1,1,1,2-tetrafluoroethane. Formulation Example D A pharmaceutical composition for use in a metered dose inhaler is prepared as follows: Representative Procedure: A suspension containing 5 % compound ofthe invention, 0.5 % lecithin, and 0.5 % trehalose is prepared by dispersing 5 g of active ingredient as micronized particles with 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 μm. The particles are loaded into canisters with pressurized 1,1,1 ,2-tetrafluoroethane.
Formulation Example E
A pharmaceutical composition for use in a nebulizer inhaler is prepared as follows: Representative Procedure: An aqueous aerosol formulation for use in a nebulizer is prepared by dissolving 0.1 mg ofthe compound ofthe invention in 1 mL of a 0.9 % sodium chloride solution acidified with citric acid. The mixture is stined and sonicated until the active ingredient is dissolved. The pH ofthe solution is adjusted to a value in the range of from 3 to 8 by the slow addition of NaOH.
Formulation Example F
Hard gelatin capsules for oral administration are prepared as follows:
Ingredients Amount
Compound of the invention 250 mg
Lactose (spray-dried) 200 mg
Magnesium stearate 10 mg
Representative Procedure: The ingredients are thoroughly blended and then loaded into a hard gelatin capsule (460 mg of composition per capsule). Formulation Example G A suspension for oral administration is prepared as follows: Ingredients Amount
Compound ofthe invention 1.0 g
Fumaric acid 0.5 g
Sodium chloride 2.0 g
Methyl paraben 0.15 g
Propyl paraben 0.05 g Granulated sugar 25.5 g
Sorbitol (70% solution) 12.85 g
Neegum k (Nanderbilt Co.) 1.0 g
Flavoring 0.035 mL
Colorings 0.5 mg Distilled water q.s. to lOO mL
Representative Procedure: The ingredients are mixed to form a suspension containing 100 mg of active ingredient per 10 mL of suspension.
Formulation Example H
An injectable formulation is prepared as follows: Ingredients Amount
Compound ofthe invention 0.2 g Sodium acetate buffer solution (0.4 M) 2.0 mL
HCI (0.5 Ν) or ΝaOH (0.5 Ν) q.s. to pH 4
Water (distilled, sterile) q.s. to 20 mL
Representative Procedure: The above ingredients are blended and the pH is adjusted to 4 ± 0.5 using 0.5 Ν HCI or 0.5 Ν ΝaOH. Utility
The biphenyl derivatives of this invention possess both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity and therefore, such compounds are useful for treating medical conditions mediated by β2 adrenergic receptors or muscarinic receptors, i.e., medical conditions that are ameliorated by treatment with a β2 adrenergic receptor agonist or a muscarinic receptor antagonist. Such medical conditions include, by way of example, pulmonary disorders or diseases associated with reversible airway obstruction, such as chronic obstructive pulmonary disease (e.g., chronic and wheezy bronchitis and emphysema), asthma, pulmonary fibrosis and the like. Other conditions which may be treated include premature labor, depression, congestive heart failure, skin diseases (e.g., inflammatory, allergic, psoriatic and proliferative skin diseases, conditions where lowering peptic acidity is desirable (e.g., peptic and gastric ulceration) and muscle wasting disease.
Accordingly, in one embodiment, this invention is directed to a method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. When used to treat a pulmonary disorder, the compounds of this 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 μg/day to about 200 μg/day.
When administered by inhalation, the compounds of this invention typically have the effect of providing bronchodilation. Accordingly, in another of its method aspects, this invention is directed to a method of providing bronchodilation in a patient, the method comprising administering to a patient requiring bronchodilation a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. Generally, the dose for providing bronchodilation will range from about 10 μg/day to about 200 μg/day.
In one embodiment, this invention is directed to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. When used to treat a COPD or asthma, the compounds of this invention will typically be administered by inhalation in multiple doses per day or in a single daily dose. Generally, the dose for treating COPD or asthma will range from about 10 μg/day to about 200 μg/day.
As used herein, COPD includes chronic obstructive bronchitis and emphysema (see, for example, Barnes, Chronic Obstructive Pulmonary Disease, NEngl JMed 2000: 343:269-78).
When used to treat a pulmonary disorder, the compounds of this invention are optionally administered in combination with other therapeutic agents. In particular, by combining the compounds of this invention with a steroidal anti-inflammatory agent (e.g. a corticosteroid), the pharmaceutical compositions of this invention can provide triple therapy, i.e., β2 adrenergic receptor agonist, muscarinic receptor antagonist and anti- inflammatory activity, using only two active components. Since pharmaceutical compositions containing two active components are typically easier to formulate compared to compositions containing three active components, such two component compositions provide a significant advantage over compositions containing three active components. Accordingly, in a particular embodiment, the pharmaceutical compositions and methods of this invention further comprise a therapeutically effective amount of a steroidal anti- inflammatory agent.
Compounds of this invention exhibit both muscarinic receptor antagonist and β2 adrenergic receptor agonist activity. Accordingly, among other properties, compounds of particular interest are those that demonstrate an inhibitory constant K; value for binding at the M3 muscarinic receptor and an EC50 value for β2 adrenergic receptor agonist activity of less than about 100 nM; particularly less than 10 nM. Among these compounds, compounds of special interest include those having muscarinic activity, expressed in terms ofthe inhibitory constant Kj for binding at the M3 muscarinic receptor, that is about equal to the compound's β2 adrenergic agonist activity, expressed in terms ofthe half maximal effective concentration EC50, as determined in the in vitro assays described herein, or in similar assays. For example, compounds of particular interest are those having a ratio of the inhibitory constant Kj for the M3 muscarinic receptor to the EC50 for the β2 adrenergic receptor ranging from about 30:1 to about 1:30; including about 20:1 to about 1:20; such as about 10:1 to about 1:10.
In one of its method aspects, the present invention also provides a method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound having both muscarinic receptor antagonist and β2 adrenergic receptor agonist activity. In a particular embodiment of this method, the compound administered has an inhibitory constant K; for the M3 muscarinic receptor that is less than about 100 nM and a half maximal effective concentration EC50 for agonism at the β2 adrenergic receptor that is less than about 100 nM. In another embodiment, the method for treating a pulmonary disorder comprises administering a therapeutically effective amount of a compound for which the ratio ofthe inhibitory constant K; for the M3 muscarinic receptor to the EC50 for agonism ofthe β2 adrenergic receptor is between about 30: 1 and about 1 :30.
Since compounds of this invention possess both β2 adrenergic agonist activity and muscarinic receptor antagonist activity, such compounds are also useful as research tools for investigating or studying biological systems or samples having β2 adrenergic receptors or muscarinic receptors, or for discovering new compounds having both β2 adrenergic agonist activity and muscarinic receptor antagonist activity. Such biological systems or samples may comprise β2 adrenergic receptors and/or muscarinic receptors. Any suitable biological system or sample having β2 adrenergic and/or 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. In this embodiment, a biological system or sample comprising a β2 adrenergic receptor or a muscarinic receptor is contacted with a β2 adrenergic receptor-agonizing or muscarinic receptor-antagonizing amount of a compound of this invention. The effects 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 ofthe enzyme adenylyl cyclase (which synthesizes cAMP), ligand- mediated changes in incorporation of guanosine 5'-O-( -thio)triphosphate ([35S]GTP S) into isolated membranes via receptor catalyzed exchange of [35S]GTP S 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 this invention will agonize or cause activation of a β2 adrenergic receptor and antagonize or decrease the activation of muscarinic receptors in any ofthe functional assays listed above, or assays of a similar nature. The amount of compound used in these studies will typically range from about 0.1 nanomolar to about 100 nanomolar.
Additionally, the compounds of this invention can be used as research tools for discovering new compounds that have both a β2 adrenergic receptor agonist and muscarinic receptor antagonist activity. In this embodiment, a β2 adrenergic receptor and muscarinic receptor binding data (for example, as determined by in vitro radioligand displacement assays) for a test compound or a group of test compounds is compared to the β2 adrenergic receptor and muscarinic receptor binding data for a compound of this invention to identify those test compounds that have about equal or superior β2 adrenergic receptor and/or 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 ofthe test data to identify test compounds of interest.
In some cases, compounds of this invention may possess either weak muscarinic receptor antagonist activity or weak β2 adrenergic receptor agonist activity. In these cases, those of ordinary skill in the art will recognize that such compounds still have utility as primarily either a β2 adrenergic receptor agonist or a muscarinic receptor antagonist, respectively.
The properties and utility ofthe compounds of this 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 are provided to illustrate specific embodiments of this invention. These specific embodiments, however, are not intended to limit the scope of this invention in any way unless specifically indicated.
The following abbreviations have the following meanings unless otherwise indicated and any other abbreviations used herein and not defined have their standard meaning: AC adenylyl cyclase
Ach acetylcholine
ATCC American Type Culture Collection
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 NN-diisopropylethylamine dPBS Dulbecco's phosphate buffered saline
DMEM Dulbecco's Modified Eagle's Medium
DMSO dimethyl sulfoxide
EDTA ethylenediaminetetraacetic acid
E ax maximal efficacy
EtOAc ethyl acetate
EtOH ethanol
FBS fetal bovine serum
FLIPR fluorometric imaging plate reader
Gly glycine
HATU <9-(7-azabenzotriazol- 1 -yl-NNN',N'-tetramethyluronium hexafluorophosphate
HBSS Hank's buffered salt solution
HEK human embryonic kidney cells
HEPES 4-(2-hydroxyethyl)- 1 -piperazineethanesulfonic acid hMi cloned human Mj receptor hM2 cloned human M2 receptor hM3 cloned human M3 receptor hM4 cloned human M4 receptor hM5 cloned human M5 receptor
HPLC high-performance liquid chromatography
IBMX 3 -isobutyl- 1 -methylxanthine
%Eff % efficacy
PBS phosphate buffered saline
PyBOP benzotriazol- 1 -yloxytripyrrolidinophosphonium hexafluorophosphate rpm rotations per minute
TFA trifluoroacetic acid
THF tetrahydrofuran
Tris tris(hydroxymethyl)aminomethane
Unless noted otherwise, reagents, starting materials and solvents were purchased from commercial suppliers (such as Aldrich, Fluka, Sigma and the like) and were used without further purification. In the examples described below, HPLC analysis was conducted using an Agilent
(Palo Alto, CA) Series 1100 instrument with Zorbax Bonus RP 2.1 x 50 mm columns, supplied by Agilent, (a C14 column), having a 3.5 micron particle size. Detection was by UN absorbance at 214 nm. HPLC 10-70 data was obtained with a flow rate of 0.5 mL/minute of 10%-70% B over 6 minutes. Mobile phase A was 2 % - 98 % - 0.1% ACΝ-H2O-TFA; and mobile phase B was 90 % - 10 % - 0.1 % ACN-H2O-TFA. Using the mobile phases A and B described above, HPLC 5-35 data and HPLC 10-90 data were obtained with 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 with a 10 % - 90 % mobile phase B over a 5 minute gradient. Small scale purification was conducted using an API 150EX Prep Workstation system from Applied Biosystems. The mobile phase was A: water + 0.05% v/v TFA; and B: acetonitrile + 0.05% v/v TFA. For anays (typically about 3 to 50 mg recovered sample size) the following conditions were used: 20 mL/min flow rate; 15 min 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 mL/min flow rate; 30 min gradients and a 41.4 mm x 250 mm Microsorb BDS column with 10 micron particles (Narian, Palo Alto, CA).
The specific rotation for chiral compounds (indicated as [α]20o) was measured using a Jasco Polarimeter (Model P- 1010) with a tungsten halogen light source and a 589 nm filter at 20°C. Samples of test compounds were typically measured at 1 mg/mL water.
Preparation 1 N-l,l'-Biphenyl-2-yl-iV-4-(l-benzyl)piperidinylurea Biphenyl-2-isocyanate (50 g, 256 mmol) was dissolved in acetonitrile (400 mL) at ambient temperature. After cooling to 0°C, a solution of 4-amino-N-benzylpiperidine (48.8 g, 256 mmol) in acetonitrile (400 mL) was added over 5 min. A precipitate was observed immediately. After 15 min, acetonitrile (600 mL) was added, and the resultant viscous mixture was stined for 12 h at 35°C. The solids were then filtered off and washed with cold acetonitrile, then dried under vacuum, yielding the title compound (100 g, 98% yield). MS m/z: [M + H+] calcd for C25H27Ν3O 386.22; found 386.3. Preparation 2 N-l,l'-Biphenyl-2-yl-iV-4-piperidinylurea
The product of Preparation 1 (20 g, 52 mmol) was dissolved in a mixture of anhydrous methanol and anhydrous DMF (3:1, 800 mL). Aqueous hydrochloric acid (0.75 mL of 37% cone, solution, 7.6 mmol) was added and nitrogen gas was bubbled through the solution vigorously for 20 min. Pearlman's catalyst (Pd(OH)2, 5 g) was added under a stream of nitrogen, before placing the reaction mixture under a hydrogen atmosphere (balloon). The reaction mixture was allowed to stir for 4 days and was then passed twice through pads of Celite to remove the catalyst. The solvent was then removed under reduced pressure to yield the title compound (13 g, 85% yield). MS m/z: [M + H+] calcd for Cι8H2IN3O 296.17; found 296.0.
Alternatively, N-1,1 '-biphenyl-2-yl-iV-4-piperidinylurea was synthesized by heating together biphenyl-2-isocyanate (50 g, 256 mmol) and 4-amino-N-benzylpiperidine (51.1 g, 269 mmol) at 70 °C for 12 h (the reaction was monitored by LCMS). The reaction mixture was cooled to 50 °C and ethanol (500 mL) added, followed by slow addition of 6M hydrochloric acid (95 mL). The reaction mixture was cooled to room temperature. Ammonium formate (48.4 g, 768 mmol) was added to the reaction mixture and nitrogen gas bubbled through the solution vigorously for 20 min, before adding palladium (10 wt. % (dry basis) on activated carbon) (10 g). The reaction mixture was heated at 40 °C for 12 h, and then filtered through a pad of Celite and the solvent was removed under reduced pressure. To the crude residue was added IM hydrochloric acid (20 mL) and ION sodium hydroxide was added to adjust the pH to 12. The aqueous layer was extracted with ethyl acetate (2 x 80 mL), dried (magnesium sulfate) and solvent removed under reduced pressure to give the title compound as a solid (71.7 g, 95% yield). MS m/z: [M + H+] calcd for C18H2ιN3O 296.17; found 296.0.
Preparation 3
N-1,1 '-Biphenyl-2-yl-N'-4-[l-(9-hydroxynonyl)]piperidinylurea
9-Bromo-l-nonanol (4.84 g, 21.7 mmol) was added to a strrred solution ofthe product of Preparation 2 (5.8 g, 19.7 mmol) and diisopropylethylamine (10.29 mL, 59.1 mmol) in acetonitrile (99 mL) at 50°C. The reaction mixture was heated at 50 °C for 8 h. The reaction mixture was then allowed to cool and the solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (100 mL), washed with saturated aqueous sodium bicarbonate (2 x 50 mL) and dried (magnesium sulfate). The solvent was removed under reduced pressure. The crude product was purified by flash chromatography (dichloromethane:methanol:ammonia system) to yield the title compound (7.1 g, 16.2 mmol, 82% yield).
Preparation 4 iV-l,l'-Biphenyl-2-yl-iV-4-[l-(9-oxononyl)]piperidinylurea
Dimethyl sulfoxide (490 μL, 6.9 mmol), followed by diisopropylethylamine (324 μL, 3.45 mmol) was added to a solution ofthe product of Preparation 3 (500 mg, 1.15 mmol) in dichloromethane (11.5 mL) at -10 °C under an atmosphere of nitrogen. The reaction mixture was stined at -15 °C for 15 min, and then sulfur trioxide pyridine complex was added portionwise (549 mg, 3.45 mmol). The reaction mixture was stirred at -15 °C for 1 h, and then water (10 mL) was added. The organic phase was then separated, washed with water (10 mL), and dried (sodium sulfate). The solvent was removed under reduced pressure to give the title compound (475 mg, 1.09 mmol, 95% yield). HPLC (10- 70) Rt = 3.39.
Preparation 5 N-l,l'-Biphenyl-2-yl-iV-4-[l-(9-aminononyl)]piperidinylurea Palladium (10 wt. % (dry basis) on activated carbon) (1.5 g) was added to a stined solution ofthe product of Preparation 4 (1.58 g, 3.63 mmol) and benzylamine (516 μL, 4.72 mmol) in methanol (36.3 mL). The reaction mixture was placed under an atmosphere of hydrogen. After stirring for 12 h, the reaction mixture was filtered through a pad of Celite and washed with methanol (10 mL). The solvent was removed under reduced pressure to give the title compound (1.50 g, 3.45 mmol, 95% yield). HPLC (10-70) Rt = 2.35; MS m/z [M + H+] calcd for C27H40N4Oι 437.06; found 437.5.
Preparation 6
8-BenzyIoxy-5-(2,2-dihydroxyacetyl)-lH-quinoIin-2-one (a) 8-Acetoxy-lH-quinolin-2-one
8-Hydroxyquinoline-iV-oxide (160.0 g, 1.0 mol), commercially-available from
Aldrich, Milwaukee, WI, and acetic anhydride (800 mL, 8.4 mol) were heated at 100 °C for 3 h and then cooled in ice. The product was collected on a Buchner funnel, washed with acetic anhydride (2xl00mL) and dried under reduced pressure to give 8-acetoxy-lH- quinolin-2-one (144 g) as a solid.
(b) 5-Acetyl-8-hydroxy-lH-quinolin-2-one A sluny of aluminum chloride (85.7 g, 640 mmol) in 1 ,2-dichloroethane (280 mL) was cooled in ice, and the product of step (a) (56.8 g, 280 mmol) was added. The mixture was warmed to room temperature and then heated at 85°C. After 30 min, acetyl chloride (1.5 mL, 21 mmol) was added and the mixture was heated an additional 60 min. The reaction mixture was then cooled and added to IN hydrochloric acid (3 L) at 0 °C with good stirring. After stirring for 2 h, the solids were collected on a Buchner funnel, washed with water (3 x 250 mL) and dried under reduced pressure. The crude product isolated from several batches (135 g) was combined and triturated with dichloromethane (4 L) for 6 h. The product was collected on a Buchner funnel and dried under reduced pressure to give 5-acetyl-8-hydroxy-2(lH)-quinolinone (121 g).
(c) 5-Acetyl-8-benzyloxy-lH-qumolin-2-one
To the product of step (b) (37.7 g, 186 mmol) was added N,N-dimethylformamide (200 mL) and potassium carbonate (34.5 g, 250 mmol) followed by benzyl bromide (31.8 g, 186 mmol). The mixture was stined at room temperature for 2.25 hour and then poured into saturated sodium chloride (3.5 L) at 0 °C and stined for 1 hour. The product was collected and dried on a Buchner funnel for 1 hour, and the resulting solids were dissolved in dichloromethane (2 L) and this mixture was dried over sodium sulfate. The solution was filtered through a pad of Celite which was then washed with dichloromethane (5 x 200 mL). The combined filtrate was then concentrated to dryness and the resulting solids were triturated with ether (500 mL) for 2 h. The product was collected on a Buchner funnel, washed with ether (2 x 250 mL) and dried under reduced pressure to give 5-acetyl-8- benzyloxy-lH-quinolin-2-one (44 g) as a powder.
(d) 8-Benzyloxy-5-(2,2-dihydroxyacetyl)-lH-quinolin-2-one To a sluny ofthe product of step (c) (10.0 g, 34.1 mmol) in DMSO (60 mL) was added a 48% w/w hydrobromic acid solution (11.8 mL, 102.3 mmol). The mixture was warmed to 60 °C for 16 h then allowed to cool to room temperature. Water (100 mL) was added and the resulting slurry stirred at room temperature for 0.5 h before being cooled to 0°C. The product was collected on a Buchner funnel then dried under reduced pressure to give 8-benzyloxy-5-(2,2-dihydroxyacetyl)-lH-quinolin-2-one (12.2 g) as a solid.
Preparation 7 l-(l-{9-[2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5-yI)-2- hydroxyethylamino]nonyl}piperidin-4-yl)-3-biphenyl-2-ylurea
The products of Preparation 5 (183 mg, 0.42 mmol) and Preparation 6 (149 mg, 0.46 mmol) were stined in dichloroethane (4.2 mL) at ambient temperature for 2 h. Sodium triacetoxyborohydride (267 mg, 1.26 mmol) was then added and the reaction mixture was stined for a further 12 h. The reaction mixture was then diluted with dichloromethane (10 mL), washed with saturated aqueous sodium bicarbonate (10 mL), dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude reaction mixture was purified by flash chromatography (5-10% methanol in dichloromethane, 0.5% ammonium hydroxide) to give the title compound (144 mg, 0.20 mmol, 48% yield). HPLC (10-70) Rt = 3.48; MS m/z: [M + H+] calcd for C45H55N5O4 730.4; found 730.7.
Example 1 l-Biphenyl-2-yl-3-(l-{9-[2-hydroxy-2-(8-hydroxy-2-oxo-l,2-dihydroquinolin-5- yl)ethylamino] nonyl} piperidin-4-yl)urea
Palladium (10 wt. % (dry basis) on activated carbon) (63 mg) was added to a stined solution ofthe product of Preparation 7 (144 mg, 0.20 mmol) in methanol (2 mL) and the reaction mixture was placed under an atmosphere of hydrogen. After 12 h stirring, the reaction mixture was filtered through a pad of Celite, washed with methanol (2 mL) and then the solvent was removed under reduced pressure. The resulting residue was purified by preparative HPLC to give the title compound (10 mg). HPLC (10-70) Rt = 2.8; MS m/z: [M + H+] calcd for C38H49N5O4 640.3; found 640.5.
Preparation 8 Biphenyl-2-ylcarbamic Acid Piperidin-4-yl Ester
Biphenyl-2-isocyanate (97.5 g, 521 mmol) and 4-hydroxy-l-benzylpiperidine (105 g, 549 mmol), both commercially-available from Aldrich, Milwaukee, WI, were heated together at 70 °C for 12 h, during which time the formation of biphenyl-2-ylcarbamic acid l-benzylpiperidin-4-yl ester was monitored by LCMS. The reaction mixture was then cooled to 50 °C and ethanol (1 L) was added, and then 6M hydrochloric acid (191 mL) was added slowly. The reaction mixture was then cooled to ambient temperature and ammonium formate (98.5 g, 1.56 mol) was added and nitrogen gas was bubbled through the solution vigorously for 20 min. Palladium (10 wt. % (dry basis) on activated carbon) (20 g) was then added. The reaction mixture was heated at 40 °C for 12 h and then filtered through a pad of Celite. The solvent was then removed under reduced pressure and IM hydrochloric acid (40 mL) was added to the crude residue. Sodium hydroxide (ION) was then added to adjust the pH to 12. The aqueous layer was extracted with ethyl acetate (2 x 150 mL) and dried (magnesium sulfate), and then the solvent was removed under reduced pressure to give the title compound (155 g, 100%). HPLC (10-70) Rt = 2.52; MS m/z: [M + H+] calc'd for C18H2oN2O2297.15; found 297.3.
Preparation 9 ΛyV-(Di-tert-butoxycarbonyl)-9-bromononylamine
A solution of di-tert-butoxycarbonylamine (3.15 g, 14.5 mmol) in N,N- dimethylformamide (0.28 mL) was cooled to 0 °C for about 10 min. Sodium hydride, 60% in mineral oil (0.58 g, 14.5 mmol) was added and the reaction mixture was stirred at 0 °C for 10 min. The reaction mixture was removed from the ice bath and allowed to wann to room temperature for about 30 min. The reaction mixture was then cooled back down to 0 °C and a solution of 1,9-dibromononane (2.46 mL, 12.1 mmol) in dimethylformamide (100 mL) was added. The reaction mixture was stined overnight at room temperature. After 24 h, MS analysis showed that the reaction was completed. The reaction mixture was concentrated to dryness and diluted with ethyl acetate (100 mL). The organic layer was washed with saturated sodium bicarbonate (2 x 100 mL), brine (100 mL), dried
(magnesium sulfate) and concentrated under reduced pressure to yield the crude product, which was purified by chromatography on silica gel using 5% ethyl acetate in hexanes to afford the title compound. MS m/z: [M + H+] calcd for C19H36N!θ4Br 423.18; found 423. Preparation 10
Biphenyl-2-ylcarbamic Acid l-(9-Di-tert-butoxycarbonylamino)nonyl]piperidin-4-yl
Ester
A mixture of 1 : 1 acetonitrile and N,N-dimethylformamide (50 mL) was added to the products of Preparation 8 (3.0 g, 10.1 mmol) and Preparation 9 (5.1 g, 12.2 mmol) and triethylamine (1.42 mL, 10.1 mmol). The reaction mixture was stined at ambient temperature for 24 h and was monitored by LCMS analysis. The reaction mixture was then concentrated and diluted with ethyl acetate (50 mL). The organic layer was washed with saturated sodium bicarbonate (2 x 50 mL) and brine (50 mL). The organic phase was then dried over magnesium sulfate and concentrated to yield 6.5 g of crude oil. The oil was purified by chromatography on silica gel using 1 : 1 hexanes/ethyl acetate to provide the title compound (3 g). MS m/z: [M + H+] calcd for C37H55N3O6 638.41; found 639.
Preparation 11 Biphenyl-2-ylcarbamic Acid l-(9-Aminononyl)piperidin-4-yl Ester
Trifluoroacetic acid (11 mL) was added to a solution ofthe product of Preparation 10 (7.2 g, 11.3 mmol) in dichloromethane (56 mL). After 2 h, LCMS analysis showed that the reaction was completed. The reaction mixture was then concentrated to dryness and diluted with ethyl acetate (75 mL). Sodium hydroxide (IN) was then added until the pH of the mixture reached 14. The organic phase was then collected and washed with saturated sodium bicarbonate (2 x 50 mL) and brine (50 mL). The organic phase was then dried over magnesium sulfate and concentrated to provide the title compound (5.5 g). MS m/z: [M + H+] calcd for C27H39N3O2 438.30; found 439.
Preparation 12
Biphenyl-2-ylcarbamic Acid l-{9-[2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5-yl)-2- hydroxyethylamino] nonyl} piperidin-4-yl Ester
The product of Preparation 11 (196 mg, 0.43 mmol) was dissolved in dichloroethane (4 mL) and sodium triacetoxyborohydride (101 mg, 0.48 mmol) was added. The reaction mixture was stined at ambient temperature for about 10 min. and then 8- benzyloxy-5-(2,2-dihydroxyacetyl)-lH-quinolin-2-one (Preparation 6) (141 mg, 0.43 mmol) was added. LCMS analysis showed that the reaction was completed after 2 h. Methanol (1 mL) was added to the reaction mixture and then sodium borohydride (18 mg, 0.48 mmol) was added slowly. After 1 hour, LCMS analysis showed that the reaction was completed. The reaction mixture was then quenched with aqueous ammonium chloride and this mixture was extracted with dichloromethane. The organic phase was washed with saturated sodium bicarbonate (2 x 50 mL) and brine (10 mL). The organic phase was then dried over magnesium sulfate and concentrated to provide 315 mg of a yellow solid. The solid was purified by silica gel chromatography using 10% methanol in dichloromethane to afford the title compound (64 mg). MS m/z: [M + H+] calcd for C43H55N4O5 730.40; found 731.
Example 2
Biphenyl-2-ylcarbamic Acid l-{9-[2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino] nonyl} piperidin-4-yl Ester Ditrifluoroacetate
A solution ofthe product of Preparation 12 (64 mg, 0.09 mmol) in methanol (450 mL) was flushed with nitrogen. Palladium on carbon (10%, 10 mg) was then added and the reaction mixture was placed under a balloon containing hydrogen and stirred. LCMS analysis showed that the reaction was completed after 9 h. The reaction mixture was then filtered and the filtrate was concentrated to provide a yellow crispy solid. The solid was purified by preparative HPLC (5-35 over 60 min) to afford the title compound (19 mg). MS m/z: [M + H+] calcd for C38H48N4O5 641.36; found 641.
Preparation 13
8-Benzyloxy-5-[(R)-2-bromo-l-(terf-butyldimethylsilanyloxy)ethyl]-lHr-quinolin-2-one (a) 8-Benzyloxy-5-(2-Bromoacetyl)-liϊ-quinolin-2-one 5-Acetyl-8-benzyloxy-lH-quinolin-2-one (Preparation 6) (20.0 g, 68.2 mmol) was dissolved in dichloromethane (200 mL) and cooled to 0°C. Boron trifluoride diethyl etherate (10.4 mL, 82.0 mmol) was added via syringe and the mixture was warmed to room temperature to give a thick suspension. The suspension was heated at 45 °C (oil bath) and a solution of bromine (11.5 g, 72.0 mmol) in dichloromethane (100 mL) was added over 40 min. The mixture was kept at 45 °C for an additional 15 min and then cooled to room temperature. The mixture was concentrated under reduced pressure and then triturated with 10% aqueous sodium carbonate (200 mL) for 1 hour. The solids were collected on a Buchner funnel, washed with water (4 x 100 mL) and dried under reduced pressure. The product of two runs was combined for purification. The crude product (52 g) was triturated with 50% methanol in chloroform (500 mL) for 1 hour. The product was collected on a Buchner funnel and washed with 50% methanol in chloroform (2 x 50 mL) and methanol (2 x 50 mL). The solid was dried under reduced pressure to give the title compound (34.1 g) as a powder.
(b) 8-Benzyloxy-5-((R)-2-bromo-l-hydroxyethyl)-lH-quinoIin-2-one (R)-(+)-α,α-Diphenylprolinol (30.0 g, 117 mmol) and trimethylboroxine (11.1 mL,
78 mmol) were combined in toluene (300 mL) and stined at room temperature for 30 min. The mixture was placed in a 150 °C oil bath and liquid was distilled off. Toluene was added in 20 mL aliquots and distillation was continued for 4 h. A total of 300 mL toluene was added. The mixture was then cooled to room temperature. A 500 μL aliquot was evaporated to dryness and weighed (246 mg) to determine that the concentration of catalyst was 1.8 M.
8-Benzyloxy 5-(2-bromoacetyl)-lH-quinolin-2-one (90.0 g, 243 mmol) was placed under nitrogen and tetrahydrofuran (900 mL) was added followed by the catalyst described above (1.8 M in toluene, 15 mL, 27 mmol). The suspension was cooled to -10±5 °C in an ice/isopropanol bath. Borane (1.0 M in THF, 294 mL, 294 mmol) was added over 4 h. The reaction was then stined an additional 45 min at -10 °C and then methanol (250 mL) was added slowly. The mixture was concentrated under vacuum and the residue was dissolved in boiling acetonitrile (1.3 L), filtered while hot and then cooled to room temperature. The crystals were filtered, washed with acetonitrile and dried under vacuum to give the title compound (72.5 g, 196 mmol, 81% yield, 95% ee, 95% pure by HPLC).
(c) 8-Benzyloxy-5-[(R)-2-bromo-l-(te/t-butyldimethylsilanyloxy)ethyl]-lHr- quinolin-2-one
To the product of step (b) (70.2 g, 189 mmol) was added NN-dimethylformamide (260 mL) and this mixture was cooled in an ice bath under nitrogen. 2,6-Lutidine (40.3 g, 376 mmol) was added over 5 min and then tert-butyldimethylsilyl trifluoromethanesulfonate (99.8 g, 378 mmol) was added slowly while maintaining the temperature below 20°C. The mixture was allowed to wann to room temperature for 45 min. Methanol (45 mL) was added to the mixture dropwise over 10 min and the mixture was partitioned between ethyl acetate/cyclohexane(l:l, 500 mL) and water/brine (1:1, 500mL). The organics were washed twice more with water/brine (1:1, 500 mL each). The combined organics were evaporated under reduced pressure to give a light yellow oil. Two separate portions of cyclohexane (400 mL) were added to the oil and distillation continued until a thick white slurry was formed. Cyclohexane (300 mL) was added to the sluny and the resulting white crystals were filtered, washed with cyclohexane (300 mL) and dried under reduced pressure to give the title compound (75.4 g, 151 mmol, 80% yield, 98.6 % ee).
Preparation 14
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-(8-benzyloxy-2-oxo-l,2-dihydroquinolin-5- yl)-2-(-!ert-butyldimethylsilanyloxy)ethylamino]nonyl}piperidin-4-yI Ester
The product of Preparation 13 (3.9 g, 8.17 mmol) was added to a solution ofthe product of Preparation 11 (5.0 g, 11.4 mmol) in THF (20 mL), followed by sodium bicarbonate (2.0 g, 24.5 mmol) and sodium iodide (1.8 g, 12.2 mmol). The reaction mixture was heated to 80 °C for 72 h. The reaction mixture was then cooled, diluted with dichloromethane (20 mL) and the organic phase was washed with saturated sodium bicarbonate (2 x 50 mL) and brine (50 mL). The organic phase was then dried (magnesium sulfate) and concentrated to give 6.5 g of a crude product. The crude product was purified by chromatography on silica gel eluting with 3% methanol in dichloromethane to provide the title compound (1.4 g, 21% yield).
Preparation 15
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-(8-benzyloxy-2-oxo-l,2-dihydroquinolin-5- yϊ)-2-hy droxy ethylamino] nonyl} piperidin-4-yl Ester
Triethylamine hydrogen fluoride (376 μL, 2.3 mmol) was added to a solution ofthe product of Preparation 14 (1.3 g, 1.5 mmol) in THF (8 mL) and the reaction mixture was stirred at ambient temperature. After 5 h, the reaction was complete as determined by LCMS analysis. The reaction mixture was then quenched with IN NaOH until the pH was 14 and then diluted with ethyl acetate (20 mL) and washed with IN NaOH (20 mL) and brine (20 mL). The organic phase was then separated, dried over magnesium sulfate, and concentrated to yield the title compound (1.1 g). Example 3
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2-dihydro- quinolin-5-yl)ethylamino]nonyl}piperidin-4-yl Ester Ditrifluoroacetate
A solution ofthe product of Preparation 15 (1.1 g, 1.5 mmol) was flushed with nitrogen and palladium on carbon (10%, 110 mg) was added. The reaction mixture was stined under hydrogen at balloon pressure. Analysis by LCMS showed that the reaction was completed after 9 h. The reaction mixture was then filtered and concentrated to yield a yellow solid. The solid was purified by preparative HPLC (5-30 over 60 min) to afford the title compound (510 mg). MS m/z: [M + H+] calcd for C38H48N4O5 641.36; found 641. HPLC method 10-70: 3.207. [α]20 D = -23.6 (c = 1.0 mg/mL, water).
Example 3A
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2-dihydro- quinolin-5-yl)ethylamino]nonyl}piperidin-4-yl Ester Ditrifluoroacetate Alternatively, the title compound was prepared as follows:
(a) 9-Bromononanal
To a 100-mL round-bottomed flask equipped with a magnetic stirrer, addition funnel and temperature controller, under nitrogen, was added 9-bromononanol (8.92 g, 40 mmol) and dichloromethane (30 mL). The resulting mixture was cooled to 5 °C and a solution of sodium bicarbonate (0.47 g, 5.6 mmol) and potassium bromide (0.48 g, 4 mmol) in water (10 mL) was added. 2,2,6,6-Tetramethyl-l-piperidinyloxy free radical (TEMPO) (63 mg, 0.4 mmol) was added and then a 10 to 13% bleach solution (27 mL) was added dropwise through the addition funnel at a rate such that the temperature was maintained at about 8 °C (+/- 2 °C) with an ice cold bath (over about 40 min.). After addition ofthe bleach was complete, the mixture was stined for 30 min. while maintaining the temperature at about 0°C. A solution of sodium bisulfite (1.54 g) in water (10 mL) was added and the resulting mixture was stirred at room temperature for 30 min. The layers of the mixture were then separated, and the milky aqueous layer was extracted with dichloromethane (1 x 20 mL). The combined dichloromethane layers were then washed with water (1 x 30 mL), dried (MgSO4), filtered and concentrated under reduced pressure to afford the title intermediate (8.3 g, 94% yield), which was used without further purification in the next step. (b) 9-Bromo-l,l-dimethoxynonane
