WO2007127196A2 - DIALKYLPHENYL COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY - Google Patents

DIALKYLPHENYL COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY Download PDF

Info

Publication number
WO2007127196A2
WO2007127196A2 PCT/US2007/009925 US2007009925W WO2007127196A2 WO 2007127196 A2 WO2007127196 A2 WO 2007127196A2 US 2007009925 W US2007009925 W US 2007009925W WO 2007127196 A2 WO2007127196 A2 WO 2007127196A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
ethyl
methyl
assay
Prior art date
Application number
PCT/US2007/009925
Other languages
French (fr)
Other versions
WO2007127196A3 (en
Inventor
Pierre-Jean Colson
Adam Hughes
Craig Husfeld
Mathai Mammen
Miroslav Rapta
Original Assignee
Theravance, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Theravance, Inc. filed Critical Theravance, Inc.
Priority to MX2008013555A priority Critical patent/MX2008013555A/en
Priority to JP2009507757A priority patent/JP2009541209A/en
Priority to ES07776093T priority patent/ES2391584T3/en
Priority to EP07776093A priority patent/EP2010489B1/en
Priority to CA002650530A priority patent/CA2650530A1/en
Priority to BRPI0710767-6A priority patent/BRPI0710767A2/en
Priority to AU2007243481A priority patent/AU2007243481A1/en
Publication of WO2007127196A2 publication Critical patent/WO2007127196A2/en
Publication of WO2007127196A3 publication Critical patent/WO2007127196A3/en
Priority to IL194334A priority patent/IL194334A0/en
Priority to NO20084603A priority patent/NO20084603L/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
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to novel dialkylphenyl compounds having ⁇ 2 adrenergic receptor agonist and muscarinic receptor antagonist activity.
  • This invention also relates to pharmaceutical compositions comprising such compounds, processes and intermediates for preparing such compounds and methods of using such compounds, for example, to treat pulmonary disorders.
  • bronchodilators Pulmonary disorders, such as asthma and chronic obstructive pulmonary disease (COPD), are commonly treated with bronchodilators.
  • bronchodilator used to treat pulmonary disorders 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 a combination of a ⁇ 2 adrenergic receptor agonist and a muscarinic receptor antagonist are known in the art for use in treating pulmonary disorders.
  • a muscarinic receptor antagonist such as tiotropium bromide
  • a ⁇ 2 adrenergic receptor agonist such as formoterol fumarate.
  • the present invention provides novel dialkylphenyl compounds having both ⁇ 2 adrenergic receptor agonist and muscarinic receptor antagonist activity.
  • a compound of this invention has been found to possess a long duration of action, i.e., a duration of at least about 24 hours, when administered to a mammal by inhalation. Accordingly, the compounds of this invention are expected to be useful and advantageous as therapeutic agents for treating pulmonary disorders.
  • the present invention relates to a compound of formula I:
  • R 1 is methyl or ethyl
  • R 2 is methyl or ethyl
  • the compound of formula I is a compound having formula II:
  • R 1 and R 2 are as defined herein (including any specific or preferred embodiments); or a pharmaceutically acceptable salt or solvate thereof.
  • this invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formula I.
  • this invention relates to a pharmaceutical composition comprising (a) a compound of formula I; and (b) a second therapeutic agent.
  • this invention relates to pharmaceutical composition comprising (a) a compound of formula I; (b) a second therapeutic agent; and (c) a pharmaceutically acceptable carrier.
  • this invention relates to a combination of therapeutic agents, the combination comprising (a) a compound of formula I; and (b) a second therapeutic agent.
  • this invention relates to a combination of pharmaceutical compositions, the combination comprising (a) a first pharmaceutical composition comprising a compound of formula I and a first pharmaceutically acceptable carrier; and (b) a second pharmaceutical composition comprising a second therapeutic agent and a second pharmaceutically acceptable carrier.
  • This invention also relates to a kit containing such pharmaceutical compositions.
  • Compounds of this invention possess both ⁇ 2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity.
  • this invention relates 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.
  • This invention also relates to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient a therapeutically effective amount of a compound of formula I.
  • this invention relates to a method of producing bronchodilation in a mammal, the method comprising administering to a mammal a bronchodilation-producing amount of a compound of formula I.
  • This invention also relates to method of antagonizing a muscarinic receptor and agonizing a ⁇ 2 adrenergic receptor in a mammal, the method comprising administering to the mammal a compound of formula I.
  • this invention relates to a method of using a compound of formula I as a research tool, the method comprising conducting a biological assay using a compound of formula I.
  • this invention relates to a method of evaluating a test compound in a biological assay, the method comprising: (a) conducting a biological assay with a test compound to provide a first assay value; (b) conducting the biological assay with a compound of formula I to provide a second assay value; wherein step (a) is conducted either before, after or concurrently with step (b); and (c) comparing the first assay value from step (a) with the second assay value from step
  • this invention also relates to processes and novel intermediates useful for preparing compounds of formula I. Accordingly, in another of its method aspects, this invention relates to a process of preparing a compound of formula I, the process comprising deprotecting a compound of formula 6 (as defined herein) to provide a compound of formula I. In another of its method aspects, this invention relates to a process of preparing a compound of formula I, the process comprising: (a) reacting a compound of formula 4 with a compound of formula 5 in the presence of a reducing agent to provide a compound of formula 6; and (b) deprotecting the compound of formula 6 to provide a compound of formula I; where compounds 4, 5 and 6 are as defined herein.
  • the compounds of formula I are prepared by deprotecting a compound of formula 6, wherein the hydroxyl-protecting group is a silyl group. Accordingly, in yet another of its method aspects, this invention relates to a process of preparing a compound of formula I, the process comprising deprotecting a compound of formula 6a:
  • R a , R b and R° are independently selected from CM alkyl, phenyl, -Ci -4 alkyl-(phenyl), or one of R la , R lb and R lc is -O-(Ci- 4 alkyl); to provide a compound of formula I.
  • the processes described herein further comprise the step of forming a pharmaceutically acceptable salt of a compound of formula I.
  • this invention relates to the other processes described herein; and to the product prepared by any of the processes described herein.
  • this invention relates to a compound of formula III:
  • Y 1 is selected from -CHO, -CN, -CH 2 OH, -CH(OR 3a )OR 3b , -C(O)OH, -C(O)OR 3c , bromo and iodo, where R 3a and R 3b are selected independently from Ci- 6 alkyl, or R 3a and R 3b are joined to form C 2-6 alkylene, R 3c is selected from Ci- ⁇ alkyl; and R 1 and R 2 are as defined herein (including any specific or preferred embodiments), which compounds are useful as intermediates in preparing compounds of formula I.
  • R 1 and R 2 are methyl.
  • Y 1 is -CHO.
  • R 1 and R 2 are methyl and Y 1 is -CHO.
  • This invention also relates to the use of a compound of formula I for therapy. Additionally, the invention relates to the use of a compound of formula I for the manufacture of a medicament for the treatment of a pulmonary disorder; and to the use of a compound of formula I as a research tool. Other aspects and embodiments of this invention are disclosed herein.
  • this invention relates to novel compounds of formula I.
  • the compounds of formula I 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.
  • 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 of the composition as a whole is not eliminated by the presence of such other isomers.
  • compounds of formula I contain a chiral center at the carbon atom indicated by the symbol * in the following partial formula:
  • the carbon atom identified by the symbol * has the (R) configuration.
  • the compounds of formula I have the (R) configuration at the carbon atom identified by the symbol * or are enriched in a stereoisomer ⁇ form having the (R) configuration at this carbon atom.
  • 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 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.
  • references to a compound herein for example, reference to a compound of formula I or compound 6, includes reference to salts and stereoisomers and solvates of that compound unless otherwise indicated.
  • R 1 is methyl and R 2 is methyl. In another embodiment, R 1 is ethyl and R 2 is ethyl. In another embodiment, R 1 is methyl and R 2 is ethyl. In another embodiment, R 1 is ethyl and R 2 is methyl.
  • the present invention relates to compounds of formula I selected from: biphenyl-2-ylcarbamic acid 1 -[2-(4- ⁇ [(i?)-2-(3-formylammo-4-hydroxyphenyl)-2- hydroxyethylamino]methyl ⁇ -2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ha); biphenyl-2-ylcarbamic acid 1 -[2-(4- ⁇ [(2?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylaminojmethyl ⁇ -2,5-diethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound lib); biphenyl-2-ylcarbamic acid 1 -[2-(4- ⁇ [(/2)-2-(3-formylamino-4-hydroxyphenyl)-2-
  • alkyl means a monovalent saturated hydrocarbon group which may be linear or branched. Unless otherwise defined, such alkyl groups typically contain from 1 to 10 carbon atoms. Representative alkyl groups include, by way of example, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, «-pentyl, ⁇ -hexyl, n-heptyl, n- octyl, n-nonyl, w-decyl and the like.
  • Carbon atoms When a specific number of carbon atoms is intended for a particular term used herein, the number of carbon atoms is shown preceding the term.
  • the term "Ci- 3 alkyl” means an alkyl group having from 1 to 3 carbon atoms wherein the carbon atoms are in any chemically-acceptable configuration.
  • alkylene means a divalent saturated hydrocarbon group that 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- 1,5 -diyl and the like.
  • ethylene ethane- 1 ,2-diyl
  • propane- 1 ,3 -diyl propane- 1 ,3 -diyl
  • butane- 1 ,4- diyl pentane- 1,5 -diyl and the like.
  • 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), benzyl, 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), /7-methoxybenzyl (PMB) 5 9-fluorenylmethyl (Fm), trimethylsilyl (TMS), tert- butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like.
  • compound of the invention or “compound of formula I” or “compound of formula II” as used herein means the specified compound(s) or a pharmaceutically acceptable salt or solvate or stereoisomer thereof unless otherwise indicated.
  • halo means fluoro, chloro, bromo and iodo.
  • 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(Ci- 6 alkyl)silyl groups, such as trimethylsilyl (TMS), triethylsilyl (TES), t ⁇ t-butyldimethylsilyl (TBS) and the like; esters (acyl groups) including Ci -e alkanoyl groups, such as formyl, acetyl and the like; arylmethyl groups, such as benzyl (Bn), /j-methoxybenzyl (PMB) 5 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.
  • leaving group means a functional group or an atom that can be displaced by another functional group or atom in a substitution reaction, such as a micleophilic substitution reaction.
  • representative leaving groups include, but are not limited to, 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.
  • MMD mass median diameter
  • micronized or “in micronized form” means particles in which at least about 90 percent of the particles have a diameter of less than about 10 ⁇ m unless otherwise indicated.
  • pharmaceutically acceptable salt or solvate or stereoisomer thereof as used herein 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.
  • pharmaceutically acceptable salt means a salt that is acceptable for administration to a patient, such as a mammal (e.g., salts having acceptable mammalian safety for a given dosage regime).
  • Representative pharmaceutically acceptable salts include salts of 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-l,5-disulfonic, naphthalene-2,6- disulfonic, nicotinic, nitric, orotic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic and xinafoic acid, and the like.
  • protected derivatives thereof means a derivative of the specified compound in which one or more functional groups of the compound are protected or blocked from undergoing undesired reactions with a protecting or blocking group.
  • Functional groups that may be protected include, by way of example, carboxy groups, amino groups, hydroxyl groups, thiol groups, carbonyl groups and the like. Suitable protecting groups for such functional groups are well known to those of ordinary skill in the art as exemplified by the teachings in T. W. Greene and G. M. Wuts, Protecting
  • 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 cation typically comprises a protonated form of a compound of formula I, i.e. where one or more amino groups having been protonated by an acid.
  • the salt is a pharmaceutically acceptable salt, although this is not required for salts of intermediate compounds that are not intended for administration to a patient.
  • solvate means a complex or aggregate formed by one or more molecules of a solute, i.e. a compound of formula I or a pharmaceutically acceptable salt thereof, and one or more molecules of a solvent.
  • solvates are typically crystalline solids having a substantially fixed molar ratio of solute and solvent.
  • Representative solvents include, by way of example, water, methanol, ethanol, isopropanol, acetic acid and the like. When the solvent is water, the solvate formed is a hydrate.
  • terapéuticaally effective amount means an amount sufficient to effect treatment when administered to a patient in need of treatment.
  • treating or treatment means the treating or treatment of a disease or medical condition (such as COPD or asthma) in a patient, such as a mammal (particularly a human) that includes:
  • the compounds 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 or known by those of ordinary skill in the art. Although the following procedures may illustrate a particular embodiment or aspect of the present invention, those skilled in the art will recognize that other embodiments or aspects of the present invention can be prepared using the same or similar methods or by using other methods, reagents and starting materials known to those skilled in the art. It will also be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. While optimum reaction conditions will typically vary depending on various reaction parameters such as the reactants, solvents and quantities used, those of ordinary skill in the art can readily determine suitable reaction conditions using routine optimization procedures.
  • process conditions i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.
  • compound I 1 can be reacted with about 0.95 to about 1.05 molar equivalents of compound 2 to provide compound 3.
  • This Michael reaction is typically conducted at a temperature ranging from about 0 0 C to about 15 0 C, typically about 40 0 C to about 45 0 C, for about 8 to about 24 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as dichloromethane or mixtures of dichloromethane and methanol or ethanol, and the like.
  • the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • reaction mixture containing compound 3 can be used directly in the next step of the synthesis.
  • Compound 3 is then reacted with aqueous acid to hydrolyze the acetal group and provide aldehyde compound 4.
  • Any suitable acid can be employed in this reaction including, byway of example, hydrochloric acid, sulfuric acid, methanesulfonic acid, p-toluenesulfonic acid and the like.
  • the hydrolysis reaction is typically conducted at a temperature ranging from about 0 0 C to about 30 0 C, typically about 20 0 C to about 25 0 C, for about 1 to about 6 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as methanol, ethanol, isopropanol, dichloromethane/ethanol, acetonitrile and the like.
  • the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • Compound 4 is then reacted with about 0.95 to about 1.5 molar equivalents of compound 5 in the presence of a reducing agent to afford compound 6.
  • a reducing agent may be used in this reaction including, by way of illustration, a metal ' hydride reagent, such as sodium borohydride, sodium triacetoxyborohydride, sodium cyanoborohydride and the like, or hydrogen and a metal catalyst, such as palladium on carbon, and the like.
  • This reductive alkylation reaction is typically conducted at a temperature ranging from about -20 0 C to about 30 0 C, typically about 0 0 C to about 5 0 C, for about 1 to about 6 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent and a protic solvent, such as dichloroethane and methanol, and the like.
  • a protic solvent such as dichloroethane and methanol, and the like.
  • the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • Compound 6 is then deprotected to provide a compound of formula I.
  • the particular conditions used to deprotect compound 6 will depend on the protecting group employed.
  • P 1 is a silyl protecting group (i.e., a compound of formula 6a as defined herein), such as tert-butyldimethylsilyl, tert-butyldiphenylsilyl, diphenylmethylsilyl, di-tert-buylmethylsilyl, tert-butoxydiphenylsilyl and the like
  • this deprotection reaction is typically conducted by contacting compound 6a with a source of fluoride ion.
  • the source of fluoride ion is triethylamme trihydrofluoride.
  • suitable sources of fluoride ion include tetrabutylammonium fluoride, potassium fluoride with 18-crown-6, hydrogen fluoride, pyridine hydrofluoride, and the like.