To a 100 mL round-bottomed flask was added 9-bromononanal (7.2 g, 32.5 mmol), methanol (30 mL) and trimethylorthoformate (4 mL, 36.5 mmol). A solution of 4 N hydrochloric acid in dioxane (0.2 mL, 0.8 mmol) was added and the resulting mixture was refluxed for 3 h. The reaction mixture was then cooled to room temperature and solid sodium bicarbonate (100 mg, 1.2 mmol) was added. The resulting mixture was concentrated to one-fourth its original volume under reduced pressure and then ethyl acetate (50 mL) was added. The organic layer was washed with water (2 x 40 mL), dried (MgSO ), filtered and concentrated under reduced pressure to afford the title intermediate (8.44 g, (97 % yield)) as a liquid, which as used in the next step without further purification.
(c) Biphenyl-2-ylcarbamic Acid l-(9,9-Dimethoxynonyl)piperidin-4-yl Ester To a 50 mL three-necked, round-bottomed flask was added biphenyl-2-ylcarbamic acid piperidin-4-yl ester (1 g, 3.38 mmol) and acetonitrile (10 mL) to form a sluny. To this slurry was added 9-bromo-l,l-dimethoxynonane (l.lg, 1.3 mmol) and triethylamine (0.57 g, 4.1 mmol) and the resulting mixture was heated at 65 °C for 6 h (the reaction was monitored by HPLC until starting material is < 5 %). The reaction mixture was then cooled to room temperature at which time the mixture formed a thick sluny. Water (5 mL) was added and the mixture was filtered to collect the solid on a coarse fritted glass filer. The solid was washed with pre-mixed solution of acetonitrile (10 mL) and water (5 mL) and then with another pre-mixed solution of acetonitrile (10 mL) and water (2 mL). The resulting solid was air dried to afford the title intermediate (1.37 g, 84 %, purity >96 % by LC, 1 H NMR) as a white solid.
(d) Biphenyl-2-ylcarbamic Acid l-(9-Oxononyϊ)piperidin-4-yl Ester
To a 500 mL round-bottomed flask with a magnetic stiner was added biphenyl-2- ylcarbamic acid l-(9,9-dimethoxynonyl)piperidin-4-yl ester (7.7 g, 15.9 mmol) and then acetonitrile (70 mL) and aqueous IM hydrochloric acid (70 mL). The resulting mixture was stined at room temperature for 1 h and then dichloromethane (200 mL) was added. This mixture was stined for 15 min. and then the layers were separated. The organic layer was dried (MgSO4), filtered and concentrated under reduced pressure to afford the title intermediate (6.8 g), which was used in the next step without further purification.
(e) Biphenyl-2-ylcarbamic Acid l-(9-{Benzyl-[(R)-2-(8-benzyloxy-2-oxo- l,2-dihydroquinolin-5-yl)-2-(ter^-butyldimethylsilanyloxy)ethyl]amino}nonyl)- piperidin-4-yl Ester
To a 300 mL round-bottomed flask was added 5-[(i?)-2-benzylamino-l-(tert- butyldimethylsilanyloxy)ethyl]-8-benzyloxy-lH-quinolin-2-one (5 g, 9.73 mmol), dichloromethane (100 mL) and glacial acetic acid (0.6 mL, 10 mmol). This mixture was cooled to 0 °C using an ice bath and biphenyl-2-ylcarbamic acid l-(9-oxononyl)piperidin- 4-yl ester (4.6 g, 9.73 mmol) was added with stining. This mixture was stined at 0 °C for 30 minutes and then sodium triacetoxyborohydride (6.15 g, 29 mmol) was added in portions over 15 minutes. The reaction mixture was stined at 0° to 10 °C for 2 hours and then aqueous saturated sodium bicarbonate solution (50 mL) was added and this mixture was stined for 15 minutes. The layers were then separated and the organic layer was washed with 5% aqueous sodium chloride solution (50 mL), dried (MgSO4), filtered and concentrated under reduced pressure to afford the title intermediate (8.5 g, 80% purity by HPLC), which was used without further purification.
(f) Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-(tert-Butyldimethylsilanyloxy)-
2-(8-hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl Ester
To a 200 mL round-bottomed flask was added the intermediate from Step E (8.5 g, 9 mmol), ethanol (100 mL) and glacial acetic acid (0.54 mL, 18 mmol) and this mixture was stined until the solid dissolved. The reaction mixture was purged with hydrogen for 5 min. and then 10% palladium on carbon (1.7 g) was added. This mixture was stined at room temperature while hydrogen was slowly bubbling through the reaction mixture until >95% conversion was observed by HPLC (about 8-9 h). The mixture was then filtered through a Celite pad and the solvent was removed under reduced pressure. The residue was purified by silica gel chromatography (15g silica/1 g crude) using 5% MeOH in DCM/0.5% NH4OH (10 x 150 mL), 8% MeOH in DCM/0.5% NH4OH (10 x 150 mL) and 10% MeOH in DCM/0.5% NH4OH (10 x 150 mL). The appropriate fractions were combined and the solvent was removed under reduced pressure while maintaining the temperature <35 °C to give the title intermediate (4.05 g , 97% purity). (g) Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroqumolin-5-yl)ethylamino]nonyl}piperidin-4-yl Ester
To a 200 mL round-bottomed flask was added the intermediate from Step F (4.05 g, 5.36 mmol) and dichloromethane (80 mL) and the resulting mixture was stirred until the solid dissolved. Triethylamine trihydrofluoride (2.6 mL, 16 mmol) was added and stirring was continued under nitrogen for 18 to 20 h. Methanol (20 mL) was added and then saturated aqueous sodium bicarbonate (50 mL) was added slowly and the mixture was stirred for 15 min. The layers were then separated and the organic layer was washed with saturated aqueous sodium chloride solution (20 mL), dried (MgSO4), filtered and concentrated under reduced pressure to afford the title compound (3.5g, 98% purity by HPLC) as a yellow solid.
Example 3B
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-Hydroxy-2-(8-Hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl Ester Naphthalene-1,5- disulfonic Acid Salt
Biρhenyl-2-ylcarbamic acid 1 - {9-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl ester (1.0 g, 1.56 mmol, free base) was dissolved in methanol (10 mL; low water content). A solution of naphthalene- 1,5- disulfonic acid (0.45 g, 1.56 mmol) in methanol (5 mL; low water content) was added and the reaction mixture was stirred at 30 °C for two hours and then at room temperature overnight (18 h). The resulting thick sluny was filtered and the filtrate cake was washed with methanol (5 mL) and then dried to give the title compound (1.16 g, 80% yield) as off- white crystalline solid.
Preparation 16 iV-{2-Benzyloxy-5-[(R)-2-bromo-l-(ter -butyldimethylsilanyloxy)ethyl]phenyl}- formamide
(i?)-2-Bromo-l-(3-formamido-4-benzyloxyphenyl)ethanol (9.9 g, 28 mmol) was dissolved in dimethylformamide (36 mL). Imidazole (2.3 g, 34 mmol) and tert-butyldimethylsilyl chloride (4.7 g, 31 mmol) were added. The solution was stined under nitrogen atmosphere for 72 h. Additional imidazole (0.39 g, 5.7 mmol) and tert-butyldimethylsilyl chloride (0.64 g, 4.3 mmol) were added and the reaction was stirred for an additional 20 h. The reaction mixture was then diluted with a mixture of isopropyl acetate (53 mL) and hexanes (27 mL) and transfened to a separatory funnel. The organic layer was washed twice with a mixture of water (27 mL) and saturated aqueous sodium chloride (27 mL) followed by a final wash with saturated aqueous sodium chloride (27 mL). The organic layer was dried over sodium sulfate. Silica gel (23.6 g) and hexanes (27 mL) were added and the suspension was stirred for 10 min. The solids were removed by filtration and the filtrate concentrated under vacuum. The residue was crystallized from hexanes (45 mL) to afford 8.85 g (19 mmol, 68 %) ofthe title compound as a solid. MS m/z: [M + H+] calcd for C22H30NO3SiBr 464.1; found 464.2. The starting material, (R)-2 -bromo- 1 -(3-formamido-4-benzyloxyphenyl)ethanol, can be prepared as described in U.S. Patent No. 6,268,533 Bl; or R. Hett et al., Organic Process Research and Development, 1998, 2:96-99; or using procedures similar to those described in Hong et al., Tetrahedron Lett, 1994, 35.-6631; or similar to those described in U.S. Patent No. 5,495,054.
Preparation 17
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-(4-Benzyloxy-3-formylaminophenyl)-2-(tert- butyldimethylsilanyloxy)ethylamino] nonyl} piperidin-4-yl Ester
The product of Preparation 16 (500 mg, 1.008 mmol) and sodium iodide (243 mg, 1.62 mmol) were stined in tetrahydrofuran (0.5 mL) for 15 min at ambient temperature. The product of Preparation 11, (564 mg, 1.29 mmol) and sodium bicarbonate (272 mg, 3.24 mmol) were then added and the reaction mixture was heated at 80 °C for 24 h. The reaction mixture was then allowed to cool. Water (2 mL) was then added and the mixture was extracted with dichloromethane (2 x 2 mL). The combined organic extracts were washed with IM hydrochloric acid (2 x 1 mL), dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by flash chromatography (5-10% methanol/dichloromethane) to give the title compound (360 mg, 0.44 mmol, 41% yield). HPLC (10-70) Rt = 4.96; MS m/z: [M + H+] calcd for C49H68N4O5 821.51; found 821.9. Preparation 18
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-(4-Benzyloxy-3-formylaminophenyl)-2- hydroxy ethylamino] nonyl} piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 17 (360 mg, 0.44 mmol) in tetrahydrofuran (2.2 mL) at ambient temperature was added triethylamine trihydrofluoride (108 μL, 0.66 mmol). The reaction mixture was stined for 24 h and then diluted with dichloromethane (5 mL) and washed with IM hydrochloric acid (2 mL) and saturated aqueous sodium bicarbonate (2 mL). The organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude title compound was used directly in the next step without further purification. HPLC ( 10-70) Rt = 4.6; MS m/z: [M + H+] calcd for C43H54N4O5 707.43; found 707.8.
Example 4
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-(3-Formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]nonyl}piperidin-4-yl Ester Ditrifluoroacetate
Palladium (10 wt. % (dry basis) on activated carbon) (124 mg) was added to a stirred solution ofthe product of Preparation 18 (311 mg, 0.44 mmol) in ethanol (4 mL) and the reaction mixture was placed under an atmosphere of hydrogen. After stirring for 12 h, the reaction mixture was filtered through a pad of Celite, washed with methanol (2 mL) and the solvent was removed under reduced pressure. The resulting residue was purified by preparative HPLC to give the title compound (41 mg). HPLC (10-70) Rt = 3.0; MS m/z: [M + H+] calcd for C36H48N4O5617.39; found 617.5.
Example 5 Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2,3,4- tetrahydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl Ester Ditrifluoroacetate
Palladium (10 wt. % (dry basis) on activated carbon) (80 mg) was added to a stined solution ofthe product of Example 3 (80 mg, 0.11 mmol) in ethanol (1.1 mL) and the reaction mixture was placed under an atmosphere of hydrogen. The reaction mixture was stined for 12 h, and then filtered through a pad of Celite, washed with methanol (2 mL) and the solvent removed under reduced pressure. The crude material was resubjected to the above conditions to ensure complete reaction. The resulting residue was purified by preparative HPLC to yield the title compound (6 mg). HPLC (10-70) Rt = 3.23; MS m/z: [M + H+] calcd for C38H50N4O5 643.39; found 643.7.
Preparation 19 3-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic Acid Methyl Ester
Methyl 3-bromopropionate (553 μL, 5.07 mmol) was added to a stined solution of the product of Preparation 8 (1.00 g, 3.38 mmol) and DIPEA (1.76 mL, 10.1 mmol) in acetonitrile (34 mL) at 50 °C and the reaction mixture was heated at 50 °C overnight. The solvent was then removed under reduced pressure and the residue was dissolved in dichloromethane (30 mL). The resulting solution was washed with saturated aqueous sodium bicarbonate solution (10 mL), dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (5-10% MeOH/DCM) to give the title compound (905 mg, 70%).
Preparation 20
3- [4-(Biphenyl-2-ylcarb amoy loxy)piperidin-l -yl] propionic Acid
A stined solution ofthe product of Preparation 19 (902 mg, 2.37 mmol) and lithium hydroxide (171 mg, 7.11 mmol) in 50% THF/H2O (24 mL) was heated at 30 °C overnight, and then acidified with concentrated hydrochloric acid and lyophilized to give the title compound (~ 100% yield, also contains LiCl salts).
Preparation 21
{5-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5-yl)-2-(fert-butyldimethyl- silanyloxy)ethylamino]pentyl}carbamic Acid tert-Butyl Ester The product of Preparation 13 (600 mg, 1.23 mmol) and N-tert-butoxycarbonyl-
1,5-diaminopentane (622 mg, 3.07 mmol) were dissolved in dimethyl sulfoxide (1.23 mL) and heated to 105 °C for 6 h. The reaction mixture was then cooled and diluted with ethyl acetate (10 mL) and washed with saturated aqueous sodium bicarbonate solution (4 mL). The organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (5-10% methanol/dichloromethane) to give the title compound (~100% yield). Preparation 22
5-[(R)-2-(5-Aminopentylamino)-l-(tert-butyldimethylsilanyloxy)ethyl]-8-benzyloxy- lH-quinolin-2-one
A solution ofthe product of Preparation 21 (800 mg, 1.31 mmol) in trifluoroacetic acid/dichloromethane (25%, 12 mL) was stined at ambient temperature for 1 hour. The solvent was then removed under reduced pressure and the crude residue was dissolved in dichloromethane (15 mL) and washed with IN sodium hydroxide (8 mL). The organic phase was separated, dried (magnesium sulfate) and the solvent was removed under reduced pressure to give the title compound (509 mg, 81% yield over 2 steps).
Preparation 23
Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5- yl)-2-(tert-butyldimethylsilanyloxy)ethylamino]pentylcarbamoyl}ethyl)piperidin-4-yl
Ester To the product of Preparation 20 (417 mg, 1.13 mmol) and H ATU (430 mg,
1.13 mmol) was added the product of Preparation 22 (458 mg, 0.90 mmol) in DMF (1.8 mL), followed by DIPEA (204 μL, 1.17 mmol). The reaction mixture was stined at 50 °C for 12 h, and then the solvent was removed under reduced pressure. The crude residue was dissolved in dichloromethane (10 mL). The resulting solution was washed with saturated aqueous sodium bicarbonate solution (4 mL), dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (5-10% methanol/dichloromethane and 0.5% NH4OH) to give the title compound (240 mg, 31% yield).
Preparation 24
Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5- yl)-2-hydroxyethylamino]pentylcarbamoyl}ethyl)piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 23 (240 mg, 0.28 mmol) in dichloromethane (2.8 mL) was added triethylamine trihydrofluoride (91 μL, 0.56 mmol). The reaction mixture was stirred for 10 h, and then diluted with dichloromethane (10 mL). The resulting solution was then washed with saturated aqueous sodium bicarbonate solution (5 mL), and then the organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure to give the title compound (209 mg, 100% yield). Example 6
Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentylcarbamoyl}ethyl)piperidin-4-yl Ester, Ditrifluoroacetate To a stined solution ofthe product of Preparation 24 (209 mg, 0.28 mmol) in ethanol (2.8 mL) was added palladium (10 wt. % (dry basis) on activated carbon) (81 mg) and the reaction mixture was placed under an atmosphere of hydrogen and stirred overnight. The reaction mixture was then filtered and solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound (58 mg). HPLC (10-70) Rt = 2.30; MS m/z: [M + H+] calcd for C37H45N5O6 656.34; found 656.6; [α]20 D = -6.5 (c=1.0mg/mL, water).
Example 6A
Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentylcarbamoyl}ethyl)piperidin-4-yl Ester
Alternatively, the title compound can be prepared as follows: (a) 5-ChloropentanaI
To a 2 L three-necked round-bottomed flask, equipped with a magnetic stirrer, addition funnel and temperature controller under nitrogen, was added 5-chloropentanol (53 g, 0.433 mol) and dichloromethane (300 mL). This mixture was cooled to 5 °C and a solution of sodium bicarbonate (5 g, 0.059 mol) and potassium bromide (5.1 g, 0.043 mol) in water (225 mL) was added. 2,2,6,6-Tetramethyl-l-piperidinyloxy free radical (TEMPO) (63 mg, 0.4 mmol) was added and then a 10 to 13 % bleach solution (275 mL) was added dropwise through the addition funnel at a rate such that the temperature was maintained at about 8 °C (+/- 2 °C) with an ice cold bath (over about 45 min.). After addition ofthe bleach was complete, the mixture was stined for 30 min. while maintaining the temperature at about 5°C. A solution of sodium bisulfite (4 g) in water (30 mL) was added and the resulting mixture was stined at room temperature for 30 min. The layers ofthe mixture were then separated, and the aqueous layer was extracted with dichloromethane (1 x 50 mL). The combined dichloromethane layers were then washed with water (1 x 50 mL), dried (MgSO4), filtered and concentrated under reduced pressure to afford the title compound (53 g). The product was distilled at 65°C/8 ton to afford the title compound (31.16 g) as an orange oil (GC purity was 70 to 80 %). The product was further purified by adding the crude material (4 g) to a mixture of ethanol (920 mL), ethyl acetate (12 mL) and water (4 mL). Sodium bisulfite (4g) was added and the mixture was heated to reflux for 4 h and then cooled to room temperature and stirred for 14 h at room temperature to form a very thick sluny. The solids were filtered on a coarse fritted filter, washed with the solvent mixture (5 mL) and the solids were dried on the filter to afford 8.4 g ofthe bisulfite adduct. This material was then added to MTBE (20 mL) and aqueous 1 N sodium hydroxide (45 mL) was added with vigorous stirring. The resulting biphasic mixture was stined vigorously until all the solids had dissolved (about 15 min) and then the layers were separated. The aqueous layer was extracted with MTBE (20 mL) and the combined MTBE layers were dried (MgSO4), filtered and concentrated to afford 3.46 g ofthe title compound as a colorless liquid (GC purity >90 %).
(b) 5-[(R)-2-[Benzyl-(5-chloropentyl)amino]-l-(tert- butyldimethylsilanyloxy)ethyl]-8-benzyloxy-lH-quinolin-2-one
To a 1 L three-necked round-bottomed flask was added the product of Preparation 28 (48.4 g, 94 mmol), dichloromethane (400 mL) and glacial acetic acid (11.3 mL). This mixture was stined at 0 °C (ice bath) and the product from step (a) (12.5 g, 103.6 mmol) was added and stirring was continued for 15 min. Sodium triacetoxyborohydride (59.8 g, 282 mmol) was then added in portions over a 15 min. period and the resulting mixture was stined at 0 °C to 10 °C for 2 h. Aqueous saturated sodium bicarbonate solution (200 mL) was then added slowly (gas evolution) and stirring was continued for 15 min. The pH of the solution was then adjusted with solid sodium carbonate to a pH of about 9 and the layers were separated. The organic layer was washed with aqueous 5% sodium chloride solution (200 mL), dried (MgSO4), filtered and concentrated under reduced pressure to afford the title compound (53 g).
(c) 5-[(R)-2-[(5-N^V-Diformylaminopentyl)benzylamino]-l-(tert- butyldimethyl-silanyloxy)ethyl]-8-benzyloxy-lH-quinolin-2-one To a stirred solution ofthe product of step (b) (26.5 g, 42.8 mmol) in l-methyl-2- pyrrolidinone (175 mL) was added sodium diformylamide (6.1 g, 64.2 mmol) and sodium iodide (2.13 g, 14.3 mmol). The reaction flask was flushed with nitrogen and then the mixture was heated at 65 °C for 8 h. The mixture was then cooled to room temperature and water (300 mL) and ethyl acetate (100 mL) were added. This mixture was stirred for 10 min. and then the layers were separated. The aqueous layer was extracted with ethyl acetate (150 mL) and the combined organic layers were washed with water (300 mL), aqueous 50 % brine solution (300 mL), water (300 mL), dried (MgSO4) filtered and concentrate to afford the title compound (23.3 g).
(d) 5-[(R)-2-[(5-Aminopentyl)benzylamino]-l-(tert- butyldimethylsilanyloxy)ethyl] -8-benzyloxy-l H-quinolin-2-one
To a stined solution ofthe product from step (c) (10.5 g, 16 mmol) in methanol (75 mL) was added /?-toluenesulfonic acid (7.42 g. 39 mmol). The resulting mixture was heated at 40 °C for 15 h and then concentrated under reduced pressure to about half its volume. Methanol (70 mL) was added and the mixture was heated at 50 °C for 2 h and then concentrated under reduced pressure. Water (100 mL), methanol (50 mL) and MTBE (100 mL) were added and this mixture was stined for 15 min and then the layers were separated. To the aqueous layer was added aqueous 1 N sodium hydroxide (45 mL) and MTBE (100 mL), and this mixture was stined for 15 min. The layers were then separated and the aqueous layer was extracted with MTBE (100 mL). The combined MTBE layers were dried (MgSO ), filtered and concentrated to afford the title compound as a yellow oil (7.3 g). This material contained about 13 % (by HPLC) ofthe conesponding des-tert- butyldimethylsilyl compound.
(e) 3-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic Acid
To a solution ofthe product of Preparation 8 (50 g, 67.6 mmol) in dichloromethane (500 mL) was added acrylic acid (15.05 mL, 100 mmol). The resulting mixture was heated at 50 °C under reflux for 18 h and then the solvent was removed. Methanol (600 mL) was added and this mixture was heated at 75 °C for 2 h and then cooled to room temperature to form a thick slurry. The solid was collected by filtration, washed with methanol (50 mL) and air dried to afford the title compound (61g, >96% purity) as white powder. (f) Biphenyl-2-ylcarbamic Acid l-[2-(5-{Benzyl-[(R)-2-(8-benzyloxy-2-oxo- l,2-dihydroquinolin-5-yl)-2-(tert-butyldimethylsilanyloxy)ethyl]amino}- pentylcarbamoyl)ethyI]piperidin-4-yI Ester
A mixture ofthe product of step (e) (3.68 g, 10 mmol) and N,N-dimethylformamide (50 mL) was heated at 60 °C until the solid completely dissolved and then cooled to room temperature. The product of step (d) (6 g, 10 mmol) and diisopropylethylamine (3.5 mL) was added and the reaction mixture was cooled to 0°C. PyBOP (6.25 g, 12 mmol) was added in one portion and the reaction mixture was stined at 0 °C to room temperature for 2 hours. The reaction mixture was then poured into cold water (500 mL) with stirring and the pH ofthe resulting mixture was adjusted to about 2 using aqueous 1 M hydrochloric acid. This mixture was stined for 15 min and then filtered to collect the solid, which was washed with water (100 mL) and dried to afford the title compound (8.7 g, HPLC purity >95%) as an off- white solid.
(g) Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-Hydroxy-2-(8-hydroxy-2- oxo-l,2-dihydroqu olin-5-yl)ethylamino]pentylcarbamoyl}ethyl)piperidin-4-yl Ester
The product of step (f) can be deprotected using essentially the same procedures as those described in Preparation 24 and Example 6 to afford the title compound.
Preparation 25
2-(iV-BenzyloxycarbonyI-N-methylamino)ethanal
(a) 2-(N-Benzyloxycarbonyl-N-methylamino)ethanol
Benzyl chloroformate (19 g, 111.1 mmol) in THF (20 mL) was added dropwise over 15 min to a stirred solution of 2-(methylamino)ethanol (10 g, 133.3 mmol) in THF (100 mL) and aqueous sodium carbonate (100 mL) at 0°C. The reaction mixture was stined at 0 °C for 12 h and then extracted with EtOAc (2 x 200 mL). The organic layer was washed with aqueous sodium carbonate (200 mL) and dried (potassium carbonate) and solvent was removed under reduced pressure to give the title compound (22.5 g, 97% yield).
(b) 2-(N-Benzyloxycarbonyl-iV-methylamino)ethanal
DMSO (71 mL,l mol) and DIPEA (87.1 mL, 0.5 mol) were added to a stined solution ofthe product of step (a) (20.9 g, 0.1 mol) in dichloromethane (200 mL) at -10°C. The reaction mixture was stined at -10 °C for 15 min and then sulfur trioxide pyridine complex (79.6 g, 0.5 mol) was added and the resulting mixture was stined for 1 hour. The reaction mixture was quenched with addition of IM hydrochloric acid (200 mL). The organic layer was separated and washed with saturated aqueous sodium bicarbonate (100 mL), brine (100 mL), dried (potassium carbonate) and solvent removed under reduced pressure to give the title compound (20.7 g, ~100% yield).
Preparation 26
Biphenyl-2-ylcarbamic Acid l-[2-(methylamino)ethyl]piperidin-4-yl Ester To a stined solution ofthe product of Preparation 25 (20.7 g, 100 mmol) and the product of Preparation 8 (25 g, 84.7 mmol) in MeOH (200 mL) was added sodium triacetoxyborohydride (21.2 g, 100 mmol). The reaction mixture was stined for 12 h at ambient temperature and then it was quenched with 2M hydrochloric acid and the solvent was removed under reduced pressure. The residue was dissolved in ethyl acetate (200 mL) and washed with saturated aqueous sodium bicarbonate solution (100 mL) and brine (50 mL), and then dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (50-90%) EtOAc/hexanes) to give biρhenyl-2-ylcarbamic acid l-[2-(benzyloxycarbonyl- methylamino)ethyl]piperidin-4-yl ester as an oil. The oil was dissolved in methanol (100 mL) and palladium (10 wt. % (dry basis) on activated carbon) (5 g) was added. The reaction mixture was stined under hydrogen (30 psi) for 12 h and then filtered through Celite, which was washed with methanol, and solvent was evaporated to give the title compound (13.2 g, 44%> yield).
Preparation 27
Biphenyl-2-ylcarbamic Acid l-{2-[(6-Bromohexanoyl)methylamino] ethyl} piperidin-4- yl Ester
6-Bromohexanoyl chloride (3.23 mL, 21.1 mmol) was added to a stined solution of the product of Preparation 26 (6.2 g, 17.6 mmol) and DIPEA (6.13 mL, 35.2 mmol) in dichloroethane (170 mL). The reaction mixture was stined for 1 hour and it was then diluted with EtOAc (250 mL) and washed with saturated aqueous sodium bicarbonate solution (2 x 200 mL) and brine (200 mL), and then dried (magnesium sulfate). The solvent was removed under reduced pressure to give the title compound (6.6 g, 73% yield). Preparation 28
8-Benzyloxy-5-[(R)-2-(N-benzylamino)-l-(tert-butyldimethylsilanyloxy)ethyl]-lH- quinolin-2-one
A stined solution ofthe product of Preparation 13 (1.00 g, 2.05 mmol) and benzylamine (493 μL, 4.51 mmol) in DMSO (1.7 mL) was heated at 105 °C for 4 h. The reaction mixture was allowed to cool and was then diluted with EtOAc (10 mL) and the organic layer was washed with saturated aqueous ammonium chloride solution (5 mL) and IN sodium hydroxide (5 mL), dried (MgSO ) and solvent removed under reduced pressure. The crude residue was purified by column chromatography (50% EtOAc/hexanes) to give the title compound (700 mg, 67%). MS m/z: [M + H+] calcd for C3 !H38N2O3Si 515.27; found 515.5.
Preparation 29
Biphenyl-2-ylcarbamic Acid l-{2-[(6-{Benzyl-[(R)-2-(8-benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-(tert-butyldimethylsilanyloxy)ethyl]amino}hexanoyl)- methylamino]ethyl}piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 28 (807 mg, 1.57 mmol) and DIPEA (819 μL, 4.7 mmol) in acetonitrile (3.14 mL) was added the product of Preparation 27 (995 mg, 1.88 mmol). The reaction mixture was heated to 80 °C for 24 h. The solvent was removed under reduced pressure and the residue was dissolved in EtOAc (10 mL) and then washed with saturated aqueous sodium bicarbonate solution (5 mL), dried (magnesium sulfate), and the solvent removed under reduced pressure. The crude material was purified by column chromatography (4-6% MeOH/DCM) to obtain the title compound (452 mg, 30% yield).
Preparation 30
Biphenyl-2-ylcarbamic Acid l-{2-[(6-{Benzyl-[(R)-2-(8-benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-hydroxyethyl]anιino}hexanoyl)methylamino]ethyl}piperidin- 4-yl Ester To a stined solution ofthe product of Preparation 29 (452 mg, 0.47 mmol) in dichloromethane (4.7 mL) was added triethylamine trihydrofluoride (116 μL, 0.71 mmol).
The reaction mixture was stirred for 10 h. and then it was diluted with dichloromethane (10 mL) and washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic phase was then dried (MgSO4) and the solvent was removed under reduced pressure to give the title compound (100%> yield).
Example 7 Biphenyl-2-ylcarbamic Acid l-[2-({6-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethyIamino]hexanoyl}methylamino)ethyl]piperidin-4-yl Ester Ditrifluoroacetate
To a stined solution ofthe product of Preparation 30 (400 mg, 0.47 mmol) in ethanol (4.7 mL) was added palladium (10 wt. % (dry basis) on activated carbon) (160 mg) and the reaction mixture was placed under an atmosphere of hydrogen and stined overnight. The reaction mixture was then filtered and solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound (73 mg). HPLC (10-70) Rt = 2.33; MS m/z: [M + H+] calcd for C38H47N5O6 670.36; found 670. [α]20 D = -9.4 (c = 1.0 mg/mL, water).
Preparation 31
Biphenyl-2-ylcarbamic Acid l-[2-(4-(Aminomethyι)phenylcarbamoyl)- ethyl]piperidin-4-yl Ester
To a stined solution of 4-(N-tert-butoxycarbonylaminomethyl)aniline (756 mg, 3.4 mmol), the product of Preparation 20 (1.5 g, 4.08 mmol) 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 stined at 50 °C overnight and then the solvent was removed under reduced pressure. The resulting residue was dissolved in dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic phase was then dried (magnesium sulfate) 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 h. The solvent was then removed under reduced pressure and the crude residue was dissolved in dichloromethane (30 mL) and washed with IN sodium hydroxide (15 mL). The organic phase was separated, dried (magnesium sulfate) and the solvent was removed under reduced pressure to give the title compound (1.5 g, 94% over 2 steps). Preparation 32
Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-
5-yl)-2-(tert-butyldimethylsilanyloxy)ethylamino]methyl}phenylcarbamoyl)ethyl]- piperidin-4-yl Ester A solution ofthe product of Preparation 31 (489 mg, 1.04 mmol), the product of
Preparation 13 (610 mg, 1.25 mmol), sodium bicarbonate (262 mg, 3.12 mmol) and sodium iodide (203 mg, 1.35 mmol) in THF (0.52 mL) were heated at 80 °C for 12 h. The reaction mixture was diluted with dichloromethane (10 mL) and washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic phase was dried (MgSO4) and the solvent was removed under reduced pressure. The crude residue was purified by flash chromatography (10% MeOH/DCM) to give the title compound as a solid (687 mg, 77% yield).
Preparation 33 Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-(8-BenzyIoxy-2-oxo-l,2-dihydroquinolin- 5-yl)-2-hydroxyethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 32 (687 mg, 0.8 mmol) in dichloromethane (8 mL) was added triethylamine trihydrofluoride (261 μL, 1.6 mmol). The reaction mixture was stirred for 10 h and then was diluted with dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic phase was then dried (magnesium sulfate) and the solvent was removed under reduced pressure to yield the title compound (500 mg, 81% yield).
Example 8 Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl Ester Ditrifluoroacetate
To a stirred solution ofthe product of Preparation 33 (500 mg, 0.65 mmol) in ethanol (6.5 mL) was added palladium (10 wt. % (dry basis) on activated carbon) (200 mg) and the reaction mixture was placed under a hydrogen atmosphere and stined overnight. The reaction mixture was then filtered and the solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound (81 mg, 2 TFA salt). HPLC (10-70) Rt = 2.41; MS m/z: [M + H+] calcd for C39H41N5O6 676.32; found 676.5.
Preparation 34 Biphenyl-2-yIcarbamic Acid l-(2-to*t-Butoxycarbonylaminoethyl)piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 8 (2.00 g, 6.76 mmol) and DIPEA (3.54 mL, 20.3 mmol) in acetonitrile (67.6 mL) at 50 °C was added 2-tert- butoxycarbonylaminoethyl bromide (1.82 g, 8.11 mmol) and the reaction mixture was heated at 50 °C overnight. The solvent was then removed under reduced pressure and the residue was dissolved in dichloromethane (60 mL) and washed with saturated aqueous sodium bicarbonate solution (30 mL). The organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by column chromatography (5 % MeOH/DCM) to yield the title compound as a solid (2.32 g, 78% yield).
Preparation 35
Biphenyl-2-ylcarbamic Acid l-(2-Aminoethyl)piperidin-4-yl Ester
The product of Preparation 34 was dissolved in TFA/DCM (25%, 52 mL) and stined at room temperature for 2 h. The solvent was then removed under reduced pressure and the crude residue dissolved in dichloromethane (30 mL) and washed with IN sodium hydroxide (15 mL). The organic phase was separated, dried (magnesium sulfate) and the solvent was removed under reduced pressure to give the title compound (1.61 g, 90% yield).
Preparation 36
Biphenyl-2-ylcarbamic Acid l-[2-(4-Aminomethylbenzoylamino)ethyl]piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 35 (339 mg, 1 mmol), 4-(tert- butoxycarbonylaminomethyl)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 and then the solvent was removed under reduced pressure. The resulting residue was dissolved in dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude product was dissolved in TFA/DCM (25%, 10 mL) and this mixture was stined at room temperature for 2 h. The solvent was removed under reduced pressure and the crude residue was dissolved in dichloromethane (15 mL) and washed with IN sodium hydroxide (5 mL). The organic phase was separated, dried (magnesium sulfate) and the solvent was removed under reduced pressure to afford the title compound (472 mg, -100% over 2 steps).
Preparation 37 Biphenyl-2-ylcarbamic Acid l-[2-(4-{ [(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin- 5-yl)-2-(tert-butyldimethylsilanyloxy)ethylammo]methyl}benzoylamino)ethyl]- piperidin-4-yl Ester
A solution ofthe product of Preparation 36 (520 mg, 1.1 mmol), the product of Preparation 13 (634 mg, 1.3 mmol), sodium bicarbonate (277 mg, 3.3 mmol) and sodium iodide (215 mg, 1.43 mmol) in THF (0.55 mL) was heated at 80 °C for.12 h. The reaction mixture was then diluted with dichloromethane (10 mL) and washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic phase was then dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by flash chromatography (5-10% MeOH/DCM) to give the title compound as a solid (316 mg, 33% yield).
Preparation 38
Biphenyl-2-ylcarbamic Acid l-[2-(4-{ [(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-
5-yl)-2-hydroxyethylamino]methyl}benzoylamino)ethyl]piperidin-4-yl Ester To a stined solution ofthe product of Preparation 37 (316 mg, 0.36 mmol) in dichloromethane (3.6 mL) was added triethylamine trihydrofluoride (117 μL, 0.72 mmol). The reaction mixture was stined for 10 h and was then diluted with dichloromethane (10 mL) and washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic phase was dried (MgSO4) and the solvent was removed under reduced pressure to give the title compound, which was used directly in the next step (100 % yield). Example 9
Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroqumoIm-5-yl)ethyIammo]methyl}benzoylanιino)ethyl]piperidin-4-yl Ester Ditrifluoroacetate To a stirred solution ofthe product of Preparation 38 (275 mg, 0.36 mmol) in ethanol (3.6 mL) was added palladium (10 wt. % (dry basis) on activated carbon) (275 mg) and the reaction mixture was placed under a hydrogen atmosphere and stined overnight. The reaction mixture was then filtered and the solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to yield the title compound (6 mg, 2 TFA salt). HPLC ( 10-70) Rt = 2.26; MS m/z: [M + H+] calcd for C39H41N5O6 676.32; found 676.5.
Preparation 39
Biphenyl-2-ylcarbamic Acid l-(2-Aminoethyl)piperidin-4-yl Ester 2-tert-Butoxycarbonylaminoethyl bromide (1.22 g, 5.44 mmol) was added to a solution of the product of Preparation 8 (1.46 g, 4.95 mmol) and diisopropylethylamine (1.03 mL, 5.94 mmol) in acetonitrile (24 mL). The reaction mixture was stirred at 65 °C for 12 hours, at which time MS analysis showed that the reaction was completed. The reaction mixture was concentrated to dryness and then dichloromethane (10 mL) was added. Trifluoroacetic acid was added to this mixture and the mixture was stirred at room temperature for 4 hours, at which time MS analysis showed that the reaction was complete. The mixture was then concentrated to half its volume and IN sodium hydroxide was added to the solution until the pH was adjusted to 14. The organic layer was washed with brine, then dried over magnesium sulfate and filtered. The filtrate was concentrated to give 1.6 g ofthe title compound as a solid. MS m/z: [M + H+] calcd for C20H25N3O2 340.2; found 340.
Preparation 40
5-[(R)-2-(5-Aminopentylamino)-l-(tert-butyldimethylsilanyloxy)ethyl]-8-benzyloxy- lH-quinolin-2-one
N-tert-butoxycarbonyl-l,5-diaminopentane (1.04 g, 5.12 mmol) was added to a solution ofthe product of Preparation 13 (1.00 g, 2.05 mmol) in dimethyl sulfoxide (2 mL). The solution was stined at 75 °C for 12 hours, at which time LCMS analysis showed that the reaction was complete. The reaction mixture was then concentrated under vacuum to dryness. To the residue was added dichloromethane (2 mL) and trifluoroacetic acid (1 mL) was then added. The solution was stined at room temperature for about 3 hours, at which time MS analysis showed that the reaction was complete. The solution was concentrated to half its volume and IN sodium hydroxide was added until the pH was adjusted to 14. The organic layer was collected, washed with brine, dried over magnesium sulfate and then concentrated to yield 782 mg ofthe title compound as an oil. MS m/z: [M + H+] calcd for C29H43N3O3Si 510.8; found 510.
Preparation 41
Biphenyl-2-ylcarbamic Acid l-[2-(3-{5-[(R)-2-(8-Benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-(tert-butyldimethylsiIanyloxy)ethylamino]pentyl}- ureido)ethyl]piperidin-4-yl Ester
Carbonyl diimidazole (127 mg, 0.78 mmol) was added to a solution ofthe product of Preparation 39 (266 mg, 0.78 mmol) in dimethyl formamide (4 mL) and the resulting mixture was stirred at room temperature for 3 hours. After 3 hours, the product of Preparation 40 (399 mg, 0.78 mmol) was added to the reaction mixture and this mixture was stined for 12 hours at room temperature, at which time LCMS analysis determined that the reaction was complete. The reaction mixture was concentrated in vacuo and the residue was diluted with ethyl acetate (5 mL). The organic layer was washed two times with saturated sodium bicarbonate (5 mL) and then brine (5 mL). The organic layer was dried over magnesium sulfate, filtered and then concentrated to afford 597 mg ofthe title compound as a solid which was used without further purification. MS m/z: [M + H+] calcd for C50H66N6O6Si 875.5; found 875.
Preparation 42
Biphenyl-2-ylcarbamic Acid l-[2-(3-{5-[(R)-2-(8-Benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-hydroxyethylamino] pentyl} ureido)ethyl] piperidin-4-yl Ester Triethylamine trihydrofluoride (0.16 mL, 1.02 mmol) was added to a solution of the product of Preparation 41 (597 mg, 0.68 mmol) in tetrahydrofuran (3.4 mL) and this mixture was stined at room temperature for about 12 hours, at which time the reaction was determined to be completed by MS analysis. The reaction mixture was diluted with ethyl acetate (5 mL) and this mixture was washed with IN sodium hydroxide (5 mL), brine, dried over magnesium sulfate and concentrated to give 417 mg ofthe title compound as a solid. MS m/z: [M + H+] calcd for C44H5ιN6O6 760.4; found 760.
Example 10 Biphenyl-2-ylcarbamic Acid l-[2-(3-{5-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentyI}ureido)ethyl]piperidin-4-yl Ester Ditrifluoroacetate
A solution ofthe product of Preparation 42 (417 mg, 0.55 mmol) in ethanol (3 mL) was purged with nitrogen for about 10 minutes. Palladium (10 wt. % (dry basis) on activated carbon) (200 mg) was added and the solution was flushed again with nitrogen for about 10 minutes. The flask was purged under vacuum and then filled with nitrogen three times and then a hydrogen-filled balloon was placed over the flask. The reaction mixture was stirred under hydrogen for 12 hours, at which time the reaction was determined to be complete by MS analysis. The reaction mixture was then filtered and the organic filtrate concentrated and purified by HPLC (10-35 % over 60 minute) to give 146 mg ofthe title compound as a powder. MS m/z: [M + H+] calcd for C37H 6N6O6 671.4; found 670. HPLC (10-70) Rt = 2.6 minutes.
Example 11 Biphenyl-2-ylcarbamic Acid l-[3-(3-{5-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentyl}ureido)propyl]piperidin-4-yl Ester Ditrifluoroacetate
Using the method described above in Preparations 39-42 and Example 10, and substituting 3-tert-Butoxycarbonylaminoprop-l-yl bromide for 2-tert- butoxycarbonylaminoethyl bromide in Preparation 39, the title compound was prepared. MS m/z: [M + H+] calcd for C38H48N6O6 685.4; found 684. HPLC (10 70) Rt = 2.6 minutes.
Preparation 43 6-(2-Bromo-(R)-l-te/*j'-butyldimethylsilyloxy)ethyl-2,2-dimethyl-l,3-benzodioxan (a) 6-Bromo-2,2-dimethyl-4H-benzo[l,3]dioxine
To 5-bromo-2-hydroxybenzyl alcohol (93 g, 0.46 mol, available from Sigma- Aldrich) in 2.0 L of 2,2-dimethoxypropane was added 700 mL of acetone, followed by zinc chloride (170 g). After stirring for 18 hours, 1.0 M aqueous sodium hydroxide was added until the aqueous phase was basic. Diethyl ether (1.5 L) was added to the slurry and the organic phase was decanted into a separatory funnel. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give the title compound as an oil.
(b) 6-Acetyl-2,2-dimethyl-4H-benzo [1 ,3] dioxine
To the product of step (a) (110 g, 0.46 mol) in 1.0 L of THF at -78 °C was added 236 mL (0.51 mol) of 2.14 M n-butyllithium in hexanes via a dropping funnel. After 30 minutes, N-methyl-N-methoxy acetamide (71 g, 0.69 mol, available from TCI) was added. After 2 hours, the reaction mixture was quenched with water, diluted with 2.0 L of 1.0 M aqueous phosphate buffer (pH = 7.0) and extracted once with diethyl ether. The diethyl ether phase was washed once with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give a light orange oil. The oil was dissolved in a minimum volume of ethyl acetate, diluted with hexanes, and to give the title compound as a crystalline solid.
(c) 6-Bromoacetyl-2,2-dimethyl-4H-benzo[l,3]dioxine
To the product of step (b) (23.4 g, 0.113 mol) in 600 mL of THF at -78 °C was added 135 mL of 1.0 M sodium hexamethyldisilazane in THF (Sigma- Aldrich). After 1 hour, trimethylsilyl chloride (15.8 mL, 0.124 mol) was added. After another 30 minutes, bromine (5.82 mL, 0.113 mol) was added. After 10 minutes, the reaction was quenched by diluting the reaction mixture with diethyl ether and pouring it onto 500 mL of 5% aqueous Na2SO3 premixed with 500 mL of 5% aqueous NaHCO3. The phases were separated and the organic phase was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give the title compound as an oil that solidified upon storage in the freezer.
(d) (R)-2-Bromo-l-(2,2-dimethyl-4H-benzo[l,3]dioxin-6-yI)ethanol To the product of step (c) (10 g, 35.1 mmol) in 100 mL of THF was added the solid catalyst of Preparation 13, step (c)(1) (0.97 g, 3.5 mmol). The solution was cooled to between -20 °C and -10 °C and BH3-THF (35 mL, 35 mmol) diluted with 50 mL THF was added dropwise via a dropping funnel. After the addition was complete, the reaction mixture was allowed to warm to ambient temperature. After 30 minutes, the reaction mixture was quenched by slow addition of 50 mL of methanol and then concentrated to a thick oil. The oil was purified by silica gel chromatography eluted with 1 :2 ethyl acetate/hexanes. The fractions were combined and concentrated to give the title compound as an off-white solid.
(e) [(R)-2-Bromo-l-(2,2-dimethyl-4H-benzo[l,3]dioxin-6-yl)ethoxy]-tert- butyldimethylsilane
To the product of step (d) (10 g, 34.8 mmol) and imidazole (4.7 g, 69.7 mmol) dissolved in 100 mL DMF was added tert-butyldimethylsilyl chloride (5.78 g, 38.3 mmol). The reaction mixture was stined for 18 hours. The reaction mixture was then partitioned between 200 mL of saturated sodium chloride and 200 mL of diethyl ether. The aqueous layer was extracted with 200 mL of diethyl ether. The organic layers were then combined, washed with saturated sodium chloride (3 x 100 mL), dried over MgSO and concentrated. The product was purified by silica gel chromatography, eluting with hexanes followed by 5% ethyl acetate in hexanes. The desired fractions were combined and concentrated to give the title compound as an oil.
Preparation 44 Biphenyl-2-ylcarbamic Acid l-{9-[2-(ter^-Butyldimethylsilanyloxy)-2-(2,2-dimethyl- 4H-benzo[l,3]dioxin-6-yl)ethylamino]nonyl}piperidin-4-yl Ester
The product of Preparation 43 (802 mg, 2.00 mmol) and sodium iodide (300 mg, 2.00 mmol) were stined in tetrahydrofuran (0.77 mL) for 15 min at ambient temperature. The product of Preparation 11 (675 mg, 1.54 mmol) and sodium bicarbonate (388 mg, 4.62 mmol) were added and the reaction mixture was heated at 80 °C for 24 h. The reaction mixture was then cooled and water (2 mL) was added. The mixture was then extracted with dichloromethane (2 x 2 mL). The combined organic extracts were dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude residue was purified by flash chromatography (5-10% methanol/dichloromethane) to give the title compound as a solid (798 mg, 1.05 mmol, 60% yield). MS m/z: [M + H+] calcd for C45H67N3O5Si 758.5; found 758.6. Preparation 45
Biphenyl-2-ylcarbamic Acid l-{9-[2-(2,2-Dimethyl-4H-benzo [1,3] dioxin-6-yl)-2- hydroxyethylamino] nonyl} piperidin-4-yl Ester
Triethylamine trihydrofluoride (342 μL, 2.10 mmol) was added to a stirred solution ofthe Product of Preparation 44 (798 mg, 1.05 mmol) in dichloromethane (10.5 mL) at ambient temperature. The reaction mixture was stined for 24 h and it was then diluted with dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate (15 mL). The organic layer was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude title compound was isolated as an oil (659 mg, 1.02 mmol), which was used in the next step without further purification. MS m/z: [M + H+] calcd for C39H53N3O5 644.4; found 644.8.
Example 12
Biphenyl-2-ylcarbamic Acid l-{9-[(R)-2-Hydroxy-2-(4-hydroxy-3- hydroxymethylphenyl)ethylamino]nonyl}piperidin-4-yl Ester Ditrifluoroacetate
Trifluoroacetic acid (2.80 mL) was added to a stined solution ofthe product of Preparation 45 (600 mg, 0.93 mmol) in THF/H2O (14 mL, 1:1) and the reaction mixture was stirred for 2 h at ambient temperature. The reaction mixture was concentrated under reduced pressure and dissolved in 20%> MeCN/H2O then purified by preparative HPLC to yield the title compound (200 mg, 2TFA salt). HPLC (10-70) R = 2.76; MS m/z: [M + H+]calcd for C36H49N3O5 604.4; found 604.8.
Preparation 46 l-[l-(9-Benzylaminononyl)piperidin-4-yl]-3-biphenyl-2-ylurea N-Benzylamine (0.903 ml, 8.30 mmol) was added to a solution ofthe product of
Preparation 4 (2.40 g, 5.52 mmol) in methanol (25 mL) and the resulting mixture was stined at ambient temperature. After 10 min, sodium triacetoxyborohydride (1.75 g, 8.30 mmol) was added to the reaction mixture. The progress ofthe reaction was followed by HPLC analysis. After 2 h at ambient temperature, the reaction was quenched with water (5 mL) and then concentrated to half its volume under vacuum. The reaction mixture was diluted with dichloromethane (15 mL) and washed with IN sodium hydroxide (2 x 10 mL) and then brine (5 mL). The organic layer was dried over magnesium sulfate and concentrated to yield the title compound. Preparation 47
2-Benzyloxy-5-(2-bromoacetyl)benzoic Acid Methyl Ester
(a) 2-Benzyloxy-5-acetylbenzoic Acid Methyl Ester Methyl 5-acetylsalicylate (100 g, 0.515 mol) was dissolved in acetonitrile (1 L) in a
2 L flask under reflux conditions and a nitrogen atmosphere. Potassium carbonate (213.5 g, 1.545 mol) was added portion-wise over 15 min. Benzyl bromide (67.4 mL, 0.566 mol) was added using a dropping funnel over 15 min. The reaction was heated to 85 °C for 9 h, and then filtered and rinsed with acetonitrile (100 mL). The solution was concentrated to about 300 mL volume under reduced pressure and partitioned between water (1 L) and ethyl acetate (1 L). The organic layer was washed with saturated sodium chloride (250 mL), dried using magnesium sulfate (75 g), and then filtered and rinsed with ethyl acetate (100 mL). The organic layer was concentrated to give 2-benzyloxy-5-acetylbenzoic acid methyl ester as a solid (100% yield).
(b) 2-Benzyloxy-5-(2-bromoacetyl)benzoic Acid Methyl Ester
The product of step (a) (10.0 g, 35.2 mmol) was dissolved in chloroform (250 mL) in a 500 mL flask under a nitrogen atmosphere. Bromine (1.63 mL, 31.7 mmol) dissolved in chloroform (50 mL) was added using a dropping funnel over 30 min. The reaction mixture was stined for 2.5 h and then concentrated to give a solid. The solid was dissolved in toluene (150 mL) with some gentle heat, followed by the addition of ethyl ether (150 mL) to yield the title compound as a crystalline solid (55% yield).
Preparation 48 5-[2-(Benzyl-{9-[4-(3-biphenyl-2-ylureido)piperidin-l-yl]nonyl}amino)acetyl]-2- benzyloxybenzoic Acid Methyl Ester
The product of Preparation 47 (371 mg, 1.00 mmol) was added to a solution ofthe product of Preparation 46 (448 mg, 0.85 mmol) in dimethyl sulfoxide (4.5 mL) followed by the addition of potassium carbonate (234 mg, 1.7 mmol). The reaction mixture was stined at 40 °C for 6 h, at which time the product of Preparation 46 was no longer observed by HPLC analysis. The reaction mixture was cooled to ambient temperature and filtered, and then diluted with ethanol (4 mL). Sodium borohydride (63 mg, 1.7 mmol) was added to the reaction mixture and the reaction was stined at ambient temperature for 24 h. The reaction mixture was quenched with 0.5 M ammonium chloride (5 mL) and extracted into ethyl acetate (2 x 10 mL). The combined organic layers were washed with saturated sodium bicarbonate (10 mL) and then with brine (5 mL). The organic layer was dried over magnesium sulfate and the solvent was removed under reduced pressure. The crude residue was purified by chromatography on silica gel (3% methanol in chloroform) to give the title compound.
Preparation 49 l-[l-(9-{Benzyl-[2-(4-benzyloxy-3-hydroxymethylphenyl)-2- hydroxyethyI]amino}nonyl)piperidin-4-yl]-3-biphenyl-2-ylurea
A solution ofthe product of Preparation 48 (163mg, 0.20 mmol) in tetrahydrofuran (1.00 mL) was cooled to 0°C. Lithium aluminium hydride (1.0 M in THF; 0.50 mL, 0.50 mmol) was added dropwise to the mixture. After 1 h, the reaction mixture was quenched with water (1 mL) and diluted with ethyl acetate (2 mL). The organic layer was washed with brine, dried over magnesium sulfate, and the organic extracts were combined and concentrated to give the title compound.
Example 13 l-Biphenyl-2-yl-3-(l-{9-[2-hydroxy-2-(4-hydroxy-3- hydroxymethylphenyl)ethylamino]nonyI}piperidin-4-yI)urea Dihydrochloride
A solution ofthe product of Preparation 49 (130 mg, 0.16 mmol) in isopropanol (0.80 ml) was flushed with nitrogen for ten minutes and then palladium (10 wt. % (dry basis) on activated carbon (60 mg) was added. The reaction flask was purged with nitrogen and then a balloon filled with hydrogen was attached to the flask and the reaction mixture was stined under an atmosphere of hydrogen. After 72 h, the reaction mixture was filtered and concentrated and the residue was purified by preparative HPLC. The resulting ditrifluoroacetic acid salt ofthe title compound was dissolved in IN hydrochloric acid (5 mL) and lyophilized to yield the title compound as its dihydrochloride salt. Preparation 50
5-[(R)-2-[(3-Aminomethylcyclohexylmethyl)amino]-l-(t'ert- butyldimethylsilanyloxy)ethyl] -8-benzyloxy-l H-quinolin-2-one
A stined solution ofthe product of Preparation 13 (1.46 g, 3 mmol) and 1,3- cyclohexanebis(methylamine) (426 mg, 3 mmol) in DMSO (3 mL) was heated at 100 °C for 6 h. The reaction mixture was allowed to cool and it was then diluted with dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic layer was dried (MgSO ) and the solvent was removed under reduced pressure. The crude residue was purified by flash chromatography (10% MeOH/DCM and 0.5% NH4OH) to give the title compound as a solid (775 mg, 50% yield). MS m/z: [M + H+] calcd for C32H47N3O3Si 550.3; found 550.6.
Preparation 51
Biphenyl-2-ylcarbamic Acid l-{2-[(3-{ [(R)-2-(8-BenzyIoxy-2-oxo-l,2-dihydro- quinolin-5-yl)-2-(tert-butyldimethylsilanyIoxy)ethylamino] methyl}- cyclohexylmethyϊ)carbamoyl] ethyl} piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 50 (552 mg, 1.01 mmol), the product of Preparation 20 (309 mg, 0.84 mmol) and HATU (384 mg, 1.01 mmol) in DMF (1.68 mL) was added DIPEA (190 μL, 1.09 mmol). The reaction mixture was stined at 50 °C overnight and then the solvent was removed under reduced pressure. The resulting residue was dissolved in dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude product was purified by flash chromatography (5-10% MeOH/DCM) to give the title compound as a solid (267 mg, 36% yield). LCMS (10-70) Rt= 5.04. MS m/z: [M + H+] calcd for C53H69N5O6Si 900.5; found 900.6.
Preparation 52
Biphenyl-2-ylcarbamic Acid l-{2-[(3-{ [(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin- 5-yl)-2-hydroxyethylamino]methyl}cyclohexylmethyl)carbamoyl]ethyl}piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 51 (267 mg, 0.30 mmol) in dichloromethane (3 mL) was added triethylamine trihydrofluoride (98 μL, 0.6 mmol). The reaction mixture was stined for 10 h and then it was diluted with dichloromethane (10 mL) and washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic phase was dried (magensium sulfate) and the solvent was removed under reduced pressure to give the title compound as a solid (236 mg, 100%) yield). MS m/z: [M + H+] calcd for C47H55N5O6786.4; found 786.5.
Example 14
Biphenyl-2-ylcarbamic Acid l-{2-[(3-{ [(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexylmethyl)carbamoyl]ethyl}- piperidin-4-yl Ester
Palladium (10 wt. %> (dry basis) on activated carbon) (120 mg) was added to a stined solution ofthe product of Preparation 52 (236 mg, 0.30 mmol) in ethanol (3 mL). The reaction mixture was placed under a hydrogen atmosphere and stined overnight. The reaction mixture was then filtered and the solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound (27 mg, 2 TFA salt). HPLC (10-70) Rt = 2.76. MS m/z: [M + H+] calcd for C40H49N5O6 696.4; found 696.6.
Preparation 53 Biphenyl-2-ylcarbamic Acid l-{2-[((lR,3S)-3-Aminocyclopentanecarbonyl)amino]- ethyl}piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 39 (318 mg, 0.94 mmol), (lR,3>S)-3-tert-butoxycarbonylaminocyclopentanecarboxylic acid (258 mg, 1.1 mmol) and HATU (428 mg, 1.1 mmol) in DMF (5 mL) was added DIPEA (245 μL, 1.09 mmol). The reaction mixture was stined at room temperature overnight and then the solvent was removed under reduced pressure. The resulting residue was dissolved in dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic layer was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude product was purified by flash chromatography (5-10% MeOH/DCM) and then dissolved in a trifluoroacetic acid/DCM mixture (1 mL/5 mL) and stirred at room temperature for lh. The solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (20 mL) and washed with IM sodium hydroxide (10 mL), dried (magnesium sulfate) and the solvent reduced to yield the title compound (167 mg, 39% yield).
Preparation 54 Biphenyl-2-ylcarbamic Acid l-[2-({(lR,3S)-3-[(R)-2-(8-benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-(tert-butyldimethyIsilanyloxy)ethylamino]- cyclopentanecarbonyl} amino)ethyl] piperidin-4-yl Ester
A stirred solution ofthe product of Preparation 53 (167 mg, 0.38 mmol) and the product of Preparation 13 (92 mg, 0.19 mmol) in DMSO (0.38 mL) was heated at 90 °C for 5 h. The solution was cooled and diluted with ethyl acetate (10 mL) and then washed with saturated aqueous sodium bicarbonate (5 mL). The organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure. The crude product was purified by flash chromatography (5-10% MeOH/DCM) to yield the title compound (343 mg, 100% yield). LCMS (10-70) Rt = 4.97. MS m/z: [M + H+] calcd for C50H63N5O6Si 858.5; found 858.8.
Preparation 55
Biphenyl-2-ylcarbamic Acid l-[2-({(lR,3S)-3-[(R)-2-(8-benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-hydroxyethylamino] cyclopentanecarbonyl} amino)ethyl] - piperidin-4-yl Ester
To a stirred solution ofthe product of Preparation 54 (343 mg, 0.4 mmol) in THF (2 mL) was added triethylamine trihydrofluoride (130 μL, 0.8 mmol). The reaction mixture was stined for 10 h and was then diluted with EtOAc (10 mL). The reaction mixture was washed with saturated aqueous sodium bicarbonate solution (5 mL) and then the organic phase was dried (magnesium sulfate) and the solvent was removed under reduced pressure to give the title compound as a solid (298 mg, 100%) yield). HPLC (10- 70) Rt = 2.8. MS m/z: [M + H+] calcd for C44H49N5O6744.4 ; found 744.4. Example 15
Biphenyl-2-ylcarbamic Acid l-[2-({(lR,3S)-3-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo- l,2,3,4-tetrahydroqumolm-5-yl)ethylan ino]-cyclopentanecarbonyl}amino)ethyl]- piperidin-4-yl Ester Ditrifluoroacetate To a stirred solution ofthe product of Preparation 55 (236 mg, 0.40 mmol) in ethanol (3 mL) was added palladium (10 wt. % (dry basis) on activated carbon (120 mg). The reaction mixture was placed under a hydrogen atmosphere and stined overnight. The reaction mixture was filtered and the solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound (3 mg, 2 TFA salt). HPLC (5-75) Rt = 2.18. MS m/z: [M + H+] calcd for C37H45N5O6 656.3; found 656.2.
Preparation 56 4-(tert-Butoxycarbonylaminomethyl)-2-chlorophenylamine
A stined solution of 4-aminomethyl-2-chlorophenylamine (940 mg, 6 mmol) and di-tert-butyl dicarbonate (1.44 g, 6.6 mmol) in dichloromethane (30 mL) was stined at room temperature for 4 h, at which time the reaction was determined to be complete by LCMS. The reaction mixture was then washed with saturated aqueous sodium bicarbonate (15 mL) and the organic layer was dried over sodium sulfate and the solvent was removed under reduced pressure. The resulting orange solid was recrystallized from ethyl acetate to give the title intermediate as a white solid (~100% yield).
Preparation 57
N- [4-(tørt-Butoxy carbonylaminomethyl)-2-chlorophenyl] acrylamide
To a stined solution ofthe product of Preparation 56 (1.54 g, 6.0 mmol) in a mixture of diethyl ether (35 mL) and 1 M sodium hydroxide (35 mL) was added dropwise acryloyl chloride (687 μL, 8.45 mmol). After 1 h, the organic layer was separated, dried (Na2SO4) and the solvent was removed under reduced pressure to give the title intermediate as a white solid (1.8 g, 96% yield). Preparation 58
Biphenyl-2-ylcarbamic Acid l-[2-(4-(tert-Butoxycarbonylaminomethyl)-2- chlorophenylcarbamoyl)ethyl]piperidin-4-yl Ester
A solution ofthe product of Preparation 8 (1.04 g, 3.5 mmol) and the product of Preparation 57 (1.19 g, 3.85 mmol) in a mixture of dichloromethane and methanol (12 mL, 1 :1) was heated at 60 °C for 12 h. The reaction mixture was allowed to cool and the solvent was removed under reduced pressure. The crude material was purified by column chromatography (5-10% MeOH/DCM) to give the title intermediate as a white solid (2.00 g, 94% yield).
Preparation 59
Biphenyl-2-ylcarbamic Acid l-[2-(4-Aminomethyl-2-chlorophenylcarbamoyl)ethyl]- piperidin-4-yl Ester
A solution ofthe product of Preparation 58 (2.00 g, 3.3 mmol) was stined in dichloromethane (24 mL) and TFA (8 mL) for 1 h and then the solvent was removed under reduced pressure. The crude reaction mixture was dissolved in dichloromethane (30 mL) and washed with 1 M sodium hydroxide (2 x 30 mL). The organic layer was dried (Na2SO ) and the solvent was removed under reduced pressure to give the title intennediate as an oily white solid (1.46 g, 88% yield).
Preparation 60
Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-
5-yl)-2-(te;^-butyldimethylsilanyloxy)ethylamino]methyl}-2-chlorophenylcarbamoyl)- ethyl]piperidin-4-yl Ester A stined solution ofthe product of Preparation 59 (1.41 g, 2.79 mmol) and the product of Preparation 13 (680 mg, 1.39 mmol) in DMSO (1.39 mL) was heated at 90 °C for 8 h and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate/chloroform (20 mL, 1/1) and the organic layer was washed with saturated aqueous sodium bicarbonate (10 mL), dried (Na2SO4) and the solvent removed under reduced pressure. The resulting crude residue was purified by column chromatography (5-10% MeOH/DCM) to give the title intermediate as a white solid (1.12 g, 88% yield). MS m/z M+H+ = 914.9. Preparation 61
Biphenyl-2-yl-Carbamic Acid l-[2-(4-{ [(R)-2-(8-Benzyloxy-2-oxo-l,2- dihydroquinolin-5-yl)-2-hydroxyethyIamino]methyl}-2-chloro- phenylcarbamoyl)ethyl]piperidin-4-yl Ester To a stirred solution ofthe product of Preparation 60 (1.12 g, 1.23 mmol) in dichloromethane (12 mL) was added Et3N.3HF (401 μL, 0.6 mmol). The reaction mixture was stined for 10 h and then diluted with dichloromethane (10 mL). This mixture was washed with saturated aqueous sodium bicarbonate solution (5 mL) and the organic layer dried (Na2SO4) and the solvent removed under reduced pressure to give the title intennediate as a white solid (959 mg, 100% yield). MS m/z M+H+ =800.5.
Example 16
Biphenyl-2-ylcarbamic Acid l-[2-(2-Chloro-4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl Ester Ditrifluoroacetate
To a stirred solution ofthe product of Preparation 61 (959 mg, 1.2 mmol) in ethanol (12 mL) was added Pd/C (290 mg) and the reaction mixture was placed under a hydrogen atmosphere and stined overnight. The reaction mixture was then filtered and the solvent removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound (67 mg, 2 TFA salt). HPLC (10-70) Rt = 2.76; MS m/z M+H+= 710.6.
Preparation 62
2-Chloroethanesulfonic Acid (5-tert-Butoxycarbonylaminopentyl)amide To a stined solution of 5-(tert-butoxycarbonylamino)pentylamine (1.00 g, 4.94 mmol) and triethylamine (689 μL g, 4.94 mmol) in dichloromethane (22 mL) at 0 °C was added 2-chloro-l -ethanesulfonyl chloride (470 μL, 4.50 mmol). The reaction mixture was stirred for 2 h at room temperature and then washed with saturated aqueous sodium bicarbonate solution (15 mL). The organic layer was dried (Na2SO4) and the solvent was removed under reduced pressure to give the title compound (100% yield), which was used in the next step without further purification. Preparation 63
Biphenyl-2-ylcarbamic Acid l-[2-(5-tcrt-Butoxycarbonylaminopentylsulfamoyl)- ethyl]piperidin-4-yl Ester
A solution ofthe product of Preparation 8 (1.33 g, 3.5 mmol) and the product of Preparation 62 (1.62 g, 4.94 mmol) in dichloromethane and methanol (22 mL, 1:1) was heated at 60 °C for 5 h. The reaction mixture was allowed to cool to room temperature and the solvent was removed under reduced pressure. The crude residue was dissolved in dichloromethane (20 mL) and washed with saturated aqueous sodium bicarbonate solution (10 mL). The organic layer was then dried (Na SO4) and solvent removed under reduced pressure. The crude residue was purified by column chromatography (5-10%
MeOH/DCM) to give the title intermediate as a white solid (1.6 g, 55%). MS m/z M+H+ = 589.6.
Preparation 64 Biphenyl-2-ylcarbamic Acid l-[2-(5-Aminopentylsulfamoyl)ethyl]piperidin-4-yl Ester
A solution ofthe product of Preparation 63 (1.6 g, 2.72 mmol) was stirred in dichloromethane (21 mL) and TFA (7 mL) for 1 h and then the solvent was removed under reduced pressure. The crude reaction mixture was dissolved in dichloromethane (30 mL) and washed with 1 M sodium hydroxide (2 x 30 mL). The organic layer was dried (Na2SO4) and the solvent was then removed under reduced pressure to give the title intermediate as an oily white solid (1.19 g, 90% yield).
Preparation 65
Biphenyl-2-yIcarbamic Acid l-(2-{5-[(R)-2-(8-BenzyIoxy-2-oxo-l,2-dihydroquinoIin-5- yl)-2-(t^rt-butyldimethylsilanyloxy)ethylamino]pentylsulfamoyl}ethyl)piperidin-4-yl Ester
A stined solution ofthe product of Preparation 64 (917 mg, 1.88 mmol) and the product of Preparation 13 (460 mg, 0.94 mmol) in DMSO (0.92 mL) was heated at 90 °C for 8 h and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate/chloroform (20 mL,l/l) and the organic layer was washed with saturated aqueous sodium bicarbonate solution (10 mL), dried (Na2SO4) and the solvent was removed under reduced pressure. The resulting crude residue was purified by column chromatography (3- 6% MeOH/DCM) to give the title intermediate as a white solid (500 mg, 60% yield). MS m/z M+H+ = 896.9.
Preparation 66 Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5- yl)-2-hydroxyethylamino]pentylsulfamoyl}ethyl)piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 65 (500 mg, 0.56 mmol) in dichloromethane (5.6 mL) was added triethylamine trihydrofluoride (183 μL, 1.12 mmol). The reaction mixture was stined for 10 h and dichloromethane (10 mL) was added. The resulting mixture was washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic layer was dried (Na2SO4) and the solvent was removed under reduced pressure to give the title intermediate as a yellow solid (437 mg, 100% yield. MS m/z M+H+ = 782.8.
Example 17
Biphenyl-2-ylcarbamic Acid l-(2-{5-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino] pentylsulf amoyl} ethyl)piperidin-4-yl Ester Ditrifluoroacetate
To a stined solution ofthe product of Preparation 66 (437 mg, 0.56 mmol) in ethanol/methanol (5.6 mL, 1/1) was added Pd/C (131 mg) and the reaction mixture placed under a hydrogen atmosphere and stined overnight. The reaction mixture was then filtered and the solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound as the ditrifluoroacetic acid salt (71 mg).
HPLC (10-70) Rt = 2.59; MS m/z M+H+= 692.6.
Preparation 67
Biphenyl-2-ylcarbamic Acid l-{2-[(4-Formylbenzenesulfonyl)methylamino]- ethyl}piperidin-4-yl Ester
To a stirred solution ofthe product of Preparation 26 (350 mg, 1 mmol) and triethylamine (167 μL, 1.2 mmol) in dichloromethane (5 mL) was added 4- formylbenzenesulfonyl chloride (225 mg, 1.1 mmol). After 1 h at room temperature, the reaction was complete by MS and the reaction mixture was then washed with saturated aqueous sodium bicarbonate solution (5 mL). The organic layer was then dried (Na2SO4) and solvent removed under reduced pressure to give the title intermediate (323 mg, 62% yield). MS m/z M+H+= 522.4.
Preparation 68 Biphenyl-2-ylcarbamic Acid l-{2-[(4-{[(R)-2-(tert-Butyldimethylsilanyloxy)-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyI}benzenesulfonyl)- methylamino] ethyl} piperidin-4-yl Ester
A solution of 5-[(R)-2-amino- 1 -(tert-butyldimethylsilanyloxy)ethyl]-8-hydroxy- lH-quinolin-2-one (293 mg, 0.74 mmol) and the product of Preparation 67 in dichloromethane and methanol (6.2 mL, 1/1) was stined at room temperature for lh and then sodium triacetoxyborohydride (394 mg, 1.86 mmol) was added. The reaction mixture was stined for 4 h at which time the reaction was detennined to be complete by MS. The reaction mixture was then acidified with concentrated hydrochloric acid and the solvent was removed under reduced pressure to provide the title compound, which was used in the next step without further purification. MS m/z M+H+ = 840.8.
Example 18
Biphenyl-2-ylcarbamic Acid l-{2-[(4-{ [(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihy droquinolin-5-yl)ethylamino] methyl} benzenesulf onyl)methylamino] ethyl} - piperidin-4-yl Ester Ditrifluoroacetate
A stined solution ofthe product of Preparation 68 (520 mg, 0.62 mmol) in IM hydrochloric acid (5 mL) and acetonitrile (5 mL) was heated at 60 °C for 8 h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude residue was purified by preparative HPLC to give the title compound as the ditrifluoroacetic acid salt (220 mg). HPLC (10-70) Rt = 2.77; MS m/z M+H+= 726.7.
Preparation 69 (3-Aminomethylphenyl)methanol Hydrochloride
(a) (3-ter -Butoxycarbonylmethylphenyϊ)methanol Borane dimethyl sulfide (2.05 mL, 21.6 mmol) was added to a solution of 3-(tert- butoxycarbonylaminomethyl)benzoic acid (1.81 g, 7.20 mmol) in tetrahydrofuran (24 mL). and the resulting mixture was stirred at room temperature for 3 hours. The reaction mixture was then diluted with ethyl acetate (20 mL) and the layers were separated. The organic layer was washed with saturated sodium bicarbonate, saturated sodium chloride, dried over magnesium sulfate and concentrated to give the title compound as a yellow oil (
1.71 g)-
(b) (3-Aminomethylphenyϊ)methanol Hydrochloride
To the product of step (a) (1.71 g, 7.2 mmol) was added a solution of 4 M hydrochloric acid in dioxane (9 mL, 36 mmol) and the resulting mixture was stined at room temperature for 1 h. The reaction mixture was then concentrated and the residue was diluted with diethyl ether (50 mL) and filtered to provide the title compound as a white solid (1.09 g).
Preparation 70
Biphenyl-2-ylcarbamic Acid l-{2-[3-(3-Hydroxymethylbenzyl)ureido]ethyl}piperidin-
4-yl Ester A 0.2 M solution ofthe product of Preparation 35 (760 mg, 2.24 mmol) in N,N- dimethylformamide was added dropwise to a solution of l,l'-carbonyldiimidazole (364 mg, 2.24 mmol) and diisopropylethylamine (0.31 mL, 2.24 mmol) in N,N- dimethylformamide (11 mL) and the resulting mixture was stirred at room temperature for 2 h. Diisopropylethylamine (0.31 mL, 2.24 mmol) and the product of Preparation 69 (578 mg, 3.4 mmol) were added and this mixture was stined at 50 °C for 12 hours. The reaction mixture was then concentrated to dryness and the residue was diluted with dichloromethane (20 mL) and this solution was washed with saturated sodium bicarbonate (2 x), saturated sodium chloride, dried over magnesium sulfate, and concentrated to provide the title compound (1.12 g). LCMS (2-90) Rt = 4.01 min.; MS m/z M+H = 503.5.
Preparation 71
Biphenyl-2-ylcarbamic Acid l-{2-[3-(3-Formylbenzyl)ureido] ethyl} piperidin-4-yl
Ester
A solution ofthe product of Preparation 70 (1.12 g, 2.23 mmol) in dichloromethane (1 1.1 mL) was cooled to 0 °C and diisopropylethylamine (1.17 mL, 6.70 mmol) and dimethyl sulfoxide (0.949 mL, 13.4 mmol) were added. After about 10 minutes, pyridine sulfur trioxide complex (1.06 g, 6.70 mmol) was added and the resulting mixture was stirred at 0 °C for 2 h. The reaction was then quenched with water (15 mL) and the organic layer was washed with cold water (3 x), dried over magnesium sulfate and concentrated to provide the title compound as a yellow crisp (609 mg). LCMS (2-90) R = 4.13 min; MS m/z M+H = 501.3.