  • This reaction is typically conducted at a temperature ranging from about 0 0 C to about 50 0 C, typically about 10 0 C to about 25 0 C, for about 24 to about 72 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as dichloroethane and the like.
  • the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • Compound 1 is known in the art or can be prepared from commercially available starting materials and reagents using known procedures. See, for example, U.S. Patent Application Publication No. U.S. 2004/0167167 Al and R. Naito et al., Chem. Pharm. Bull., 46(8) 1286-1294 (1998).
  • compound 1_ can be prepared as illustrated in Scheme II:
  • biphenyl-2-isocyanate (7) is reacted with an //-protected 4- hydroxypiperidine 8, where P 2 is an amino-protecting group such as benzyl, to provide carbamate intermediate 9.
  • This reaction is typically conducted at a temperature ranging from about 20 0 C to about 100 0 C, typically about 60 0 C to about 80 0 C, for about 6 to about 24 hours or until the reaction is substantially complete.
  • this reaction can be conducted in a suitable diluent, such as dichloromethane, toluene and the like. Alternatively, this reaction can be conducted in the absence of a diluent.
  • the product 9 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like. Alternatively, the reaction mixture containing compound 9 is used directly in the next step of the synthesis.
  • the amino-protecting group, P 2 is then removed from compound 9 using conventional procedures to afford compound I 1 .
  • P 2 is a benzyl group
  • compound 9 can be deprotected using hydrogen or ammonium formate, in the presence of a catalyst, such as a palladium catalyst.
  • Representative catalysts include, by way of illustration, palladium on carbon, palladium hydroxide on carbon and the like.
  • This reaction is typically conducted at a temperature ranging from about 20 0 C to about 50 0 C, typically about 40 0 C, for about 6 to about 24 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as methanol, ethanol, isopropanol and the like.
  • compound 1 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • an aniline compound of formula 10, where X 1 is bromo or iodo is first protected at the amino group to provide a compound of formula IjL-, where P a is an amino-protecting group and P 3b is hydrogen or an arnino-protecting group.
  • Any suitable amino-protecting group may be used, such as benzyl, 4-methoxybenzyl, trifluoroacetyl and the like.
  • a compound of formula 10 can be reacted with about 2 or more molar equivalents, preferably about 2.5 to about 3.0 molar equivalents, of a benzyl halide, such as benzyl chloride, bromide or iodide, to afford compound 11 , where P 3a and P 3b are both benzyl.
  • This reaction is typically conducted at a temperature ranging from about 0 0 C to about 50 0 C, typically about 30 0 C, for about 18 to about 24 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as methanol, ethanol, isopropanol and the like.
  • this reaction is also conducted in the presence of a suitable base, such as potassium carbonate, sodium carbonate and the like.
  • a suitable base such as potassium carbonate, sodium carbonate and the like.
  • compound 11. is typically isolated using conventional procedures, such as extraction, recrystallization, . chromatography and the like.
  • Representative compounds of formula 10 that can be employed in this reaction include 2,5-d ⁇ methyl-4-iodoaniline, 2,5-diethyl-4-iodoaniline, 2-ethyl-4-iodo-5- methylaniline, 5-ethyl-4-iodo-2-methylaniline, 4-bromo-2,5-dimethylaniline, 4-bromo-2,5- diethylaniline, 4-bromo-2-ethyl-5-methylaniline, 4-bromo-5-ethyl-2-methylaniline and the like.
  • Such compounds are commercially available (for example, from Spectra Group Limited, Inc., Millbury, OH) or can be prepared from commercially available starting materials and reagents using conventional procedures.
  • Compound IJ is then contacted with about 1 to about 2 molar equivalents of an alkyl lithium reagent, such as «-butyllithium or t ⁇ rt-butyllithium, to form the corresponding anion in which the X 1 group has been exchanged for lithium.
  • an alkyl lithium reagent such as «-butyllithium or t ⁇ rt-butyllithium
  • This reaction is typically conducted at a temperature ranging from about -70 0 C to about 0 °C, typically about -20 0 C, for about 0.25 to about 1 hour or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as toluene, xylene, tetrahydrofuran and the like.
  • the resulting lithium anion is not isolated, but is reacted in situ with a molar excess of TVyV-dimethylformamide to provide compound IJ2.
  • a molar excess of TVyV-dimethylformamide to provide compound IJ2.
  • about 2 to about 4 molar equivalents of iV ⁇ V-dimethylformamide are used.
  • This reaction is typically conducted at a temperature ranging from about -70 0 C to about 0 0 C, typically about -20 0 C to about 0 0 C, for about 0.5 to about 2 hours or until the reaction is substantially complete.
  • compound 12 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • the aldehyde group of compound 12_ is then protected as an acetal by reacting compound 12_ with an alcohol or a diol in the presence of an acid catalyst.
  • Any suitable alcohol or diol can be used in this reaction.
  • representative alcohols and diols include methanol, ethanol, n-propanol, ethylene glycol, propylene glycol and the like.
  • a molar excess of the alcohol or diol are employed in this reaction, preferably about 2 to about 4 molar equivalents.
  • Any suitable acid catalyst may be used in this reaction to facilitate formation of the acetal.
  • Representative acid catalysis include, by way of example, />-toluenesulfonic acid, benzenesulfonic acid, hydrochloric acid and the like.
  • the reaction is typically conducted at a temperature ranging from about 50 0 C to about 100 0 C, typically about 60 0 C to about 80 0 C, for about 12 to about 24 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as toluene, xylene and the like.
  • the reaction is conducted in a manner which allows the water produced to be removed, such as by azeotropic distillation or by the use of molecular sieves.
  • compound ,13 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • the reaction mixture containing compound jL3 can be used directly in the next step of the synthesis.
  • compound 13 After formation of the acetal, the amino group of compound 13 is deprotected using standard reagents and conditions to form compound 14.
  • a catalyst such as a palladium catalyst.
  • Representative catalysts include, by way of example, palladium on carbon, palladium hydroxide on carbon, and the like. This reaction is typically conducted at a temperature ranging from about 20 0 C to about 50 0 C, typically about 25 0 C to about 30 0 C, for about 4 to about 12 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as methanol, ethanol, ethanol/ethyl acetate mixtures and the like.
  • a suitable diluent such as methanol, ethanol, ethanol/ethyl acetate mixtures and the like.
  • compound 14 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
  • Compound 14 is then reacted with an acryloyl halide 15, where X 2 is chloro, bromo or iodo, to form compound 2.
  • This reaction is typically conducted at a temperature ranging from about -20 0 C to about 25 0 C 5 typically about 0 0 C to about 5 0 C, for about 0.5 to about 6 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as dichloromethane and the like, in the presence of a suitable base, such as diisopropylethylamine, triethylamine and the like.
  • a suitable base such as diisopropylethylamine, triethylamine and the like.
  • a suitable base such as diisopropylethylamine, triethylamine and the like.
  • a suitable base such as diisopropylethylamine, triethylamine and the like.
  • a suitable base such as diisopropylethylamine, triethylamine and the like.
  • compounds of formula 5 can be prepared from compounds of formula 16, where P 4 is a hydroxyl -protecting group, such as benzyl, and X 3 is a leaving group, such as chloro, bromo or iodo.
  • P 4 is a hydroxyl -protecting group, such as benzyl
  • X 3 is a leaving group, such as chloro, bromo or iodo.
  • Compounds of formula 16 are known in the art. For example, U.S. Patent No. 6,268,533 Bl, issued July 31, 2001; and R.
  • the hydroxyl group of compound 16 is protected using conventional procedures and reagents to provide compound 17, where P 1 is a hydroxyl-protecting group.
  • the hydroxyl-protecting group is a silyl protecting group, such as dimethylisopropylsilyl, diethylisopropylsilyl, dimethylhexylsilyl, tert-butyldimethylsilyl, terf-butyldiphenylsilyl, diphenylmethylsilyl and the like.
  • compound 16 can be reacted with about 0.95 to about 1.2 molar equivalent of tert-butyldimethylsilyl chloride in the presence of about 1.1 to about 1.3 molar equivalents of imidazole to provide compound , 17 where P 1 is t ⁇ rt-butyldimethylsilyl.
  • This reaction is typically conducted at a temperature ranging from about 0 0 C to about 50 0 C, typically at room temperature, for about 24 to about 48 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as A ⁇ -dimethylformamide and the like.
  • compound 17 is typically isolated using conventional procedures, such as extraction, chromatography and the like.
  • benzyl amines include, benzylamine, 3,4- dimethoxybenzylamine, 4-methoxybenzylamine, 4-methylbenzylamine and the like.
  • This reaction is typically conducted by contacting compound 17 with about 2 to about 4 molar equivalents of the benzyl amine at a temperature ranging from about 40 0 C to about 100 0 C, typically from about 80 0 C to about 90 0 C, for about 5 to about 24 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as N-methyl-2-pyrrolidone (NMP) and the like.
  • NMP N-methyl-2-pyrrolidone
  • compound 18 is typically isolated using conventional procedures, such as extraction, chromatography, recrystallization and the like.
  • both Bn 1 and P 4 are benzyl groups that are removed in the same reaction mixture.
  • this reaction is conducted by contacting compound 5 with hydrogen in the presence of a catalyst, such as a palladium catalyst.
  • catalysts include palladium hydroxide on carbon, palladium on carbon, and the like.
  • this debenzylation reaction is conducted in the presence of an acid, such as acetic acid, formic acid and the like. This reaction is typically conducted at a temperature ranging from about 10 0 C to about 50 0 C, typically at room temperature, for about 6 to about 24 hours or until the reaction is substantially complete.
  • this reaction is conducted in a suitable diluent, such as methanol, ethanol and the like.
  • a suitable diluent such as methanol, ethanol and the like.
  • compound 5 is typically isolated using conventional procedures, such as extraction, chromatography and the like.
  • compound 5 is isolated as the acetic acid salt.
  • Y 1 is -CN, -C(O)OH or -C(O)OR 30 can be reduced using conventional procedures and reagents to provide aldehyde 4 (i.e., where Y 1 is -CHO). Additionally, such compounds can be reduced to the alcohol, i.e., where Y 1 is -CH 2 OH, and the alcohol can then be oxidized using standard procedures and reagents to provide aldehyde 4 (i.e., where Y 1 is -CHO).
  • pharmaceutically acceptable salts of the compounds of formula I can be prepared by contacting the free base form of a compound of formula I with a pharmaceutically acceptable acid.
  • the compounds 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 of a compound of this invention, (i.e., free base, pharmaceutically acceptable salt, solvate, etc.) that is suitable for a particular mode of administration can be used in the pharmaceutical compositions discussed herein.
  • this invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a compound of formula I.
  • 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 of formula I.
  • a pharmaceutical composition may contain more than a therapeutically effective amount, i.e., bulk compositions, or less than a therapeutically effective amount, i.e., individual unit doses designed for multiple administration to achieve a therapeutically effective amount.
  • the pharmaceutical composition will contain from about 0.01 to about 95 percent by weight of the therapeutic agent; including, from about 0.01 to about 30 percent by weight; such as from about 0.01 to about 10 percent by weight of the therapeutic 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 of ordinary skill in the pharmaceutical arts.
  • the carriers or excipients used in the pharmaceutical compositions of this invention are commercially available. For example, such materials can be purchased from Sigma (St. Louis, MO).
  • 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 of the invention with a pharmaceutically acceptable carrier and any 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.
  • MDI metered-dose inhaler
  • DPI dry powder inhaler
  • the pharmaceutical composition comprising the therapeutic agent is administered by inhalation using a nebulizer inhaler.
  • nebulizer devices typically produce a stream of high velocity air that causes the pharmaceutical composition comprising the therapeutic agent to spray as a mist that is carried into the patient's respiratory tract.
  • the therapeutic agent when formulated for use in a nebulizer inhaler, is typically dissolved in a suitable carrier to form a solution.
  • the therapeutic agent can be micronized and combined with a suitable carrier to form a suspension of micronized particles.
  • Nebulizer devices suitable for administering therapeutic agents by inhalation are well known to those of ordinary skill in the art or such devices are commercially available.
  • representative nebulizer devices or products include the Respimat Softmist Inhalaler (Boehringer Ingelheim); the AERx Pulmonary Delivery System (Aradigm Corp.); the PARI LC Plus Reusable Nebulizer (Pari GmbH); and the like.
  • 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. In one embodiment, such a solution has a pH of about 4 to about 6.
  • the pharmaceutical composition comprising the therapeutic agent is administered by inhalation using a dry powder inhaler.
  • dry powder inhalers typically administer the therapeutic agent as a free-flowing powder that is dispersed in a patient's air-stream during inspiration.
  • the therapeutic agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid (PLA), polylactide-co-glycolide (PLGA) or combinations thereof.
  • the therapeutic agent is micronized and combined with a suitable carrier to form a blend suitable for inhalation.
  • the compound of formula I is in micronized form.
  • a representative pharmaceutical composition for use in a dry powder inhaler comprises dry milled lactose and micronized particles of a compound of formula I.
  • Such a dry powder formulation can be made, for example, by combining the lactose with the therapeutic agent and then dry blending the components.
  • the therapeutic agent can be formulated without an excipient.
  • the pharmaceutical composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
  • Dry powder inhaler delivery devices suitable for administering therapeutic agents by inhalation are well known to those of ordinary skill in the art or such devices are commercially available.
  • representative dry powder inhaler delivery devices or products include Aeolizer (Novartis); Airmax (IVAX); ClickHaler (Ennovata Biomed); Diskhaler (GlaxoSmithKline); Diskus/Accuhaler (GlaxoSmithKline); Easyhaler (Orion Pharma); Eclipse (Aventis); FlowCaps (Hovione); Handihaler (Boehringer Ingelheim); Pulvinal (Chiesi); Rotahaler (GlaxoSmithKline); SkyeHaler/Certihaler (SkyePharma); Twisthaler (Schering-Plough); Turbuhaler (AstraZeneca); Ultrahaler (Aventis); and the like.
  • the pharmaceutical composition comprising the therapeutic agent is administered by inhalation using a metered-dose inhaler.
  • metered-dose inhalers typically discharge a measured amount of the therapeutic agent using a compressed propellant gas.
  • pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension of the therapeutic agent in a liquefied propellant.
  • Any suitable liquefied propellant may be employed including chlorofluorocarbons, such as CCl 3 F, and hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoro-n-propane, (HFA 227).
  • HFA formulations containing HFAs are generally preferred. 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.
  • co-solvents such as ethanol or pentane
  • surfactants such as sorbitan trioleate, oleic acid, lecithin, and glycerin.
  • 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; 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 hydrofluoroalkane to a suitable container containing the therapeutic agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the therapeutic 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.
  • Metered-dose inhaler devices suitable for administering therapeutic agents by inhalation are well known to those of ordinary skill in the art or such devices are commercially available.
  • representative metered-dose inhaler devices or products include AeroBid Inhaler System (Forest Pharmaceuticals); Atrovent Inhalation Aerosol (Boehringer Ingelheim); Flovent (GlaxoSmithKline); Maxair Inhaler (3M); Proventil Inhaler (Schering); Serevent Inhalation Aerosol (GlaxoSmithKline); and the like.
  • compositions of this invention are suitable for oral administration.
  • suitable pharmaceutical compositions for oral administration maybe in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • 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 of the present invention as the active ingredient and one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate.
  • pharmaceutically acceptable carriers 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 (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acidlmethacrylic acid ester copolymers, cellulose acetate trimellitate (CAT), carboxymethyl ethyl cellulose (CMEC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and the like.
  • enteric coatings such as cellulose acetate phthalate (CAP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acidlmethacrylic acid ester copolymers, cellulose acetate trimellitate (CAT), carboxymethyl ethyl cellulose (CMEC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and the like.
  • enteric coatings such as cellulose acetate phthal
  • the pharmaceutical compositions of the present invention may also be formulated to provide slow or controlled release of the active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres.
  • 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 of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro- encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • 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-butylene 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 emulsifier
  • Suspensions in addition to the active ingredient, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • the pharmaceutical compositions of this invention may be packaged in a unit dosage form.
  • 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.
  • 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 glycol monolaurate, azacycloalkan-2-ones and the like, and incorporated into a patch or similar delivery system. Additional excipients including gelling agents, emulsifiers and buffers, may be used in such transdermal compositions if desired.
  • the compounds of this invention can be administered parenterally, i.e., intravenously, subcutaneously or intramuscularly.
  • an intravenous composition typically comprises a sterile aqueous solution of a compound of formula I, wherein the solution has a pH in the range of about 4 to about 7.
  • the compounds of this invention may be administered in combination with one or more other therapeutic agents.
  • a compound of this invention is either physically mixed with the other therapeutic agent to form a composition containing both agents; or each agent is present in separate and distinct compositions which are administered to the patient simultaneously or sequentially.
  • a compound of formula I can be combined with second therapeutic agent using conventional procedures and equipment to form a composition comprising a compound of formula I and a second therapeutic agent.
  • the therapeutic agents may be combined with a pharmaceutically acceptable carrier to form a pharmaceutical composition comprising a compound of formula I, a second therapeutic agent and a pharmaceutically acceptable carrier.
  • the components of the composition are typically mixed or blended to create a physical mixture. The physical mixture is then administered in a therapeutically effective amount using any of the routes described herein.
  • the therapeutic agents may remain separate and distinct before administration to the patient.
  • the therapeutic agents are not physically mixed together before administration but are administered simultaneously or sequentially as separate compositions.
  • a compound of formula I can be administered by inhalation simultaneously or sequentially with another therapeutic agent using an inhalation delivery device that employs separate compartments (e.g. blister packs) for each therapeutic agent.
  • the combination may be administered using separate delivery devices, i.e., one delivery device for each therapeutic agent.
  • the therapeutic agents can be delivered by different routes of administration, i.e., one by inhalation and the other by oral administration.
  • any therapeutic agent compatible with the compounds of the present invention may be used in combination with such compounds.
  • the second therapeutic agent is one that is effectively administered by inhalation.
  • therapeutic agents include, but are not limited to, anti-inflammatory agents, such as steroidal anti-inflammatory agents (including corticosteroids and glucocorticoids), nonsteroidal anti-inflammatory agents (NSAIDs), and PDE 4 inhibitors; bronchodilators, such as PDE 3 inhibitors, adenosine 2b modulators and ⁇ 2 adrenergic receptor agonists; antiinfective agents, such as Gram-positive antibiotics, Gram-negative antibiotics, and antiviral agents; antihistamines; protease inhibitors; afferent blockers, such as D 2 agonists and neurokinin modulators; and muscarinic receptor antagonists (antichlolinergic agents).
  • anti-inflammatory agents such as steroidal anti-inflammatory agents (including corticosteroids and glucocorticoids), nonsteroidal anti-inflammatory agents (NSA
  • Suitable doses for the other therapeutic agents administered in combination with a compound of the invention are typically in the range of about 0.05 ⁇ g/day to about 500 mg/day.
  • a compound of formula I is administered in combination with a steroidal anti-inflammatory agent.
  • steroidal anti-inflammatory agents that can be used in combination with the compounds of this invention include, but are not limited to, beclomethasone dipropionate; budesonide; butixocort propionate; 20i2-16 ⁇ ,17 ⁇ -[butylidenebis(oxy)]-6 ⁇ ,9 ⁇ -difluoro-l l ⁇ - hydroxy-17 ⁇ -(methylthio)androsta-4-en-3-one (RPR-106541); ciclesonide; dexamethasone; 6 ⁇ ,9 ⁇ -difiuoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-l l ⁇ -hydroxy-16 ⁇ -methyl- 3-oxoandrosta-l,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester; 6 ⁇ ,9 ⁇ -difluoro-l l ⁇ - hydroxy- 16 ⁇ -methyl- 17 ⁇ -[(4-methytl- 1 ,3-thiazole-5-carbonyl)
  • steroidal antiinflammatory agents are commercially available or can be prepared using conventional procedures and reagents.
  • preparation and use of steroidal antiinflammatory agents is described in U.S. Patent No. 6,750,210 B2, issued June 15, 2004; U.S. Patent No. 6,759,398 B2, issued July 6, 2004; U.S. Patent No. 6,537,983, issued March 25, 2003; U.S. Patent Application Publication No. US 2002/0019378 Al, published February 14, 2002; and references cited therein.
  • the steroidal anti-inflammatory agent When employed, the steroidal anti-inflammatory agent is typically administered in an amount that produces a therapeutically beneficial effect when co-administered with a compound of the invention. Typically, the steroidal anti-inflammatory agent will be administered in an amount sufficient to provide from about 0.05 ⁇ g to about 500 ⁇ g per dose.
  • a micronized compound of the invention (100 mg) is blended with milled lactose (25 g) (e.g., lactose in which not greater than about 85% of the particles have a MMD of about 60 ⁇ m to about 90 ⁇ m and not less than 15% of the particles have a MMD of less then 15 ⁇ m).
  • This blended mixture is then loaded into individual blisters of a peelable blister pack in an amount sufficient to provide about 10 ⁇ g to about 500 ⁇ g of the compound of the invention per dose.
  • the contents of the blisters are administered using a dry powder inhaler.
  • a micronized compound of the invention (1 g) is blended with milled lactose (200 g) to form a bulk composition having a weight ratio of compound to milled lactose of 1 :200.
  • the blended composition is packed into a dry powder inhalation device capable of delivering between about 10 ⁇ g to about 500 ⁇ g of the compound of the invention per dose.
  • a micronized compound of the invention (100 mg) and a micronized steroidal antiinflammatory agent (500 mg) are blended with milled lactose (30 g). This blended mixture is then loaded into individual blisters of a peelable blister pack in an amount sufficient to provide about 10 ⁇ g to about 500 ⁇ g of the compound of the invention per dose. The contents of the blisters are administered using a dry powder inhaler.
  • Example D
  • a micronized compound of the invention (10 g) is dispersed in a solution prepared by dissolving lecithin (0.2 g) in demineralized water (200 mL).
  • the resulting suspension is spray dried and then micronized to form a micronized composition comprising particles having a mean diameter less than about 1.5 ⁇ m.
  • the micronized composition is then loaded into metered-dose inhaler cartridges containing pressurized 1,1,1,2- tetrafluoroethane in an amount sufficient to provide about 10 ⁇ g to about 500 ⁇ g of the compound of the invention per dose when administered by the metered dose inhaler.
  • a compound of the invention (25 mg) is dissolved in citrate buffered (pH 5) isotonic saline (125 mL). The mixture is stirred and sonicated until the compound is dissolved. The pH of the solution is checked and adjusted, if necessary, to pH 5 by slowly adding aqueous IN sodium hydroxide. The solution is administered using a nebulizer device that provides about 10 ⁇ g to about 500 ⁇ g of the compound of the invention per dose.
  • a compound of the invention 50 g
  • spray-dried lactose 440 g
  • magnesium stearate 10 g
  • the resulting composition is loaded into a hard gelatin capsule (500 mg of composition per capsule) that are administered orally.
  • VEEGUM ® K magnesium aluminum silicate
  • the resulting suspension contains 100 mg of active ingredient per 10 mL of suspension.
  • the suspension is administered orally.
  • Injectable Composition A compound of the invention (0.2 g) is blended with 0.4 M sodium acetate buffer solution (2.0 mL). The pH of the resulting solution is adjusted to pH 4 using 0.5 N aqueous hydrochloric acid or 0.5 N aqueous sodium hydroxide, as necessary, and then sufficient water for injection is added to provide a total volume of 20 mL. The mixture is then filtered through a sterile filter (0.22 micron) to provide a sterile solution suitable for administration by injection.
  • the compounds of this invention possess both p 2 adrenergic receptor agonist and muscarinic receptor antagonist activity and therefore, such compounds are expected to be useful as therapeutic agents 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.
  • 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 whez bronchitis and emphysema), asthma, pulmonary fibrosis and the like.
  • chronic obstructive pulmonary disease e.g., chronic and whez bronchitis and emphysema
  • asthma pulmonary fibrosis
  • Other conditions 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.
  • this invention relates 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.
  • 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 dose per day or a single dose per week.
  • the dose for treating a pulmonary disorder will range from about 10 ⁇ g/day to about 500 ⁇ g/day.
  • this invention relates to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient a therapeutically effective amount of a compound of formula I.
  • the dose for treating COPD or asthma will range from about 10 ⁇ g/day to about 500 ⁇ g/day.
  • COPD chronic obstructive pulmonary disease
  • the term "COPD" is understood by those of ordinary skill in the art to include a variety of respiratory conditions, including chronic obstructive bronchitis and emphysema, as exemplified by the teachings of Barnes, Chronic Obstructive Pulmonary Disease, N. Engl. J. Med., 2000: 343:269-78, and references cited therein.
  • this invention When administered by inhalation, the compounds of this invention typically have the effect of producing bronchodilation. Accordingly, in another of its method aspects, this invention relates to a method of producing bronchodilation in a mammal, the method comprising administering to a mammal a bronchodilation-producing amount of a compound of formula I. Generally, the dose for producing bronchodilation will range from about 10 ⁇ g/day to about 500 ⁇ g/day.
  • the compounds of this invention are optionally administered in combination with another therapeutic agent or agents.
  • triple therapy i.e., ⁇ 2 adrenergic receptor agonist activity, muscarinic receptor antagonist activity and anti-inflammatory activity
  • triple therapy i.e., ⁇ 2 adrenergic receptor agonist activity, muscarinic receptor antagonist activity and anti-inflammatory activity
  • pharmaceutical compositions (and combinations) containing two therapeutic agents are typically easier to formulate and/or administer compared to compositions containing three therapeutic agents, such two component compositions provide a significant advantage over compositions containing three therapeutic agents.
  • the pharmaceutical compositions, combinations and methods of this invention further comprise a steroidal anti-inflammatory agent.
  • 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. Additionally, such compounds are useful in screening assays to discover, for example, new compounds having both ⁇ 2 adrenergic agonist activity and muscarinic receptor antagonist activity.
  • Such 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 and/or a muscarinic receptor is typically contacted with a ⁇ 2 adrenergic receptor-agonizing or muscarinic receptor-antagonizing amount of a compound of this invention.
  • the effects on the biological system or sample caused by the compound are then determined or measured using conventional procedures and equipment, such as by measuring binding in a radioligand binding assays or ligand-mediated changes in a " functional assay or by determining the amount of bronchoprotection provided by the compound in a bronchoprotection assay in a mammal. .
  • Representative ligand-mediated changes in a functional assay include ligand-mediated changes in intracellular cyclic adenosine monophosphate (cAMP); ligand-mediated changes in activity of the enzyme adenylyl cyclase (which synthesizes cAMP); ligand-mediated changes in incorporation of guanosine 5'-O-(thio)triphos ⁇ hate ([ 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 FLJPR ® from Molecular Devices, Inc.); and the like.
  • cAMP cyclic adenosine monophosphate
  • adenylyl cyclase which synthesizes cAMP
  • ligand-mediated changes in incorporation of guanosine 5'-O-(thio)triphos ⁇ hate [ 35 S
  • Compounds of this invention are expected to agonize or cause activation of a ⁇ 2 adrenergic receptor and antagonize or decrease the activation of muscarinic receptors in the functional assays listed herein or in assays of a similar nature.
  • the compounds of this invention will typically be used in these studies at a concentration ranging from about 0.1 nanomolar to about 100 nanomolar.
  • the compounds of this invention can be used as research tools for evaluating other chemical compounds.
  • a compound of formula I is used as a standard in an assay to allow comparison of the results obtained with a test compound and the compound of formula I.
  • ⁇ 2 adrenergic receptor and/or muscarinic receptor binding data (as determined, for example, by in vitro radioligand displacement assays) for a test compound or a group of test compounds is compared to the ⁇ 2 adrenergic receptor and/or muscarinic receptor binding data for a compound of formula I to identify those test compounds that have desirable binding, i.e. test compounds having binding about equal or superior to a compound of formula I, if any.
  • bronchoprotective effects can be determined for test compounds and a compound of formula I in a bronchoprotection assay in a mammal and this data compared to identify test compounds providing about equal or superior bronchoprotective effects.
  • This aspect of the invention includes, as separate embodiments, both (i) the generation of comparison data (using the appropriate assays) and (ii) the analysis of the test data to identify test compounds of interest.
  • HPLC analysis was typically conducted using an Agilent (Palo Alto, CA) Series 1100 instrument with Zorbax Bonus RP 2.1 x 50 mm columns, supplied by Agilent, (a C 14 column), having a 3.5 micron particle size. Detection was by UV absorbance at 214 nm.
  • Mobile phase "A” was 2 % acetonitrile, 97.9 % water, and 0.1% trifluoro acetic acid (v/v/v); and mobile phase "B” was 89.9 % acetonitrile, 10 % water, and 0.1 % trifluoroacetic acid (v/v/v).
  • HPLC (10-70) data were obtained with a flow rate of 0.5 mL/minute of 10% to 70% mobile phase B gradient over a 6-minute period; HPLC (5-35) data were obtained with a flow rate of 0.5 mL/minute of 5% to 35% mobile phase B gradient over a 5-minute period; and HPLC (10-90) data were obtained with a flow rate of 0.5 mL/minute of 10% to 90% mobile phase B gradient over a 5-minute period.
  • LCMS data typically were obtained with an Applied Biosystems (Foster City, CA) Model API-150EX instrument.
  • LCMS 10- 90 data were obtained with a 10% to 90% mobile phase B gradient over a 5-minute period.
  • Small-scale purifications were typically conducted using an API 150EX Prep Workstation system from Applied Biosystems.
  • the mobile phase "A” was water containing 0.05% trifluoroacetic acid (v/v); and mobile phase “B” was acetonitrile containing 0.05% trifluoroacetic acid (v/v).
  • the following conditions were typically used: 20 mIVmin flow rate; 15 min gradients and a 20 mm x 50 mm Prism RP column with 5 micron particles (Thermo Hypersil-Keystone, Bellefonte, PA).
  • Biphenyl-2-isocyanate 97.5 g, 521 mmol
  • 4-hydroxy-l-benzylpiperidine 105 g, 549 mmol
  • the reaction mixture was then cooled to 50 0 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 0 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 (10N) was then added to adjust the pH to 12.