Preparation 72
Biphenyl-2-ylcarbamic Acid l-{2-[3-(3-{ [(R)-2-(tert-butyldimethylsilanyloxy)-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}benzyl)ureido]ethyl}- piperidin-4-yl Ester
5-[(/?)-2-Amino- 1 -(tert-butyldimethylsilanyloxy)ethyl]-8-hydroxy- 1 H-quinolin-2- one (575 mg, 1.40 mmol) was added to a solution ofthe product of Preparation 71 (609 mg, 1.2 mmol) and diisopropylamine (0.25 mL, 1.40 mmol) in dichloromethane (6 mL) and the resulting mixture was stined at room temperature for 45 min. Sodium triactetoxyborohydride (385 mg, 1.80 mmol) was then added and this mixture was stirred at room temperature for 12 h. The reaction was then quenched with 10%o aqueous hydrochloric acid (5 mL) and the layers were separated. The organic layer was washed with saturated sodium bicarbonate, saturated sodium chloride, dried over magnesium sulfate and concentrated to give the title compound (1.1 g). HPLC (10-70) Rt = 3.55 min; MS m/z M+H = 819.7.
Example 19
BiphenyI-2-y arbamic Acid l-{2-[3-(3-{[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}benzyl)ureido]ethyl}piperidin-4-yl Ester Ditrifluoroacetate
Triethylamine trihydrofluoride (2.4 mL, 13.6 mmol) was added to a solution ofthe product of Preparation 72 (1.1 g, 1.36 mmol) in dichloromethane (2 mL) and the resulting mixture was stined at room temperature for 15 h. The reaction mixture was then concentrated under vacuum to dryness and the residue was dissolved in a 1 : 1 mixture of water and acetonitrile with 0.1% TFA and this mixture was purified by HPLC (5-35 over 60 min) to provide the title compound as the ditrifluroacetate salt (296 mg, 99% purity). MS m/z M+H = 705.6. Preparation 73
Biphenyl-2-ylcarbamic Acid l-[(E)-3-(4-Nitrophenyl)allyl]piperidin-4-yl Ester
The product of Preparation 8 (2.96 g, 0.01 mol) and^-nitrocinnamaldehyde (1.77 g, 0.01 mol) were stirred in 50 mL of dichloromethane for 2 h. Sodium triacetoxyborohydride (6.33 g, 0.03 mol) was added and the resulting mixture was stirred for 2 h. The reaction was then quenched with 10 mL of water and this mixture was diluted with dichloromethane (100 mL). The organic layer was washed with saturated sodium bicarbonate (2 x), brine, dried over Na2SO4, filtered and concentrated to provide the title compound as a yellow foam (3.8 g, 80%> yield).
Preparation 74
Biphenyl-2-ylcarbamic Acid l-[3-(4-Aminophenyl)propyl]piperidin-4-yl Ester
The product of Preparation 73 (2.5 g, 5.4 mmol) was dissolved in 100 mL of ethanol and the resulting solution was purged with nitrogen for 30 min. Palladium on carbon (2.5g; 50% w/w water; 10%> Pd; l.lmmol Pd) was then added while degassing with nitrogen. This mixture was then placed under hydrogen (50 psi) until hydrogen was no longer consumed (~30 minutes). The mixture was then purged with nitrogen, filtered through Celite and concentrated. The residue was dissolved in ethyl acetate and this mixture was washed with saturated sodium bicarbonate (2 x), brine, dried (Na2SO ), filtered and concentrated to provide the title compound (2.08 g, 90%) yield). MS m/z M+H = 430.5.
Preparation 75
Biphenyl-2-ylcarbamic Acid l-{3-[4-(4-{2-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydro- quinolin-5-yl)-2-(ter^butyldimethylsilanyloxy)ethylamino]ethyl}phenyIanιino)- phenyl]propyl}piperidin-4-yl Ester
To a 25 mL round-bottomed flask was added the product of Preparation 74 (400 mg, 0.8 mmol); 8-benzyloxy-5-[(R)-2-[2-(4-bromophenyl)ethylamino]-l-(tert- butyldimexhylsilanyloxy)ethyl]-lH-quinolin-2-one (769 mg, 1.2 mmol); tris(dibenzylideneacetone)dipalladium(0) (73 mg, 0.08 mmol, 20% Pd); and 2-
(dicyclohexylphosphino)biphenyl (84 mg, 0.24 mmol). This mixture was purged with nitrogen and then dry, degassed toluene (8 mL, 0.1 M) was added and the resulting mixture was heated at 70°C for 30 min. Sodium tert-butoxide (382 mg, 4.0 mmol) was then added, and the temperature was raised to 95 °C for 4 h, at which time LCMS showed complete consumption ofthe product of Preparation 74 and a large product peak (M+H = 956.7). The reaction mixture was then cooled to room temperature and diluted with ethyl acetate. This mixture was washed with saturated sodium bicarbonate (2 x), brine, dried' (Na2SO ), filtered and concentrated to provide the title compound (1.5g), which was used without further purification.
Preparation 76
Biphenyl-2-ylcarbamic Acid l-{3-[4-(4-{2-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydro- quinolm-5-yl)-2-hydroxyethylamino] ethyl} phenylamino)phenyl]propyl}piperidin-4-yl Ester
The product of Preparation 75 was dissolved in dichloromethane (10 mL) and triethylamine trihydrofluoride (10 eq.) was added. The reaction mixture was stined overnight and then diluted with dichloromethane and the organic layer was washed with saturated sodium bicarbonate (2 x), brine, dried (Na2SO4), filtered and concentrated to provide 1.3 g of crude product. This material was purified by silica gel chromatography (DCM, incrementally to 50% methanol) to provide the title compound (300 mg, about 75% purity), which was used without further purification.
Example 20
Biphenyl-2-ylcarbamic acid l-{3-[4-(4-{2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)phenyl]propyl}piperidin-4-yl ester Ditrifluoroacetate
The product of Preparation 76 (300 mg) was dissolved in 10 mL of ethanol and this mixture was purged with nitrogen for 15 minutes. Palladium on carbon (10% Pd, 50%> w/w water, 0.2 eq. Pd) was added while degassing. The resulting mixture was then placed under 1 atm. of hydrogen for 2 h, at which time the reaction was complete by LCMS. The solution was then purged with nitrogen for 15 min and then filtered through Celite and concentrated. The resulting residue was purified by prep HPLC to afford the title compound as the ditrifluoroacetate salt (59 mg, >95% purity). MS m/z M+H = 752.8. Preparation 77
Biphenyl-2-ylcarbamic Acid l-[2-Fluoro-3-(4-hydroxymethylpiperidin-l-ylmethyl)- benzyl]piperidin-4-yl Ester The product of Preparation 8 (500 mg, 1.69 mmol), 2,6-bis(bromomethyl)-l- fluorobenzene (476 mg, 1.69 mmol, piperidin-4-ylmethanol (195 mg, 1.69 mmol) and potassium carbonate (466 mg, 3.37 mmol) were suspended in acetonitrile (5 mL) and stined at room temperature for 18 h. The reaction mixture was then concentrated and the residue was dissolved in dichloromethane/water. The layers were separated and the organic layer was washed with water (2 x), brine, dried (MgSO4) and concentrated. The crude material was purified by silica gel column chromatography eluting with 3% methanol/chloroform to give the title compound as a white foam (282 mg). MS m/z M+H = 532.3.
Preparation 78
Biphenyl-2-ylcarbamic Acid l-[2-Fluoro-3-(4-formylpiperidin-l-ylmethyl)benzyl]- piperidin-4-yl Ester
The product of Preparation 77 (282 mg, 0.53 mmol) was dissolved in dichloromethane and to this mixture was added diisopropylethylamine (280 μL, 1.6 mmol) and dimethyl sulfoxide (115 μL, 1.6 mmol). The reaction mixture was cooled to -15 °C under nitrogen and pyridine sulfur trioxide complex (255 mg, 1.6 mmol) was added and the resulting mixture was stined for 40 min. The reaction was then quenched with water and the layers were separated. The organic layer was washed with aqueous NaH2PO4 (IM x 3), brine, dried (MgSO4) and concentrated to provide the title compound as a foam (253 mg). MS m/z M+H = 530.4.
Example 21
Biphenyl-2-ylcarbamic Acid l-[2-Fluoro-3-(4-{ [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}piperidin-l-ylmethyl)benzyl]piperidin- 4-yl Ester Ditrifluoroacetate
The product of Preparation 78 (253 mg, 0.48 mmol) was dissolved in a 1:1 mixture of dichloromethane and methanol (6 mL) and to this mixture was added 5-[(R)-2-amino-l- (tert-butyldimethylsilanyloxy)ethyl]-8-hydroxy-lH-quinolin-2-one acetate (228 mg, 0.58 mmol) and sodium triacetoxyborohydride (317 mg, 1.5 mmol). The reaction mixture was stirred under nitrogen at room temperature for 18 h and then concentrated. The residue was dissolved in a 2:3 mixture of acetonitrile and aqueous 6 N hydrochloric acid, and this mixture was heated at 55 °C for 4 hours. The reaction mixture was then concentrated and the residue was dissolved in water/acetonitrile/trifluoroacetic acid (1:1 :0.005) and purified by reverse phase column chromatography to afford the title compound as a white solid (175 mg). MS m/z M+H = 734.5.
Preparation 79 2- [4-(3-Bromopropoxy)phenyl] ethanol
To a solution of 4-hydroxyphenethyl alcohol (4.37 g, 31.0 mmol) and potassium carbonate (6.55 g, 47.0 mmol) in acetonitrile (62.0 mL) was added 1,3 dibromopropane (31.0 mL, 316 mmol). The reaction mixture was heated to 70 °C for 12 hours and then cooled to room temperature, filtered and concentrated under vacuum. The resulting oil was purified by silica gel chromatography using a mixture of 4:1 hexanes and ethyl acetate to give the title compound (6.21 g) as a white solid.
Preparation 80
Biphenyl-2-ylcarbamic Acid l-{3-[4-(2-Hydroxyethyl)phenoxy]propyl}piperidin-4-yl Ester
To a solution ofthe product of Preparation 79 (1.11 g, 4.30 mmol) and diisopropylethylamine (0.90 mL, 5.10 mmol) in acetonitrile (21.5 mL) was added the product of Preparation 8 (1.27 g, 4.30 mmol) and the resulting mixture was stirred at 60°C. for 12 h. The reaction mixture was then diluted with dichloromethane (20 mL) and washed with saturated sodium bicarbonate (25 mL), saturated sodium chloride (25 mL), dried over magnesium sulfate and concentrated to provide the title compound (1.98 g, 85% purity). MS m/z M+H = 475.5.
Preparation 81 Biphenyl-2-ylcarbamic Acid l-{3-[4-(2-Oxoethyl)phenoxy]propyl}piperidin-4-yl Ester
A solution ofthe product of Preparation 80 (723 mg, 1.53 mmol) and dichloromethane (75 mL) was cooled to about 5 °C and diisopropylethylamine (798 mL, 4.58 mmol) and dimethyl sulfoxide (649 mL, 9.15 mmol) were added. Pyridine sulfur trioxide (728 mg, 4.58 mmol) was then added and the resulting mixture was stirred at 5 °C for 45 min. The reaction mixture was then diluted with dichloromethane (20 mL) and washed with saturated sodium bicarbonate (25 mL), saturated sodium chloride (25 mL), dried over magnesium sulfate and concentrated to provide the title compound (604 mg). MS m/z M+H = 473.4.
Preparation 82
Biphenyl-2-ylcarbamic Acid l-[3-(4-{2-[(R)-2-(to*t-butyldimethylsilanyloxy)-2-(8- hydroxy-2-oxo-l,2-dihydroquinoIin-5-yl)ethylamino]ethyl}- phenoxy)propyl] piperidin-4-yl Ester
The product of Preparation 81 (604 mg, 1.28 mmol) was dissolved in methanol (6.4 mL) and 5-[(R)-2-amino- 1 -(tert-butyldimethylsilanyloxy)ethyl]-8-hydroxy- 1 H-quinolin-2- one (605 mg, 1.53 mmol) and diisopropylethylamine (0.27 mL, 1.53 mmol) were added. Sodium triacetoxyborohydride (405 mg,1.91 mmol) was then added and the reaction mixture was stirred at room temperature for 3 h. The reaction mixture was then concentrated to dryness and the residue was diluted with ethyl acetate (20 mL) and this solution was washed with saturated sodium bicarbonate (25 mL), saturated sodium chloride (25 mL), dried over magnesium sulfate and concentrated to give the title compound (704 mg). MS m/z M+H = 791.8.
Example 22
Biphenyl-2-yIcarbamic Acid l-[3-(4-{2-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino] ethyl} phenoxy)propyl] piperidin-4-yl Ester Ditrifluoroacetate Triethylamine trihydrofluoride (1.5 mL, 8.87 mmol) was added to a solution ofthe product of Preparation 82 (702 mg, 0.89 mmol) in dichloromethane (4.5 mL) and the resulting mixture was stined at room temperature for 24 h. The mixture was then concentrated under vacuum and purified by HPLC (2-35 over 90 min) to give the title compound (92 mg) as a white powder. MS m/z M+H = 677.4. Preparation 83
Methyl 4-Iodophenylacetate
To a stirred solution of 4-iodophenylacetic acid (5.0 g, 19.1 mmol) in MeOH (200 mL) was added 4N hydrochloric acid in dioxane (10 mL). The reaction mixture was stined for 24 h at room temperature and then the solvent was removed under reduced pressure to give the title compound (5.17 g, 98% yield), which was used without further purification.
Preparation 84
Methyl [4-(4-Hydroxybut-l-ynyl)phenyl] acetate To a stirred solution ofthe product of Preparation 83 (4.5 g, 16.3 mmol) in diethylamine (100 mL) was added but-3-yn-l-ol (1.9 mL, 32.6 mmol), Pd(PPh3)2Cl2 (500 mg, 1.63 mmol) and Cul (154 mg, 0.815 mmol) and resulting mixture was stined for 17 h at room temperature. The solvent was then removed under reduced pressure and the residue was dissolved in diethyl ether (200 mL) and this solution was filtered to remove salts. The solvent was then removed under reduced pressure and the crude product was purified by silica gel chromatography (60 % EtOAc/Hexane) to afford the title intermediate (3.03 g, 91% yield).
Preparation 85 Methyl [4-(4-Hydroxybutyl)phenyl] acetate
A stined solution ofthe product of Preparation 84 (2.8 g, 12.8 mmol) in methanol (50 mL) was flushed with nitrogen and then 10% palladium on carbon (400 mg, 20% wt/wt) was added. The reaction flask was then alternately placed under vacuum and flushed with hydrogen for cycles and then stined under hydrogen for 14 h. The reaction mixture was flushed with nitrogen and then filtered and the solvent removed under reduced pressure to give the title compound (2.75 g, 97%> yield), which was used without further purification.
Preparation 86 Methyl (4-{4-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]butyl}phenyl)acetate (a) Methyl {4-[4-(Toluene-4-sulfonyloxy)butyl]phenyl}acetate
To a stined solution ofthe product of Preparation 85 (2.6 g, 12.5 mmol) in THF (100 mL) was added DABCO (2.6 g, 25.0 mmol) and then p-toluenesulfonyl chloride (2.44 g, 13.75 mmol). The reaction mixture was stined at room temperature for 23 h and then solvent was removed under reduced pressure and the residue was dissolved in dichloromethane (200 mL). The organic layer was then washed with water (2 X 100 mL), IN hydrochloric acid (100 mL), aqueous saturated sodium chloride solution (100 mL), dried (MgSO4), filtered and the solvent removed under reduced pressure to give the title compound, which was used without further purification.
(b) Methyl (4-{4-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l- yl] butyl} phenyl)acetate To the crude product from step (a) was added DMF (50 mL), diisopropylethylamine (3.0 mL, 17.3 mmol) and the product of Preparation 8 (2.4 g, 8.1 mmol). The reaction mixture was stined at room temperature for 18 h and then the solvent was removed under reduced pressure to give the title compound (3.5 g, 86.3% yield). MS m/z 501.6 (MH+), £,-4.89 min (10-70% ACN: H2O, reverse phase HPLC).
Preparation 87
Biphenyl-2-ylcarbamic Acid l-{4-[4-(2-Hydroxyethyl)phenyl]butyl}piperidin-4-yl
Ester
To a stined solution ofthe product of Preparation 86 (2.0 g, 4.0 mmol) in THF (100 mL) was added dropwise DIBAL (24 mL, 24 mmol, 1.0 M in THF). After the addition was complete, the reaction mixture was stined for 3 h and then quenched by slow addition of methanol (until gas evolution ceased). The mixture was then stined for 30 min. and then ethyl acetate (200 mL) and aqueous IN sodium hydroxide (200 mL) were added. The organic layer was separated and washed with aqueous saturated sodium chloride solution (100 mL), dried (MgSO ), filtered and the solvent removed under reduced pressure to give the title compound (1.3 g, 69%) yield), which was used without further purification. MS m/z 473.4 (MH+), i? 4.53 min (10-70% ACN: H2O, reverse phase HPLC). Example 23
Biphenyl-2-ylcarbamic Acid l-[2-(4-{2-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenyl)ethyl]piperidin-4-yl Ester
To a stined solution ofthe product of Preparation 87 (500 mg, 1.06 mmol) in dichloromethane (25 mL) was added dimethyl sulfoxide (0.60 mL, 10.6 mmol) and diisopropylethylamine (0.921 mL, 5.3 mmol). The reaction mixture was then cooled to -10 °C and pyridine sulfur trioxide (842 mg, 5.3 mmol) was added. The reaction mixture was stined for 1 h and then quenched by adding water (100 mL). This mixture was stined for 10 min and then the organic layer was removed and washed with aqueous saturated sodium chloride solution (100 mL), dried (MgSO4) and then filtered.
To the filtrate was added methanol (25 mL), 5-[(R)-2-amino-l-(tert- butyldimethylsilanyloxy)ethyl]-8-hydroxy-lH-quinolin-2-one acetate (419 mg, 1.06 mmol) and sodium triacetoxyborohydride (468 mg, 2.12 mmol). This mixture was stirred for 16 h and then condensed and, to the resulting mixture, was added a 1 :1 mixture of acetonitrile and aqueous 4N hydrochloric acid (20 mL). This mixture was heated at 50 °C for 17 h and then the solvent was removed under reduced pressure. To the residue was added a 1 : 1 mixture of acetic acid and water (8.0 mL) and the mixture was chromatographed on reverse-phase silica gel (gradient elution, 10-50% ACN/H2O) to afford the title compound (67 mg, 7% yield over 3 steps). MS m/z (MH+) 675.5; Λ 3.07 (10-70% ACN: H2O, reverse phase HPLC).
Preparation 88
Ethyl 3-[5-(2-Ethoxycarbonylvinyl)thiophen-2-yl] acrylate To a stined solution of sodium hydride (2.1 g, 53 mmol, 60%> in mineral oil) in
THF (200 mL) was slowly added triethylphosphonoacetate (10 mL, 50 mmol) Hydrogen gas evolution was observed and the reaction was stined until gas evolution ceased (about 30 min). To this reaction mixture was added 2,5-thiophenedicarboxaldehyde (3 g, 21 mmol) and the reaction mixture was stined for 1 h. The solvent was removed under reduced pressure and the residue was dissolved in dichloromethane (200 mL). The organic layer was washed with water (100 mL), aqueous IN hydrochloric acid (100 mL), aqueous saturated sodium chloride solution (100 mL), dried (MgSO4), filtered and the solvent removed under reduced pressure to give the title compound (5.8 g, 98% yield), which was used without further purification. Preparation 89
Ethyl 3-[5-(2-Ethoxycarbonylethyl)thiophen-2-yl]propionate
A stined solution ofthe product of Preparation 88 (5.8 g, 21 mmol) in methanol (200 mL) was flushed with nitrogen and 10% palladium on carbon (576 mg, 10%> wt/wt) was added. The reaction flask was alternately placed under vacuum and flushed with hydrogen for 3 cycles and then the reaction mixture was stined under hydrogen for lh. The mixture was then was flushed with nitrogen, filtered and the solvent removed under reduced pressure to give the title compound (5.8 g, 99% yield), which was used without further purification.
Preparation 90 3-[5-(3-Hydroxypropyl)thiophen-2-yl]propan-l-ol
To a stined solution of DIBAL (88 mL, 88 mmol, l.OM in cyclohexane) in THF (300 mL) at -78 °C was added dropwise the product of Preparation 89 (5.0 g, 17.6 mmol). After the addition was complete, the reaction mixture was warmed to room temperature over 30 min and then quenched by slow addition of aqueous IN hydrochloric acid (200 mL). Dichloromethane (400 mL) was added and the layers were separated. The aqueous layer was washed with dichloromethane (4 x 100 mL) and the combined organic layers were washed with aqueous saturated sodium chloride solution (100 mL), dried (MgSO ), filtered and the solvent removed under reduced pressure to give the title compound (3.0 g, 85% yield), which was used without further purification.
Preparation 91 Biphenyl-2-ylcarbamic Acid l-{3-[5-(3-Hydroxypropyϊ)thiophen-2- yl]propyl}piperidin-4-yl Ester
(a) Toluene-4-sulfonic Acid 3-[5-(3-Hydroxypropyf)thiophen-2-yl]propyl Ester
To a stined solution ofthe product of Preparation 90 (423 mg, 2.1 mmol) in THF (20 mL) was added DABCO (420 mg, 4.2 mmol) and then ?-toluenesulfonyl chloride (442 mg, 2.3 mmol). The reaction mixture was stined at room temperature for 2 h and then the solvent was removed under reduced pressure and the residue was dissolved in dichloromethane (200 mL). The organic layer was washed with water (2 x 100 mL), aqueous saturated sodium chloride solution (100 mL), dried MgSO4), filtered and the solvent removed under reduced pressure to give the title compound, which was used without further purification.
(b) Biphenyl-2-ylcarbamic Acid l-{3-[5-(3-Hydroxypropyl)thiophen-2- yl]propyl}piperidin-4-yl Ester
To the product from step (a) was added acetonitrile (20 mL), diisopropylethylamine (0.5 mL, 2.8 mmol) and the product of Preparation 8 (626 mg, 2.11 mmol). The reaction mixture was heated to 50 °C for 20 h and then cooled to room temperature and the solvent was removed under reduced pressure. The residue was purified by silica gel chromatography (5% MeOH/DCM with 0.6% NH3 (aq)) to afford the title compound (450 mg, 44% yield). MS m/z (MH+) 479.6; R 4.15 min (10-70% ACN: H2O, reverse phase HPLC).
Preparation 92
Biphenyl-2-ylcarbamic Acid l-[3-(5-{3-[(R)-2-(tert-Butyldimethylsilanyloxy)-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]propyl}thiophen-2-yl)propyl]- piperidin-4-yl Ester
To a stirred solution ofthe product of Preparation 91 (450 mg, 0.94 mmol) in dichloromethane (20 mL) was added dimethyl sulfoxide (0.21 mL, 3.7 mmol) and diisopropylethylamine (0.65 mL, 3.7 mmol). This mixture was cooled to -10 °C and pyridine sulfur trioxide (444 mg, 2.8 mmol) was added. The reaction mixture was stirred for 3 h and then was quenched by adding water (100 mL). This mixture was stined for 10 min and then the organic layer was removed and was washed with aqueous saturated sodium chloride solution (100 mL), dried (MgSO ) and filtered.
To the filtrate was added methanol (20 mL), 5-[(i?)-2-amino-l-(tert- butyldimethylsilanyloxy)ethyl]-8-hydroxy-lH-quinolin-2-one acetate (368 mg, 0.93 mmol) and then sodium triacetoxyborohydride (412 mg, 1.86 mmol). This mixture was stined for 19 h and then the mixture was condensed to give the title compound, which was used without further purification. MS m/z (MH+) 795.8; R/4.93 min (10-70% ACN: H2O, reverse phase HPLC). Example 24
Biphenyl-2-ylcarbamic Acid l-[3-(5-{3-[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]propyl}thiophen-2-yl)propyl]piperidin-4-yl Ester
To the crude product from Preparation 92 was added a 1:1 mixture of acetonitrile and aqueous 4 N hydrochloric acid (25 mL). This mixture was heated at 50 °C for 17 h and then the solvent was then removed under reduced pressure. To the residue was added a 1 : 1 mixture of acetic acid and water (8.0 mL) and this mixture was chromatographed on reverse-phase silica gel (gradient elution, 10-50% ACN/H2O) to afford the title compound (135 mg, 16 % yield for 3 steps). MS m/z (MH+) 681.5; R 3.03 (10-70% ACN: H2O, reverse phase HPLC).
Preparation 93
Methyl 4-Amino-5-chIoro-2-methoxybenzoate
To a solution of 4-amino-5-chloro-3-methoxybenzoic acid (1.008 g, 5.0 mmol) in a mixture of toluene (9 mL) and methanol (1 mL) at 0 °C was added
(trimethylsilyl)diazomethane (2.0 M in hexane, 3.0 mL, 6.0 mmol) dropwise. The reaction mixture was then warmed to room temperature and stined for 16 h. Excess (trimethylsilyl)diazomethane was quenched by adding acetic acid until the bright yellow color ofthe reaction mixture disappeared. The mixture was then concentrated in vacuo to give the title compound as an off-white solid, which was used without further purification.
Preparation 94
Methyl 4-Acryloylamino-5-chloro-2-methoxybenzoate
To crude product of Preparation 93 was added dichloromethane (10 mL, 0.5 M) and triethylamine (2.1 mL, 15 mmol). This mixture was cooled to 0 °C and acryloyl chloride (812 μL, 10 mmol) was added dropwise with stirring. After 2 h, the reaction was quenched by adding methanol (about 2 mL) at 0 °C and the resulting mixture was stined at room temperature for 15 min and then concentrated in vacuo. Dichloromethane (30 mL) and water (30 mL) were added to the residue and this mixture was mixed thoroughly. The layers were separated and the aqueous layer was extracted with dichloromethane (20 mL). The organic layers were combined, dried (Na2SO4), filtered and the solvent was removed in vacuo to give the title compound as a brown foamy solid, which was used without further purification. Preparation 95
Methyl 4-{3-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionylanιino}-5- chloro-2-methoxybenzoate To the crude product from Preparation 94 was added the product of Preparation 8
(1.33 g, 4.5 mmol) and a mixture of THF (22.5 mL) and methanol (2.5 mL). This mixture was heated at 50 °C with stirring for 16 h and then the solvent was removed in vacuo. The residue was chromatographed (silica gel; EtOAc) to give the title compound (0.82 g; Rf = 0.4, 29% yield over 3 steps) as an off-white foamy solid. MS mlz 566.4 (M+H, expected 565.20 for C30H32C1N3O6).
Preparation 96
Biphenyl-2-ylcarbamic Acid l-[2-(2-Chloro-4-hydroxymethyl-5-methoxy- phenylcarbamoyι)ethyl]piperidin-4-yl Ester To a solution ofthe product of Preparation 95(0.82 mg, 1.45 mmol) in a mixture of
THF (4.5 mL) and methanol (0.5 mL) at 0 °C was added lithium borohydride (32 mg, 1.45 mmol). The reaction mixture was allowed to warm to room temperature and was stined for 41 h. The reaction was then quenched by adding IN aqueous hydrochloric acid at 0 °C until no more bubbling was observed and this mixture was stined for 10 min. The solvent was removed in vacuo and the residue was dissolved in acetonitrile (about 2 mL). This solution was purified by prep-RP-HPLC (gradient: 2 to 50% acetonitrile in water with 0.05%) TFA). The appropriate fractions were collected and combined and lyophilized to give the title compound as a trifluoroacetate salt. This salt was treated with isopropyl acetate (10 mL) and IN aqueous sodium hydroxide (10 mL) and the organic layer was collected, dried (Na2SO4), filtered and the solvent was removed in vacuo to give the title compound (161 mg, 21% yield) as a white foamy solid. MS m/z 538.4 (M+H, expected 537.20 for C29H32ClN3O5).
Preparation 97 Biphenyl-2-ylcarbamic Acid l-[2-(2-Chloro-4-formyl-5-methoxyphenylcarbamoyl)- ethyl]piperidin-4-yl Ester
To a solution ofthe product of Preparation 96 (161 mg, 0.3 mmol) in dichloromethane (3 mL) was added dimethyl sulfoxide (213 μL, 3.0 mmol) and diisopropylethylamine (261 μL, 1.5 mmol). This mixture was cooled to -20 °C and sulfur trioxide pyridine complex (238 mg, 1.5 mmol) was added slowly. After 30 min, the reaction mixture was quenched by adding water (about 3 mL). The layers were separated and the organic layer was dried (Na2SO ), filtered and the solvent was removed in vacuo to give the title compound as a light yellow solid. MS mlz 536.3 (M+H, expected 535.19 for C29H30C1N3O5).
Preparation 98
Biphenyl-2-ylcarbamic Acid l-[2-(4-{ [(R)-2-(tert-Butyldimethylsilanyloxy)-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}-2-chloro-5-methoxy- phenylcarbamoyl)ethyl]piperidin-4-yl Ester
To the product from Preparation 97 in a mixture of dichloromethane (0.5 mL) and methanol (0.5 mL) was added 5-[(i?)-2-amino-l-(tert-butyldimethylsilanyloxy)ethyl]-8- hydroxy-lH-quinolin-2-one acetate (124.1 mg, 3.1 mmol) and the resulting mixture was stirred at room temperature for 1.5 h. Sodium triacetoxyborohydride (190.7 mg, 0.9 mmol) was added and the resulting mixture was stined at room temperature for 15 h. The reaction was quenched by adding water (about 0.2 mL) and the mixture was concentrated in vacuo to give the title compound, which was used without further purification. MS m/z 854.5 (M+H, expected 853.36 for C46H56ClN5O7Si).
Example 25
Biphenyl-2-ylcarbamic Acid l-[2-(2-Chloro-4-{ [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}-5- methoxyphenylcarbamoyl)ethyl]piperidin-4-yl Ester Ditrifluoroacetate
To a suspension ofthe product of Preparation 98 in dichloromethane (1.0 mL, 0.3 M) was added triethylamine trihydrofluoride (245 μL, 1.5 mmol). This mixture was stined at room temperature for 45 h and then the mixture was concentrated in vacuo. The residue was dissolved in a mixture of DMF (0.5 mL), acetonitrile/water (1:1, with 0.1% TFA, 0.6 mL), TFA (0.3 mL) and acetonitrile (about 1 mL) and this mixture was purified by prep- RP-HPLC (gradient: 2 to 50% acetonitrile in water with 0.05% TFA). The appropriate fractions were collected and combined and lyophilized to give the title compound (100 mg, 34% yield, 98.7% pure by HPLC) as an off-white solid. MS mlz 740.5 (M+H, expected 739.28 for C40H42C1N5O7). Using the methods described above and the appropriate starting materials, the following compounds were prepared.
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Preparation 99
Biphenyl-2-ylcarbamic Acid l-[2-(4-[l,3]dioxolan-2-ylphenylcarbamoyl)-ethyl]-4- methylpiperidin-4-yl Ester
A mixture of biphenyl-2-yl carbamic acid 4-methylpiperidin-4-yl ester (2.73 g, 8.79 mmol) and N-(4-[ 1 ,3]dioxolan-2-yl-phenyl)acrylamide (2.05 g, 8.80 mmol) were heated in 100 mL of 1 : 1 methanol/dichloromethane at 50 °C under nitrogen for 1 h. The solution was then diluted with ethyl acetate and the organic layer was washed with water, brine, dried (MgSO ) and concentrated under reduced pressure to give the title compound. MS m/z calcd for C3ιH35N3O5 (M+H)+ 530.6; found 530.4.
Preparation 100
Biphenyl-2-ylcarbamic Acid l-[2-(4-FormylphenyIcarbamoyl)ethyl]-4- methylpiperidin-4-yl Ester
The product of Preparation 99 was redissolved in 40 mL of methanol and 25 mL of aqueous 1 N hydrochloric acid was added. The resulting mixture was stined at room temperature overnight and the organic solvent was removed under reduced pressure. The residue was dissolved in ethyl acetate and the organic layer was washed with water, brine, dried (MgSO4) and the solvent removed under reduced pressure. The product was triturated with dichloromethane to give the title compound as a white powder (2.47 g). LCMS (2-90) Rt = 4.27 min; MS m/z calcd for C29H31N3O4 (M+H)+ 486.6, found 486.5.
Preparation 101
Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-(te^-butyldimethylsilanyloxy)-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]- 4-methyl-piperidin-4-yl Ester
A mixture ofthe product of Preparation 100 (1.70 g, 3.51 mmol) and 5-[(R)-2- amino- 1 -(tert-butyldimethylsilanyloxy)ethyl] -8-hydroxy- 1 H-quinolin-2-one acetate (1.65 g, 4.19mmol) in 40 mL of 1:1 methanol and dichloromethane was stined at room temperature overnight. Sodium triacetoxyborohydride (2.23 g, 10.5 mmol) was then added in one portion and the reaction mixture was stined at room temperature for 6 hr. The reaction was then quenched with water and diluted with ethyl acetate. The layers were separated and the organic layer was washed with saturated sodium bicarbonate, brine, dried (MgSO4) and the solvent removed under reduced pressure to give the title compound (2.9 g). MS m/z calcd for C46H57N5O6Si (M+H)+ 805.0, found 804.6.
Example 170 Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(R)-2-Hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-phenylcarbamoyl)ethyl]-4- methylpiperidin-4-yl Ester
The product of Preparation 101 (2.9 g, 3.6mmol) was dissolved in 75 mL of dichloromethane and triethylamine trihydrofluoride (0.85 mL , 5.2mmol) was added. The resulting mixture was stined at room temperature overnight and then the solvent was removed under reduced pressure to give the crude product as an oil. The product was then dissolved in acetic acid/water (1:1) and purified by prep HPLC to give the title compound as an off-white solid. LCMS (2-90) Rt = 3.67 min.; MS m/z calcd C40H43N5O6 (M+H)+ 690.8, found 690.3. Using the methods described herein and the appropriate starting materials, the following compounds can be prepared.
Figure imgf000153_0001
Figure imgf000154_0001
Preparation 102
Biphenyl-2-ylcarbamic acid (R)-(l-azabicyclo[3.2.1]oct-4-yl) Ester
2-Biphenyl isocyanate (1.00 g, 5.12 mmol) and (i?)-(-)-3-quinuclidinol hydrochloride (921 mg, 5.63 mmol) were heated together in N,N-dimethylformamide (2.06 mL) at 110 °C for 12 h. The reaction mixture was cooled and diluted with ethyl acetate (15 mL) and then washed with saturated aqueous sodium bicarbonate (2 x 10 mL). The organic layer was extracted with 1 M hydrochloric acid (3 x 20 mL) and the combined aqueous extracts were made basic to pH 8-9 with potassium carbonate. The aqueous layer was then extracted with ethyl acetate (3 x 20 mL) and the combined organic layers were dried (magnesium sulfate) and solvent was removed under reduced pressure to give the title compound as a yellow oil (1.64 g, 99%> yield).
Preparation 103
(R)-4-(Biphenyl-2-ylcarbamoyloxy)-l-(9-bromononyl)-l-azoniabicyclo[3.2.1]octane
Bromide
To a stined solution ofthe product of Preparation 102 (1.21 g, 3.76 mmol) and triethylamine (1.05 mL, 7.52 mmol) in acetonitrile (18.8 mL) was added 1,9- dibromononane (994 μL, 4.89 mmol) and the reaction mixture was heated at 50 °C for 4 h.
The reaction mixture was then cooled and the solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (20 mL) and the organic layer was washed with saturated aqueous sodium bicarbonate (10 mL), dried (magnesium sulfate) and solvent removed under reduced pressure. The crude product was purified by flash chromatography (10% methanol/dichloromethane, 0.5 % ammonium hydroxide) to give the title compound (1.04 g, 1.97 mmol, 52% yield).
Preparation 104 (R)-l-(9-i\ V-Di(tert-butoxycarbonyl)aminononyl)-4-(biphenyl-2-ylcarbamoyloxy)-l- azoniabicyc!o[3.2.1]octane Bromide
To a stirred solution of sodium hydride (60% dispersion in mineral oil) (126 mg, 3.15 mmol) in N,N-dimethylformamide (10 mL) under an atmosphere of nitrogen at 0°C, was added di-tert-butyl iminodicarboxylate (513 mg, 2.36 mmol) in N,N- dimethylformamide (5 mL). The reaction mixture was stined at room temperature for 15 min and then it was cooled to 0 °C and the product of Preparation 103 (1.04 g, 1.97 mmol) in N,N-dimethylformamide (5 mL) was added. The reaction mixture was allowed to warm to room temperature over a 12 h period and then the solvent was removed under reduced pressure to give the title compound, which was used without further purification. Preparation 105
(R)-l-(9-Aminononyl)-4-(biphenyl-2-ylcarbamoyloxy)-l-azoniabicyclo[3.2.1]octane
Bromide
The product of Preparation 104 (1.31 g, 1.97 mmol) was dissolved in dichloromethane (15 mL) and trifluoroacetic acid (5 mL) was added slowly. The reaction mixture was stined at room temperature for 1 h and then the solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (20 mL) and washed with aqueous 1 M sodium hydroxide (20 mL). The organic layer was extracted with 1 M hydrochloric acid (3 x 20 mL) and the combined aqueous extracts were made basic with potassium carbonate and extracted with dichloromethane (3 x 20 mL). The combined organic layers were dried (magnesium sulfate) and solvent was removed under reduced pressure to give the title compound (210 mg, 23% yield over 2 steps).
Preparation 106 (R)-l-{9-[(R)-2-(8-Benzyloxy-2-oxo-l,2-dihydroquinolin-5-yl)-2-hydroxyethylamino]- nonyl}-4-(biphenyl-2-ylcarbamoyloxy)-l-azoniabicyclo[3.2.1]octane Bromide
The product of Preparation 105 (210 mg, 0.45 mmol) and sodium triacetoxyborohydride (286 mg, 1.35 mmol) were stined in dichloroethane (4.5 mL) at room temperature for 2 h and then the product of Preparation 6 (163 mg, 0.50 mmol) was added. The reaction mixture was stined for 12 h and then diluted with dichloromethane (10 mL) and washed with saturated aqueous sodium bicarbonate (10 mL), dried (magnesium sulfate) and solvent removed under reduced pressure. The crude reaction product was purified by flash chromatography (10-50% methanol/dichloromethane, 0.5% ammonium hydroxide) to give the title compound (131 mg, 38% yield).
Example 183