  • Step A - iV- ⁇ S-[(/?)-2-Benzylainino-l-(tert-butyIdiinethylsiIanyloxy)ethyI]-2- benzyloxyphenyl ⁇ formamide
  • Step B i ⁇ r - ⁇ 5-[( J R)-2-Amino-l-(ter ⁇ '-butyIdimethylsiIanyloxy)ethyl]-2-hydroxyphenyI ⁇ - formamide Acetic Acid Salt
  • Step C Methyl 4- ⁇ 3-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l- yl]propionylamino ⁇ -2,5-dimethylbenzoate
  • 3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic acid 670 mg, 1.82 mmol
  • methyl 4-amino-2,5-dimethylbenzoate (390 mg, 2.18 mmol) in dichloromethane (3.6 mL) and diisopropylethylamine (0.413 mL) was added 0-(7- azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate (HATU) (829 mg, 2.18 mmol).
  • HATU 0-(7- azabenzotriazol-l-yl)-l,l,3,3-tetra
  • Step D BiphenyI-2-yIcarbamic Acid l-[2-(4-HydroxymethyI-2,5- dimethyIphenylcarbamoyl)ethyl]piperidin-4-yl Ester
  • Step E Biphenyl-2-ylcarbamic Acid l-[2-(4-Formyl-2,5- dimethylphenyIcarbamoyl)ethy]]p ⁇ peridin-4-yI Ester
  • Step F Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [(i ⁇ )-2-(tert-ButyIdimethylsilanyloxy)-2- (3-formylamino-4-hydroxyphenyl)ethylamino]methyl ⁇ -2,5- dimethyIphenylcarbamoyI)ethyl]piperidin-4-yl Ester
  • a solution of biphenyl-2-ylcarbamic acid l-[2-(4-fo ⁇ nyl-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester 150 mg, 0.30 ⁇ unol
  • N- ⁇ 5-[(i?)- 2-amino-l-(tert-butyldimethylsilanyloxy)ethyl]-2-hydroxyphenyl ⁇ formamide (112 mg, 0.36 mmol) in a 1:1 mixture of dichloromethane and methanol (3.0
  • Step G Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [(/?)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5- dimethyIphenylcarbamoyI)ethyl]piperidin-4-yl Ester
  • To a suspension of biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [(R)-2-(tert- butyldimethylsilanyloxy)-2-(3-formylamino-4-hydroxyphenyl)ethylamino]methyl ⁇ -2,5- dimethylphenylcarbamoyl)-ethyl]piperidin-4-yl ester (238 mg, 0.30 mmol) in dichloromethane (3.0 mL) was added triethylamine trihydrofiuoride (147 ⁇ L, 0.90 mmol). This mixture was stirred at room
  • dibenzyl-(4-iodo-2,5-dimethylphenyl)amine hydrochloride was transferred to a 3 L flask and toluene (1 L) and 1 M aqueous sodium hydroxide (1 L) were added. The resulting mixture was stirred for 1 hour and then the layers were separated. The organic layer was washed with dilute brine (500 mL) and the solvent was removed by rotoevaporation to provide dibenzyl-(4-iodo-2,5-dimethylphenyl)amine (80 g) as a semi- solid thick oil. (Alternatively, dichloromethane can be used in place of toluene in this step).
  • Step B 4-Dibenzylamino-2,5-diniethylbenzaldehyde Hydrochloride
  • dibenzyl-(4-iodo-2,5-dimethylphenyl)amine 15 g, 35 mmol
  • Toluene (300 mL) was added and the resulting mixture was stirred for about 15 minutes.
  • the reaction flask was purged with dry nitrogen and cooled to about —20 0 C and 1.6 M n-butyllithium in hexanes (33 mL, 53 mmol) was added dropwise via the addition funnel.
  • Step C 4-[l,3]Dioxolan-2-yI-2,5-dimethyIphenylamine
  • the crude dibenzyl-(4-[l,3]dioxolan-2-yl-2,5-dimethyl- ⁇ henyl)amine was dissolved in a 2:1 mixture of ethanol and water (150 mL total) and the resulting mixture was purged with dry nitrogen for 5 minutes. Palladium on carbon (2.3 g, 10 Wt. % containing about 50% water) and solid sodium bicarbonate (1.0 g) were added and the resulting mixture was hydrogenated at about 1 atm of hydrogen at 25 0 C to 30 0 C for about 8 hours.
  • Step D -7V-(4-[l,3]Dioxolan-2-yl-2,5-dimethylphenyl)acrylaini(Ie
  • reaction mixture was stirred at 0 0 C to 5 0 C for 1 hour and then water (50 mL) was added and stirring was continued for about 30 minutes at which time fine solids had formed.
  • the mixture was filtered to collect the solids. The layers of the filtrate were then separated and the organic layer was concentrated under reduced pressure to dryness.
  • Step E Biphenyl-2-yIcarbamic Acid 1 -[2-(4-Formy 1-2,5- dimethylphenylcarbamoyI)ethyl]piperidin-4-yI Ester Hydrochloride
  • biphenyl-2-ylcarbamic acid piperidin-4- yl ester 1.2 g, 4.04 mmol
  • iV-(4-[l,3]dioxolan-2-yl-2 5 5-dimethylphenyl)acrylamide 1.0 g, 4.04 mmol
  • Ethanol (10 mL) and dichloromethane (10 mL) were added to form a slurry.
  • the reaction mixture was heated at 45 0 C to 50 0 C for about 18 hours and then cooled to room temperature.
  • Aqueous IM hydrochloric acid (10 mL) was added and the resulting mixture was stirred vigorously for about 3 hours.
  • Step F Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [(jR)-2-(ter/-ButyldimethylsiIanyloxy)-2- (3-formylamino-4-hydroxyphenyl)ethylamino]raethyl ⁇ -2,5-dimethyIphenyI- carbamoyl)ethyl]piperidin-4-yl Ester
  • biphenyl-2-ylcarbamic acid 1- [2-(4-formyI-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester hydrochloride 38 g, 70 mmol
  • N- ⁇ 5-[(i?)-2-amino-l-(rer/-butyldimethylsilanyloxy)ethyl]-2- hydroxyphenyl ⁇ formamide acetic acid salt (33.6 g, 91 mmol).
  • Dichloromethane (500 mL) and methanol (500 mL) were added and the resulting mixture was stirred at room temperature under dry nitrogen for about 3 hours.
  • reaction mixture was then cooled to 0 0 C to 5 0 C and solid sodium triacetoxyborohydride (44.5 g, 381 mmol) was added in portions over a 10 minute period.
  • the reaction mixture was slowly warmed from 0 °C to room temperature over a period of about 2 hours and then cooled to 0 0 C.
  • Saturated aqueous sodium bicarbonate (500 mL) and dichloromethane (500 mL) were added. This mixture was stirred thoroughly and then the layers were separated.
  • the crude product (30 g) was dissolved in dichlorometha ⁇ e containing 2% methanol (150 mL total) and loaded onto a silica gel column (300 g) that had been packed and equilibrated with dichloromethane containing 2% methanol and 0.5% ammonium hydroxide.
  • the product was eluted from the column using dichloromethane containing 2% methanol and 0.5% ammonium hydroxide (1 L); dichloromethane containing 4% methanol and 0.5% ammonium hydroxide (1 L) and dichloromethane containing 5% methanol and 0.5% ammonium hydroxide (about 3 L).
  • Step G Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [(if)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylaminolmethyl ⁇ -2,5-dimethylphenyl- carbamoyI)ethyl]piperidin-4-yl Ester Hydrofluoride Salt
  • Step H BiphenyI-2-ylcarbamic Acid l-[2-(4- ⁇ [(i ⁇ )-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5-dimelhyIphenylcarbamoyl)- ethyl]piperidin-4-yl Ester
  • Biphenyl-2-ylcarbamic acid 1 -[2-(4- ⁇ [( ⁇ )-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl ⁇ -2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester hydrofluoride salt 25 g was purified on a 6-inch reverse-phase column (Microsorb solid phase) in three equal batches using a 10 % to 50 % mixture of acetonitrile in water containing 1% trifluoroacetic acid as the mobile phase. Fractions with greater than 99% purity were combined and then diluted with one volume of water.
  • biphenyl-2-ylcarbamic acid piperidin- 4-yl ester (17.0 g, 58 mmol) andiV-(4-[l,3]dioxolan-2-yl-2,5-dimethylphenyl)acrylamide (13.1 g, 52.9 mmol).
  • Ethanol (150 mL) and dicloromethane (150 mL) were added to form a slurry.
  • the reaction mixture was heated at 50 0 C to 55 0 C for about 24 hours and then cooled to room temperature. Most of the solvent was removed on a rotary evaporator, resulting in a thick slurry.
  • biphenyl-2-ylcarbamic acid l-[2-(4- [l,3]dioxolan-2-yl-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester 15 g, 27.6 mmol
  • acetonitrile 150 mL
  • Aqueous 2 M hydrochloric acid 75 mL was added and the resulting mixture was stirred at 30 0 C for 1 hour. The mixture was then cooled to room temperature and ethyl acetate (150 mL) was added.
  • Step C Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [(i?)-2-(te/t-ButyldimethylsilanyIoxy)-2- (3-formyIamino-4-hydroxyphenyl)ethylimino]methyI ⁇ -2,5-dimethyl- phenylcarbamoyl)ethyl]piperidin-4-yl Ester
  • Step D Biphenyl-2-yIcarbamic Acid l-[2-(4- ⁇ [(i?)-2-( ⁇ ert-ButyldimethylsiIanyloxy)-2- (3-formylamino-4-hydroxyphenyl)ethyIamino]methyl ⁇ -2,5-dimethylphenyl- carbamoyl)ethyI]piperidin-4-yI Ester
  • Step E Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [CR)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethyIamino]methyI ⁇ -2,5-dimetbyIphenyI- carbamoyl)ethyl]piperidin-4-yI Ester L-Tartrate Salt
  • Step F Biphenyl-2-ylcarbamic Acid l-[2-(4- ⁇ [( ⁇ )-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5-dimethyIphenylcarbamoyI)- ethyl]piperidin-4-yI Ester
  • the 1 H and 13 C NMR spectra showed the presence of a major isomer (about 82 mole percent) and a minor isomer (about 18 mole percent) believed to be rotational isomers resulting from hindered rotation about the -NH-C(O)H bond.
  • the phenyl group is believed to be syn to the carbonyl oxygen in the major isomer and anti in the minor isomer.
  • This crystalline free base was determined to have a differential scanning calorimetry (DSC) trace that exhibit a peak in endothermic heat flow at about 142 C C to about 150 0 C; and a powder x-ray diffraction (PXRD) pattern having significant diffraction peaks, among other peaks, at 2 ⁇ values of about 20.7 ⁇ 0.3, 21.6 ⁇ 0.3, 22.5 ⁇ 0.3 and 23.2 ⁇ 0.3.
  • DSC differential scanning calorimetry
  • PXRD powder x-ray diffraction
  • Step A 4-Iodo-2,5-diraethylphenylamine
  • 2,5-dimethylaniline (2Og, 165 ramol) in a 1:1 mixture of dichloromethane and methanol (400 mL) was added sodium bicarbonate (20.8g, 250 mmol) and tetramethylammonium dichloroiodate(I) (44.7g, 165 mmol).
  • the resulting mixture was stirred at room temperature for 1 hour and then water was added (500 mL).
  • the organic layer was removed and washed with 5% aqueous sodium thiosulfate (500 mL) and brine (500 mL).
  • the organic layer was then dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide 4-iodo-2,5-dimethylphenylamine (39.6 g, 98% yield).
  • the product was used without further purification.
  • Step B -iV-(4-Iodo-2,5-dimethylphenyl)acrylamide
  • 4-iodo-2,5-dimethylphenylamine 37.2 g, 151 mmol
  • dichloromethane 500 mL
  • sodium bicarbonate 25.4 g, 302 mmol
  • the resulting mixture was cooled to 0 0 C and acryolyl chloride (12.3 mL, 151 mmol) was added slowly over a period of 25 minutes.
  • the resulting mixture was stirred at room temperature overnight and then filtered. The volume of the filtrate was reduced to about 100 mL and a precipitate formed.
  • Step D 4- ⁇ 3-[4-(BiphenyI-2-ylcarbamoyloxy)piperidi ⁇ -l-yI]propionylamino ⁇ -2,5- dimethylbenzoic Acid Methyl Ester
  • the resulting mixture was purged with carbon monoxide and then stirred overnight at 70 0 C to 80 0 C under a carbon monoxide atmosphere (balloon pressure).
  • the reaction mixture was concentrated under vacuum and the residue was dissolved in dichloromethane (500 mL). This mixture was washed with IN aqueous hydrochloric acid (500 mL), water (500 mL) and then brine (500 mL). The organic layer was then dried over anhydrous magnesium sulfate, filtered and then concentrated under vacuum.
  • the residue was mixed with ethanol (about 5:1 v/w ethanol to residue) and the mixture was heated until all solid material dissolved.
  • Step E Biphenyl-2-ylcarbamic Acid l-[2-(4-Hydroxymethyl-2,5-dimethyl- phenyIcarbamoyl)ethyl]piperidin-4-yl Ester
  • Step F Biphenyl-2-ylcarbamic Acid l-[2-(4-Formyl-2,5- dimethyIphenyIcarbamoyl)ethyl]piperidin-4-yl Ester
  • Radioligand binding assays for cloned muscarinic receptors were performed in 96- well microtiter plates in a total assay volume of 100 ⁇ L.
  • CHO cell membranes stably expressing either the hMi, I1M 2 , hM 3 , hJVLj or I1M 5 muscarinic subtype were diluted in assay buffer to the following specific target protein concentrations ( ⁇ g/well): 10 ⁇ g for hMi, 10- 15 ⁇ g for hM 2 , 10-20 ⁇ g for hM 3 , 10-20 ⁇ g for TaM 4 , and 10- 12 ⁇ g for hM 5 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 K ⁇ values of the radioligand were performed using L-
  • Displacement assays for determination of K,- values of test compounds were performed with [ 3 H]-NMS at 1 nM and eleven different test compound concentrations.
  • test compounds were initially dissolved to a concentration of 400 ⁇ M in dilution buffer and then serially diluted 5x with dilution buffer to final concentrations ranging from 10 pM to 100 ⁇ M.
  • the addition order and volumes to the assay plates were as follows: 25 ⁇ L radioligand, 25 ⁇ L diluted test compound, and 50 ⁇ L membranes. Assay plates were incubated for 60 minutes at 37 0 C. Binding reactions were terminated by rapid filtration over GF/B glass fiber filter plates (PerkinElmer, Inc.) pre-treated in 1% BSA. Filter plates were rinsed three times with wash buffer (10 mM HEPES) to remove unbound radioactivity.
  • Biphenyl-2-yicarbamic acid l-[2-(4- ⁇ [(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester was found to have a Kj value of less than 10 nM for the Mi, M 2 , M 3 , M 4 and M 5 muscarinic receptor subtypes.
  • Human embryonic kidney (HEK-293) cell lines stably expressing cloned human ⁇ i and ⁇ 2 adrenergic receptors or Chinese hamster ovarian (CHO) cell lines stably expressing cloned human ⁇ 3 adrenergic receptors were grown to near confiuency in DMEM or Hams F- 12 media with 10% FBS in the presence of 500 ⁇ g/mL Geneticin. The cell monolayer was lifted with 2 mM 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.
  • ⁇ i and ⁇ 2 receptor expressing membranes For preparation of ⁇ i and ⁇ 2 receptor expressing membranes, cell pellets were re-suspended in lysis buffer (10 mM HEPES/HC1, 10 mM EDTA, pH 7.4 at 4 0 C) and homogenized using a tight-fitting Dounce glass homogenizer (30 strokes) on ice.
  • lysis buffer 10 mM Tris/HCl, pH 7.4
  • ⁇ 3 receptor expressing membranes cell pellets were homogenated in lysis buffer (10 mM Tris/HCl, pH 7.4) supplemented with one tablet of "Complete Protease Inhibitor Cocktail Tablets with 2 mM EDTA" per 50 mL buffer (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, 12.5 mM MgCl 2 , 1 mM EDTA).
  • the protein concentration of the 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 0 C or used immediately.
  • Binding assays were performed in 96-well microliter plates in a total assay volume of 100 ⁇ L with 10-15 ⁇ g of membrane protein containing the human ⁇ i, ⁇ 2 or ⁇ 3 adrenergic receptors in assay buffer (75 mM Tris/HCl pH 7.4 at 25 0 C, 12.5 mM MgCl 2 , 1 mM EDTA, 0.2% BSA).
  • Displacement assays for determination of K 1 - values of test compounds were done with [ 3 H]-dihydroalprenolol at 1 nM and [ 125 I]-(-)-iodocyanopindolol at 0.5 nM for 10 or 11 different concentrations of test compound ranging from 10 pM to 10 ⁇ M. Non-specific binding was determined in the presence of 10 ⁇ M propranolol.
  • Assays were incubated for 1 hour at 37 0 C, and then binding reactions were terminated by rapid filtration over GF/B for the ⁇ i and ⁇ 2 receptors or GF/C glass fiber filter plates for the ⁇ 3 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 0 C, 12.5 mM MgCl 2 , 1 mM EDTA) to remove unbound radioactivity.
  • filtration buffer 75 mM Tris/HCl pH 7.4 at 4 0 C, 12.5 mM MgCl 2 , 1 mM EDTA
  • K/ values for test compounds were calculated from observed IC 50 values and the Kd value of the 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.
  • Biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [(i?)-2-(3- formylarnino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester was found to have a Kj value of less than 10 nM for the ⁇ 2 adrenergic receptor and Ki values greater than 1000 nM for the ⁇ i and ⁇ 3 adrenergic receptors.
  • the functional potency of a test compound as an antagonist for the hM 2 receptor was determined by measuring the ability of the test compound to block oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM 2 receptor.
  • cAMP assays were performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with l25 I-cAMP (NEN SMP004B, PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturer's instructions.
  • the cells were centrifuged again at 650 x g for five minutes and re-suspended in stimulation buffer to an assay concentration of 1.6 x 10 6 to 2.8 x 10 6 cells/mL.
  • the test compound was initially dissolved to a concentration of 400 ⁇ M in dilution buffer (dPBS supplemented with 1 mg/mL BSA (0.1%)), and then serially diluted with dilution buffer to final molar concentrations ranging from 100 ⁇ M to 0.1 nM.
  • Oxotremorine was diluted in a similar manner.
  • oxotremorine inhibition of adenylyl cyclase activity 25 ⁇ L forskolin (25 ⁇ M final concentration diluted in dPBS), 25 ⁇ L diluted oxotremorine, and 50 ⁇ L cells were added to agonist assay wells.
  • 25 ⁇ L forskolin and oxotremorine 25 ⁇ M and 5 ⁇ M final concentrations, respectively, diluted in dPBS
  • 25 ⁇ L diluted test compound, and 50 ⁇ L cells were added to remaining assay wells.
  • Biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ila) was found to have a Kobs value of less than about 10 nM for blockade of oxotremorine-inhibition of forskolin- mediated cAMP accumulation in CHO-Kl cells expressing the hM 2 receptor.
  • the functional potency of a test compound as an antagonist of the hM2 receptor was determined by measuring the ability of the test compound to block oxotremorine-stimulated [ 35 S]GTPyS binding in CHO-Kl cells expressing the hM 2 receptor.
  • frozen membranes were thawed and then diluted in assay buffer with a final target tissue concentration of 5 to 10 ⁇ g protein per well.
  • the membranes were briefly homogenized using a Polytron PT-2100 tissue disrupter and then added to the assay plates.
  • the EC 9 0 value (effective concentration for 90% maximal response) for stimulation of [ 35 S]GTPyS binding by the agonist oxotremorine was determined in each experiment.
  • test compound To determine the ability of a test compound to inhibit oxotremorine-stimulated [ 35 S]GTPyS binding, the following was added to each well of 96 well plates: 25 ⁇ L of assay buffer with [ 35 S]GTPyS (0.4nM), 25 ⁇ L of oxotremorine(EC9o) and GDP (3uM), 25 ⁇ L of diluted test compound and 25 ⁇ L CHO cell membranes expressing the hM 2 receptor. The assay plates were then incubated at 37 0 C for 60 minutes. The assay plates were filtered over 1% BSA-pretreated GF/B filters using a PerkinElmer 96-well harvester.
  • Biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester was found to have a Kobs value of less than about 10 nM for blockade of oxotremorine-stimulated [ 35 S]GTPyS binding in CHO-Kl cells expressing the I1M 2 receptor.
  • the functional potency of a test compound as an antagonist of ItM 1 , 11M 3 and cMs receptors was determined by measuring the ability of the test compound to inhibit agonist-mediated increases in intracellular calcium.
  • CHO cells stably expressing the receptors were seeded into 96-well FLIPR plates the night before the assay was done. Seeded cells were washed twice 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) using Cellwash (MTX Labsystems, Inc.) to remove growth media. After washing, each well contained 50 ⁇ L of FLIPR buffer. The cells were then incubated with 50 ⁇ L/well of 4 ⁇ M FLUO-4AM (a 2X solution was made) for 40 minutes at 37 0 C, 5% carbon dioxide.
  • 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
  • An oxotremorine concentration of 3 x EC F was prepared in stimulation plates such that an EC 9 0 concentration of oxotremorine was added to each well in test assay plates.
  • the parameters used for the FLIPR were: exposure length of 0.4 seconds, laser strength of 0.5 watts, excitation wavelength of 488 nm, and emission wavelength of
  • Baseline was determined by measuring the change in fluorescence for 10 seconds prior to addition of oxotremorine. Following oxotremorine 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 was expressed as maximum fluorescence minus baseline fluorescence for each well. The raw data was analyzed against the logarithm of test compound concentration by nonlinear regression with GraphPad Prism (GraphPad Software, Inc., San Diego, CA) using the built-in model for sigmoidal dose-response.
  • Antagonist Kob S values were determined by Prism using the oxotremorine EC 50 value as the Ko and the oxotremorine EC 90 for the ligand concentration according to the Cheng-Prusoff equation (Cheng & Prusoff, 1973).
  • Adrenergic Receptors cAMP assays were performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with [ 125 I]-CAMP (NEN SMP004,
  • ⁇ i and ⁇ 2 receptor agonist potency HEK-293 cell lines stably expressing cloned human P 1 and ⁇ 2 receptors were grown to near confluency in DMEM supplemented with 10% FBS and Geneticin (500 ⁇ g/mL).
  • ⁇ 3 receptor agonist potency CHO-Kl cell line stably expressing cloned human or ⁇ 3 adrenergic receptors was grown to near confluency in Hams F- 12 media supplemented with 10% FBS and Geneticin (250 ⁇ g/mL).
  • 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 ⁇ M to 1 pM. Reactions were incubated for 10 min at 37 0 C and stopped by adding 100 ⁇ L of cold detection buffer containing [ 125 I]-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.
  • Biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5-dimethylphenyl- carbamoyl)ethyl]piperidin-4-yl ester (compound Ila) was found to have an EC50 value less than about 10 nM for the ⁇ 2 adrenergic receptor; an EC 50 value of about 30 nM for the P 1 adrenergic receptor; and an EC5 0 value greater than 700 nM for the ⁇ 3 adrenergic receptor.
  • Example 16 Whole-cell cAMP Flashplate Assay With a Lung Epithelial Cell Line Endogenously Expressing Human ⁇ z Adrenergic Receptor
  • the agonist potency and intrinsic activity of a test compound were determined using a cell line expressing endogenous levels of the ⁇ 2 adrenergic receptor.
  • Cells from a human lung epithelial cell line (BEAS-2B) (ATCC CRL-9609, American Type Culture Collection, Manassas, VA) (January B, et al., British Journal of
  • test compounds were serially diluted into assay buffer (75 mM Tris/HCl pH 7.4 at 25 0 C, 12.5 mM MgCl 2 , 1 mM EDTA, 0.2% BSA) in Beckman Biomek-2000.
  • Test compounds were tested in the assay at 11 different concentrations, ranging from 10 ⁇ M to 10 pM. Reactions were incubated for 10 min at 37 0 C and stopped by addition of 100 ⁇ L of ice L cold detection buffer. Plates were sealed, incubated over night at 4 0 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.
  • Biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5-dimethylphenyl- carbamoyl)ethyl]pi ⁇ eridin-4-yl ester was found to have an ECs 0 value of less than 10 nM with intrinsic activity value of greater than 0.3 compared with a full ⁇ 2 agonist isoproterenol (1.0),
  • the assay was conducted using male Duncan-Hartley guinea pigs (Harlan, Indianapolis, IN), weighing between 250 and 400 g.
  • a test compound or vehicle i.e., sterile water
  • IH inhalation
  • Animals were exposed to an aerosol generated from an LC Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA) driven by Bioblend (a mixture of 5% CO 2 ; 21% O 2 ; and 74% N 2 ) at a pressure of 22 psi.
  • Pulmonary function was evaluated at various time points after inhalation dosing.
  • the guinea pigs were anesthetized with an intramuscular (IM) injection of a mixture of ketamine (43.7 mg/kg/xylazine (3.5 mg/kg)/acepromazine (1.05 mg/kg).
  • IM intramuscular
  • 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 niethacholine or histamine.
  • the trachea was then dissected free and cannulated with a 14G needle (#NE-014, Small Parts, Miami Lakes, FL).
  • 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 was measured in the tracheal cannula using a Biopac transducer connected to a Biopac (TSD 137C) pre-amplifier. Body temperature was maintained at 37 0 C using a heating pad. Prior to initiating data collection, pentobarbital (25 mg/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 of the bronchoconstrictor (methacholine or histamine).
  • IP intraperitoneally
  • mice When methacholine was used as the bronchoconstrictor agent, animals were pre-treated with propranolol (5 mg/kg, IV) to isolate the antimuscarinic effects of the test compound. The propranolol was administered 30 minutes prior to construction of the dose-response curve to methacholine or histamine. Changes in VP were recorded using the Acknowledge Data Collection Software (Santa Barbara, CA). After the completion of study, the animals were euthanized.
  • propranolol 5 mg/kg, IV
  • the propranolol was administered 30 minutes prior to construction of the dose-response curve to methacholine or histamine. 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 H 2 O) peak pressure (after bronchoconstrictor challenge) - peak baseline pressure.
  • the dose-response curve to methacholine or histamine was fitted to a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California). The following equation was used:
  • Y is the response. Y starts at Min and approaches asymptotically to Max with a sigmoidal shape.
  • % 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) x 100). Inhibition curves were fitted using the four parameter logistic equation from GraphPad software. ID 50 (dose required to produce 50% inhibition of the bronchoconstrictor response) and Emax (maximal inhibition) were also estimated wherever appropriate.
  • PD T 1 Z 2 The magnitude of bronchoprotection at different time-points after inhalation of the test compound was used to estimate the pharmacodynamic half-life (PD T 1 Z 2 ).
  • biphenyl-2-ylcarbamic acid l-[2-(4- ⁇ [( ⁇ )-2-(3- ' formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl ⁇ -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (Ha) was found to have an ID50 of less than about 50 ⁇ g/mL for both methacholine-induced bronchoconstriction and histamine- induced bronchoconstriction.
  • this compound produced significant bronchoprotection for up to about 72 hours when administered as a single sub-maximal dose (100 ⁇ g/mL).
  • salmeterol (3 ⁇ g/mL) (a ⁇ 2 adrenergic receptor agonist) exhibited significant bronchoprotection for 6 to 14 hours; and tiotropium (10 ⁇ g/mL) (a muscarinic receptor antagonist) exhibited significant bronchoprotection for greater than 72 hours.
  • bronchoprotective efficacy and duration of test compounds was determined using a guinea pig assay.
  • Groups of 6 male guinea pigs (Duncan-Hartley (HsdPoctDH) 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 arranged 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).
  • WFI test compound or vehicle
  • the gas flow through the nebulizer at this operating pressure was approximately 3 L per minute.
  • the generated aerosols were driven into the chambers by positive pressure. No dilution air was used during the delivery of aerosolized solutions.
  • approximately 1.8 mL of solution was nebulized. This value was measured gravimetrically by comparing pre-and post-nebulization weights of the filled nebulizer.
  • bronchoprotective effects of test compounds administered via inhalation were evaluated using whole body plethysmography at 1.5, 24, 48 and 72 hours post-dose. Forty-five minutes prior to the start of the pulmonary evaluation, each guinea pig was anesthetized with an intramuscular injection of ketamine (43.75 mg/kg), xylazine (3.50 mg/kg) and acepromazine (1.05 mg/kg). The surgical site was shaved and cleaned with 70% alcohol and a 2 to 3 cm midline incision of the ventral aspect of the neck was made.
  • 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 or histamine in saline.
  • PE-50 Becton Dickinson, Sparks, MD
  • the trachea was then dissected free and cannulated with a 14G teflon tube (#NE- 014, Small Parts, Miami Lakes, FL).
  • anesthesia was maintained by additional intramuscular injections of the anesthetic mixture. The depth of anesthesia was monitored and adjusted if the animal responded to pinching of its paw or if the respiration rate was greater than 100 breaths per minute.
  • the animal was placed into a plethysmo graph (#PLY3114, Buxco Electronics, Inc., Sharon, CT) and an esophageal pressure cannula (PE- 160, Becton Dickinson, Sparks, MD) was inserted to measure pulmonary driving pressure.
  • PE- 160 Becton Dickinson, Sparks, MD
  • the teflon tracheal tube was attached to the opening of the 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 three 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 had not collapsed and that the animal did not suffer from hyperventilation.
  • the pulmonary evaluation was initiated after determining that baseline values were within the range of 0.3 to 0.9 mL per cm H 2 O for compliance and within the range of 0.1 to 0.199 cm H 2 O per mL per second for resistance.
  • a Buxco pulmonary measurement computer pro gam was used for the collection and derivation of pulmonary values.
  • Starting the program initiated the experimental protocol and data collection.
  • the changes in volume over time that occurred 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 breth.
  • This signal and the pulmonary driving pressure changes collected using a Sensym pressure transducer (TRD4100), were connected via a Buxco (MAX 2270) preamplifier to a data collection interface (SFT3400 and SFT3813). AU other pulmonary parameters are derived from these two inputs.
  • Baseline values were collected for 5 minutes, after which time the guinea pigs were challenged with either acetylcholine or histamine.
  • propanolol (5 mg/Kg, iv) (Sigma- Aldrich, St. Louis, MO) was administered 15 minutes prior to challenge with acetylcholine.
  • Acetylcholine (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 of the experiment: 1.9 ⁇ g/minute at 5 minutes, 3.8 ⁇ g/minute at 10 minutes, 7.5 ⁇ g/minute at 15 minutes, 15.0 ⁇ g/minute at 20 minutes, 30 ⁇ g/minute at 25 minutes and 60 ⁇ g/minute at 30 minutes.
  • bronchoprotective effects of the test compound was assessed in the acetylcholine challenge model without pretreatment with propanolol.