4-(Biphenyl-2-ylcarbamoyloxy)-l-{9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroqumolm-5-yl)-ethylamήιo]nonyl}-l-azoniabicyclo[2.2.2]octane Bromide Ditrifluoroacetate To a stined .solution ofthe product of Preparation 105 (131 mg, 0.17 mmol) in methanol (1.8 mL) was added palladium (10 wt. % dry basis on activated carbon; 39 mg) and the reaction mixture was placed under an atmosphere of hydrogen. After stirring for 12 h, the reaction mixture was filtered through a pad of Celite, washing with methanol (2 mL) and solvent removed under reduced pressure. The resulting residue was purified by preparative HPLC to give the title compound as its ditrifluoroacetate salt (8 mg). MS m/z 667.5.
Using the methods described herein and the appropriate starting materials, the following compounds can be prepared.
Figure imgf000157_0001
Preparation A Cell Culture and Membrane Preparation From Cells Expressing Human βi, β2 or β3 Adrenergic Receptors
Chinese hamster ovarian (CHO) cell lines stably expressing cloned human β1} β2 or β3 adrenergic receptors, respectively, were grown to near confluency in Hams F-12 media with 10%) FBS in the presence of 500 μg/mL Geneticin. The cell monolayer was lifted with 2mM EDTA in PBS. Cells were pelleted by centrifugation at 1 ,000 rpm, and cell pellets were either stored frozen at -80 °C or membranes were prepared immediately for use. For preparation of β] and β2 receptor expressing membranes, cell pellets were resuspended in lysis buffer (10 mM HEPES/HC1, lOmM EDTA, pH 7.4 at 4°C) and homogenized using a tight-fitting Dounce glass homogenizer (30 strokes) on ice. For the more protease-sensitive β3 receptor expressing membranes, cell pellets were homogenated in lysis buffer (lOmM Tris/HCl, pH 7.4) supplemented with one tablet of "Complete Protease Inhibitor Cocktail Tablets with 2 mM EDTA" per 50 mL buffer (Roche Catalog No. 1697498, Roche Molecular Biochemicals, Indianapolis, IN). The homogenate was centrifuged at 20,000 x g, and the resulting pellet was washed once with lysis buffer by re- suspension and centrifugation as above. The final pellet was then re-suspended in ice-cold binding assay buffer (75 mM Tris/HCl pH 7.4, 12.5mM MgCl2, 1 mM EDTA). The protein concentration ofthe membrane suspension was determined by the methods described in Lowry et al., 1951, Journal of Biological Chemistry, 193, 265; and Bradford, Analytical Biochemistry, 1976, 72, 248-54. All membranes were stored frozen in aliquots at -80 °C or used immediately.
Preparation B
Cell Culture and Membrane Preparation From Cells Expressing Human Mi, M2, M3 and M Muscarinic Receptors
CHO cell lines stably expressing cloned human hMi, hM2, hM3 and hM4 muscarinic receptor subtypes, respectively, were grown to near confluency in HAM's F-12 media supplemented with 10% FBS and 250 μg/mL Geneticin. The cells were grown in a 5% CO2, 37 °C incubator and lifted with 2 mM 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 use. For membrane preparation, cell pellets were resuspended in lysis buffer and homogenized with a Polytron PT-2100 tissue disrupter (Kjnematica AG; 20 seconds x 2 bursts). Crude membranes were centrifuged at 40,000 x g for 15 minutes at 4°C. The membrane pellet was then resuspended with re- suspension buffer and homogenized again with the Polytron tissue disrupter. The protein concentration ofthe membrane suspension was detennined by the method described in Lowry et al., 1951, Journal of Biochemistry, 193, 265. 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 °C until use.
Assay Test Procedure A
Radioligand Binding Assay for Human βi, β2 and β3 Adrenergic Receptors Binding assays were performed in 96-well microtiter plates in a total assay volume of 100 μL with 10-15 μg of membrane protein containing the human βi, β2or β3 adrenergic receptors in assay buffer (75 mM Tris/HCl pH 7.4 at 25°C, 12.5 mM MgCl2, 1 mM EDTA, 0.2% BSA). Saturation binding studies for determination of K^ values ofthe radioligand were done using [3H]-dihydroalprenolol (NET- 720, 100 Ci/mmol, PerkinElmer Life Sciences Inc., Boston, MA) for the βi and β2 receptors and [125I]-(-)- iodocyanopindolol (NEX-189, 220 Ci/mmol, PerkinElmer Life Sciences Inc., Boston, MA) at 10 or 11 different concentrations ranging from 0.01 nM to 20 nM. Displacement assays for determination of K,- values of test compounds were done with [3H]-dihydroalprenolol at 1 nM and [125I]-(-)-iodocyanopindolol at 0.5 nM for 10 or 11 different concentrations of test compound ranging from 10 pM to 10 μM. Non-specific binding was determined in the presence of 10 μM propranolol. Assays were incubated for 1 hour at 37°C, and then binding reactions were terminated by rapid filtration over GF/B for the β[ and β2 receptors or GF/C glass fiber filter plates for the β3 receptors (Packard BioScience Co., Meriden, CT) presoaked in 0.3% polyethyleneimine. Filter plates were washed three times with filtration buffer (75 mM Tris/HCl pH 7.4 at 4°C, 12.5 mM MgCl2, 1 mM EDTA) to remove unbound radioactivity. The plates were then dried and 50 μL of Microscint-20 liquid scintillation fluid (Packard BioScience Co., Meriden, CT) was added and plates were counted in a Packard Topcount liquid scintillation counter (Packard BioScience Co., Meriden, CT). Binding data were analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the 3 -parameter model for one-site competition. The curve minimum was fixed to the value for nonspecific binding, as determined in the presence of 10 μM propranolol. K,- values for test compounds were calculated from observed IC50 values and the I value ofthe radioligand using the Cheng-Prusoff equation (Cheng Y, and Prusoff WH., Biochemical Pharmacology, 1973, 22, 23, 3099-108).
In this assay, a lower K; value indicates that a test compound has a higher binding affinity for the receptor tested. Exemplified compound of this invention that were tested in this assay typically were found to have a K; value of less than about 300 nM for the β2 adrenergic receptor. For example, the compounds of Examples 3 and 6 were found to have Kj values of less than 10 nM.
If desired, the receptor subtype selectivity for a test compound can be calculated as the ratio of Ki(βι)/K;(β2) or Ki(β3)/K;(β2). Typically, compounds of this invention demonstrated greater binding at the β2 adrenergic receptor compared to the βi or β3 adrenergic receptor, i.e. Kj(β or K;(β3) is typically greater than Kj(β2). Generally, compounds having selectivity for the β2 adrenergic receptor over the βi or β3 adrenergic receptors are preferred; especially compounds having a selectivity greater than about 5; and in particular, greater than about 8. By way of example, the compounds of Examples 3 and 6 had a ratio of Ki(βι)/Ki(β2) greater than 8.
Assay Test Procedure B
Radioligand Binding Assay for Muscarinic Receptors
Radioligand binding assays for cloned human muscarinic receptors were performed in 96-well microtiter plates in a total assay volume of 100 μL. CHO cell membranes stably expressing either the hMi, hM2, hM3, hM4 or hM5 muscarinic subtype were diluted in assay buffer to the following specific target protein concentrations (μg/well): 10 μg for hMi, 10-15 μg for hM2, 10-20 μg for hM3, 10-20 μg for hM4, and 10-12 μg for hM5 to get similar signals (cpm). The membranes were briefly homogenized using a Polytron tissue disruptor (10 seconds) prior to assay plate addition. Saturation binding studies for determining KD values ofthe radioligand were performed using L-[N-methyl- 3H]scoρolamine 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 GF/B 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. The 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 KD value ofthe radioligand using the Cheng-Prusoff equation (Cheng Y; PrusoffWH. (1973) Biochemical Pharmacology, 22(23):3099-108). K; values were converted to pKj values to determine the geometric mean and 95% confidence intervals. These summary statistics were then converted back to Kj 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. Exemplified compound of this invention that were tested in this assay typically were found to have a K,- value of less than about 300 nM for the M3 muscarinic receptor. For example, the compounds of Examples 3 and 6 were found to have Kj values of less than 10 nM.
Assay Test Procedure C
Whole-cell cAMP Flashplate Assay in CHO Cell Lines Heterologously Expressing
Human βi, β2 or β3 Adrenergic Receptors cAMP assays were performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with [ 125I]-cAMP (NEN SMP004,
PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturers instructions. For the determination of β receptor agonist potency (EC50), CHO-K1 cell lines stably expressing cloned human βi, β2 or β adrenergic receptors were grown to near confluency in HAM's F-12 media supplemented with 10% FBS and Geneticin (250 μg/mL). Cells were rinsed with PBS and detached in dPBS (Dulbecco's Phosphate Buffered Saline, without CaCl2and MgCl2) containing 2 mM EDTA or Trypsin-EDTA solution (0.05% tryρsin/0.53 mM EDTA). After counting cells in Coulter cell counter, cells were pelleted by centrifugation at 1,000 rpm and re-suspended in stimulation buffer containing IB MX (PerkinElmer Kit) pre-warmed to room temperature to a concentration of 1.6 x 106 to 2.8 x 106 cells/mL. About 60,000 to 80,000 cells per well were used in this assay. Test compounds (10 mM in DMSO) were diluted into PBS containing 0.1 % BSA in Beckman Biomek-2000 and tested at 11 different concentrations ranging from 100 μM to 1 pM. Reactions were incubated for 10 min at 37 °C and stopped by adding 100 μL of cold detection buffer containing [125I]-cAMP (NEN SMP004, PerkinElmer Life Sciences, Boston, MA). The amount of cAMP produced (pmol/well) was calculated based on the counts observed for the samples and cAMP standards as described in the manufacturer's user manual. Data were analyzed by nonlinear regression analysis with the GraphPad
Prism Software package (GraphPad Software, Inc., San Diego, CA) with the sigmoidal equation. The Cheng-Prusoff equation (Cheng Y, and Prusoff WH., Biochemical Pharmacology, 1973, 22, 23, 3099-108) was used to calculate the EC50 values.
In this assay, a lower EC50 value indicates that the test compound has a higher functional activity at the receptor tested. Exemplified compound of this invention that were tested in this assay typically were found to have a EC50 value of less than about 300 nM for the β2 adrenergic receptor. For example, the compounds of Examples 3 and 6 were found to have EC50 values of less than 10 nM.
If desired, the receptor subtype selectivity for a test compound can be calculated as the ratio of EC501)/EC502) or EC503)/EC5o(β2). Typically, compounds of this invention demonstrated greater functional activity at the β2 adrenergic receptor compared to the or β3 adrenergic receptor, i.e. EC50(βι) or EC503) is typically greater than EC502). Generally, compounds having selectivity for the β adrenergic receptor over the βj or β adrenergic receptors are preferred; especially compounds having a selectivity greater than about 5; and in particular, greater than about 10. By way of example, the compounds of Examples 3 and 6 had ratios of EC501)/EC5o(β2) greater than 10.
Assay Test Procedure D
Functional Assays of Antagonism for Muscarinic Receptor Subtypes
A. Blockade of Agonist-Mediated Inhibition of cAMP Accumulation In this assay, the functional potency of a test compound is determined by measuring the ability ofthe test compound to block oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-K1 cells expressing the hM2 receptor. cAMP assays are performed in a radioimmunoassay fonnat 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 50 mL 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 Inc., Wellesley, MA). The amount of cAMP 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 is 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 ofthe oxotremorine concentration-response curve and the oxotremorine assay concentration as the KD and [L], respectively.
In this assay, a lower K; value indicates that the test compound has a higher functional activity at the receptor tested. Exemplified compound of this invention that were tested in this assay typically were found to have a Kj value of less than about 300 nM for blockade of oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM2 receptor. For example, the compound of Example 3 was found to have a K; value of less than 10 nM.
B. Blockade of Agonist-Mediated [35S] GTPγS Binding
In a second functional assay, the functional potency of test compounds can be detennined by measuring the ability of the compounds to block oxotremorine-stimulated [35S]GTPγS_binding in CHO-Kl cells expressing the hM2 receptor.
At the time of use, frozen membranes were thawed and then diluted in assay buffer with a final target tissue concentration of 5-10 μg protein per well. The membranes were briefly homogenized using a Polytron PT-2100 tissue disrupter and then added to the assay plates.
The EC9o value (effective concentration for 90%> maximal response) for stimulation of [35S]GTPγS binding by the agonist oxotremorine was determined in each experiment. To determine the ability of a test compound to inhibit oxotremorine-stimulated [35S]GTPγS binding, the following was added to each well of 96 well plates: 25 μL of assay buffer with [35S]GTPγS (0.4nM), 25 μL of oxotremorine(EC90) and GDP (3uM), 25 μL of diluted test compound and 25 μL CHO cell membranes expressing the hM2 receptor. The assay plates were then incubated at 37 °C for 60 minutes. The assay plates were filtered over 1% BSA-pretreated GF/B filters using a PerkinElmer 96-well harvester. The plates were rinsed with ice-cold wash buffer for 3 x 3 seconds and then air or vacuum dried. Microscint-20 scintillation liquid (50 μL) was added to each well, and each plate was sealed and radioactivity counted on a Topcounter (PerkinElmer). Data were 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 was used to calculate the Kj, using the IC50 values ofthe concentration-response curve for the test compound and the oxotremorine concentration in the assay as the Kp and [L], ligand concentration, respectively. In this assay, a lower K; value indicates that the test compound has a higher functional activity at the receptor tested. Exemplified compound of this invention that were tested in this assay typically were found to have a K; value of less than about 300 nM for blockade of oxotremorine-stimulated [35S]GTPγS_binding in CHO-Kl cells expressing the hM2 receptor. For example, the compound of Example 3 was found to have a K; value of less than 10 nM.
C. Blockade of Agonist-Mediated Calcium Release via FLIPR Assays
Muscarinic receptor subtypes (Mi, 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), which 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 Mj 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 hMi, hM3 and cMs 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 EC90 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)Λ1/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 K; values are determined by Prism using the oxotremorine EC50 value as the KD and the oxotremorine EC o for the ligand concentration according to the Cheng-Prusoff equation (Cheng & Prusoff, 1973).
In this assay, a lower Kj value indicates that the test compound has a higher functional activity at the receptor tested. Exemplified compound of this invention that were tested in this assay typically were found to have a K; value of less than about 300 nM for blockade of agonist-mediated calcium release in CHO cells stably expressing the hMls hM3 and cM5 receptors. For example, the compound of Example 3 was found to have a K; value of less than 10 nM for the hMls hM3 and cM5 receptors.
Assay Test Procedure E
Whole-cell cAMP Flashplate Assay With a Lung Epithelial Cell Line Endogenously Expressing Human β2 Adrenergic Receptor
For the determination of agonist potencies and efficacies (intrinsic activities) in a cell line expressing endogenous levels ofthe β2 adrenergic receptor, a human lung epithelial cell line (BEAS 2B) was used (ATCC CRL-9609, American Type Culture Collection, Manassas, NA) (January B, et al, British Journal of Pharmacology, 1998, 123, 4, 701-11). Cells were grown to 75-90% confluency in complete, serum- free medium (LHC-9 MEDIUM containing Epinephrine and Retinoic Acid, cat # 181-500, Biosource International, Camarillo, CA). The day before the assay, medium was switched to LHC-8 (no epinephrine or retinoic acid, cat # 141-500, Biosource International, Camarillo, CA). cAMP assays were performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with [125I]-cAMP (ΝEΝ SMP004,
PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturers instructions. On the day ofthe assay, cells were rinsed with PBS, lifted by scraping with 5mM EDTA in PBS, and counted. Cells were pelleted by centrifugation at 1,000 rpm and re-suspended in stimulation buffer pre-warmed to 37 °C at a final concentration of 600,000 cells/mL. Cells were used at a final concentration of 100,000 to 120,000 cells/well in this assay. Test compounds were serially diluted into assay buffer (75 mM Tris/HCl pH 7.4 at 25°C, 12.5 mM MgCl2, 1 mM EDTA, 0.2% BSA) in Beckman Biomek-2000. Test compounds were tested in the assay at 11 different concentrations, ranging from 10 μM to 10 pM. Reactions were incubated for 10 min at 37 °C and stopped by addition of 100 μL of ice-cold detection buffer. Plates were sealed, incubated over night at 4 °C and counted the next morning in a Topcount scintillation counter (Packard BioScience Co., Meriden, CT). The amount of cAMP produced per mL of reaction was calculated based on the counts observed for samples and cAMP standards, as described in the manufacturer's user manual. Data were analyzed by nonlinear regression analysis with the GraphPad Prism Software package (GraphPad Software, Inc., San Diego, CA) using the 4-parameter model for sigmoidal dose-response.
In this assay, a lower EC50 value indicates that the test compound has a higher functional activity at the receptor tested. Exemplified compound of this invention that were tested in this assay typically were found to have a EC50 value of less than about 300 nM for the β2 adrenergic receptor. For example, the compounds of Examples 3 and 6 were found to have EC50 values of less than 10 nM.
If desired, test compound efficacy (%>Eff) was calculated from the ratio ofthe observed Emax (TOP ofthe fitted curve) and the maximal response obtained for isoproterenol dose response curve and was expressed as %Eff relative to isoproterenol. Exemplified compounds of this invention tested in this assay typically demonstrated a %Eff greater than about 40.
Assay Test Procedure F
Duration of Bronchoprotection In Guinea Pig Models of Acetylcholine-induced or Histamine-Induced Bronchoconstriction
These in vivo assays were used to assess the bronchoprotective effects of test compounds exhibiting both muscarinic receptor antagonist and β2 adrenergic receptor agonist activity. To isolate muscarinic antagonist activity in the acetylcholine-induced bronchoconstriction model, the animals were administered propanolol, a compound that blocks β receptor activity, prior to the administration of acetylcholine. Duration of bronchoprotection in the histamine-induced bronchoconstriction model reflects β2 adrenergic receptor agonist activity. Groups of 6 male guinea pigs (Duncan-Hartley (HsdPoc:DH) Harlan,
Madison, WI) weighing between 250 and 350 g were individually identified by cage cards. Throughout the study, animals were allowed access to food and water ad libitum. Test compounds were administered via inhalation over 10 minutes in a whole-body exposure dosing chamber (R&S Molds, San Carlos, CA). The dosing chambers were ananged so that an aerosol was simultaneously delivered to 6 individual chambers from a central manifold. Guinea pigs were exposed to an aerosol of a test compound or vehicle (WFI). These aerosols were generated from aqueous solutions using an LC Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, NA) driven by a mixture of gases (CO2 = 5%, O2 = 21 % and Ν2 = 74%) at a pressure of 22 psi. The gas flow through the nebulizer at this operating pressure was approximately 3 L/ minute. The generated aerosols were driven into the chambers by positive pressure. No dilution air was used during the delivery of aerosolized solutions. During the 10 minute nebulization, approximately 1.8 mL of solution was nebulized. This value was measured gravimetrically by comparing pre-and post-nebulization weights ofthe filled nebulizer.
The bronchoprotective effects of test compounds administered via inhalation were evaluated using whole body plefhysmography at 1.5, 24, 48 and 72 hours post-dose. Forty-five minutes prior to the start ofthe pulmonary evaluation, each guinea pig was 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 was shaved and cleaned with 70% alcohol, a 2-3 cm midline incision ofthe ventral aspect ofthe neck was made. Then, the jugular vein was isolated and cannulated with a saline-filled polyethylene catheter (PE-50, Becton Dickinson, Sparks, MD) to allow for intravenous infusions of acetylcholine (Ach) or histamine in saline. The trachea was then dissected free and cannulated with a 14G teflon tube (#NE- 014, Small Parts, Miami Lakes, FL). If required, anesthesia was maintained by additional intramuscular injections ofthe aforementioned anesthetic mixture. The depth of anesthesia was monitored and adjusted if the animal responds to pinching of its paw or if the respiration rate was greater than 100 breaths/minute.
Once the cannulations were completed, the animal was placed into a plethysmograph (#PLY3114, Buxco Electronics, Inc., Sharon, CT) and an esophageal pressure cannula (PE-160, Becton Dickinson, Sparks, MD) was inserted to measure pulmonary driving pressure (pressure). The teflon tracheal tube was attached to the opening ofthe plethysmograph to allow the guinea pig to breathe room air from outside the chamber. The chamber was then sealed. A heating lamp was used to maintain body temperature and the guinea pig's lungs were 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 did not collapse and that the animal did not suffer from hyperventilation.
Once it was determined that baseline values were within the range of 0.3 - 0.9 mL/cm H O for compliance and within the range of 0.1 - 0.199 cm H2O/mL per second for resistance, the pulmonary evaluation was initiated. A Buxco pulmonary measurement computer progam enabled the collection and derivation of pulmonary values.
Starting this program initiated the experimental protocol and data collection. The changes in volume over time that occur within the plethysmograph with each breath were measured via a Buxco pressure transducer. By integrating this signal over time, a measurement of flow was calculated for each breath. This signal, together with the pulmonary driving pressure changes, which were collected using a Sensym pressure transducer (#TRD4100), was connected via a Buxco (MAX 2270) preamplifier to a data collection interface (#'s SFT3400 and SFT3813). All other pulmonary parameters were derived from these two inputs. Baseline values were collected for 5 minutes, after which time the guinea pigs were challenged with Ach or histamine. When evaluating the muscarinic antagonist effects, propanolol (5 mg/Kg, iv) (Sigma- Aldrich, St. Louis, MO) was administered 15 minutes prior to challenge with Ach. Ach (Sigma- Aldrich, St. Louis, MO) (0.1 mg/mL) was 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 ofthe 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. Alternatively, bronchoprotection of test compounds was assessed in the acetylcholine challenge model without pretreatment with a beta blocking compound. When evaluating the β2 adrenergic receptor agonist effects of test compounds, histamine (25 μg/mL) (Sigma- Aldrich, St. Louis, MO) was infused intravenously for 1 minute from a syringe pump at the following doses and prescribed times from the start of the experiment: 0.5 μg/minute at 5 minutes, 0.9 μg/minute at 10 minutes, 1.9 μg/minute at 15 minutes, 3.8 μg/minute at 20 minutes, 7.5 μg/minute at 25 minutes and 15 μg/minute at 30 minutes. If resistance or compliance had not returned to baseline values at 3 minutes following each Ach or histamine dose, the guinea pig's lungs were inflated 3 times with 4 mL of air from a 10 mL calibration syringe. Recorded pulmonary parameters include respiration frequency (breaths/minute), compliance (mL/cm H2O) and pulmonary resistance (cm H2O/ mL per second). Once the pulmonary function measurements were completed at minute 35 of this protocol, the guinea pig was removed from the plethysmograph and euthanized by carbon dioxide asphyxiation. The data were evaluated in one of two ways: (a) Pulmonary resistance (RL, cm H2O/mL per second) was calculated from the ratio of "change in pressure" to "the change in flow." The RL response to ACh (60 μg/min, IH) was computed for the vehicle and the test compound groups. The mean ACh response in vehicle-treated animals, at each pre-treatment time, was calculated and used to compute % inhibition of ACh response, at the conesponding pre-treatment time, at each test compound dose. Inhibition dose-response curves for 'RL' were fitted with a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) to estimate bronchoprotective ID50 (dose required to inhibit the ACh (60 μg/min) bronchocontrictor response by 50%). The equation used was as follows:
Y = Min + (Max-Min)/(1 + 10 ((log ID5°-χ)* Hillslope) )
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 ofthe baseline pulmonary resistance, was 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 (derived from the equation used to calculate PC20 values in the clinic (see^w. Thoracic Soc, 2000):
pn n n r + (log - log C ^Ro - R
PD2= antilog log Cι+
L R2 - R1
where: Ci = concentration of Ach or histamine preceding C2
C2 = concentration of Ach or histamine resulting in at least a 2-fold increase in pulmonary resistance (RJ R0 = Baseline RL value
Rj = RL value after Cj
R2 = RL value after C2
Statistical analysis ofthe data was performed using a two tailed - Students t-test. A P-value <0.05 was considered significant.
Exemplified compounds of this invention that were tested in this assay typically produced a dose-dependent bronchoprotective effect against MCh-induced bronchoconstriction and His-induced bronchoconstriction. Generally, test compounds having a potency (ID50 at 1.5 h post-dose) of less than about 300 μg/mL for ACh-induced bronchoconstriction and less than about 300 μg/mL for His-induced bronchoconstriction in this assay are generally prefened. For example, the compounds of Examples 3 and 6 were found to have an ID50 less than about 100 μg/mL for ACh-induced bronchoconstriction and an ID50 less than about 100 μg/mL for His-induced bronchoconstriction at 1.5 hours post- dose. Additionally, test compounds having a duration (PD T^2) of brochoprotective activity of at least about 24 hours in this assay are generally prefened. By way of example, the compounds of Examples 3 and 6 were found to have a PD Tj/2 of at least about 24 hours post-dose.
Assay Test Procedure G
Einthoven Model for Measuring Changes in Ventilation in Guinea Pigs
The bronchodilator activity of test compounds was evaluated in an anesthetized guinea pig model (the Einthoven model), which uses ventilation pressure as a surrogate measure of airway resistance. See, for example, Einthoven (1892) Pfugers Arch. 51: 367 - 445; and Mohammed et al. (2000) Pulm Pharmacol Ther.13(6) ;287-92. In this model, muscarinic antagonist and β2 agonist activity was assessed by determining the protective effects against methacholine (MCh) and histamine (His)-induced bronchoconstriction.
This assay was conducted using Duncan-Hartley guinea pigs (Harlan, Indianapolis, IN), weighing between 300 and 400 g. The test compound or vehicle (i.e., sterile water) was dosed by inhalation (IH) over a 10 minute time period in a whole body exposure dosing chamber (R+S Molds, San Carlos, CA) using 5 mL of dosing solution. Animals were exposed to an aerosol, which was generated from an LC Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, NA) driven by Bioblend a mixture of gasses (5% CO2; 21% O2; and 74% Ν2) at a pressure of 22 psi. Pulmonary function was evaluated at various time-points after inhalation dosing. Forty five minutes prior to the start of pulmonary function evaluation, the guinea pigs were anesthetized with an intramuscular (IM) injection of a mixture of ketamine (13.7 mg/kg/xylazine (3.5 mg/kg)/acepromazine (1.05 mg/kg). A supplemental dose of this mixture (50% of initial dose) was administered as needed. The jugular vein and carotid artery were isolated and cannulated with saline-filled polyethylene catheters (micro- renathane and PE-50, respectively, Beckton Dickinson, Sparks, MD). The carotid artery was connected to a pressure transducer to allow the measurement of blood pressure and the jugular vein cannula was used for IV injection of either MCh or His. The trachea was then dissected free and cannulated with a 14G needle (#NE-014, Small Parts, Miami Lakes, FL). Once the cannulations were complete, the guinea pigs were ventilated using a respirator (Model 683, Harvard Apparatus, Inc., MA) set at a stroke volume of lmL/100 g body weight but not exceeding 2.5 mL volume, and at a rate of 100 strokes per minute. Ventilation pressure (VP) was measured in the tracheal cannula using a Biopac transducer that was connected to a Biopac (TSD 137C) pre-amplifier. Body temperature was maintained at 37 °C using a heating pad. Prior to initiating data collection, pentobarbital (25mg/kg) was administered intraperitoneally (IP) to suppress spontaneous breathing and obtain a stable baseline. The changes in VP were recorded on a Biopac Windows data collection interface. Baseline values were collected for at least 5 minutes, after which time guinea pigs were challenged IV non-cumulatively with 2-fold incremental doses ofthe bronchoconstrictor (MCh or His). When MCh was used as the bronchoconstrictor agent, animals were pre-treated with propranolol (5 mg/kg, IV) to isolate the antimuscarinic effects ofthe test compound. Changes in VP were recorded using the Acknowledge Data Collection Software (Santa Barbara, CA). After the completion of study, the animals were euthanized.
Change in VP was measured in cm of water. Change in VP (cm H2O) = peak pressure (after bronchoconstrictor challenge) - peak baseline pressure. The dose-response curve to MCh or His was fitted to a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) The equation used was as follows: Y = Min + (Max-Min)/(1 + 10 ((lo ID5°-χ)* Hil,slo e) )
where X is the logarithm of dose, Y is the response. Y starts at Min and approaches asymptotically to Max with a sigmoidal shape.
The percent inhibition ofthe bronchoconstrictor response to a submaximal dose of MCh or His was calculated at each dose ofthe test compound using the following equation: % Inhibition of response = 100-((peak pressure (after bronchoconstrictor challenge, treated) - peak baseline pressure (treated) * 100%> / (peak pressure (after bronchoconstrictor challenge, water) - peak baseline pressure (water)). Inhibition curves were fitted using the four parameter logistic equation from GraphPad software. ID50 (dose required to produce 50% inhibition ofthe bronchoconstrictor response) and Emax (maximal inhibition) were also estimated wherever appropriate.
The magnitude of bronchoprotection at different time-points after inhalation ofthe test compound was used to estimate the pharmacodynamic half-life (PD T^). PD Tι/2 was determined using a non-linear regression fit using a one-phase exponential decay equation (GraphPad Prism, Version 4.00): Y=Span*exp(-K*X) + Plateau; Starts at Span+Plateau and decays to Plateau with a rate constant K. The PD T[/2= 0.69/K. Plateau was constrained to 0. Exemplified compounds of this invention that were tested in this assay typically produced a dose-dependent bronchoprotective effect against MCh-induced bronchoconstriction and His-induced bronchoconstriction. Generally, test compounds having an ID50 less than about 300 μg/mL for MCh-induced bronchoconstriction and an ID50 less than about 300 μg/mL for His-induced bronchoconstriction at 1.5 hours post-dose in this assay are preferred. Additionally, test compounds having a duration (PD Tι/2) of brochoprotective activity of at least about 24 hours in this assay are generally preferred.
Assay Test Procedure H
Inhalation Guinea Pig Salivation Assay Guinea pigs (Charles River, Wilmington, MA) weighing 200-350 g were acclimated to the in-house guinea pig colony for at least 3 days following arrival. Test compound or vehicle were dosed via inhalation (IH) over a 10 minute time period in a pie shaped dosing chamber (R+S Molds, San Carlos, CA). Test solutions were dissolved in sterile water and delivered using a nebulizer filled with 5.0 mL of dosing solution. Guinea pigs were restrained in the inhalation chamber for 30 minutes. During this time, guinea pigs were restricted to an area of approximately 110 sq. cm. This space was adequate for the animals to turn freely, reposition themselves, and allow for grooming. Following 20 minutes of acclimation, guinea pigs were 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 were evaluated at 1.5, 6, 12, 24, 48, or 72 hrs after treatment.
Guinea pigs were anesthetized one hour before testing with an intramuscular (IM) injection of a mixture of ketamine 43.75 mgkg, xylazine 3.5 mg/kg, and acepromazine 1.05 mg/kg at an 0.88 mL/kg volume. Animals were 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) was inserted in the guinea pig's mouth. Five minutes later, the muscarinic agonist pilocarpine (3.0 mg/kg, s.c.) was administered and the gauze pad was immediately discarded and replaced by a new pre- weighed gauze pad. Saliva was collected for 10 minutes, at which point the gauze pad was 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 was calculated. The vehicle group mean was considered to be 100%> salivation. Results were calculated using result means (n = 3 or greater). Confidence intervals (95%) were calculated for each dose at each time point using two-way ANOVA. This model is a modified version ofthe procedure described in Rechter, "Estimation of anticholinergic drug effects in mice by antagonism against pilocarpine-induced salivation" Ata Pharmacol Toxicol, 1996, 24:243-254.
The mean weight of saliva in vehicle-treated animals, at each pre-treatment time, was calculated and used to compute % inhibition of salivation, at the conesponding pre- treatment time, at each dose. The inhibition dose-response data were fitted to a a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) to estimate anti-sialagogue ID50 (dose required to inhibit 50%) of pilocarpine-evoked salivation). The equation used was as follows:
Y = Min + (Max-Min)/(1 + 10 ((log ID50~ )* Hillslope) ) 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 ofthe anti-sialagogue ID50 to bronchoprotective ID50 was used to compute the apparent lung-selectivity index ofthe test compound. Generally, compounds having an apparent lung-selectivity index greater than about 5 are preferred. In this assay, the compound of Example 3 had an apparent lung-selectivity index greater than 5.
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 ofthe 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

WHAT IS CLAIMED IS:
1. A compound of fonnula I:
Figure imgf000176_0001
I wherein: a is 0 or an integer of from 1 to 3; each R1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -ORla, -C(O)ORl , -SRlc, -S(O)Rld, -S(O)2Rle and -NRIfRIg; each of Rla, Rlb, Rlc, Rld, Rle, Rlf and Rl is independently hydrogen, (l-4C)alkyl or phenyl-(l-4C)alkyl; b is 0 or an integer of from 1 to 3; each R2 is independently selected from ( 1 -4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl,
(3-6C)cycloalkyl, cyano, halo, -OR2a, -C(O)OR2b, -SR2°, -S(O)R2d, -S(O)2R2e and -NR2fR2g; each of R2a, R2b, R2c, R2d, R2e, R2f and R2g is independently hydrogen, (l-4C)alkyl or phenyl-(l-4C)alkyl; W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring and represents O or NWa; Wa is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R3 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR3a, -C(O)OR3b, -SR3°, -S(O)R3d, -S(O)2R3e and
-NR3fR3g; or two R3 groups are joined to form (l-3C)alkylene, (2-3C)alkenylene or oxiran- 2,3-diyl; each of R3a, R3b, R3c, R3d, R3e, R3f and R3g is independently hydrogen or (1- 4C)alkyl;
R4 is a divalent group ofthe formula:
-(R4 -(A%^R4b)t-Q-(R -(A -(R4 -
wherein d, e, f, g, h and i are each independently selected from 0 and 1 ; R4a, R4b, R4c and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2- 10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl-(l-4C)alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, -O-(6-10C)arylene, (6-10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy; Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Q°)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q , -N(O')C(O)O- and -N(Qk);
Qa, Qb, Qc, Qd, Qe, Qf, Qg, Qh, Q!, Q1' and Qk are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 16;
R5 represents hydrogen or (l-4C)alkyl;
R6 is -NR6aCR6b(O) or -CR6oR6dOR6e and R7 is hydrogen; or R6 and R7 together form -NR7aC(O)-CR7b=CR7c- , -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7i-CR7jR7k- or - CR71R7m-CR7nR70- C(O) -NR7p-; each of R6a, R6b, R6c, R6d and R6e is independently hydrogen or (l-4C)alkyl; and each of R7a, R7b, R7c, R7d, R7e, R7f, R7g, R7h, R7i, R7j, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or (l-4C)alkyl; or a phannaceutically acceptable salt or solvate or stereoisomer thereof.
2. The compound of Claim 1 , wherein the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of
Figure imgf000178_0001
3. The compound of Claim 2, wherein the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is 8, 9, 10 or 11.
4. The compound of any one of Claims 1 to 3, wherein W is attached to the piperidine ring at the 4-position with respect to the nitrogen atom.
5. The compound of any one of Claims 1 to 4, wherein a, b, and c are each 0, and Wa and R5 are both hydrogen.
6. The compound of any one of Claims 1 to 5, wherein W represents O.
7. The compound of any one of Claims 1 to 6, wherein R6 is -NHCHO or - CH2OH and R7 is hydrogen; or R6 and R7 together form -NHC(O)-CH=CH-, -CH=CH- C(O)-NH-, -CH2-CH2-C(O)NH- or -NHC(O)-CH2-CH2-.
8. The compound of any one of Claims 1 to 7, wherein R4 is a divalent group ofthe formula: -(R4a)d- where R4a is (4-10C)alkylene.
9. The compound of Claim 8, wherein R4 is -(CH2)8-, -(CH2)9, and -(CH2) io-
10. The compound of any one of Claims 1 to 7, wherein R4 is a divalent group ofthe formula:
-(R4a)d-(A2)h-(R4d)i-
wherein R4a is (l-lOC)alkylene; A2 is (6-10C)arylene or (2-9C)heteroarylene; and R4d is (l-10C)alkylene.
11. The compound of any one of Claims 1 to 7, wherein R4 is a divalent group ofthe formula:
-(R4a)d-Q-(A2)h-(R4d)i-
wherein Q is -O- or -N(Qk)-; Qk is hydrogen or (l-3C)alkyl; R4a is (1- 10C)alkylene; A2 is (6-10C)arylene or (2-9C)heteroarylene; and R4d is (l-lOC)alkylene.
12. The compound of any one of Claims 1 to 7, wherein Q is -N(Qa)C(O)- or -C(O)N(Qb)-.
13. The compound of Claim 12, wherein R4 is selected from:
— (CH2)m-C-N— (CH2)n
wherein m is an integer from 2 to 10; and n is an integer from 2 to 10; provided that m + n is an integer from 4 to 12;
Figure imgf000179_0001
wherein o is an integer from 2 to 7; and p is an integer from 1 to 6; provided that o + p is an integer from 3 to 8; and wherein the phen-l,4-ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
Figure imgf000180_0001
wherein q is an integer from 2 to 6; r is an integer from 1 to 5; and s is an integer from 1 to 5; provided that q + r + s is an integer from 4 to 8; and wherein the phen- 1,4- ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l- 4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
Figure imgf000180_0002
wherein t is an integer from 2 to 10; and u is an integer from 2 to 10; provided that t + u is an integer from 4 to 12;
Figure imgf000180_0003
wherein v is an integer from 2 to 7; and w is an integer from 1 to 6; provided that v + w is an integer from 3 to 8; and wherein the phen-l,4-ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy; and
Figure imgf000181_0001
wherein x is an integer from 2 to 6; y is an integer from 1 to 5; and z is an integer from 1 to 5; provided that x + y + z is an integer from 4 to 8; and wherein the phen- 1,4- ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l- 4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy.
14. A compound of formula II:
Figure imgf000181_0002
II wherein
W represents O or NWa;
Wa is hydrogen or (l-4C)alkyl;
R4 is a divalent group ofthe formula:
-(R (A')r(R ( -(A (R4
wherein d, e, f, g, h and i are each independently selected from 0 and 1;
R4a, R4b, R4c and R4d are each independently selected from (l-lOC)alkylene, (2-
10C)alkenylene and (2-10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl-(l-4C)alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, -O-(6-10C)arylene, (6-10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroaryl ene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy; Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Q°)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q , ^( ^)0(0)0- and -N(Qk);
QΛ Q , Q°, θΛ Qe, Qf, Q8, Qh, Q\ and Qk are each independently selected from hydrogen, ( 1 -6C)alkyl, A3 and ( 1 -4C)alkylene- A4, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 14; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
15. A compound of formula III :
Figure imgf000183_0001
III
wherein
W represents O or NWa;
Wa is hydrogen or (l-4C)alkyl;
R4 is a divalent group ofthe formula:
-(RV(A')e -(R b)r-Q- (R4c)g-(A2)h-(R4d)i-
wherein d, e, f, g, h and i are each independently selected from 0 and 1 ; R4a, R4b, R4c and R4d are each independently selected from (1-10C)alkylene, (2-
10C)alkenylene and (2-10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl-(l-4C)alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 1 OQarylene, -O-(6- 10C)arylene, (6- 10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-, -N(Qa)C(O)-, -C(O)N(Q )-, -N(Qc)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q , -N(Q,')C(O)O- and -N(Qk);
Qa, Qb, Q°, Qd, Qe, Qf, Qg, Qh, Q1, Q> and Qk are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4 or R4c to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 14; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof
16. A compound of formula IV :
Figure imgf000184_0001
IV
wherein
W represents O or NW ;
Wa is hydrogen or (l-4C)alkyl; R4 is a divalent group ofthe formula:
RV )e )r(HR ( 2HR d)i-
wherein d, e, f, g, h and i are each independently selected from 0 and 1 ; R4a, R4b, R4c and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from ( 1 -4C)alkyl, fluoro, hydroxy, phenyl and phenyl-( 1 -4C)alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, -O-(6-10C)arylene, (6-10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl,
-S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy; Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-,
-N(Qa)C(O)-, -C(O)N(Qb)-, -N(Qc)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Q )S(O)2N(Qh)-, -OC(O)N(Q , -N(Qi)C(O)O- and -N(Qk);
Qa, Qb, Q°, Qd, Qe, Qf, Qg, Qh, Q and Qk are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4b or R4° to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 14; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
17. The compound of any one of Claims 14, 15 or 16, wherein the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 8 to 14.
18. The compound of any one of Claims 14, 15 or 16, wherein the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is 8, 9, 10 or 11.
19. The compound of any one of Claims 14, 15 or 16, wherein W represents O.
20. The compound of any one of Claims 14, 15 or 16, wherein R4 is a divalent group ofthe formula: -(R4a)d- where R4a is (4-10C)alkylene.
21. The compound of Claim 20, wherein R4 is -(CH2)8-, -(CH )9, and -(CH2)10-
22. The compound of any one of Claims 14, 15 or 16, wherein R4 is a divalent group ofthe formula:
-(R4V(A2)h- (R4d)i-
wherein R >4aa i •s ( 1 - 10C)alkylene; Az is (6- 10C)arylene or (2-9C)heteroarylene; and
R4d is (l-10C)alkylene.
23. The compound of any one of Claims 14, 15 or 16, wherein R4 is a divalent group ofthe formula:
-(R4a)d-Q-(A2)h-(R4d)i- wherein Q is -O- or -N(Qk)-; Qk is hydrogen or (l-3C)alkyl; R4a is (1-
10C)alkylene; A is (6-10C)arylene or (2-9C)heteroarylene; and R 4dQ is (l-lOC)alkylene
24. The compound of any one of Claims 14, 15 or 16, wherein Q is -N(Qa)C(O)- or -C(O)N(Qb)-.
25. The compound of Claim 24 wherein R is selected from:
O
II
(CH2 2),m -c- -N— (CH2)n-
wherein m is an integer from 2 to 10; and n is an integer from 2 to 10; provided that m + n is an integer from 4 to 12;
Figure imgf000187_0001
wherein o is an integer from 2 to 7; and p is an integer from 1 to 6; provided that o + p is an integer from 3 to 8; and wherein the phen-l,4-ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
-
Figure imgf000187_0002
wherein q is an integer from 2 to 6; r is an integer from 1 to 5; and s is an integer from 1 to 5; provided that q + r + s is an integer from 4 to 8; and wherein the phen- 1,4- ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l- 4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
Figure imgf000188_0001
wherein t is an integer from 2 to 10; and u is an integer from 2 to 10; provided that t + u is an integer from 4 to 12;
Figure imgf000188_0002
wherein v is an integer from 2 to 7; and w is an integer from 1 to 6; provided that v + w is an integer from 3 to 8; and wherein the phen-l,4-ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy; and
Figure imgf000188_0003
wherein x is an integer from 2 to 6; y is an integer from 1 to 5; and z is an integer from 1 to 5; provided that x + y + z is an integer from 4 to 8; and wherein the phen- 1 ,4- ylene group is optionally substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l- 4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy.
26. The compound of any one of Claims 14, 15 or 16, wherein R is selected from: -(CH2)7- -(CH2)8- -(CH2)9- -(CH2)10-;
-(CH2)n-;
-(CH2)2C(O)NH(CH2)5-;
-(CH2)2N(CH3)C(O)(CH2)5-; -(CH2)2C(O)NH(phen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(phen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)NH(CH2)5-;
-(CH2)3NHC(O)NH(CH2)5-;
-(CH2)2C(O)NHCH2(cyclohex- 1 ,3-ylene)CH2-; -(CH2)2NHC(O)( -cyclopent- 1 ,3-ylene)-;
-(CH2)2NHC(O)NH(phen- 1 ,4-ylene)(CH2)2-; l-[-(CH2)2C(O)](piperidin-4-yl)(CH2)2-;
-(CH2)2NHC(O)(trarø-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(c^-cyclopent- 1 ,3-ylene)-; -(CH2)2NH(phen- 1 ,4-ylene)(CH2)2-; l-[-(CH2)2NHC(O)](piperidin-4-yl)(CH2)2-;
-CH2(phen- 1 ,4-ylene)NH(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NHCH2(phen- 1 ,3-ylene)CH2-;
-(CH2)2C(O)NHCH2(pyrid-2,6-ylene)CH2-; -(CH2)2C(O)NH(c/5-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(trα/rø-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(cw-cyclopent- 1 ,3-ylene)CH2-;
-(CH2)2N(CH3)C(O)(phen- 1 ,3-ylene)CH2-;
-(CH2)2N(CH3)C(O)(trα^-cyclohex- 1 ,4-ylene)CH2-; -(CH2)2C(O)NH(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)C*H(CH3)- ((5)-isomer);
-(CH2)2C(O)NH(phen- 1 ,4-ylene)C*H(CH3)- ((R)-isomer);
2-[(S)-(-CH2-](pyrrolidin-l-yl)C(O)(CH2)4-; 2-[(S)-(-CH2-] (pyrrolidin- 1 -yl)C(O)(phen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(4-chloroρhen-l,3-ylene)CH2-;
-CH2(2-fluorophen-l,3-ylene)CH2-;
-(CH2)2C(O)NH(4-methylphen- 1 ,3-ylene)CH2-; -(CH2)2C(O)NH(6-chloroρhen- 1 ,3-ylene)CH2-;
-(CH2)2C(O)NH(2-chlorophen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(2,6-dichlorophen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)NHCH2(phen- 1 ,3-ylene)CH2-;
4-[-CH2-](piperidin-l-yl)C(O)(phen-l,4-ylene)CH2-; -(CH2)2C(O)N(CH2CH3)(phen- 1 ,4-ylene)CH2-; l-[-(CH2)2NHC(O)](piperidin-4-yl)-;
-(CH2)2C(O)NH(phen-l,4-ylene)(CH2)2-;
-(CH2)2NHC(O)(thien-2,5-ylene)CH2-;
-(CH2)2N(CH3)C(O)(3-nitrophen- 1 ,4-ylene)CH2-; -(CH2)2N(CH3)C(O)(tr rø-cyclohex- 1 ,4-ylene)-;
1 -[-CH2(2-fluoroρhen- 1 ,3-ylene)CH2](piperidin-4-yl)-;
5-[-(CH2)2NHC(O)](pyrid-2-yl)CH2-;
-(CH2)2(phen-l,4-ylene)(CH2)2-;
-(CH2)3(thien-2,5-ylene)(CH2)3-; -(CH2)2(phen- 1 ,4-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-CH2(phen-l,2-ylene)NH(phen-l,4-ylene)(CH2)2-; l-[-CH2(2-fluorophen-l,3-ylene)CH2](piperidin-4-yl)(CH2)2-;
1 -[-CH2(2-fluorophen- 1 ,3-ylene)CH2](piperidin-4-yl)CH2-;
-(CH2)2C(O)NH(3-chlorophen- 1 ,4-ylene)CH2-; -(CH2)2C(O)NH(2-(CF3O-)phen-l,4-ylene)CH2-;
-(CH2)3(phen- 1 ,3-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2S(O)2NH(CH2)5-;
-CH2(phen- 1 ,3-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(2-iodophen- 1 ,4-ylene)CH2-; -(CH2)2C(O)NH(2-chloro-5-methoxyphen-l,4-ylene)CH2-;
-(CH2)2C(O)NH(2-chloro-6-methylphen- 1 ,4-ylene)CH2-;
-(CH2)2C(O)NH(CH2)5-;
-(CH2)2N(CH3)S(O)2(phen- 1 ,4-ylene)CH2-; -(CH2)2C(O)NH(2-bromophen- 1 ,4-ylene)CH2-;
-(CH2)3(phen- 1 ,4-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)3(phen- 1 ,2-ylene)NH(phen- 1 ,4-ylene)(CH2)2-; l-[-CH2(2-fluorophen-l,3-ylene)CH2](piperidin-4-yl)(CH2)3-; -(CH2)2C(O)NH(2-methoxyphen- 1 ,4-ylene)CH2-;
-(CH2)5NH(phen-l,4-ylene)(CH2)2-;
4-[-(CH2)2-](piperidin- 1 -yl)(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(phen- 1 ,4-ylene)CH(CH3)CH2-;
-(CH2)2-(trarø-cyclohex- 1 ,4-ylene)NH(phen- 1 ,4-ylene)(CH2)2-; -(CH2)2C(O)NH(2-fluorophen- 1 ,4-ylene)CH2-;
-(CH2)2(phen- 1 ,3-ylene)NH(phen- 1 ,4-ylene)(CH2)2-;
-(CH2)2C(O)NH(2,5-difluorophen- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(phen-l,4-ylene)(CH2)2-; l-[-CH2(pyrid-2,6-ylene)CH2](piperidin-4-yl)CH2-; -(CH2)3NH(phen-l ,4-ylene)(CH2)2-;
-(CH2)2NH(naphth- 1 ,4-ylene)(CH2)2-;
-(CH2)3O(phen- 1 ,4-ylene)CH2-;
1 -[-(CH2)3](piperidin-4-yl)CH2-;
4-[-(CH2)2](piperidin-l-yl)C(O)(phen-l,4-ylene)CH2-; -(CH2)3(phen- 1 ,4-ylene)NHC(O)(CH2)2-;
-(CH2)3O(phen-l,4-ylene)(CH2)2-;
2-[-(CH2)2](benzimidazol-5-yl)CH2-;
-(CH2)2-(trα/rø-cyclohex- 1 ,4-ylene)NHC(O)(CH2)2-;
-(CH2)2-(tra/w-cyclohex- 1 ,4-ylene)NHC(O)(CH2)4-; -(CH2)2-(trarø-cyclohex- 1 ,4-ylene)NHC(O)(CH2)5-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)2-;
-(CH2)2NHC(O)NH(phen- 1 ,4-ylene)CH2-;
-(CH2)2N(CH3)(CH2)2(cw-cyclohex- 1 ,4-ylene)-;
-(CH2)2C(O)NH(2,3,5,6-tetrafluorophen- 1 ,4-ylene)CH2-; -(CH2)2C(O)NH(2,6-diiodophen- 1 ,4-ylene)CH2-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)3-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)4-;
4-[-(CH2)2](piperidin-l-yl)C(O)(CH2)5-; to t o on o o, x
Figure imgf000192_0001
-(CH2)2C(O)NH(phen- 1 ,4-ylene)CH2NHC(0)CH 12: -
-(CH2)2C(O)NH(phen- 1 ,4-ylene)(CH2)2NHC(O)CH2-;
-(CH2)2C(O)NHCH2(trαπs-cyclohex- 1 ,4-ylene)CH2-;
-(CH2)2NHC(O)(CH2)5-;
-(CH2)2O(phen- 1 ,3-ylene)O(CH2)2-;
-(CH2)2O(phen-l,2-ylene)O(CH2)2-;
-CH2(phen- 1 ,2-ylene)O(phen- 1 ,2-ylene)CH2-;
-(CH2)2C(O)NH(CH2)6-;
-(CH2)3(phen-l,4-ylene)(CH2)3-;
-(CH2)3(phen-l,4-ylene)(CH2)2-;
-(CH2)4(phen-l,4-ylene)(CH2)2-;
-(CH2)3(furan-2,5-ylene)(CH2)3-;
-(CH2)2N(CH3)C(O)NH(phen- 1 ,4-ylene)(CH2)2-;
4-[-(CH2)2] (piperidin- 1 -yl)C(O)NH(phen- 1 ,4-ylene)(CH2):
-(CH2)3(phen-l,3-ylene)(CH2)3-;
-(CH2)3(tetrahydrofuran-2,5-ylene)(CH2) -; and
-(CH2)2O(phen-l,4-ylene)C(O)(CH2)2-.
27. A compound of formula I:
Figure imgf000193_0001
I wherein: a is 0 or an integer of from 1 to 3; each R1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -ORIa, -C(O)ORlb, SRlc, -S(O)Rld, -S(O)2Rle, and -NRlfRlg; each of Rla, Rlb, Rlc, Rld, Rle, RIf and RI is independently hydrogen or (1- 4C)alkyl; b is 0 or an integer of from 1 to 3; each R2 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR2a, -C(O)OR2b, SR2c, -S(O)R2d, -S(O)2R2e, and -NR2fR2g; each of R2a, R2b, R2c, R2d, R2e, R2f and R2g is independently hydrogen or (1- 4C)alkyl;
W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring, and represents O or NWa;
Wa is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R3 is a substituent on carbon independently selected from (l-4C)alkyl, (2- 4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR3a, -C(O)OR3b, SR3°, -S(O)R3d, -S(O)2R3e, and -NR3fR3g; each of R3a, R3b, R3c, R3d, R3e, R3f and R3g is independently hydrogen or (1- 4C)alkyl;
R4 is a divalent group of the formula:
Figure imgf000194_0001
wherein d, e, f, g, h and i are each independently selected from 0 and 1 ;
R4a, R4b, R4c and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2- 10C)alkynylene wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl(l-4C)-alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, (2-9C)heteroarylene and (3-6C)heterocyclene; wherein each cycloalkyl ene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (1- 4C)alkyl and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Q°)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q and -N(Q')C(O)O-;
Qa, Qb, Qc, Qd, Qβ, Qf, Qg, Qh, Q1 and are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4; wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group;
A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl, (2-9C)heteroaryl and (3-6C)heterocyclyl; wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 8 to 14;
R5 represents hydrogen or (l-4C)alkyl;
R6 is -NR6aCR6b(O) and R7 is hydrogen, or R6 and R7 together form -NR7aC(O)-CR7b=CR7c-, -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7l-CR7jR7k- or - CR71R7m-CR7nR7o-C(O)-NR7p-; each of R6a and R6b is independently hydrogen or ( 1 -4C)alkyl; and each of R7a, R7b, R7c, R7d, R7e, R7f, R7g, R7h, R7', R7j, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or (l-4C)alkyl; or a phannaceutically acceptable salt or solvate or stereoisomer thereof.
28. A compound selected from:
1 -biphenyl-2-yl-3-( 1 - {9-[2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5- yl)ethylamino]nonyl}piperidin-4-yl)urea; biphenyl-2-ylcarbamic acid 1 - {9-[2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{9-[(R)-2-(3-formylamino-4-hydroxy-phenyl)-2- hydroxyethyl amino] nonyl } piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{9-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2,3,4- tetrahydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]pentylcarbamoyl} ethyl)piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 -[2-( {6-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]hexanoyl}methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- { [( )-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzoylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentyl}-ureido)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[3-(3- {5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]pentyl} -ureido)propyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-{9-[(R)-2-hydroxy-2-(4-hydroxy-3- hydroxymethylρhenyl)ethylamino]nonyl}piperidin-4-yl ester; 1 -biphenyl-2-yl-3-(l - {9-[2-hydroxy-2-(4-hydroxy-3- hydroxymethylphenyl)ethylamino]nonyl}piperidin-4-yl)urea; biphenyl-2-yl carbamic acid 1 - {2-[(3- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexylmethyl)carbamoyl]ethyl}piperidin-4- yl ester; biphenyl-2-ylcarbamic acid l-[2-({(li?,3>S)-3-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2,3,4-tetrahydroquinolin-5-yl)ethylamino]cyclopentanecarbonyl}amino)ethyl]piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2-chloro-4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]pentylsulfamoyl} -ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzenesulfonyl)methylamino]ethyl}piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(3-{[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}benzyl)ureido]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {3-[4-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)phenyl]propyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-fluoro-3-(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl}piperidin- 1 -ylmethyl)benzyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[3-(4- {2-[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenoxy)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino] ethyl} phenyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[3-(4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}piperidin- 1 -yl)-propyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(2-chloro-4- { [( ?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}-5-methoxyphenylcarbamoyl)ethyl]- piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {7- [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]heptyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{8-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]octyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[3-(4- {2-[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenyl)ureido]ethyl}piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-[3-(4-{2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethyl}piperidin-l-yl)-3-oxoρropyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexanecarbonyl)amino]ethyl}piperidin-4- yl ester; biphenyl-2-ylcarbamic acid l-[2-({(lR,3S)-3-[(Λ)-2-(3-formylamino-4- hydroxyphenyl)-2-hydroxyethylamino] -cyclopentanecarbonyl } amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {5-[(7?)-2-(3-formylamino-4-hydroxyphenyl)- 2-hydroxyethylamino]pentyl}ureido)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(7?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[3-(3- {5-[2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]pentyl}ureido)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- {2-[(β)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}piρeridine-l-carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[4-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}-phenylamino)benzyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzylcarbamoyl)ethyl]piperidin-4-yl ester;
3-[4-(3-biρhenyl-2-yl-ureido)piperidin-l-yl]-N-(4-{[(Λ)-2-hydroxy-2-(8-hydroxy- 2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenyl)propionamide; biphenyl-2-ylcarbamic acid 1 - {2-[(6- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-l)ethylamino]methyl}pyridin-2-ylmethyl)carbamoyl]ethyl}piperidin-4- yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({(li?,3ιS)-3-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]-cyclopentanecarbonyl} amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(3- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzoyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {2-[(4- { [(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexanecarbonyl)methylamino]- ethyl}piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(4- { [2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {(S)- 1 -[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]ethyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {(R)- 1 -[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]ethyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(( )-l-{5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentanoyl}pyrrolidin-2-ylmethyl)piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 -[(S)- 1 -(4- {[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzoyl)pyrrolidin-2-ylmethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(R)-2-(3-fonnylamino-4-hydroxy-phenyl)-2- hydroxy-ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{(i?)-l-[(R)-2-(3-formylamino-4-hydroxy- phenyl)-2-hydroxy-ethylamino]ethyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4-chloro-3- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester;
N- {2-[4-(3-biρhenyl-2-yl-ureido)-piρeridin- 1 -yl]ethyl} -4- {[(i?)-2-hydroxy-2-(8- hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl}benzamide;
1 -biphenyl-2-yl-3- { 1 -[3-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5 -yl)ethylamino] ethyl } piperidin- 1 -yl)-3 -oxo-propyl]piperidin-4-yl } urea; 3-[4-(3-biphenyl-2-yl-ureido)piperidin-l-yl]-N-(3-{[(i?)-2-hydroxy-2-(8-hydroxy-
2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl} -benzyl)propionamide; biphenyl-2-ylcarbanιic acid 1 -(2-fluoro-3- {[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl} -benzyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-4-methyl-phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2-chloro-5-{[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2,6-dichloro-4- {[(i?)-2-hydroxy-2-(8-hydroxy-2- oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[ 1 -(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}benzoyl)-piperidin-4-ylmethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-(3-formylamino-4-hydroxy-phenyl)-2- hydroxyethylamino]methyl}-benzoylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[ethyl-(4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl}phenyl)carbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(3- {4-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]piperidin-l-yl}-3-oxo-propyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(5- { [( ?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-thiophene-2-carbonyl)amino]ethyl}piperidin-4- yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- {[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroqumolin-5-yl)ethylamino]methyl}-3-nitro-benzoyl)methylamino]ethyl}piperidin-4- yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl}-cyclohexylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-({4-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]cyclohexanecarbonyl}-methylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2-fluoro-3- {4-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]piperidin- 1 -ylmethyl }benzyl)piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {2-[(6- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}pyridine-3-carbonyl)amino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[3-(5-{3-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]propyl}-thiophen-2-yl)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(i?)-2-(3-formylamino-4-hydroxyphenyι)- 2-hydroxy-ethylamino]ethyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)benzyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-fluoro-3-(4-{2-[(R)-2-hydroxy-2-(8-hydroxy-2- oxo-l,2-dihydroquinolin-5-yl)ethylamino]ethyl}piperidin-l-ylmethyl)benzyl]piperidin-4- yl ester; biphenyl-2-ylcarbamic acid l-[3-(4-{2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)propyl]piρeridin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-chloro-4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-2-trifluoromethoxy- phenylcarbamoyl)ethyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 - {3-[3-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)phenyl]propyl}piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-[3-(4-{2-[(5)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)benzyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4- {[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l ,2- dihydroquinolin-5-yl)ethylamino]methyl}-2-iodo-phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2-chloro-4- {[(R^-hydroxy^-^-hydroxy^-oxo- l^-dihydroquinolin-S-y^ethylaminojmethylJ-ό-methyφ^ 4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{5-[2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentylcarbamoyl } ethyl)-piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2-bromo-4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]ρiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {3-[2-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)-phenyl]propyl}ρiperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-fluoro-3-(4- {3-[(R)-2-hydroxy-2-(8-hydroxy-2- oxo-l,2-dihydroquinolin-5-yl)ethylamino]propyl}piperidin-l-ylmethyl)benzyl]piperidin-4- yl ester; biphenyl-2-yl carbamic acid 1 -[2-(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-2-methoxy-phenylcarbamoyl)ethyl]piperidin-4- yl ester; biphenyl-2-ylcarbamic acid 1 -[5-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino] ethyl } phenylamino)pentyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-{2-[l-(4-{2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenyl)-piperidin-4-yl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]-l-methyl-ethyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[4-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)cyclohexyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2-fluoro-4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl} phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[3-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)phenyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2,5-difluoro-4-{[(R)-2-hydroxy-2-(8-hydroxy-2- oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}-benzoylamino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[6-(4-{[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}piperidin-l-ylmethyl)pyridin-2- ylmethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}-naphthalen-l-ylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[ 1 -(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzoyl)piperidin-4-yl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[3-(4- {3-[(fl)-2-hydroxy-2-(8-hydroxy-2-oxo-l ,2- dihydroquinolin-5-yl)ethylamino]propionylamino}phenyl)propyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[3 -(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(5-{[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl} - 1 H-benzoimidazol-2-yl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{3-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]propionylamino } cyclohexyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]pentanoylamino}cyclohexyl)ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(4- {6-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]-hexanoylamino} cyclohexyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(l-{3-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]propionyl}piperidin-4-yl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(4-{[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}phenyl)ureido]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(2- {4-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]cyclohexyl}-ethyl)methylamino]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2,3,5,6-tetrafluoro-4- { [(R)-2-hydroxy-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)- ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - [2-(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl} -2,6-diiodo-phenylcarbamoyl)ethyl]piperidin-4- yl ester; biphenyl-2-ylcarbamic acid 1 -[2-( 1 - {4-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]-butyryl}piperidin-4-yl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(l - {5-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ρentanoyl}piperidin-4-yl)ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(l - {6-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]-hexanoyl}piperidin-4-yl)ethyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 -[2-(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-(3-formylamino-4-hydroxy-phenyl)-2- hydroxyethylamino]methyl}-benzylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {2-[3-(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzyl)ureido]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[3-(4-{[(.2?)-2-(3-formylamino-4-hydroxyphenyl)- 2-hydroxyethylamino]methyl}benzyl)-ureido]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl } -2-methyl-phenylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(3- {4-[2-(4- {[(7?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl}piperidin- 1 -yl)ethyl]phenoxy}propyl)- piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(3- {2-[(7?)-2-hydroxy-2-(8-hydroxy-2-oxo-l ,2- dihydroquinolin-5-yl)ethylamino]ethyl}-benzylcarbamoyl)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(3-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)ethyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 -(2- { [2-(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}ρhenoxy)acetyl]methylamino}ethyl)piperidin-4- yl ester; biρhenyl-2-ylcarbamic acid 1 -(2- {[2-(3- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)acetyl]methylamino}ethyl)-piperidin-4- yl ester; biphenyl-2-ylcarbamic acid l-{2-[(5-{[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}furan-2-carbonyl) methylamino] ethyl }piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-{2-[(5-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-thiophene-2-carbonyl)methylamino]ethyl}- piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino] ethoxy} phenoxy)ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {2-[4-(4- { [( ?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-benzoylamino)cyclohexyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[2-(2- { [(7?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)acetylamino]cyclohexyl} ethyl)- piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[2-(3- {[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)acetylamino]cyclohexyl}ethyl)- piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -(2- {4-[2-(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)acetylamino]cyclohexyl}ethyl)- piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {4-[(5- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}furan-2-carbonyl)amino]cyclohexyl}- ethyl)piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{4-[(5-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}-thiophene-2-carbonyl)amino]cyclohexyl}- ethyl)piperidin-4-yl ester; biρhenyl-2-ylcarbarnic acid 1 -(2- { 1 -[2-(2- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)acetyl]piperidin-4-yl} ethyl)- piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2 { 1 -[2-(3- {[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]mei thyl}phenoxy)acetyl]ρiperidin-4-yl}ethyl)- piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-(2- l-[2-(4-{[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]me' :hyl } phenoxy)acetyl]piperidin-4-yl } ethyl)- piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-{2- 1 -(5- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl furan-2-carbonyl) piρeridin-4-yl]ethyl}piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-{2- 1 -(5- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl -thiophene-2-carbonyl)piperidin-4- yl] ethyl } piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-{2- ;4-(3 - { [(i?)-2-hydroxy-2-(8 -hydroxy-2-oxo- 11,,2: - dihydroquinolin-5-yl)ethylamino]methyl -benzoylamino)phenyl]ethyl}piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-{2- 4-(4- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl -benzoylamino)phenyl] ethyl } piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-(2- 4-[2-(2- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]meι hyl}phenoxy)acetylamino]phenyl}ethyl)- piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-(2 4-[2-(3-{[(7?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]meι :hyl}phenoxy)acetylamino]phenyl}ethyl)- piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-(2 {4-[2-(4- { )-2-hydroxy-2-(8-hydroxy f--22--oxo- l,2-dihydroquinolin-5-yl)ethylamino]me thyl}phenoxy)acetylamino]phenyl}ethyl)- piperidin-4-yl ester; biphenyl-2-yl carbamic acid l-(2- 4-[(5- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl furan-2-carbonyl)amino]ρhenyl}ethyl)piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-(2- 4-[(5-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl -thiophene-2-carbonyl)amino]phenyl}- ethyl)piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-{2- 4-(3- {[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl -b enzoylamino)cyclohexyl] ethyl } piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[3-(3-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}ρhenoxy)-propyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-[2-hydroxy-3-(4-{2-[(R)-2-hydroxy-2-(8-hydroxy-2- oxo-l,2-dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[4-(4- {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino] ethyl} phenylamino)butyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 - {2-[4-( {2-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]acetylamino}-methyl)phenylcarbamoyl]ethyl}piperidin- 4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[4-(2- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]acetylamino} ethyl)phenylcarbamoyl]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[(4- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-cyclohexylmethyl)carbamoyl]ethyl}piperidin-4- yl ester; biphenyl-2-yl carbamic acid 1 -(2- {6-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]hexanoylamino } -ethyl)piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 -[2-(3- {2-[(ft)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethoxy}phenoxy)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[2-(2- {2-[(,S)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethoxy}phenoxy)ethyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(2- { [(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}phenoxy)benzyl]piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -(2- {6-[(R)-2 -hydro xy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]hexylcarbamoyl} ethyl) piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid l-[2-({(lR,3iS)-3-[(Λ)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]cyclopentanecarbonyl}amino)ethyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[3-(4-{3-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]propyl}phenyl)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[3-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenyl)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[4-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenyl)butyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[3-(5-{3-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]propyl } furan-2-yl)propyl]piperidin-4-yl ester; biρhenyl-2-ylcarbamic acid 1 - {2-[3-(4- {2-[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenyl)-l-methylureido]ethyl}piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {2-[ 1 -(4- {2-[(tf)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylcarbamoyl)piperidin-4-yl]ethyl}piperidin-4- yl ester; biphenyl-2-ylcarbamic acid 1 -[3-(3- {3-[(7?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]propyl}phenyl)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -[3-(5- {3-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]propyl} tetrahydro furan-2-yl)propyl]piperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]ethylcarbamoyl}phenoxy)ethyl]piperidin-4-yl ester; (5-bromobiphenyl-2-yl)carbamic acid 1 - {9-[2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl} -piperidin-4-yl ester;
(2'-fluorobiphenyl-2-yl)carbamic acid 1 - {9-[(ft)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]nonyl} -piperidin-4-yl ester;
(3'-chloro-3,5-difluorobiphenyl-2-yl)carbamic acid 1 - {9-[(i?)-2-hydroxy-2-(8- hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)-ethylamino]nonyl}piperidin-4-yl ester;
(3',5'-dichloro-3,5-difluorobiphenyl-2-yl)carbamic acid 1 - {9-[(R)-2- hydroxy-2-(8-hydroxy-2-oxo-l,2-dihydroquinolin-5-yl)-ethylamino]nonyl}piperidin-4-yl ester;
(3,5-difluorobiphenyl-2-yl)carbamic acid l-{9-[(i?)-2-hydroxy-2-(8-hydroxy-2- oxo- 1 ,2-dihydroquinolin-5-yl)ethylamino]nonyl}piperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -[2-(4- { [(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]methyl}-phenylcarbamoyl)ethyl]-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl}phenylcarbamoyl)-ethyl]-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl}-4-methylpiperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 - {9-[(-2?)-2-(3-formylamino-4-hydroxy-phenyl)-2- hydroxyethylamino]nonyl} -4-methylpiperidin-4-yl ester; biphenyl-2-yl carbamic acid 1 -(2- {5-[(ft)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]pentylcarbamoyl}-ethyl)-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {5-[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]pentylcarbamoyl} ethyl)-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 -(2- {6-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo-l ,2- dihydroquinolin-5-yl)ethylamino]hexanoylamino } ethyl)-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-(2-{6-[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]hexanoylamino } ethyl)-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)ethylamino]methyl}benzoylamino)ethyl]-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl}benzoylamino)ethyl]-4-methylpiperidin-4-yl ester; biphenyl-2-ylcarbamic acid 1 - {3-[4-(4- {2-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]ethyl}phenylamino)phenyl]propyl}-4-methylpiperidin-4- yl ester; biphenyl-2-ylcarbamic acid l-[2-(2-chloro-4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}phenylcarbamoyl)ethyl]-4-methylpiperidin- 4-yl ester; biphenyl-2-ylcarbamic acid l-[2-(2-chloro-4-{[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]methyl}-5-methoxyphenylcarbamoyl)ethyl]-4- methylpiperidin-4-yl ester;
4-(biphenyl-2-ylcarbamoyloxy)-l-{9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo-l,2- dihydroquinolin-5-yl)-ethylamino]nonyl}-l-azoniabicyclo[2.2.2]octane; biphenyl-2-ylcarbamic acid 8- {9-[(i?)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl}-8-azabicyclo[3.2.1]oct-3-yl ester;
7-(biphenyl-2-ylcarbamoyloxy)-9- {9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)-ethylamino]nonyl}-9-methyl-3-oxa-9- azoniatricyclo[3.3.1.0*2,4*]nonane; and biphenyl-2-yl carbamic acid 9- {9-[(K)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydroquinolin-5-yl)ethylamino]nonyl}-3-oxa-9-aza-tricyclo[3.3.1.0*2,4*]non-7-yl ester; or a pharmaceutically acceptable salt or solvate thereof.
29. Biρhenyl-2-ylcarbamic acid 1 - {9-[(R)-2-hydroxy-2-(8-hydroxy-2-oxo- 1 ,2- dihydro-quinolin-5-yl)ethylamino]nonyl}piperidin-4-yl ester or a pharmaceutically acceptable salt or solvate thereof.
30. Biphenyl-2-ylcarbamic acid l-(2-{5-[(/?)-2-hydroxy-2-(8-hydroxy-2-oxo- l,2-dihydroquinolin-5-yl)ethylamino]pentylcarbamoyl}ethyl)piperidin-4-yl ester or a pharmaceutically acceptable salt or solvate thereof.
31. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of any one of Claims 1 to 30.
32. The pharmaceutical composition of Claim 31 , wherein the composition further comprises a therapeutically effective amount of a steroidal anti-inflammatory agent.
33. The pharmaceutical composition of Claim 31 , wherein the composition further comprises a therapeutically effective amount of a PDE4 inhibitor.
34. A method for treating a pulmonary disorder, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of any one of Claims 1 to 30.
35. A method of providing bronchodilation in a patient, the method comprising administering to a patient requiring bronchodilation a therapeutically effective amount of a compound of any one of Claims 1 to 30.
36. A method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a compound of any one of Claims 1 to 30.
37. A method for treating a pulmonary disorder, the method comprising administering a compound having both muscarinic receptor antagonist and β2-adrenergic receptor agonist activity.
38. The method of Claim 37, wherein the compound has a K; for the M3 muscarinic receptor of less than about 100 nM and an EC50 for the β2 adrenergic receptor of less than about 100 nM.
39. The method of Claim 37 or Claim 38, wherein the compound has a ratio of the Kj for the M3 muscarinic receptor to the EC50 for the β2 adrenergic receptor is from about 30:1 to about 1 :30.
40. A method of studying a biological system or sample comprising a muscarinic receptor or a β2 adrenergic receptor, the method comprising:
(a) contacting the biological system or sample with a compound of any one of Claims 1 to 30; and
(b) determining the effects caused by the compound on the biological system or sample.
41. A process for preparing a compound of formula I:
Figure imgf000210_0001
wherein: a is 0 or an integer of from 1 to 3; each R1 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -ORla, -C(O)ORlb, -SRlc, -S(O)Rld, -S(O)2Rle and -NRlfRIg; each of RIa, Rlb, Rlc, Rld, RIe, Rlf and Rlg is independently hydrogen, (l-4C)alkyl or phenyl-(l-4C)alkyl; b is 0 or an integer of from 1 to 3; each R2 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR2a, -C(O)OR2b, -SR2°, -S(O)R2d, -S(O)2R2e and - NR2fR2g; each of R2a, R2b, R2°, R2d, R2e, R2f and R2g is independently hydrogen, ( 1 -4C)alkyl or phenyl-(l-4C)alkyl;
W is attached to the 3- or 4-position with respect to the nitrogen atom in the piperidine ring and represents O or NWa;
Wa is hydrogen or (l-4C)alkyl; c is 0 or an integer of from 1 to 4; each R3 is independently selected from (l-4C)alkyl, (2-4C)alkenyl, (2-4C)alkynyl, (3-6C)cycloalkyl, cyano, halo, -OR3a, -C(O)OR3b, -SR3c, -S(O)R3d, -S(O)2R3e and -NR3fR3g; or two R3 groups are joined to form (l-3C)alkylene, (2-3C)alkenylene or oxiran- 2,3-diyl; each of R3a, R3b, R3c, R3d, R3e, R3f and R3g is independently hydrogen or ( 1 -
4C)alkyl;
R4 is a divalent group ofthe formula:
wherein d, e, f, g, h and i are each independently selected from 0 and 1 ; R4a, R4b, R4c and R4d are each independently selected from (l-lOC)alkylene, (2- 10C)alkenylene and (2-10C)alkynylene, wherein each alkylene, alkenylene or alkynylene group is unsubstituted or substituted with from 1 to 5 substituents independently selected from (l-4C)alkyl, fluoro, hydroxy, phenyl and phenyl-(l-4C)alkyl;
A1 and A2 are each independently selected from (3-7C)cycloalkylene, (6- 10C)arylene, -O-(6-10C)arylene, (6-10C)arylene-O-, (2-9C)heteroarylene, -O-(2- 9C)heteroarylene, (2-9C)heteroarylene-O- and (3-6C)heterocyclene, wherein each cycloalkylene is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl, and each arylene, heteroarylene or heterocyclene group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl, (l-4C)alkoxy, -S-(l-4C)alkyl, -S(O)-(l-4C)alkyl, -S(O)2-(l-4C)alkyl, -C(O)O(l-4C)alkyl, carboxy, cyano, hydroxy, nitro, trifluoromethyl and trifluoromethoxy;
Q is selected from a bond, -O-, -C(O)O-, -OC(O)-, -S-, -S(O)-, -S(O)2-, -N(Qa)C(O)-, -C(O)N(Qb)-, -N(Qc)S(O)2-, -S(O)2N(Qd)-, -N(Qe)C(O)N(Qf)-, -N(Qg)S(O)2N(Qh)-, -OC(O)N(Q , -N( )C(O)O- and -N(Qk); Qa, Qb, Qc, Qd, Qe, Qf, Qg, Qh, Q;, OJ and Qk are each independently selected from hydrogen, (l-6C)alkyl, A3 and (l-4C)alkylene-A4, wherein the alkyl group is unsubstituted or substituted with from 1 to 3 substituents independently selected from fluoro, hydroxy and (l-4C)alkoxy; or together with the nitrogen atom and the group R4b or R4c to which they are attached, form a 4-6 membered azacycloalkylene group; A3 and A4 are each independently selected from (3-6C)cycloalkyl, (6-10C)aryl,
(2-9C)heteroaryl and (3-6C)heterocyclyl, wherein each cycloalkyl is unsubstituted or substituted with from 1 to 4 substitutents selected independently from (l-4C)alkyl and each aryl, heteroaryl or heterocyclyl group is unsubstituted or substituted with from 1 to 4 substituents independently selected from halo, (l-4C)alkyl and (l-4C)alkoxy; provided that the number of contiguous atoms in the shortest chain between the two nitrogen atoms to which R4 is attached is in the range of from 4 to 14;
R5 represents hydrogen or (l-4C)alkyl;
R6 is -NR6aCR6b(O) or -CR6oR6dOR6e and R7 is hydrogen; or R6 and R7 together form -NR7aC(O)-CR7b=CR7c- , -CR7d=CR7e-C(O)-NR7f-, -NR7gC(O)-CR7hR7i-CR7jR7k- or - CR71R7m-CR7nR70- C(O) -NR7p-; each of R6a, R6b, R6°, R6d and R6e is independently hydrogen or (l-4C)alkyl; and each of R7a, R7b, R7c, R7d, R7e, R7f, R7g, R7h, R7i, R7j, R7k, R71, R7m, R7n, R7° and R7p is independently hydrogen or (l-4C)alkyl; or a stereoisomer thereof; the process comprising: (a) reacting a compound of formula 1:
Figure imgf000212_0001
or a salt thereof; with a compound of formula 2:
Figure imgf000213_0001
wherein X1 represents a leaving group, and P1 and P2 each independently represent a hydrogen atom or a hydroxyl-protecting group; (b) reacting a compound of formula 3 :
Figure imgf000213_0002
or salt thereof; with a compound of fonnula 4 :
Figure imgf000213_0003
wherein X represents a leaving group, and P3 and P4 each independently represent a hydrogen atom or a hydroxyl-protecting group; (c) coupling a compound of formula 5 :
Figure imgf000214_0001
with a compound of formula 6:
Figure imgf000214_0002
wherein XQa and XQb each independently represent functional groups that couple to form a group Q, P5a represents a hydrogen atom or an amino-protecting group; and P5 and P6 each independently represent a hydrogen atom or a hydroxyl-protecting group;
(d) for a compound of formula I wherein R5 represents a hydrogen atom, reacting a compound of formula 3 with a compound of formula 7:
Figure imgf000214_0003
or a hydrate thereof (e.g., a glyoxal), in the presence of a reducing agent, wherein P7 represents a hydrogen atom or a hydroxyl-protecting group; (e) reacting a compound of formula 1 with a compound of formula 8:
Figure imgf000215_0001
or a hydrate thereof, in the presence of a reducing agent, wherein P8 and P9 each independently represent a hydrogen atom or a hydroxyl-protecting group, P 10 . represents a hydrogen atom or an amino-protecting group, and R 4\ represents a residue that, together with the carbon to which it is attached, affords a group R4 upon completion ofthe reaction; (f) reacting a compound of formula 9:
Figure imgf000215_0002
wherein X3 represents a leaving group, with a compound of formula 10:
Figure imgf000215_0003
10 wherein P11 and P12 each independently represent a hydrogen atom or a hydroxyl- protecting group, and P13 represents a hydrogen atom or an amino-protecting group; or (g) reacting a compound of formula 11:
Figure imgf000216_0001
11
or a hydrate thereof; wherein R4 represents a residue that, together with the carbon to which it is attached, affords a group R4 upon completion ofthe reaction; with a compound of formula 10 in the presence of a reducing agent; and then removing any protecting group P1, P2, P3, P4, P5a, P5b, P6, P7, P8, P9, P10, P11, P12 or P13 to provide a compound of formula I.
42. The process of Claim 41 , wherein the process further comprises forming a phannaceutically acceptable salt ofthe compound of fonnula I.
43. The product prepared by the process of Claim 41 or 42.
44. A compound of any one of Claims 1 to 30 for use in therapy or as a medicament.
45. A compound of any one of Claims 1 to 30 for treatment of a pulmonary disorder.
46. Medicament containing a compound of any one of Claims 1 to 30.
47. Use of a compound of any one of Claims 1 to 30 for the manufacture of a medicament.
48. The use of Claim 45, wherein the medicament is for the treatment of a pulmonary disorder.
49. Use of a compound of any one of Claims 1 to 30 for treatment of a pulmonary disorder.
50, The use of Claim 48 or 49, wherein the pulmonary disorder is chronic obstructive pulmonary disease or asthma.
PCT/US2004/004449 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity WO2004074246A2 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
JP2006503604A JP4555283B2 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity
DK04711253.7T DK1615889T3 (en) 2003-02-14 2004-02-13 Biphenyl derivatives with beta2-adrenergic receptor agonist and muscarinic receptor antagonist activity
AU2004213411A AU2004213411B2 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
KR1020057014929A KR101223991B1 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
MXPA05008528A MXPA05008528A (en) 2003-02-14 2004-02-13 Biphenyl derivatives.
BRPI0407508A BRPI0407508B8 (en) 2003-02-14 2004-02-13 biphenyl derivative compounds, pharmaceutical composition and use thereof
AT04711253T ATE482934T1 (en) 2003-02-14 2004-02-13 BIPHENYL DERIVATIVES WITH BETA2 ADRENERG RECEPTOR AGONISTIC AND MUSCARIN RECEPTOR ANTAGONISTIC ACTIVITY
CA2515777A CA2515777C (en) 2003-02-14 2004-02-13 Biphenyl derivatives
DE602004029347T DE602004029347D1 (en) 2003-02-14 2004-02-13 BIPHENYL DERIVATIVES WITH BETA2 ADRENERG RECEPTOR AGONISTIC AND MUSCARIN RECEPTOR OF ANTAGONISTIC ACTIVITY
SI200431566T SI1615889T1 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
NZ541579A NZ541579A (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
EP04711253A EP1615889B1 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
KR1020117010078A KR101224497B1 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
IL169922A IL169922A (en) 2003-02-14 2005-07-27 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
IS7964A IS2847B (en) 2003-02-14 2005-07-28 Biphenyl derivatives with beta2 adrenergic receptor antagonist and muscarinic receptor antagonist activity
NO20054206A NO331947B1 (en) 2003-02-14 2005-09-09 Biphenyl derivative, its use and pharmaceutical composition comprising the compound.
HK06106230.1A HK1086266A1 (en) 2003-02-14 2006-05-29 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
AU2010219338A AU2010219338B2 (en) 2003-02-14 2010-09-08 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US44784303P 2003-02-14 2003-02-14
US60/447,843 2003-02-14
US46703503P 2003-05-01 2003-05-01
US60/467,035 2003-05-01