  • histamine 25 ⁇ g/mL
  • histamine 25 ⁇ g/mL
  • histamine 25 ⁇ g/mL
  • 0.5 ⁇ g/minute at 5 minutes 0.9 ⁇ g/minute at 10 minutes
  • 1.9 ⁇ g/minute at 15 minutes 3.8 ⁇ g/minute at 20 minutes
  • 7.5 ⁇ g/minute at 25 minutes and 15 ⁇ g/minute at 30 minutes 0.5 ⁇ g/minute at 5 minutes
  • 0.9 ⁇ g/minute at 10 minutes 1.9 ⁇ g/minute at 15 minutes
  • 3.8 ⁇ g/minute at 20 minutes 7.5 ⁇ g/minute at 25 minutes and 15 ⁇ g/minute at 30 minutes.
  • the guinea pig's lungs were inflated 3 times with 4 mL of air from a 10 mL calibration syringe.
  • the pulmonary parameters recorded included respiration frequency (breaths per minute), compliance (mL per cm H 2 O) and pulmonary resistance (cm H 2 O per 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.
  • Pulmonary resistance (cm H 2 O per mL per second) was calculated from the ratio of change in pressure to the change in flow.
  • the RL response to acetylcholine (60 ⁇ g/min, IH) was computed for the vehicle and the test compound.
  • Inhibition dose-response curves for 'R L ' were fitted with a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California) to estimate bronchoprotective BD 50 (dose required to inhibit the acetylcholine (60 ⁇ g/min) bronchoconstriction response by 50%).
  • bronchoprotective BD 50 dose required to inhibit the acetylcholine (60 ⁇ g/min) bronchoconstriction response by 50%. The following equation was used:
  • Y is the response (percent inhibition of acetylcholine- induced increase in R L ). Y starts at Min and approaches asymptotically to Max with a sigmoidal shape.
  • the quantity PD 2 which is defined as the amount of acetylcholine or histamine needed to cause a doubling of the baseline pulmonary resistance, was calculated using the pulmonary resistance values derived from the flow and the pressure over a range of acetylcholine or histamine challenges using the following equation (which was derived from an equation used to calculate PC2 0 values described in American Thoracic Society. Guidelines for methacholine and exercise challenge testing - 1999. Am J Respir Crit Care Med. 2000; 161: 309-329):
  • Ci concentration of acetylcholine or histamine preceding C 2
  • C 2 concentration of acetylcholine or histamine resulting in at least a 2-fold increase in pulmonary resistance (R 1 )
  • R 0 Baseline
  • R 2 R L value after C 2
  • This compound lacks the alkyl groups present on phenyl ring of the compounds of the present invention.
  • compound 50 showed no significant bronchoprotection at 24 hours post-dose for doses ranging from 3 ⁇ g/mL to 300 ⁇ g/mL.
  • the PD2x valves for compound 50 at 24 hours were similar to the vehicle (water) group.
  • salmeterol 100 ⁇ g/mL
  • tiotropium (10 ⁇ g/mL) (a muscarinic receptor antagonist) exhibited significant bronchoprotection for at least 24 hours.

Abstract

This invention relates to compounds of formula (I) Wherein R1 and R2 are as defined in the specification, or a pharmaceutically acceptable salt or solvate or stereoisomer thereof. The invention also relates to pharmaceutical compositions and combinations comprising such compounds, processes and intermediates for preparing such compounds, and methods of using such compound to for example, treat pulmonary disorders, such as chronic obstructive pulmonary disease and asthma.

Description

DIALKYLPHENYL COMPOUNDS
HAVING P2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY
BACKGROUND OF THE INVENTION Field of the Invention
The present invention relates to novel dialkylphenyl compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity. This invention also relates to pharmaceutical compositions comprising such compounds, processes and intermediates for preparing such compounds and methods of using such compounds, for example, to treat pulmonary disorders.
State of the Art
Pulmonary disorders, such as asthma and chronic obstructive pulmonary disease (COPD), are commonly treated with bronchodilators. One type of bronchodilator used to treat pulmonary disorders consists of β2 adrenergic receptor (adrenoceptor) agonists, such as albuterol, formoterol and salmeterol. These compounds are generally administered by inhalation. Another type 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 a combination of a β2 adrenergic receptor agonist and a muscarinic receptor antagonist are known in the art for use in treating pulmonary disorders. For example, U.S. Patent No. 6,433,027, issued August 13, 2002, discloses medicament compositions containing a muscarinic receptor antagonist, such as tiotropium bromide, and a β2 adrenergic receptor agonist, such as formoterol fumarate.
__!-_ Additionally, compounds having both p2 adrenergic receptor agonist and muscarinic receptor antagonist activity are known in the art. For example, U.S. Patent No. 7,141,671, issued November 28, 2006, discloses biphenyl compounds having both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity. Compounds possessing both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity are highly desirable since such compounds provide bronchodilation through two independent modes of action while having single molecule pharmacokinetics.
When treating pulmonary disorders, it is particularly useful to provide therapeutic agents that have a long duration of action, i.e., a duration of at least about 24 hours, when administered by inhalation so that patients only need to administer the therapeutic agent once a day or less. Not all dual-acting compounds disclosed previously in the art possess this desirable property.
Accordingly, a need exists for novel compounds having both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity that possess a long duration of action when administered to a patient by inhalation.
SUMMARY OF THE INVENTION
The present invention provides novel dialkylphenyl compounds having both β2 adrenergic receptor agonist and muscarinic receptor antagonist activity. Among other properties, a compound of this invention has been found to possess a long duration of action, i.e., a duration of at least about 24 hours, when administered to a mammal by inhalation. Accordingly, the compounds of this invention are expected to be useful and advantageous as therapeutic agents for treating pulmonary disorders.
Accordingly, in one of its composition aspects, the present invention relates to a compound of formula I:
Figure imgf000003_0001
wherein
R1 is methyl or ethyl; R2 is methyl or ethyl; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
In a particular aspect of this invention, the compound of formula I is a compound having formula II:
Figure imgf000004_0001
wherein R1 and R2 are as defined herein (including any specific or preferred embodiments); or a pharmaceutically acceptable salt or solvate thereof.
In another of its composition aspects, this invention relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formula I.
If desired, the compounds of the present invention can be administered in combination with other therapeutic agents, such as a steroidal anti-inflammatory agent. Accordingly, in another of its composition aspects, this invention relates to a pharmaceutical composition comprising (a) a compound of formula I; and (b) a second therapeutic agent. In yet another of its composition aspects, this invention relates to pharmaceutical composition comprising (a) a compound of formula I; (b) a second therapeutic agent; and (c) a pharmaceutically acceptable carrier.
In still another of its composition aspects, this invention relates to a combination of therapeutic agents, the combination comprising (a) a compound of formula I; and (b) a second therapeutic agent. Ih another of its composition aspects, this invention relates to a combination of pharmaceutical compositions, the combination comprising (a) a first pharmaceutical composition comprising a compound of formula I and a first pharmaceutically acceptable carrier; and (b) a second pharmaceutical composition comprising a second therapeutic agent and a second pharmaceutically acceptable carrier. This invention also relates to a kit containing such pharmaceutical compositions. Compounds of this invention possess both β2 adrenergic receptor agonist activity and muscarinic receptor antagonist activity. Accordingly, the compounds of formula I are expected to be useful as therapeutic agents for treating pulmonary disorders, such as asthma and chronic obstructive pulmonary disease. Accordingly, in one of its method aspects, this invention relates 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. This invention also relates to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient a therapeutically effective amount of a compound of formula I. Additionally, in another of its method aspects, this invention relates to a method of producing bronchodilation in a mammal, the method comprising administering to a mammal a bronchodilation-producing amount of a compound of formula I. This invention also relates to method of antagonizing a muscarinic receptor and agonizing a β2 adrenergic receptor in a mammal, the method comprising administering to the mammal a compound of formula I.
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 relates to a method of using a compound of formula I as a research tool, the method comprising conducting a biological assay using a compound of formula I.
The compounds of this invention can also be used to evaluate new chemical compounds. Accordingly, in another of its method aspects, this invention relates to a method of evaluating a test compound in a biological assay, the method comprising: (a) conducting a biological assay with a test compound to provide a first assay value; (b) conducting the biological assay with a compound of formula I to provide a second assay value; wherein step (a) is conducted either before, after or concurrently with step (b); and (c) comparing the first assay value from step (a) with the second assay value from step
(b).
This invention also relates to processes and novel intermediates useful for preparing compounds of formula I. Accordingly, in another of its method aspects, this invention relates to a process of preparing a compound of formula I, the process comprising deprotecting a compound of formula 6 (as defined herein) to provide a compound of formula I. In another of its method aspects, this invention relates to a process of preparing a compound of formula I, the process comprising: (a) reacting a compound of formula 4 with a compound of formula 5 in the presence of a reducing agent to provide a compound of formula 6; and (b) deprotecting the compound of formula 6 to provide a compound of formula I; where compounds 4, 5 and 6 are as defined herein.
In a particular embodiment of this invention, the compounds of formula I are prepared by deprotecting a compound of formula 6, wherein the hydroxyl-protecting group is a silyl group. Accordingly, in yet another of its method aspects, this invention relates to a process of preparing a compound of formula I, the process comprising deprotecting a compound of formula 6a:
Figure imgf000006_0001
wherein Ra, Rb and R° are independently selected from CM alkyl, phenyl, -Ci-4 alkyl-(phenyl), or one of Rla, Rlb and Rlc is -O-(Ci-4 alkyl); to provide a compound of formula I.
In other embodiments, the processes described herein further comprise the step of forming a pharmaceutically acceptable salt of a compound of formula I. In other embodiments, this invention relates to the other processes described herein; and to the product prepared by any of the processes described herein.
In a particular embodiment, this invention relates to a compound of formula III:
Figure imgf000006_0002
or a salt or stereoisomer thereof, wherein Y1 is selected from -CHO, -CN, -CH2OH, -CH(OR3a)OR3b, -C(O)OH, -C(O)OR3c, bromo and iodo, where R3a and R3b are selected independently from Ci-6 alkyl, or R3a and R3b are joined to form C2-6 alkylene, R3c is selected from Ci-β alkyl; and R1 and R2 are as defined herein (including any specific or preferred embodiments), which compounds are useful as intermediates in preparing compounds of formula I. In a particular embodiment of formula in, R1 and R2 are methyl. In another particular embodiment of formula III, Y1 is -CHO. In still another particular embodiment of formula III, R1 and R2 are methyl and Y1 is -CHO.
This invention also relates to the use of a compound of formula I for therapy. Additionally, the invention relates to the use of a compound of formula I for the manufacture of a medicament for the treatment of a pulmonary disorder; and to the use of a compound of formula I as a research tool. Other aspects and embodiments of this invention are disclosed herein.
DETAILED DESCRIPTION OF THE INVENTION
In one of its composition aspects, this invention relates to novel compounds of formula I. The compounds of formula I 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 of the 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 partial formula:
Figure imgf000007_0001
In a particular embodiment of this invention, the carbon atom identified by the symbol * has the (R) configuration. In this embodiment, the compounds of formula I have the (R) configuration at the carbon atom identified by the symbol * or are enriched in a stereoisomer^ form having the (R) configuration at this carbon atom.
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 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.
Accordingly, those skilled in the art will recognize that reference to a compound herein, for example, reference to a compound of formula I or compound 6, includes reference to salts and stereoisomers and solvates of that compound unless otherwise indicated.
Additionally, as used herein, the singular forms "a," "an" and "the" include the corresponding plural forms unless the context of use clearly dictates otherwise.
The nomenclature used herein to name the compounds of this invention and intermediates thereof has generally been derived using the commercially available
AutoNom software (MDL, San Leandro, California). Typically, compounds of formula I have been named as piperidin-4-yl ester derivatives of biphenyl-2-yl carbamic acid.
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 one embodiment, R1 is methyl and R2 is methyl. In another embodiment, R1 is ethyl and R2 is ethyl. In another embodiment, R1 is methyl and R2 is ethyl. In another embodiment, R1 is ethyl and R2 is methyl.
Thus, in one of its composition aspects, the present invention relates to compounds of formula I selected from: biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(i?)-2-(3-formylammo-4-hydroxyphenyl)-2- hydroxyethylamino]methyl}-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ha); biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(2?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylaminojmethyl} -2,5-diethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound lib); biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(/2)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl}-2-methyl-5-ethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound lie); biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl} -2-ethyl-5 -methylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Hd); or a pharmaceutically acceptable salt thereof.
Definitions
When describing the compounds, compositions, methods and processes of this invention, the following terms have the following meanings unless otherwise indicated.
The term "alkyl" means a monovalent saturated hydrocarbon group which may be linear or branched. Unless otherwise defined, such alkyl groups typically contain from 1 to 10 carbon atoms. Representative alkyl groups include, by way of example, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, «-pentyl, π-hexyl, n-heptyl, n- octyl, n-nonyl, w-decyl and the like.
When a specific number of carbon atoms is intended for a particular term used herein, the number of carbon atoms is shown preceding the term. For example, the term "Ci-3 alkyl" means an alkyl group having from 1 to 3 carbon atoms wherein the carbon atoms are in any chemically-acceptable configuration.
The term "alkylene" means a divalent saturated hydrocarbon group that 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- 1,5 -diyl and the like.
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), benzyl, 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), /7-methoxybenzyl (PMB)5 9-fluorenylmethyl (Fm), trimethylsilyl (TMS), tert- butyldimethylsilyl (TBS), diphenylmethyl (benzhydryl, DPM) and the like.
The term "compound of the invention" or "compound of formula I" or "compound of formula II" as used herein means the specified compound(s) or a pharmaceutically acceptable salt or solvate or stereoisomer thereof unless otherwise indicated.
The term "halo" means fluoro, chloro, bromo and iodo.
The term "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(Ci-6 alkyl)silyl groups, such as trimethylsilyl (TMS), triethylsilyl (TES), tøt-butyldimethylsilyl (TBS) and the like; esters (acyl groups) including Ci -e alkanoyl groups, such as formyl, acetyl and the like; arylmethyl groups, such as benzyl (Bn), /j-methoxybenzyl (PMB)5 9-fluorenylmethyl (Fm), diphenylmethyl (benzhydryl, DPM) and the like. Additionally, two hydroxyl groups can also be protected as an alkylidene group, such as prop-2-ylidine, formed, for example, by reaction with a ketone, such as acetone.
The term "leaving group" means a functional group or an atom that can be displaced by another functional group or atom in a substitution reaction, such as a micleophilic substitution reaction. By way of example, representative leaving groups include, but are not limited to, 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 "mass median diameter" or "MMD" when used to refer to particles means the diameter such that half the mass of the particles is contained in particles with larger diameter and half is contained in particles with smaller diameter.
The term "micronized" or "in micronized form" means particles in which at least about 90 percent of the particles have a diameter of less than about 10 μm unless otherwise indicated. The term "or a pharmaceutically acceptable salt or solvate or stereoisomer thereof as used herein 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 "pharmaceutically acceptable salt" means a salt that is acceptable for administration to a patient, such as a mammal (e.g., salts having acceptable mammalian safety for a given dosage regime). Representative pharmaceutically acceptable salts include salts of 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-l,5-disulfonic, naphthalene-2,6- disulfonic, nicotinic, nitric, orotic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic and xinafoic acid, and the like.
The term "protected derivatives thereof means a derivative of the specified compound in which one or more functional groups of the compound are protected or blocked from undergoing undesired reactions with a protecting or blocking group. Functional groups that may be protected include, by way of example, carboxy groups, amino groups, hydroxyl groups, thiol groups, carbonyl groups and the like. Suitable protecting groups for such functional groups are well known to those of ordinary skill in the art as exemplified by the teachings 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 "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. In the present invention, the cation typically comprises a protonated form of a compound of formula I, i.e. where one or more amino groups having been protonated by an acid. 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.
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 or asthma) in a patient, such as a mammal (particularly a human) that includes:
(a) preventing the disease or medical condition from occurring, i.e., prophylactic treatment of a patient; (b) ameliorating the disease or medical condition, i.e., eliminating or causing regression of the disease or medical condition in a patient;
(c) suppressing the disease or medical condition, i.e., slowing or arresting the development of the disease or medical condition in a patient; or
(d) alleviating the symptoms of the disease or medical condition in a patient. All other terms used herein are intended to have their ordinary meaning as understood by those of ordinary skill in the art to which they pertain.
General Synthetic Procedures
The compounds 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 or known by those of ordinary skill in the art. Although the following procedures may illustrate a particular embodiment or aspect of the present invention, those skilled in the art will recognize that other embodiments or aspects of the present invention can be prepared using the same or similar methods or by using other methods, reagents and starting materials known to those skilled in the art. It will also be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. While optimum reaction conditions will typically vary depending on various reaction parameters such as the reactants, solvents and quantities used, those of ordinary skill in the art can readily determine suitable reaction conditions using 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
-H-- undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions and reagents 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.
In one embodiment, the compounds of formula I are synthesized as illustrated in Scheme I:
Scheme I
Figure imgf000013_0001
Compound of Formula I wherein P1 is a hydroxyl-protecting group; and R3a and R3b are selected independently from Ci-6 alkyl, or R3a and R3b are joined to form C2-6 alkylene.
As shown in Scheme I, compound I1 can be reacted with about 0.95 to about 1.05 molar equivalents of compound 2 to provide compound 3. This Michael reaction is typically conducted at a temperature ranging from about 0 0C to about 15 0C, typically about 40 0C to about 45 0C, for about 8 to about 24 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as dichloromethane or mixtures of dichloromethane and methanol or ethanol, and the like. Upon completion of the reaction, the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Alternatively, the reaction mixture containing compound 3 can be used directly in the next step of the synthesis.
Compound 3 is then reacted with aqueous acid to hydrolyze the acetal group and provide aldehyde compound 4. Any suitable acid can be employed in this reaction including, byway of example, hydrochloric acid, sulfuric acid, methanesulfonic acid, p-toluenesulfonic acid and the like. The hydrolysis reaction is typically conducted at a temperature ranging from about 0 0C to about 30 0C, typically about 20 0C to about 25 0C, for about 1 to about 6 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as methanol, ethanol, isopropanol, dichloromethane/ethanol, acetonitrile and the like. Upon completion of the reaction, the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Compound 4 is then reacted with about 0.95 to about 1.5 molar equivalents of compound 5 in the presence of a reducing agent to afford compound 6. Any suitable reducing agent may be used in this reaction including, by way of illustration, a metal ' hydride reagent, such as sodium borohydride, sodium triacetoxyborohydride, sodium cyanoborohydride and the like, or hydrogen and a metal catalyst, such as palladium on carbon, and the like. This reductive alkylation reaction is typically conducted at a temperature ranging from about -20 0C to about 30 0C, typically about 0 0C to about 5 0C, for about 1 to about 6 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent and a protic solvent, such as dichloroethane and methanol, and the like. Upon completion of the reaction, the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Compound 6 is then deprotected to provide a compound of formula I. The particular conditions used to deprotect compound 6 will depend on the protecting group employed. For example, when P1 is a silyl protecting group (i.e., a compound of formula 6a as defined herein), such as tert-butyldimethylsilyl, tert-butyldiphenylsilyl, diphenylmethylsilyl, di-tert-buylmethylsilyl, tert-butoxydiphenylsilyl and the like, this deprotection reaction is typically conducted by contacting compound 6a with a source of fluoride ion. In a particular embodiment, the source of fluoride ion is triethylamme trihydrofluoride. Other suitable sources of fluoride ion include tetrabutylammonium fluoride, potassium fluoride with 18-crown-6, hydrogen fluoride, pyridine hydrofluoride, and the like. This reaction is typically conducted at a temperature ranging from about 0 0C to about 50 0C, typically about 10 0C to about 25 0C, for about 24 to about 72 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as dichloroethane and the like. Upon completion of the reaction, the product is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Compound 1 is known in the art or can be prepared from commercially available starting materials and reagents using known procedures. See, for example, U.S. Patent Application Publication No. U.S. 2004/0167167 Al and R. Naito et al., Chem. Pharm. Bull., 46(8) 1286-1294 (1998). By way of example, compound 1_ can be prepared as illustrated in Scheme II:
Scheme II
Figure imgf000015_0001
As shown in Scheme II, biphenyl-2-isocyanate (7) is reacted with an //-protected 4- hydroxypiperidine 8, where P2 is an amino-protecting group such as benzyl, to provide carbamate intermediate 9. This reaction is typically conducted at a temperature ranging from about 20 0C to about 100 0C, typically about 60 0C to about 80 0C, for about 6 to about 24 hours or until the reaction is substantially complete. If desired, this reaction can be conducted in a suitable diluent, such as dichloromethane, toluene and the like. Alternatively, this reaction can be conducted in the absence of a diluent. Upon completion of the reaction, the product 9 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like. Alternatively, the reaction mixture containing compound 9 is used directly in the next step of the synthesis. The amino-protecting group, P2, is then removed from compound 9 using conventional procedures to afford compound I1. For example, when P2 is a benzyl group, compound 9 can be deprotected using hydrogen or ammonium formate, in the presence of a catalyst, such as a palladium catalyst. Representative catalysts include, by way of illustration, palladium on carbon, palladium hydroxide on carbon and the like. This reaction is typically conducted at a temperature ranging from about 20 0C to about 50 0C, typically about 40 0C, for about 6 to about 24 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as methanol, ethanol, isopropanol and the like. Upon completion of the reaction, compound 1 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Compounds of formula 2 are known in the art or can be prepared from commercially available starting materials and reagents using known procedures. By way of illustration, compounds of formula 2 can be prepared as shown in Scheme III:
Scheme HI
Figure imgf000016_0001
As shown in Scheme HI, an aniline compound of formula 10, where X1 is bromo or iodo, is first protected at the amino group to provide a compound of formula IjL-, where P a is an amino-protecting group and P3b is hydrogen or an arnino-protecting group. Any suitable amino-protecting group may be used, such as benzyl, 4-methoxybenzyl, trifluoroacetyl and the like. For example, a compound of formula 10 can be reacted with about 2 or more molar equivalents, preferably about 2.5 to about 3.0 molar equivalents, of a benzyl halide, such as benzyl chloride, bromide or iodide, to afford compound 11, where P3a and P3b are both benzyl. This reaction is typically conducted at a temperature ranging from about 0 0C to about 50 0C, typically about 30 0C, for about 18 to about 24 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as methanol, ethanol, isopropanol and the like. Typically, this reaction is also conducted in the presence of a suitable base, such as potassium carbonate, sodium carbonate and the like. Upon completion of the reaction, compound 11. is typically isolated using conventional procedures, such as extraction, recrystallization, . chromatography and the like.
Representative compounds of formula 10 that can be employed in this reaction include 2,5-dϊmethyl-4-iodoaniline, 2,5-diethyl-4-iodoaniline, 2-ethyl-4-iodo-5- methylaniline, 5-ethyl-4-iodo-2-methylaniline, 4-bromo-2,5-dimethylaniline, 4-bromo-2,5- diethylaniline, 4-bromo-2-ethyl-5-methylaniline, 4-bromo-5-ethyl-2-methylaniline and the like. Such compounds are commercially available (for example, from Spectra Group Limited, Inc., Millbury, OH) or can be prepared from commercially available starting materials and reagents using conventional procedures.
Compound IJ, is then contacted with about 1 to about 2 molar equivalents of an alkyl lithium reagent, such as «-butyllithium or tørt-butyllithium, to form the corresponding anion in which the X1 group has been exchanged for lithium. This reaction is typically conducted at a temperature ranging from about -70 0C to about 0 °C, typically about -20 0C, for about 0.25 to about 1 hour or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as toluene, xylene, tetrahydrofuran and the like. The resulting lithium anion is not isolated, but is reacted in situ with a molar excess of TVyV-dimethylformamide to provide compound IJ2. Generally, about 2 to about 4 molar equivalents of iV^V-dimethylformamide are used. This reaction is typically conducted at a temperature ranging from about -70 0C to about 0 0C, typically about -20 0C to about 00C, for about 0.5 to about 2 hours or until the reaction is substantially complete. Upon completion of the reaction, compound 12 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
The aldehyde group of compound 12_ is then protected as an acetal by reacting compound 12_ with an alcohol or a diol in the presence of an acid catalyst. Any suitable alcohol or diol can be used in this reaction. For example, representative alcohols and diols include methanol, ethanol, n-propanol, ethylene glycol, propylene glycol and the like. Generally, a molar excess of the alcohol or diol are employed in this reaction, preferably about 2 to about 4 molar equivalents. Any suitable acid catalyst may be used in this reaction to facilitate formation of the acetal. Representative acid catalysis include, by way of example, />-toluenesulfonic acid, benzenesulfonic acid, hydrochloric acid and the like.
The reaction is typically conducted at a temperature ranging from about 50 0C to about 100 0C, typically about 60 0C to about 80 0C, for about 12 to about 24 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as toluene, xylene and the like. Typically, the reaction is conducted in a manner which allows the water produced to be removed, such as by azeotropic distillation or by the use of molecular sieves. Upon completion of the reaction, compound ,13 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like. Alternatively, the reaction mixture containing compound jL3 can be used directly in the next step of the synthesis.
After formation of the acetal, the amino group of compound 13 is deprotected using standard reagents and conditions to form compound 14. For example, if P3a and P3b are benzyl groups, compound 13 can be deprotected using hydrogen and a catalyst, such as a palladium catalyst. Representative catalysts include, by way of example, palladium on carbon, palladium hydroxide on carbon, and the like. This reaction is typically conducted at a temperature ranging from about 20 0C to about 50 0C, typically about 25 0C to about 30 0C, for about 4 to about 12 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as methanol, ethanol, ethanol/ethyl acetate mixtures and the like. Upon completion of the reaction, compound 14 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like. Compound 14 is then reacted with an acryloyl halide 15, where X2 is chloro, bromo or iodo, to form compound 2. This reaction is typically conducted at a temperature ranging from about -20 0C to about 25 0C5 typically about 0 0C to about 5 0C, for about 0.5 to about 6 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as dichloromethane and the like, in the presence of a suitable base, such as diisopropylethylamine, triethylamine and the like. Generally, about 1 to 1.2 molar equivalents of the acryloyl halide and about 1 to about 2 molar equivalent of the base are used in this reaction. Upon completion of the reaction, compound 2 is typically isolated using conventional procedures, such as extraction, recrystallization, chromatography and the like.
Compounds of formula 5 are known in the art or can be prepared from commercially available starting materials and reagents using known procedures. By way of illustration, compounds of formula 5 can be prepared as shown in Scheme IV:
Scheme IV
Figure imgf000019_0001
18
As illustrated in Scheme IV, compounds of formula 5 can be prepared from compounds of formula 16, where P4 is a hydroxyl -protecting group, such as benzyl, and X3 is a leaving group, such as chloro, bromo or iodo. Compounds of formula 16 are known in the art. For example, U.S. Patent No. 6,268,533 Bl, issued July 31, 2001; and R. Hett et al., Organic Process Research & Development 1998, 2, 96-99; describe the preparation of iV-[2-benzyloxy-5-((i?)-2-bromo-l-hydroxyethyl)phenyl]formamide (i.e., the (R) enantiomer of compound 16, where P4 is benzyl and X3 is bromo) starting from 2-bromo- 4'-benzyloxy-3'-nitroacetophenone, the synthesis of which is described in K. Murase et al., Chem. Pharm. Bull, 25(6) 1368-1377 (1977). If desired, the racemic form of compound 16 can be prepared by using a non-chiral reducing agent, such as borane dimethylsulfide complex, to reduce 2-bromo-4'-benzyloxy-3'-nitroacetophenone.
The hydroxyl group of compound 16 is protected using conventional procedures and reagents to provide compound 17, where P1 is a hydroxyl-protecting group. In a particular embodiment, the hydroxyl-protecting group is a silyl protecting group, such as dimethylisopropylsilyl, diethylisopropylsilyl, dimethylhexylsilyl, tert-butyldimethylsilyl, terf-butyldiphenylsilyl, diphenylmethylsilyl and the like. For example, compound 16 can be reacted with about 0.95 to about 1.2 molar equivalent of tert-butyldimethylsilyl chloride in the presence of about 1.1 to about 1.3 molar equivalents of imidazole to provide compound ,17 where P1 is tørt-butyldimethylsilyl. This reaction is typically conducted at a temperature ranging from about 0 0C to about 50 0C, typically at room temperature, for about 24 to about 48 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as Aζ^-dimethylformamide and the like. Upon completion of the reaction, compound 17 is typically isolated using conventional procedures, such as extraction, chromatography and the like. By way of further illustration, the synthesis of iV-{2-benzyloxy-5-[(i?)-2-bromo-l-(tert- butyldimethylsilanyloxy)ethyl]phenyl}formamide is described in Example 2 of U.S. Patent Publication No. US 2004/0248985 Al, published December 9, 2004. Compound 17 is then reacted with a benzyl amine (i.e., Bn^NH2) to afford compound IJJ, where Bn1 is an unsubstituted benzyl group or a substituted benzyl group having 1 to 3 substituents on the phenyl ring of the benzyl group selected independently from CM alkyl or Ci-4 alkoxy. Representative benzyl amines include, benzylamine, 3,4- dimethoxybenzylamine, 4-methoxybenzylamine, 4-methylbenzylamine and the like. This reaction is typically conducted by contacting compound 17 with about 2 to about 4 molar equivalents of the benzyl amine at a temperature ranging from about 40 0C to about 100 0C, typically from about 80 0C to about 90 0C, for about 5 to about 24 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as N-methyl-2-pyrrolidone (NMP) and the like. Upon completion of the reaction, compound 18 is typically isolated using conventional procedures, such as extraction, chromatography, recrystallization and the like.
Removal of the benzyl group, Bn1, and P4 using conventional procedures and reagents then affords compound 5. In one embodiment, both Bn1 and P4 are benzyl groups that are removed in the same reaction mixture. Typically, this reaction is conducted by contacting compound 5 with hydrogen in the presence of a catalyst, such as a palladium catalyst. Representative catalysts include palladium hydroxide on carbon, palladium on carbon, and the like. Generally, this debenzylation reaction is conducted in the presence of an acid, such as acetic acid, formic acid and the like. This reaction is typically conducted at a temperature ranging from about 10 0C to about 50 0C, typically at room temperature, for about 6 to about 24 hours or until the reaction is substantially complete. Generally, this reaction is conducted in a suitable diluent, such as methanol, ethanol and the like. Upon completion of the reaction, compound 5 is typically isolated using conventional procedures, such as extraction, chromatography and the like. In a particular embodiment, compound 5 is isolated as the acetic acid salt.