Publications (2)

Publication Number Publication Date
WO2004074246A2 true WO2004074246A2 (en) 2004-09-02
WO2004074246A3 WO2004074246A3 (en) 2004-11-18

Family

ID=32912274

Family Applications (3)

Application Number Title Priority Date Filing Date
PCT/US2004/004449 WO2004074246A2 (en) 2003-02-14 2004-02-13 Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
PCT/US2004/004224 WO2004074276A1 (en) 2003-02-14 2004-02-13 BIPHENYL DERIVATIVES HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY
PCT/US2004/004273 WO2004074812A2 (en) 2003-02-14 2004-02-13 Library of biphenyl derivatives useful for identifying compounds having both beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity

Family Applications After (2)

Application Number Title Priority Date Filing Date
PCT/US2004/004224 WO2004074276A1 (en) 2003-02-14 2004-02-13 BIPHENYL DERIVATIVES HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY
PCT/US2004/004273 WO2004074812A2 (en) 2003-02-14 2004-02-13 Library of biphenyl derivatives useful for identifying compounds having both beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity

Country Status (28)

Country Link
US (15) US20040209915A1 (en)
EP (5) EP1615889B1 (en)
JP (6) JP4555283B2 (en)
KR (3) KR101223991B1 (en)
AR (1) AR043176A1 (en)
AT (1) ATE482934T1 (en)
AU (2) AU2004213411B2 (en)
BR (1) BRPI0407508B8 (en)
CA (1) CA2515777C (en)
CY (2) CY1111276T1 (en)
DE (1) DE602004029347D1 (en)
DK (2) DK1615889T3 (en)
ES (2) ES2566155T3 (en)
HK (3) HK1086266A1 (en)
HU (1) HUE027380T2 (en)
IL (1) IL169922A (en)
IS (1) IS2847B (en)
MX (1) MXPA05008528A (en)
MY (1) MY148487A (en)
NO (2) NO331947B1 (en)
NZ (1) NZ541579A (en)
PE (1) PE20040950A1 (en)
PL (1) PL216397B1 (en)
PT (1) PT1615889E (en)
RU (1) RU2330841C2 (en)
SI (2) SI2246345T1 (en)
TW (1) TWI331995B (en)
WO (3) WO2004074246A2 (en)

Cited By (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006023460A3 (en) * 2004-08-16 2006-04-06 Theravance Inc COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY
WO2006056471A1 (en) 2004-11-29 2006-06-01 Novartis Ag 5-hydroxy-benzothiazole derivatives having beta-2-adrenorecptor agonist activity
WO2006050239A3 (en) * 2004-10-29 2006-10-12 Glaxo Group Ltd Muscarinic acetylcholine receptor antagonists
WO2006108643A2 (en) 2005-04-14 2006-10-19 Novartis Ag Organic compounds
WO2007090859A1 (en) * 2006-02-10 2007-08-16 Glaxo Group Limited Succinic acid salt of biphenyl-2-ylcarbamic acid 1-[2-(2-chloro-4-{ [ (r)-2-hydroxy-2- (8-hydroxy-2-oxo-1, 2-dihydroquinolin-5-yl) eth ylaminoimethyl] } -5-methoxyphenylcarbamoyl) ethyl] piperidin-4-yl ester and its use for the treatment of pulmonary disorders
WO2007121920A2 (en) 2006-04-21 2007-11-01 Novartis Ag Purine derivatives for use as adenosin a2a receptor agonists
WO2007127297A2 (en) * 2006-04-25 2007-11-08 Theravance, Inc. Crystalline forms of a dimethylphenyl compound
JP2008510014A (en) * 2004-08-16 2008-04-03 セラヴァンス, インコーポレーテッド Compounds having β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity
WO2008041095A1 (en) * 2006-10-04 2008-04-10 Pfizer Limited Sulfonamide derivatives as adrenergic agonists and muscarinic antagonists
WO2009087224A1 (en) 2008-01-11 2009-07-16 Novartis Ag Pyrimidines as kinase inhibitors
US7612084B2 (en) 2006-03-20 2009-11-03 Pfizer Inc Amine derivatives for the treatment of asthma and COPD
JP2009541209A (en) * 2006-04-25 2009-11-26 セラヴァンス, インコーポレーテッド Dialkylphenyl compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2009150137A2 (en) 2008-06-10 2009-12-17 Novartis Ag Organic compounds
WO2010004517A1 (en) * 2008-07-11 2010-01-14 Pfizer Limited Triazole derivatives useful for the treatment of diseases
WO2010119064A1 (en) 2009-04-14 2010-10-21 Glaxo Group Limited Process for the preparation of a biphenyl-2-ylcarbamic acid ester
EP2279777A2 (en) 2007-01-10 2011-02-02 Irm Llc Compounds and compositions as channel activating protease inhibitors
EP2280006A1 (en) 2005-08-08 2011-02-02 Pulmagen Therapeutics (Synergy) Limited Pharmaceutical composition for inhalation comprising an oxazole or thiazole m3 muscarinic receptor antagonist
EP2281813A1 (en) 2005-08-08 2011-02-09 Pulmagen Therapeutics (Synergy) Limited Bicyclo[2.2.1]hept-7-ylamine derivatives and their uses
EP2286813A2 (en) 2006-01-31 2011-02-23 Novartis AG Use of naphthyridine derivatives as medicaments
EP2292619A1 (en) 2004-10-22 2011-03-09 Novartis AG Purine derivatives for use as adenonsin A-2A receptor agonists
WO2011050325A1 (en) 2009-10-22 2011-04-28 Vertex Pharmaceuticals Incorporated Compositions for treatment of cystic fibrosis and other chronic diseases
EP2332933A1 (en) 2007-05-07 2011-06-15 Novartis AG Epithelial sodium channel (ENaC) inhibitors
WO2011081937A1 (en) 2009-12-15 2011-07-07 Gilead Sciences, Inc. Corticosteroid-beta-agonist-muscarinic antagonist compounds for use in therapy
WO2011113894A1 (en) 2010-03-19 2011-09-22 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf
WO2012007539A1 (en) 2010-07-14 2012-01-19 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2012034095A1 (en) 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
WO2012034091A1 (en) 2010-09-09 2012-03-15 Irm Llc Imidazo [1, 2] pyridazin compounds and compositions as trk inhibitors
WO2012035158A1 (en) 2010-09-17 2012-03-22 Novartis Ag Pyrazine derivatives as enac blockers
EP2444120A1 (en) 2007-12-10 2012-04-25 Novartis AG Spirocyclic amiloride analogues as ENac blockers
WO2012032546A3 (en) * 2010-09-08 2012-06-28 Cadila Healthcare Limited Process for the preparation of salmeterol and its intermediates
WO2012116217A1 (en) 2011-02-25 2012-08-30 Irm Llc Compounds and compositions as trk inhibitors
EP2532679A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
WO2012168359A1 (en) 2011-06-10 2012-12-13 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2012168349A1 (en) 2011-06-10 2012-12-13 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2013030802A1 (en) 2011-09-01 2013-03-07 Novartis Ag Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
WO2013038386A1 (en) 2011-09-16 2013-03-21 Novartis Ag Heterocyclic compounds for the treatment of cystic fibrosis
WO2013038390A1 (en) 2011-09-16 2013-03-21 Novartis Ag N-substituted heterocyclyl carboxamides
WO2013038378A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013038381A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine/pyrazine amide derivatives
WO2013038373A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013105061A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused dihydropyrido [2,3 -b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105058A1 (en) 2012-01-13 2013-07-18 Novartis Ag 7,8- dihydropyrido [3, 4 - b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105065A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pah and related disorders
WO2013105063A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105057A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused pyrroles as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105066A1 (en) 2012-01-13 2013-07-18 Novartis Ag Salts of an ip receptor agonist
WO2013140319A1 (en) 2012-03-19 2013-09-26 Novartis Ag Crystalline form of a succinate salt
WO2014086927A1 (en) 2012-12-06 2014-06-12 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2014086924A1 (en) 2012-12-06 2014-06-12 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2014125413A1 (en) 2013-02-13 2014-08-21 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2014132220A1 (en) 2013-03-01 2014-09-04 Novartis Ag Solid forms of bicyclic heterocyclic derivatives as pdgf receptor mediators
US8883800B2 (en) 2011-07-15 2014-11-11 Boehringer Ingelheim International Gmbh Substituted quinazolines, the preparation thereof and the use thereof in pharmaceutical compositions
US8883805B2 (en) 2004-11-05 2014-11-11 Boehringer Ingelheim International Gmbh Process for the preparation of chiral 8-(3-aminopiperidin-1-yl)-xanthines
WO2015008230A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors comprising a heteroaromatic ring-benzyl-amide-cycle core
WO2015008229A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors
US9034883B2 (en) 2010-11-15 2015-05-19 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
US9108964B2 (en) 2002-08-21 2015-08-18 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
WO2015162456A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162459A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162461A1 (en) 2014-04-24 2015-10-29 Novartis Ag Pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US9173859B2 (en) 2006-05-04 2015-11-03 Boehringer Ingelheim International Gmbh Uses of DPP IV inhibitors
US9212183B2 (en) 2008-12-23 2015-12-15 Boehringer Ingelheim International Gmbh Salt forms of 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine
US9233108B2 (en) 2011-11-11 2016-01-12 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9266888B2 (en) 2006-05-04 2016-02-23 Boehringer Ingelheim International Gmbh Polymorphs
US9315463B2 (en) 2010-05-13 2016-04-19 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9415016B2 (en) 2008-04-03 2016-08-16 Boehringer Ingelheim International Gmbh DPP-IV inhibitor combined with a further antidiabetic agent, tablets comprising such formulations, their use and process for their preparation
WO2016128456A1 (en) 2015-02-12 2016-08-18 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US9457029B2 (en) 2009-11-27 2016-10-04 Boehringer Ingelheim International Gmbh Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US9486526B2 (en) 2008-08-06 2016-11-08 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients inappropriate for metformin therapy
WO2016193241A1 (en) 2015-06-01 2016-12-08 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US9518050B2 (en) 2012-12-18 2016-12-13 Almirall, S.A. Cyclohexyl and quinuclidinyl carbamate derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activity
US9526730B2 (en) 2012-05-14 2016-12-27 Boehringer Ingelheim International Gmbh Use of a DPP-4 inhibitor in podocytes related disorders and/or nephrotic syndrome
US9526728B2 (en) 2014-02-28 2016-12-27 Boehringer Ingelheim International Gmbh Medical use of a DPP-4 inhibitor
US9555001B2 (en) 2012-03-07 2017-01-31 Boehringer Ingelheim International Gmbh Pharmaceutical composition and uses thereof
US9562039B2 (en) 2013-02-27 2017-02-07 Almirall, S.A. Salts of 2-amino-1-hydroxyethyl-8-hydroxyquinolin-2(1H)-one derivatives having both β2 adrenergic receptor agonist and M3 muscarinic receptor antagonist activities
US9603851B2 (en) 2010-05-05 2017-03-28 Boehringer Ingelheim International Gmbh Combination therapy
US9643961B2 (en) 2010-05-13 2017-05-09 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic antagonist and M3 muscarinic antagonist activities
WO2017093208A1 (en) 2015-12-03 2017-06-08 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US9713618B2 (en) 2012-05-24 2017-07-25 Boehringer Ingelheim International Gmbh Method for modifying food intake and regulating food preference with a DPP-4 inhibitor
WO2018011090A1 (en) 2016-07-13 2018-01-18 Chiesi Farmaceutici S.P.A. Hydroxyquinolinone compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2018108089A1 (en) 2016-12-14 2018-06-21 北京硕佰医药科技有限责任公司 Class of bifunctional compounds with quaternary ammonium salt structure
US10005771B2 (en) 2014-09-26 2018-06-26 Almirall, S.A. Bicyclic derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US10040765B2 (en) 2012-09-21 2018-08-07 Crystal Pharma S.A.U. Methods for the preparation of indacaterol and pharmaceutically acceptable salts thereof
US10155000B2 (en) 2016-06-10 2018-12-18 Boehringer Ingelheim International Gmbh Medical use of pharmaceutical combination or composition
US10456390B2 (en) 2013-07-25 2019-10-29 Almirall, S.A. Combinations comprising MABA compounds and corticosteroids
WO2020250116A1 (en) 2019-06-10 2020-12-17 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf, copd, and bronchiectasis
WO2021038426A1 (en) 2019-08-28 2021-03-04 Novartis Ag Substituted 1,3-phenyl heteroaryl derivatives and their use in the treatment of disease
US11033552B2 (en) 2006-05-04 2021-06-15 Boehringer Ingelheim International Gmbh DPP IV inhibitor formulations
US11911388B2 (en) 2008-10-16 2024-02-27 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients with insufficient glycemic control despite therapy with an oral or non-oral antidiabetic drug

Families Citing this family (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6693202B1 (en) * 1999-02-16 2004-02-17 Theravance, Inc. Muscarinic receptor antagonists
ES2165768B1 (en) 1999-07-14 2003-04-01 Almirall Prodesfarma Sa NEW DERIVATIVES OF QUINUCLIDINE AND PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.
PE20050130A1 (en) * 2002-08-09 2005-03-29 Novartis Ag ORGANIC COMPOUNDS
PE20040950A1 (en) 2003-02-14 2005-01-01 Theravance Inc BIPHENYL DERIVATIVES AS AGONISTS OF ß2-ADRENERGIC RECEPTORS AND AS ANTAGONISTS OF MUSCARINAL RECEPTORS
US7317102B2 (en) 2003-04-01 2008-01-08 Theravance, Inc. Diarylmethyl and related compounds
WO2004106333A1 (en) * 2003-05-28 2004-12-09 Theravance, Inc. Azabicycloalkane compounds as muscarinic receptor antagonists
PT1631295E (en) * 2003-06-06 2010-05-24 Arexis Ab Use of condensed heterocyclic compounds as scce inhibitors for the treatment of skin diseases
US20050026948A1 (en) * 2003-07-29 2005-02-03 Boehringer Ingelheim International Gmbh Medicaments for inhalation comprising an anticholinergic and a betamimetic
ES2329586T3 (en) * 2003-11-21 2009-11-27 Theravance, Inc. COMPOUNDS THAT HAVE AGONIST ACTIVITY OF THE BETA2 ADRENERGIC RECEIVER AND ANTAGONIST OF THE MUSCARINE RECEIVER.
TW200531692A (en) 2004-01-12 2005-10-01 Theravance Inc Aryl aniline derivatives as β2 adrenergic receptor agonists
WO2005080375A1 (en) * 2004-02-13 2005-09-01 Theravance, Inc. Crystalline form of a biphenyl compound
EP1723115A1 (en) * 2004-03-11 2006-11-22 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
WO2005087735A1 (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
JP2007528408A (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
EP1723113A1 (en) * 2004-03-11 2006-11-22 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
ES2317245T3 (en) * 2004-05-31 2009-04-16 Laboratorios Almirall, S.A. COMBINATIONS THAT INCLUDE ANTIMUSCARINIC AGENTS AND BETA-ADRENERGIC AGONISTS.
ES2257152B1 (en) * 2004-05-31 2007-07-01 Laboratorios Almirall S.A. COMBINATIONS THAT INCLUDE ANTIMUSCARINIC AGENTS AND BETA-ADRENERGIC AGONISTS.
TWI374883B (en) * 2004-08-16 2012-10-21 Theravance Inc Crystalline form of a biphenyl compound
TW200714587A (en) * 2005-03-10 2007-04-16 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
US7642355B2 (en) 2005-03-10 2010-01-05 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
US7659403B2 (en) * 2005-03-10 2010-02-09 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
JP2008533028A (en) * 2005-03-10 2008-08-21 セラヴァンス, インコーポレーテッド Biphenyl compounds useful as muscarinic receptor antagonists
ES2265276B1 (en) 2005-05-20 2008-02-01 Laboratorios Almirall S.A. DERIVATIVES OF 4- (2-AMINO-1-HYDROXYETHYL) Phenol as agonists of the BETA2 ADRENERGIC RECEIVER.
JP2008546695A (en) 2005-06-13 2008-12-25 セラヴァンス, インコーポレーテッド Biphenyl compounds useful as muscarinic receptor antagonists
US8429052B2 (en) * 2005-07-19 2013-04-23 Lincoln National Life Insurance Company Method and system for providing employer-sponsored retirement plan
TW200738658A (en) 2005-08-09 2007-10-16 Astrazeneca Ab Novel compounds
US7973055B2 (en) * 2006-03-09 2011-07-05 Theravance, Inc. Crystalline forms of a biphenyl compound
TW200745067A (en) 2006-03-14 2007-12-16 Astrazeneca Ab Novel compounds
ES2296516B1 (en) * 2006-04-27 2009-04-01 Laboratorios Almirall S.A. DERIVATIVES OF 4- (2-AMINO-1-HYDROXYETHYL) Phenol as agonists of the BETA2 ADRENERGIC RECEIVER.
GB0613154D0 (en) * 2006-06-30 2006-08-09 Novartis Ag Organic Compounds
ES2302447B1 (en) * 2006-10-20 2009-06-12 Laboratorios Almirall S.A. DERIVATIVES OF 4- (2-AMINO-1-HYDROXYETHYL) Phenol as agonists of the BETA2 ADRENERGIC RECEIVER.
TW200833670A (en) 2006-12-20 2008-08-16 Astrazeneca Ab Novel compounds 569
PT2119700E (en) * 2007-01-31 2012-10-02 Toray Industries Benzylamine derivative or pharmaceutically acceptable acid addition salt thereof, and use thereof for medical purposes
GB0702458D0 (en) 2007-02-08 2007-03-21 Astrazeneca Ab Salts 668
ES2306595B1 (en) 2007-02-09 2009-09-11 Laboratorios Almirall S.A. NAPADISYLATE SALT OF 5- (2 - ((6- (2,2-DIFLUORO-2-PHENYLETOXI) HEXIL) AMINO) -1-HYDROXYETHYL) -8-HYDROXYCHINOLIN-2 (1H) -ONE AS ADRENERGIC RECEIVER AGONIST BETA2 .
US8682466B2 (en) * 2007-05-04 2014-03-25 Taiwan Semiconductor Manufacturing Company, Ltd. Automatic virtual metrology for semiconductor wafer result prediction
ES2320961B1 (en) * 2007-11-28 2010-03-17 Laboratorios Almirall, S.A. DERIVATIVES OF 4- (2-AMINO-1-HYDROXYETHYL) PHENOL AS BETA2 ADRENERGIC RECEIVER AGONISTS.
DK2242759T3 (en) * 2008-02-06 2012-12-17 Astrazeneca Ab compounds
EP2096105A1 (en) * 2008-02-28 2009-09-02 Laboratorios Almirall, S.A. Derivatives of 4-(2-amino-1-hydroxyethyl)phenol as agonists of the b2 adrenergic receptor
EP2100599A1 (en) 2008-03-13 2009-09-16 Laboratorios Almirall, S.A. Inhalation composition containing aclidinium for treatment of asthma and chronic obstructive pulmonary disease
EP2100598A1 (en) 2008-03-13 2009-09-16 Laboratorios Almirall, S.A. Inhalation composition containing aclidinium for treatment of asthma and chronic obstructive pulmonary disease
AU2009260899B2 (en) 2008-06-18 2012-02-23 Astrazeneca Ab Benzoxazinone derivatives acting as beta2-adrenoreceptor agonist for the treatment of respiratory disorders
WO2010007561A1 (en) * 2008-07-15 2010-01-21 Pfizer Limited Novel compounds active as muscarinic receptor antagonists
US20100016366A1 (en) * 2008-07-15 2010-01-21 Paul Alan Glossop Novel Compounds Active as Muscarinic Receptor Antagonists
UY32297A (en) 2008-12-22 2010-05-31 Almirall Sa MESILATE SALT OF 5- (2 - {[6- (2,2-DIFLUORO-2-PHENYLITOXI) HEXIL] AMINO} -1-HYDROXYETHYL) -8-HYDROXYCHINOLIN-2 (1H) -ONA AS A RECEIVER AGONIST B (BETA ) 2 ACRENERGIC
TW201036957A (en) 2009-02-20 2010-10-16 Astrazeneca Ab Novel salt 628
EP2228368A1 (en) 2009-03-12 2010-09-15 Almirall, S.A. Process for manufacturing 5-(2-{[6-(2,2-difluoro-2-phenylethoxy) hexyl]amino}-1-hydroxyethyl)-8-hydroxyquinolin-2(1H)-one
EP2409977A4 (en) 2009-03-17 2012-09-26 Daiichi Sankyo Co Ltd Amide derivative
ES2715965T3 (en) 2009-04-23 2019-06-07 Theravance Respiratory Co Llc Diamine compounds that have muscarinic receptor antagonist activity and beta 2 adrenergic receptor agonist
AU2015234331B2 (en) * 2009-04-23 2017-08-03 Theravance Biopharma R&D Ip, Llc Diamide compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
EP2426121A4 (en) * 2009-04-30 2012-10-31 Teijin Pharma Ltd Quaternary ammonium salt compound
WO2011061527A1 (en) 2009-11-17 2011-05-26 Astrazeneca Ab Combinations comprising a glucocorticoid receptor modulator for the treatment of respiratory diseases
KR101742253B1 (en) * 2010-07-13 2017-05-31 세라밴스 바이오파마 알앤디 아이피, 엘엘씨 Process for preparing a biphenyl-2-ylcarbamic acid
AR083115A1 (en) * 2010-09-30 2013-01-30 Theravance Inc CRYSTAL OXALATE SALTS OF A DIAMID COMPOUND
GB201016912D0 (en) 2010-10-07 2010-11-24 Astrazeneca Ab Novel combination
JP6060242B2 (en) * 2010-11-30 2017-01-11 株式会社日立国際電気 Substrate processing apparatus, semiconductor device manufacturing method, and baffle structure
EP2510928A1 (en) 2011-04-15 2012-10-17 Almirall, S.A. Aclidinium for use in improving the quality of sleep in respiratory patients
EP2578570A1 (en) 2011-10-07 2013-04-10 Almirall, S.A. Novel process for preparing 5-(2-{[6-(2,2-difluoro-2-phenylethoxy)hexyl]amino}-1(r)-hydroxyethyl)-8-hydroxyquinolin-2(1h)-one via novel intermediates of synthesis.
EP2641900A1 (en) 2012-03-20 2013-09-25 Almirall, S.A. Novel polymorphic Crystal forms of 5-(2-{[6-(2,2-difluoro-2-phenylethoxy) hexyl]amino}-1-(R)-hydroxyethyl)-8-hydroxyquinolin-2(1h)-one, heminapadisylate as agonist of the ß2 adrenergic receptor.
CZ306252B6 (en) 2013-03-15 2016-10-26 Zentiva, K.S. Process for preparing 5-[(R)-2-(5,6-diethylindan-2-ylamino)-1-hydroxyethyl]-8-hydroxy-(1H)-quinolin-2-one (indacaterol)
TWI641373B (en) 2013-07-25 2018-11-21 阿爾米雷爾有限公司 SALTS OF 2-AMINO-1-HYDROXYETHYL-8-HYDROXYQUINOLIN-2(1H)-ONE DERIVATIVES HAVING BOTH MUSCARINIC RECEPTOR ANTAGONIST AND β2 ADRENERGIC RECEPTOR AGONIST ACTIVITIES
WO2017001907A1 (en) * 2015-06-29 2017-01-05 Teva Pharmaceuticals International Gmbh Biocatalytic processes for the preparation of vilanterol
MX2018008799A (en) * 2016-01-22 2019-01-14 Sichuan Haisco Pharmaceutical Co Ltd Nitrogenous heterocyclic amide derivative, preparation method thereof, and pharmaceutical application.
CN106706868B (en) * 2016-11-15 2018-12-04 河南省农业科学院 Application of the artificial expression HSP27 albumen in terms of detecting beta-2-adrenoreceptor agonists class medicament residue
CN106632257B (en) * 2016-12-15 2019-02-12 上海市奉贤区中心医院 The synthetic method of GSK961081 and its intermediate
US11866482B2 (en) 2018-02-07 2024-01-09 Duke University System and method for homogenous GPCR phosphorylation and identification of beta-2 adrenergic receptor positive allosteric modulators
US11484531B2 (en) 2018-08-30 2022-11-01 Theravance Biopharma R&D Ip, Llc Methods for treating chronic obstructive pulmonary disease
WO2022175982A1 (en) * 2021-02-19 2022-08-25 Gbr Laboratories Private Limited A process for preparing batefenterol and intermediates thereof
CN114409890A (en) * 2022-02-28 2022-04-29 中国科学院长春应用化学研究所 Amino-functionalized polyethylene glycol derivative and preparation method thereof

Family Cites Families (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4026897A (en) * 1974-01-31 1977-05-31 Otsuka Pharmaceutical Company 5-[1-Hydroxy-2-(substituted-amino)]alkyl-8-hydroxycarbostyril derivatives
BR8007911A (en) 1979-12-06 1981-06-16 Glaxo Group Ltd PERFECTED INHALER
CY1492A (en) 1981-07-08 1990-02-16 Draco Ab Powder inhalator
DE3134590A1 (en) 1981-09-01 1983-03-10 Boehringer Ingelheim KG, 6507 Ingelheim NEW BENZO HETEROCYCLES
US4460581A (en) * 1982-10-12 1984-07-17 Boehringer Ingelheim Kg (1-Hydroxy-2-amino-alkyl)-substituted benzoxazinones and benzoxazolinones
ZW6584A1 (en) * 1983-04-18 1985-04-17 Glaxo Group Ltd Phenethanolamine derivatives
PT83094B (en) 1985-07-30 1993-07-30 Glaxo Group Ltd DEVICES PROPER FOR THE ADMINISTRATION OF MEDICINES TO PATIENTS
US4894219A (en) 1988-03-29 1990-01-16 University Of Florida Beta-agonist carbostyril derivatives, assay method and pharmacological composition
US5225183A (en) 1988-12-06 1993-07-06 Riker Laboratories, Inc. Medicinal aerosol formulations
DK443489D0 (en) 1989-09-08 1989-09-08 Ferrosan As SUBSTITUTED URINARY COMPOUNDS, THEIR PREPARATION AND USE
SK280967B6 (en) 1990-03-02 2000-10-09 Glaxo Group Limited Inhalation device
US6006745A (en) 1990-12-21 1999-12-28 Minnesota Mining And Manufacturing Company Device for delivering an aerosol
US5874063A (en) 1991-04-11 1999-02-23 Astra Aktiebolag Pharmaceutical formulation
AU2178392A (en) 1991-06-12 1993-01-12 Minnesota Mining And Manufacturing Company Albuterol sulfate suspension aerosol formulations
US5337740A (en) 1991-08-01 1994-08-16 New England Pharmaceuticals, Inc. Inhalation devices
ES2216800T3 (en) 1991-12-18 2004-11-01 Minnesota Mining And Manufacturing Company AEROSOL FORMULATIONS IN SUSPENSION.
HUT72321A (en) 1992-03-31 1996-04-29 Glaxo Group Ltd Substituted phenylcarbamate and phenyl-urea derivatives, pharmaceutical compositions containing them and process for their preparation
US5239993A (en) 1992-08-26 1993-08-31 Glaxo Inc. Dosage inhalator providing optimized compound inhalation trajectory
US5497763A (en) 1993-05-21 1996-03-12 Aradigm Corporation Disposable package for intrapulmonary delivery of aerosolized formulations
CA2123728A1 (en) * 1993-05-21 1994-11-22 Noriyoshi Sueda Urea derivatives and their use as acat inhibitors
WO1995006635A1 (en) 1993-09-02 1995-03-09 Yamanouchi Pharmaceutical Co., Ltd. Carbamate derivative and medicine containing the same
US5415162A (en) 1994-01-18 1995-05-16 Glaxo Inc. Multi-dose dry powder inhalation device
WO1995021820A1 (en) 1994-02-10 1995-08-17 Yamanouchi Pharmaceutical Co., Ltd. Novel carbamate derivative and medicinal composition containing the same
US5495054A (en) 1994-05-31 1996-02-27 Sepracor, Inc. Tetrahydroindeno[1,2-D][1,3,2]oxazaboroles and their use as enantioselective catalysts
US5983956A (en) 1994-10-03 1999-11-16 Astra Aktiebolag Formulation for inhalation
SE9501384D0 (en) 1995-04-13 1995-04-13 Astra Ab Process for the preparation of respirable particles
SK284447B6 (en) 1995-04-14 2005-04-01 Glaxo Wellcome Inc. Metered dose inhaler
DE19536902A1 (en) 1995-10-04 1997-04-10 Boehringer Ingelheim Int Miniature fluid pressure generating device
EP0863141B1 (en) 1995-10-13 2001-09-12 Banyu Pharmaceutical Co., Ltd. Substituted heteroaromatic derivatives
PE92198A1 (en) 1996-08-01 1999-01-09 Banyu Pharma Co Ltd DERIVATIVES OF FLUORINE-CONTAINED 1,4-PIPERIDINE
US6040344A (en) 1996-11-11 2000-03-21 Sepracor Inc. Formoterol process
AU9281298A (en) 1997-10-01 1999-04-23 Kyowa Hakko Kogyo Co. Ltd. Benzodioxole derivatives
JP2002508366A (en) 1997-12-12 2002-03-19 スミスクライン・ビーチャム・パブリック・リミテッド・カンパニー Quinoline piperazine and quinoline piperidine derivatives, methods for their preparation, and their use as complex 5-HT1A, 5-HT1B and 5-HT1D receptor antagonists
SI1070056T1 (en) 1998-03-14 2004-12-31 Altana Pharma Ag Phthalazinone pde iii/iv inhibitors
PT1073417E (en) 1998-04-18 2003-04-30 Glaxo Group Ltd PHARMACEUTICAL FORMULA IN AEROSOL
GB9808802D0 (en) 1998-04-24 1998-06-24 Glaxo Group Ltd Pharmaceutical formulations
GB9812088D0 (en) * 1998-06-05 1998-08-05 Celltech Therapeutics Ltd Chemical compounds
US6713651B1 (en) * 1999-06-07 2004-03-30 Theravance, Inc. β2-adrenergic receptor agonists
AU763638B2 (en) 1998-06-08 2003-07-31 Theravance Biopharma R&D Ip, Llc Muscarinic receptor antagonists
US6541669B1 (en) 1998-06-08 2003-04-01 Theravance, Inc. β2-adrenergic receptor agonists
SE9804000D0 (en) 1998-11-23 1998-11-23 Astra Ab New composition of matter
WO2000035877A1 (en) * 1998-12-18 2000-06-22 Du Pont Pharmaceuticals Company Heterocyclic piperidines as modulators of chemokine receptor activity
US6693202B1 (en) 1999-02-16 2004-02-17 Theravance, Inc. Muscarinic receptor antagonists
DE19921693A1 (en) 1999-05-12 2000-11-16 Boehringer Ingelheim Pharma Pharmaceutical composition for treating respiratory disorders, e.g. asthma, comprises combination of anticholinergic and beta-mimetic agents having synergistic bronchospasmolytic activity and reduced side-effects
GB9913083D0 (en) 1999-06-04 1999-08-04 Novartis Ag Organic compounds
DK1169019T3 (en) 1999-04-14 2003-06-02 Glaxo Group Ltd Pharmaceutical aerosol formulation
US6593497B1 (en) * 1999-06-02 2003-07-15 Theravance, Inc. β2-adrenergic receptor agonists
MXPA02005596A (en) 1999-12-07 2004-09-10 Theravance Inc Carbamate derivatives having muscarinic receptor antagonist activity.
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
UA73965C2 (en) * 1999-12-08 2005-10-17 Theravance Inc b2 ADRENERGIC RECEPTOR ANTAGONISTS
US6933296B2 (en) 2000-06-05 2005-08-23 Altana Pharma B.V. Compounds effective as β2-adrenoreceptor agonists as well as PDE4-inhibitors
IL156499A0 (en) 2000-12-22 2004-01-04 Almirall Prodesfarma Ag Quinuclidine carbamate derivatives and their use as m3 antagonists
GB0103630D0 (en) 2001-02-14 2001-03-28 Glaxo Group Ltd Chemical compounds
DE60220887T2 (en) 2001-03-08 2008-02-28 Glaxo Group Ltd., Greenford AGONISTS OF BETA ADRENORE RECEPTORS
JP4143413B2 (en) 2001-03-22 2008-09-03 グラクソ グループ リミテッド Formanilide derivatives as β2-adrenergic receptor agonists
US20030018019A1 (en) 2001-06-23 2003-01-23 Boehringer Ingelheim Pharma Kg Pharmaceutical compositions based on anticholinergics, corticosteroids and betamimetics
US7361787B2 (en) 2001-09-14 2008-04-22 Glaxo Group Limited Phenethanolamine derivatives for treatment of respiratory diseases
TWI249515B (en) * 2001-11-13 2006-02-21 Theravance Inc Aryl aniline beta2 adrenergic receptor agonists
US6653323B2 (en) 2001-11-13 2003-11-25 Theravance, Inc. Aryl aniline β2 adrenergic receptor agonists
BR0309185A (en) 2002-04-12 2005-02-15 Boehringer Ingelheim Pharma Beta-mimetic drug and a new anticholinergic
WO2003099764A1 (en) * 2002-05-28 2003-12-04 Theravance, Inc. ALKOXY ARYL β2 ADRENERGIC RECEPTOR AGONISTS
US6711451B2 (en) * 2002-07-02 2004-03-23 3D Systems, Inc. Power management in selective deposition modeling
GB0217225D0 (en) 2002-07-25 2002-09-04 Glaxo Group Ltd Medicinal compounds
TW200410951A (en) 2002-08-06 2004-07-01 Glaxo Group Ltd M3 muscarinic acetylcholine receptor 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
US7317102B2 (en) * 2003-04-01 2008-01-08 Theravance, Inc. Diarylmethyl and related compounds
WO2004106333A1 (en) * 2003-05-28 2004-12-09 Theravance, Inc. Azabicycloalkane compounds as muscarinic receptor antagonists
TW200510298A (en) * 2003-06-13 2005-03-16 Theravance Inc Substituted pyrrolidine and related compounds
JP2007524641A (en) * 2003-07-11 2007-08-30 セラヴァンス, インコーポレーテッド Substituted 4-amino-1-benzylpiperidine compounds
ES2329586T3 (en) 2003-11-21 2009-11-27 Theravance, Inc. COMPOUNDS THAT HAVE AGONIST ACTIVITY OF THE BETA2 ADRENERGIC RECEIVER AND ANTAGONIST OF THE MUSCARINE RECEIVER.
WO2005080375A1 (en) * 2004-02-13 2005-09-01 Theravance, Inc. Crystalline form of a biphenyl compound
US7501442B2 (en) * 2004-03-11 2009-03-10 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
JP2007528408A (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
WO2005087735A1 (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
EP1723113A1 (en) * 2004-03-11 2006-11-22 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
US7071224B2 (en) * 2004-03-11 2006-07-04 Theravance, Inc. Diphenylmethyl compounds useful as muscarinic receptor antagonists
TW200538095A (en) * 2004-03-11 2005-12-01 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
WO2006023460A2 (en) * 2004-08-16 2006-03-02 Theravance, Inc. COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY
TWI374883B (en) * 2004-08-16 2012-10-21 Theravance Inc Crystalline form of a biphenyl compound
JP2008510014A (en) * 2004-08-16 2008-04-03 セラヴァンス, インコーポレーテッド Compounds having β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity
TW200811105A (en) * 2006-04-25 2008-03-01 Theravance Inc Dialkylphenyl compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
TW200811104A (en) * 2006-04-25 2008-03-01 Theravance Inc Crystalline forms of a dimethylphenyl compound

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (137)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9108964B2 (en) 2002-08-21 2015-08-18 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US9556175B2 (en) 2002-08-21 2017-01-31 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and thier use as pharmaceutical compositions
US9321791B2 (en) 2002-08-21 2016-04-26 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US10202383B2 (en) 2002-08-21 2019-02-12 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US10023574B2 (en) 2002-08-21 2018-07-17 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
JP2008510014A (en) * 2004-08-16 2008-04-03 セラヴァンス, インコーポレーテッド Compounds having β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity
WO2006023460A3 (en) * 2004-08-16 2006-04-06 Theravance Inc COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY
US7569586B2 (en) 2004-08-16 2009-08-04 Theravance, Inc. Compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity
US8143277B2 (en) 2004-08-16 2012-03-27 Theravance, Inc. Method of using compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity
EP2292619A1 (en) 2004-10-22 2011-03-09 Novartis AG Purine derivatives for use as adenonsin A-2A receptor agonists
WO2006050239A3 (en) * 2004-10-29 2006-10-12 Glaxo Group Ltd Muscarinic acetylcholine receptor antagonists
US8883805B2 (en) 2004-11-05 2014-11-11 Boehringer Ingelheim International Gmbh Process for the preparation of chiral 8-(3-aminopiperidin-1-yl)-xanthines
US9499546B2 (en) 2004-11-05 2016-11-22 Boehringer Ingelheim International Gmbh Process for the preparation of chiral 8-(3-aminopiperidin-1-yl)-xanthines
US9751855B2 (en) 2004-11-05 2017-09-05 Boehringer Ingelheim International Gmbh Process for the preparation of chiral 8-(3-aminopiperidin-1-yl)-xanthines
EP2305659A1 (en) 2004-11-29 2011-04-06 Novartis AG 5-hydroxy-benzothiazole derivatives having beta-2-adrenoreceptor agonist activity
WO2006056471A1 (en) 2004-11-29 2006-06-01 Novartis Ag 5-hydroxy-benzothiazole derivatives having beta-2-adrenorecptor agonist activity
EP2253612A1 (en) 2005-04-14 2010-11-24 Novartis AG Organic compounds
WO2006108643A2 (en) 2005-04-14 2006-10-19 Novartis Ag Organic compounds
US7994211B2 (en) 2005-08-08 2011-08-09 Argenta Discovery Limited Bicyclo[2.2.1]hept-7-ylamine derivatives and their uses
EP2280006A1 (en) 2005-08-08 2011-02-02 Pulmagen Therapeutics (Synergy) Limited Pharmaceutical composition for inhalation comprising an oxazole or thiazole m3 muscarinic receptor antagonist
EP2281813A1 (en) 2005-08-08 2011-02-09 Pulmagen Therapeutics (Synergy) Limited Bicyclo[2.2.1]hept-7-ylamine derivatives and their uses
EP2532679A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
EP2532677A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
EP2286813A2 (en) 2006-01-31 2011-02-23 Novartis AG Use of naphthyridine derivatives as medicaments
WO2007090859A1 (en) * 2006-02-10 2007-08-16 Glaxo Group Limited Succinic acid salt of biphenyl-2-ylcarbamic acid 1-[2-(2-chloro-4-{ [ (r)-2-hydroxy-2- (8-hydroxy-2-oxo-1, 2-dihydroquinolin-5-yl) eth ylaminoimethyl] } -5-methoxyphenylcarbamoyl) ethyl] piperidin-4-yl ester and its use for the treatment of pulmonary disorders
AU2007213767B2 (en) * 2006-02-10 2012-04-26 Theravance Biopharma R&D Ip, Llc Succinic acid salt of biphenyl-2-ylcarbamic acid 1-[2-(2-chloro-4-{ [ (R)-2-hydroxy-2- (8-hydroxy-2-oxo-1, 2-dihydroquinolin-5-yl) ethylaminoimethyl] } -5-methoxyphenylcarbamoyl) ethyl] piperidin-4-yl ester and its use for the treatment of pulmonary disorders
US7960551B2 (en) 2006-02-10 2011-06-14 Glaxo Group Limited Compound
NO341343B1 (en) * 2006-02-10 2017-10-16 Glaxo Group Ltd Succinic acid salt of biphenyl-2-ylcarbamic acid 1- [2- (2-chloro-4 - {[(R) -2-hydroxy-2- (8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl) ethylamino] methyl] - 5-methoxyphenylcarbamoyl) ethyl] piperidin-4-yl ester, a pharmaceutical composition comprising the succinic salt, its use, method of preparation thereof and combination thereof with an anti-inflammatory agent
EA016580B1 (en) * 2006-02-10 2012-06-29 Глаксо Груп Лимитед Succinic acid salt 1-[2-(2-chloro-4-{[(r)-2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethylamino]methyl]}-5-methoxyphenylcarbamoyl)ethyl]piperidin-4-yl ester biphenyl-2-ylcarbamic acid and its use for the treatment of pulmonary
KR101488691B1 (en) * 2006-02-10 2015-02-02 글락소 그룹 리미티드 Succinic acid salt of biphenyl-2-ylcarbamic acid 1-〔2-(2-chloro-4-{[(r)-2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethylamino]methyl}-5-methoxyphenylcarbamoyl)ethyl〕piperidin-4-yl ester and its use for the treatment of pulmonary disorders
US7612084B2 (en) 2006-03-20 2009-11-03 Pfizer Inc Amine derivatives for the treatment of asthma and COPD
EP2322525A1 (en) 2006-04-21 2011-05-18 Novartis AG Purine derivatives for use as adenosin A2A receptor agonists
WO2007121920A2 (en) 2006-04-21 2007-11-01 Novartis Ag Purine derivatives for use as adenosin a2a receptor agonists
JP2009535340A (en) * 2006-04-25 2009-10-01 セラヴァンス, インコーポレーテッド Crystalline form of dimethylphenyl compounds
US7880010B2 (en) 2006-04-25 2011-02-01 Theravance, Inc. Crystalline forms of a dimethylphenyl compound
JP2009541209A (en) * 2006-04-25 2009-11-26 セラヴァンス, インコーポレーテッド Dialkylphenyl compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2007127297A3 (en) * 2006-04-25 2007-12-21 Theravance Inc Crystalline forms of a dimethylphenyl compound
WO2007127297A2 (en) * 2006-04-25 2007-11-08 Theravance, Inc. Crystalline forms of a dimethylphenyl compound
US9815837B2 (en) 2006-05-04 2017-11-14 Boehringer Ingelheim International Gmbh Polymorphs
US11919903B2 (en) 2006-05-04 2024-03-05 Boehringer Ingelheim International Gmbh Polymorphs
US9266888B2 (en) 2006-05-04 2016-02-23 Boehringer Ingelheim International Gmbh Polymorphs
US9173859B2 (en) 2006-05-04 2015-11-03 Boehringer Ingelheim International Gmbh Uses of DPP IV inhibitors
US9493462B2 (en) 2006-05-04 2016-11-15 Boehringer Ingelheim International Gmbh Polymorphs
US10080754B2 (en) 2006-05-04 2018-09-25 Boehringer Ingelheim International Gmbh Uses of DPP IV inhibitors
US10301313B2 (en) 2006-05-04 2019-05-28 Boehringer Ingelheim International Gmbh Polymorphs
US11033552B2 (en) 2006-05-04 2021-06-15 Boehringer Ingelheim International Gmbh DPP IV inhibitor formulations
US11084819B2 (en) 2006-05-04 2021-08-10 Boehringer Ingelheim International Gmbh Polymorphs
US11291668B2 (en) 2006-05-04 2022-04-05 Boehringer Ingelheim International Gmbh Uses of DPP IV inhibitors
WO2008041095A1 (en) * 2006-10-04 2008-04-10 Pfizer Limited Sulfonamide derivatives as adrenergic agonists and muscarinic antagonists
EP2279777A2 (en) 2007-01-10 2011-02-02 Irm Llc Compounds and compositions as channel activating protease inhibitors
EP2332933A1 (en) 2007-05-07 2011-06-15 Novartis AG Epithelial sodium channel (ENaC) inhibitors
EP2444120A1 (en) 2007-12-10 2012-04-25 Novartis AG Spirocyclic amiloride analogues as ENac blockers
EP2520574A1 (en) 2007-12-10 2012-11-07 Novartis AG Amiloride analogues substituted on the cyclic guanidine moiety as ENaC blockers for treating respiratory diseases
WO2009087224A1 (en) 2008-01-11 2009-07-16 Novartis Ag Pyrimidines as kinase inhibitors
US9415016B2 (en) 2008-04-03 2016-08-16 Boehringer Ingelheim International Gmbh DPP-IV inhibitor combined with a further antidiabetic agent, tablets comprising such formulations, their use and process for their preparation
US10973827B2 (en) 2008-04-03 2021-04-13 Boehringer Ingelheim International Gmbh DPP-IV inhibitor combined with a further antidiabetic agent, tablets comprising such formulations, their use and process for their preparation
US10022379B2 (en) 2008-04-03 2018-07-17 Boehringer Ingelheim International Gmbh DPP-IV inhibitor combined with a further antidiabetic agent, tablets comprising such formulations, their use and process for their preparation
WO2009150137A2 (en) 2008-06-10 2009-12-17 Novartis Ag Organic compounds
KR101235962B1 (en) * 2008-07-11 2013-02-21 화이자 리미티드 Triazole derivatives useful for the treatment of diseases
WO2010004517A1 (en) * 2008-07-11 2010-01-14 Pfizer Limited Triazole derivatives useful for the treatment of diseases
JP2011527672A (en) * 2008-07-11 2011-11-04 ファイザー・リミテッド Triazole derivatives useful for treating diseases
US8263623B2 (en) 2008-07-11 2012-09-11 Pfizer Inc. Triazol derivatives useful for the treatment of diseases
US10034877B2 (en) 2008-08-06 2018-07-31 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients inappropriate for metformin therapy
US9486526B2 (en) 2008-08-06 2016-11-08 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients inappropriate for metformin therapy
US11911388B2 (en) 2008-10-16 2024-02-27 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients with insufficient glycemic control despite therapy with an oral or non-oral antidiabetic drug
US9212183B2 (en) 2008-12-23 2015-12-15 Boehringer Ingelheim International Gmbh Salt forms of 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine
WO2010119064A1 (en) 2009-04-14 2010-10-21 Glaxo Group Limited Process for the preparation of a biphenyl-2-ylcarbamic acid ester
WO2011050325A1 (en) 2009-10-22 2011-04-28 Vertex Pharmaceuticals Incorporated Compositions for treatment of cystic fibrosis and other chronic diseases
EP2813227A1 (en) 2009-10-22 2014-12-17 Vertex Pharmaceuticals Incorporated Compositions for treatment of cystic fibrosis and other chronic diseases
US10092571B2 (en) 2009-11-27 2018-10-09 Boehringer Ingelheim International Gmbh Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US9457029B2 (en) 2009-11-27 2016-10-04 Boehringer Ingelheim International Gmbh Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
WO2011081937A1 (en) 2009-12-15 2011-07-07 Gilead Sciences, Inc. Corticosteroid-beta-agonist-muscarinic antagonist compounds for use in therapy
EP2845593A1 (en) 2010-03-19 2015-03-11 Novartis AG Pyridine and pyrazine derivative for the treatment of chronic obstructive pulmonary disease
WO2011113894A1 (en) 2010-03-19 2011-09-22 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf
US10004747B2 (en) 2010-05-05 2018-06-26 Boehringer Ingelheim International Gmbh Combination therapy
US9603851B2 (en) 2010-05-05 2017-03-28 Boehringer Ingelheim International Gmbh Combination therapy
US9643961B2 (en) 2010-05-13 2017-05-09 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic antagonist and M3 muscarinic antagonist activities
US9315463B2 (en) 2010-05-13 2016-04-19 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
WO2012007539A1 (en) 2010-07-14 2012-01-19 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2012032546A3 (en) * 2010-09-08 2012-06-28 Cadila Healthcare Limited Process for the preparation of salmeterol and its intermediates
WO2012034095A1 (en) 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
WO2012034091A1 (en) 2010-09-09 2012-03-15 Irm Llc Imidazo [1, 2] pyridazin compounds and compositions as trk inhibitors
WO2012035158A1 (en) 2010-09-17 2012-03-22 Novartis Ag Pyrazine derivatives as enac blockers
US9034883B2 (en) 2010-11-15 2015-05-19 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
US11911387B2 (en) 2010-11-15 2024-02-27 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
WO2012116217A1 (en) 2011-02-25 2012-08-30 Irm Llc Compounds and compositions as trk inhibitors
WO2012168349A1 (en) 2011-06-10 2012-12-13 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2012168359A1 (en) 2011-06-10 2012-12-13 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US9012644B2 (en) 2011-06-10 2015-04-21 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US8883800B2 (en) 2011-07-15 2014-11-11 Boehringer Ingelheim International Gmbh Substituted quinazolines, the preparation thereof and the use thereof in pharmaceutical compositions
US8962636B2 (en) 2011-07-15 2015-02-24 Boehringer Ingelheim International Gmbh Substituted quinazolines, the preparation thereof and the use thereof in pharmaceutical compositions
US9199998B2 (en) 2011-07-15 2015-12-01 Boehringer Ingelheim Internatioal Gmbh Substituted quinazolines, the preparation thereof and the use thereof in pharmaceutical compositions
WO2013030802A1 (en) 2011-09-01 2013-03-07 Novartis Ag Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
WO2013038386A1 (en) 2011-09-16 2013-03-21 Novartis Ag Heterocyclic compounds for the treatment of cystic fibrosis
WO2013038390A1 (en) 2011-09-16 2013-03-21 Novartis Ag N-substituted heterocyclyl carboxamides
WO2013038378A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013038381A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine/pyrazine amide derivatives
WO2013038373A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
US9233108B2 (en) 2011-11-11 2016-01-12 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9549934B2 (en) 2011-11-11 2017-01-24 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US9757383B2 (en) 2011-11-11 2017-09-12 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
US10300072B2 (en) 2011-11-11 2019-05-28 Almirall, S.A. Cyclohexylamine derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
WO2013105061A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused dihydropyrido [2,3 -b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105058A1 (en) 2012-01-13 2013-07-18 Novartis Ag 7,8- dihydropyrido [3, 4 - b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105065A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pah and related disorders
WO2013105063A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105057A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused pyrroles as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105066A1 (en) 2012-01-13 2013-07-18 Novartis Ag Salts of an ip receptor agonist
US9555001B2 (en) 2012-03-07 2017-01-31 Boehringer Ingelheim International Gmbh Pharmaceutical composition and uses thereof
WO2013140319A1 (en) 2012-03-19 2013-09-26 Novartis Ag Crystalline form of a succinate salt
US10195203B2 (en) 2012-05-14 2019-02-05 Boehringr Ingelheim International GmbH Use of a DPP-4 inhibitor in podocytes related disorders and/or nephrotic syndrome
US9526730B2 (en) 2012-05-14 2016-12-27 Boehringer Ingelheim International Gmbh Use of a DPP-4 inhibitor in podocytes related disorders and/or nephrotic syndrome
US9713618B2 (en) 2012-05-24 2017-07-25 Boehringer Ingelheim International Gmbh Method for modifying food intake and regulating food preference with a DPP-4 inhibitor
US10040765B2 (en) 2012-09-21 2018-08-07 Crystal Pharma S.A.U. Methods for the preparation of indacaterol and pharmaceutically acceptable salts thereof
EP3345904A1 (en) 2012-12-06 2018-07-11 Chiesi Farmaceutici S.p.a. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2014086927A1 (en) 2012-12-06 2014-06-12 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2014086924A1 (en) 2012-12-06 2014-06-12 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US9518050B2 (en) 2012-12-18 2016-12-13 Almirall, S.A. Cyclohexyl and quinuclidinyl carbamate derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activity
WO2014125413A1 (en) 2013-02-13 2014-08-21 Novartis Ag Ip receptor agonist heterocyclic compounds
US9562039B2 (en) 2013-02-27 2017-02-07 Almirall, S.A. Salts of 2-amino-1-hydroxyethyl-8-hydroxyquinolin-2(1H)-one derivatives having both β2 adrenergic receptor agonist and M3 muscarinic receptor antagonist activities
WO2014132220A1 (en) 2013-03-01 2014-09-04 Novartis Ag Solid forms of bicyclic heterocyclic derivatives as pdgf receptor mediators
WO2015008229A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors
WO2015008230A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors comprising a heteroaromatic ring-benzyl-amide-cycle core
US10456390B2 (en) 2013-07-25 2019-10-29 Almirall, S.A. Combinations comprising MABA compounds and corticosteroids
US9526728B2 (en) 2014-02-28 2016-12-27 Boehringer Ingelheim International Gmbh Medical use of a DPP-4 inhibitor
WO2015162461A1 (en) 2014-04-24 2015-10-29 Novartis Ag Pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162456A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162459A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US10005771B2 (en) 2014-09-26 2018-06-26 Almirall, S.A. Bicyclic derivatives having β2 adrenergic agonist and M3 muscarinic antagonist activities
WO2016128456A1 (en) 2015-02-12 2016-08-18 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2016193241A1 (en) 2015-06-01 2016-12-08 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2017093208A1 (en) 2015-12-03 2017-06-08 Chiesi Farmaceutici S.P.A. Compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
US10155000B2 (en) 2016-06-10 2018-12-18 Boehringer Ingelheim International Gmbh Medical use of pharmaceutical combination or composition
WO2018011090A1 (en) 2016-07-13 2018-01-18 Chiesi Farmaceutici S.P.A. Hydroxyquinolinone compounds having muscarinic receptor antagonist and beta2 adrenergic receptor agonist activity
WO2018108089A1 (en) 2016-12-14 2018-06-21 北京硕佰医药科技有限责任公司 Class of bifunctional compounds with quaternary ammonium salt structure
WO2020250116A1 (en) 2019-06-10 2020-12-17 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf, copd, and bronchiectasis
WO2021038426A1 (en) 2019-08-28 2021-03-04 Novartis Ag Substituted 1,3-phenyl heteroaryl derivatives and their use in the treatment of disease

Also Published As

Publication number Publication date
CA2515777A1 (en) 2004-09-02
RU2330841C2 (en) 2008-08-10
JP2006517978A (en) 2006-08-03
AU2010219338A1 (en) 2010-09-30
KR20110132634A (en) 2011-12-08
US20060229334A1 (en) 2006-10-12
ES2636480T3 (en) 2017-10-05
NO333394B1 (en) 2013-05-27
DE602004029347D1 (en) 2010-11-11
JP2010159291A (en) 2010-07-22
US8618131B2 (en) 2013-12-31
CA2515777C (en) 2013-01-22
DK2246345T3 (en) 2016-05-02
US20070088054A1 (en) 2007-04-19
NO20054206L (en) 2005-10-19
IS7964A (en) 2005-07-28
US20040209915A1 (en) 2004-10-21
MXPA05008528A (en) 2005-10-20
US7507751B2 (en) 2009-03-24
HUE027380T2 (en) 2016-09-28
AU2004213411B2 (en) 2010-06-24
JP5247757B2 (en) 2013-07-24
ES2566155T3 (en) 2016-04-11
IL169922A (en) 2013-08-29
JP2007119496A (en) 2007-05-17
EP1594860A2 (en) 2005-11-16
US20040209860A1 (en) 2004-10-21
TWI331995B (en) 2010-10-21
HK1216892A1 (en) 2016-12-09
US20070037984A1 (en) 2007-02-15
BRPI0407508B8 (en) 2021-05-25
JP2006518739A (en) 2006-08-17
US20140235864A1 (en) 2014-08-21
HK1149548A1 (en) 2011-10-07
WO2004074246A3 (en) 2004-11-18
KR20050102114A (en) 2005-10-25
WO2004074276A1 (en) 2004-09-02
US7524959B2 (en) 2009-04-28
US8969571B2 (en) 2015-03-03
PE20040950A1 (en) 2005-01-01
US20040167167A1 (en) 2004-08-26
AR043176A1 (en) 2005-07-20
US7345175B2 (en) 2008-03-18
KR101223991B1 (en) 2013-01-21
US7141671B2 (en) 2006-11-28
PL378025A1 (en) 2006-02-20
NO20120150L (en) 2005-10-19
US20070276003A1 (en) 2007-11-29
EP2246345A1 (en) 2010-11-03
JP5436503B2 (en) 2014-03-05
US20080015220A1 (en) 2008-01-17
US20060223859A1 (en) 2006-10-05
EP3012254A1 (en) 2016-04-27
EP1615889A2 (en) 2006-01-18
US8242135B2 (en) 2012-08-14
US20070208176A1 (en) 2007-09-06
RU2005128557A (en) 2006-03-20
ATE482934T1 (en) 2010-10-15
US7521561B2 (en) 2009-04-21
WO2004074812A2 (en) 2004-09-02
AU2010219338B2 (en) 2012-07-12
BRPI0407508A (en) 2006-02-14
CY1117459T1 (en) 2017-04-26
SI2246345T1 (en) 2016-06-30
US20060223858A1 (en) 2006-10-05
US7355046B2 (en) 2008-04-08
EP3012254B1 (en) 2017-05-17
US7829583B2 (en) 2010-11-09
HK1086266A1 (en) 2006-09-15
NO331947B1 (en) 2012-05-07
US20110312994A1 (en) 2011-12-22
PL216397B1 (en) 2014-03-31
US20130150404A1 (en) 2013-06-13
KR101224497B1 (en) 2013-01-23
EP1615889B1 (en) 2010-09-29
CY1111276T1 (en) 2015-08-05
NO20054206D0 (en) 2005-09-09
PT1615889E (en) 2010-12-09
KR20110053393A (en) 2011-05-20
JP4555283B2 (en) 2010-09-29
IS2847B (en) 2013-09-15
WO2004074276B1 (en) 2004-10-07
BRPI0407508B1 (en) 2015-12-01
AU2004213411A1 (en) 2004-09-02
JP2006517971A (en) 2006-08-03
US20060223860A1 (en) 2006-10-05
US7514558B2 (en) 2009-04-07
WO2004074812A3 (en) 2004-11-04
US7879879B2 (en) 2011-02-01
MY148487A (en) 2013-04-30
TW200424173A (en) 2004-11-16
JP2011225617A (en) 2011-11-10
EP1592685A1 (en) 2005-11-09
SI1615889T1 (en) 2011-01-31
EP2246345B1 (en) 2016-02-10
DK1615889T3 (en) 2010-12-20
NZ541579A (en) 2008-08-29

Similar Documents

Publication Publication Date Title
EP2246345B1 (en) Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
EP1778626A1 (en) Compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 169922

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2004213411

Country of ref document: AU

Ref document number: 3375/DELNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 541579

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2005/06215

Country of ref document: ZA

Ref document number: 200506215

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 05079299

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2515777

Country of ref document: CA

Ref document number: PA/a/2005/008528

Country of ref document: MX

Ref document number: 378025

Country of ref document: PL

WWE Wipo information: entry into national phase

Ref document number: 2006503604

Country of ref document: JP

Ref document number: 1020057014929

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2004213411

Country of ref document: AU

Date of ref document: 20040213

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004213411

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004711253

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20048065284

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2005128557

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 1020057014929

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1-2005-501486

Country of ref document: PH

WWP Wipo information: published in national office

Ref document number: 2004711253

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0407508

Country of ref document: BR