Those of ordinary skill in the art will recognize that the compounds of formula I can also be prepared by other synthetic procedures. For example, the particular order in which the synthetic steps are conducted can be changed or different intermediates can be employed. By way of illustration, compounds of formula III:
Figure imgf000021_0001
where Y1 is -CN, -C(O)OH or -C(O)OR30 can be reduced using conventional procedures and reagents to provide aldehyde 4 (i.e., where Y1 is -CHO). Additionally, such compounds can be reduced to the alcohol, i.e., where Y1 is -CH2OH, and the alcohol can then be oxidized using standard procedures and reagents to provide aldehyde 4 (i.e., where Y1 is -CHO).
If desired, pharmaceutically acceptable salts of the compounds of formula I can be prepared by contacting the free base form of a compound of formula I with a pharmaceutically acceptable acid.
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, Combinations and Formulations
The compounds 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 of a compound of this invention, (i.e., free base, pharmaceutically acceptable salt, solvate, etc.) that is suitable for a particular mode of administration can be used in the pharmaceutical compositions discussed herein.
Accordingly, in one of its compositions aspects, this invention relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a compound of formula I. 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 of formula I. Those skilled in the art will recognize, however, that a pharmaceutical composition may contain more than a therapeutically effective amount, i.e., bulk compositions, or less than a therapeutically effective amount, i.e., individual unit doses designed for multiple administration to achieve a therapeutically effective amount.
Typically, the pharmaceutical composition will contain from about 0.01 to about 95 percent by weight of the therapeutic agent; including, from about 0.01 to about 30 percent by weight; such as from about 0.01 to about 10 percent by weight of the therapeutic 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 of ordinary skill in the pharmaceutical arts. The carriers or excipients used in the pharmaceutical compositions of this invention are commercially available. For example, such materials can be purchased from Sigma (St. Louis, MO). By way of further illustration, conventional formulation techniques are well known to those of ordinary skill in the art as exemplified by the teachings in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: (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 hydrofiuorocarbons; 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 of the invention with a pharmaceutically acceptable carrier and any 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 therapeutic 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 therapeutic 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 therapeutic agent is typically dissolved in a suitable carrier to form a solution. Alternatively, the therapeutic agent can be micronized and combined with a suitable carrier to form a suspension of micronized particles. Nebulizer devices suitable for administering therapeutic agents by inhalation are well known to those of ordinary skill in the art or such devices are commercially available. For example, representative nebulizer devices or products include the Respimat Softmist Inhalaler (Boehringer Ingelheim); the AERx Pulmonary Delivery System (Aradigm Corp.); the PARI LC Plus Reusable Nebulizer (Pari GmbH); and the like.
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. In one embodiment, such a solution has a pH of about 4 to about 6.
In another specific embodiment of this invention, the pharmaceutical composition comprising the therapeutic agent is administered by inhalation using a dry powder inhaler. Such dry powder inhalers typically administer the therapeutic 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 therapeutic agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid (PLA), polylactide-co-glycolide (PLGA) or combinations thereof. Typically, the therapeutic agent is micronized and combined with a suitable carrier to form a blend suitable for inhalation. Accordingly, in one embodiment of the invention, the compound of formula I is in micronized form. A representative pharmaceutical composition for use in a dry powder inhaler comprises dry milled lactose and micronized particles of a compound of formula I.
Such a dry powder formulation can be made, for example, by combining the lactose with the therapeutic agent and then dry blending the components. Alternatively, if desired, the therapeutic agent can be formulated without an excipient. The pharmaceutical composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
Dry powder inhaler delivery devices suitable for administering therapeutic agents by inhalation are well known to those of ordinary skill in the art or such devices are commercially available. For example, representative dry powder inhaler delivery devices or products include Aeolizer (Novartis); Airmax (IVAX); ClickHaler (Ennovata Biomed); Diskhaler (GlaxoSmithKline); Diskus/Accuhaler (GlaxoSmithKline); Easyhaler (Orion Pharma); Eclipse (Aventis); FlowCaps (Hovione); Handihaler (Boehringer Ingelheim); Pulvinal (Chiesi); Rotahaler (GlaxoSmithKline); SkyeHaler/Certihaler (SkyePharma); Twisthaler (Schering-Plough); Turbuhaler (AstraZeneca); Ultrahaler (Aventis); and the like.
In yet another specific embodiment of this invention, the pharmaceutical composition comprising the therapeutic agent is administered by inhalation using a metered-dose inhaler. Such metered-dose inhalers typically discharge a measured amount of the therapeutic agent using a compressed propellant gas. Accordingly, pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension of the therapeutic agent in a liquefied propellant. Any suitable liquefied propellant may be employed including chlorofluorocarbons, such as CCl3F, and hydrofluoroalkanes (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 preferred. 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.
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; 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 hydrofluoroalkane to a suitable container containing the therapeutic agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the therapeutic 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. Metered-dose inhaler devices suitable for administering therapeutic agents by inhalation are well known to those of ordinary skill in the art or such devices are commercially available. For example, representative metered-dose inhaler devices or products include AeroBid Inhaler System (Forest Pharmaceuticals); Atrovent Inhalation Aerosol (Boehringer Ingelheim); Flovent (GlaxoSmithKline); Maxair Inhaler (3M); Proventil Inhaler (Schering); Serevent Inhalation Aerosol (GlaxoSmithKline); and the like.
In another embodiment, the pharmaceutical compositions of this invention are suitable for oral administration. Suitable pharmaceutical compositions for oral administration maybe in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present invention as an active ingredient. When intended for oral administration in a solid dosage form (i.e., as capsules, tablets, pills and the like), the pharmaceutical compositions of this invention will typically comprise a compound of the present invention as the active ingredient and one or more pharmaceutically acceptable carriers, 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 (PVAP), hydroxypropyl methylcellulose phthalate, methacrylic acidlmethacrylic 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 of the present invention may also be formulated to provide slow or controlled release of the active ingredient using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres. In addition, the 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 of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro- encapsulated form, if appropriate, with one or more of the above-described excipients.
Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. 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-butylene 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 may be 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 glycol monolaurate, azacycloalkan-2-ones and the like, and incorporated into a patch or similar delivery system. Additional excipients including gelling agents, emulsifiers and buffers, may be used in such transdermal compositions if desired. Additionally, the compounds of this invention can be administered parenterally, i.e., intravenously, subcutaneously or intramuscularly. For parenteral administration, a compound of formula I is typically dissolved in a carrier acceptable for parenteral administration, such as sterile water, saline, vegetable oil and the like. By way of illustration, an intravenous composition typically comprises a sterile aqueous solution of a compound of formula I, wherein the solution has a pH in the range of about 4 to about 7.
If desired, the compounds of this invention may be administered in combination with one or more other therapeutic agents. In this embodiment, a compound of this invention is either physically mixed with the other therapeutic agent to form a composition containing both agents; or each agent is present in separate and distinct compositions which are administered to the patient simultaneously or sequentially.
For example, a compound of formula I can be combined with second therapeutic agent using conventional procedures and equipment to form a composition comprising a compound of formula I and a second therapeutic agent. Additionally, the therapeutic agents may be combined with a pharmaceutically acceptable carrier to form a pharmaceutical composition comprising a compound of formula I, a second therapeutic agent and a pharmaceutically acceptable carrier. In this embodiment, the components of the composition are typically mixed or blended to create a physical mixture. The physical mixture is then administered in a therapeutically effective amount using any of the routes described herein.
Alternatively, the therapeutic agents may remain separate and distinct before administration to the patient. In this embodiment, the therapeutic agents are not physically mixed together before administration but are administered simultaneously or sequentially as separate compositions. For example, a compound of formula I can be administered by inhalation simultaneously or sequentially with another therapeutic agent using an inhalation delivery device that employs separate compartments (e.g. blister packs) for each therapeutic agent. Alternatively, the combination may be administered using separate delivery devices, i.e., one delivery device for each therapeutic agent. Additionally, the therapeutic agents can be delivered by different routes of administration, i.e., one by inhalation and the other by oral administration.
Any therapeutic agent compatible with the compounds of the present invention may be used in combination with such compounds. In a particular embodiment, the second therapeutic agent is one that is effectively administered by inhalation. By way of illustration, representative types of therapeutic agents that may be used with the compounds of this invention include, but are not limited to, anti-inflammatory agents, such as steroidal anti-inflammatory agents (including corticosteroids and glucocorticoids), nonsteroidal anti-inflammatory agents (NSAIDs), and PDE4 inhibitors; bronchodilators, such as PDE3 inhibitors, adenosine 2b modulators and β2 adrenergic receptor agonists; antiinfective agents, such as Gram-positive antibiotics, Gram-negative antibiotics, and antiviral agents; antihistamines; protease inhibitors; afferent blockers, such as D2 agonists and neurokinin modulators; and muscarinic receptor antagonists (antichlolinergic agents). Numerous examples of such therapeutic agents are well known in the art. Suitable doses for the other therapeutic agents administered in combination with a compound of the invention are typically in the range of about 0.05 μg/day to about 500 mg/day. hi a particular embodiment of this invention, a compound of formula I is administered in combination with a steroidal anti-inflammatory agent. Representative examples of steroidal anti-inflammatory agents that can be used in combination with the compounds of this invention include, but are not limited to, beclomethasone dipropionate; budesonide; butixocort propionate; 20i2-16α,17α-[butylidenebis(oxy)]-6α,9α-difluoro-l lβ- hydroxy-17β-(methylthio)androsta-4-en-3-one (RPR-106541); ciclesonide; dexamethasone; 6α,9α-difiuoro-17α -[(2-furanylcarbonyl)oxy]-l lβ-hydroxy-16α-methyl- 3-oxoandrosta-l,4-diene-17β-carbothioic acid S-fluoromethyl ester; 6α,9α-difluoro-l lβ- hydroxy- 16α-methyl- 17α -[(4-methytl- 1 ,3-thiazole-5-carbonyl)oxy]-3-oxoandrosta-l ,4- diene-17β-carbothioic acid S-fluoromethyl ester; 6α,9α-difluoro-l lβ-hydroxy-16α-methyl- 3-oxo- 17α-propionyloxyandrosta-l ,4-diene-l 7β-carbothioic acid (S)-(2- oxotetrahydrofuran-3iS'-yl) ester; flunisolide; fluticasone propionate; methyl prednisolone; mometasone furoate; prednisolone; prednisone; rofleponide; ST-126; triamcinolone acetonide; and the like, or pharmaceutically acceptable salts thereof. Such steroidal antiinflammatory agents are commercially available or can be prepared using conventional procedures and reagents. For example, the preparation and use of steroidal antiinflammatory agents is described in U.S. Patent No. 6,750,210 B2, issued June 15, 2004; U.S. Patent No. 6,759,398 B2, issued July 6, 2004; U.S. Patent No. 6,537,983, issued March 25, 2003; U.S. Patent Application Publication No. US 2002/0019378 Al, published February 14, 2002; and references cited therein.
When employed, the steroidal anti-inflammatory agent is typically administered in an amount that produces a therapeutically beneficial effect when co-administered with a compound of the invention. Typically, the steroidal anti-inflammatory agent will be administered in an amount sufficient to provide from about 0.05 μg to about 500 μg per dose.
The following examples illustrate representative pharmaceutical compositions of the present invention:
Example A
Dry Powder Composition
A micronized compound of the invention (100 mg) is blended with milled lactose (25 g) (e.g., lactose in which not greater than about 85% of the particles have a MMD of about 60 μm to about 90 μm and not less than 15% of the particles have a MMD of less then 15 μm). This blended mixture is then loaded into individual blisters of a peelable blister pack in an amount sufficient to provide about 10 μg to about 500 μg of the compound of the invention per dose. The contents of the blisters are administered using a dry powder inhaler.
Example B
Dry Powder Composition
A micronized compound of the invention (1 g) is blended with milled lactose (200 g) to form a bulk composition having a weight ratio of compound to milled lactose of 1 :200. The blended composition is packed into a dry powder inhalation device capable of delivering between about 10 μg to about 500 μg of the compound of the invention per dose.
Example C
Dry Powder Composition
A micronized compound of the invention (100 mg) and a micronized steroidal antiinflammatory agent (500 mg) are blended with milled lactose (30 g). This blended mixture is then loaded into individual blisters of a peelable blister pack in an amount sufficient to provide about 10 μg to about 500 μg of the compound of the invention per dose. The contents of the blisters are administered using a dry powder inhaler. Example D
Metered-Dose Inhaler Composition
A micronized compound of the invention (10 g) is dispersed in a solution prepared by dissolving lecithin (0.2 g) in demineralized water (200 mL). The resulting suspension is spray dried and then micronized to form a micronized composition comprising particles having a mean diameter less than about 1.5 μm. The micronized composition is then loaded into metered-dose inhaler cartridges containing pressurized 1,1,1,2- tetrafluoroethane in an amount sufficient to provide about 10 μg to about 500 μg of the compound of the invention per dose when administered by the metered dose inhaler.
Example E
Nebulizer Composition
A compound of the invention (25 mg) is dissolved in citrate buffered (pH 5) isotonic saline (125 mL). The mixture is stirred and sonicated until the compound is dissolved. The pH of the solution is checked and adjusted, if necessary, to pH 5 by slowly adding aqueous IN sodium hydroxide. The solution is administered using a nebulizer device that provides about 10 μg to about 500 μg of the compound of the invention per dose.
Example F
Hard Gelatin Capsules
A compound of the invention (50 g), spray-dried lactose (440 g) and magnesium stearate (10 g) are thoroughly blended. The resulting composition is loaded into a hard gelatin capsule (500 mg of composition per capsule) that are administered orally.
Example G Oral Suspension
The following ingredients are thoroughly mixed to form a suspension for oral administration: Ingredients Amount
Compound of the invention 1.0 g
Fumaric acid 0.5 g
Sodium chloride 2.O g
Methyl paraben 0.15 g
Propyl paraben 0.05 g
Granulated sugar 25.5 g
Sorbitol (70% solution) 12.85 g
VEEGUM® K (magnesium aluminum silicate) 1.0 g
Flavoring 0.035 mL
Coloring 0.5 mg
Distilled water q.s. to 10O mL
The resulting suspension contains 100 mg of active ingredient per 10 mL of suspension. The suspension is administered orally.
Example H
Injectable Composition A compound of the invention (0.2 g) is blended with 0.4 M sodium acetate buffer solution (2.0 mL). The pH of the resulting solution is adjusted to pH 4 using 0.5 N aqueous hydrochloric acid or 0.5 N aqueous sodium hydroxide, as necessary, and then sufficient water for injection is added to provide a total volume of 20 mL. The mixture is then filtered through a sterile filter (0.22 micron) to provide a sterile solution suitable for administration by injection. Utility
The compounds of this invention possess both p2 adrenergic receptor agonist and muscarinic receptor antagonist activity and therefore, such compounds are expected to be useful as therapeutic agents 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 are well known to those of ordinary skill in the art as exemplified by the teachings of Eglen et al., Muscarinic Receptor Subtypes: Pharmacology and Therapuetic Potential, DN&P 10(8), 462-469 (1997); Emilien et al., Current Therapeutic Uses and Potential of beta-Adrenoceptor Agonists and Antagonists, European J. Clinical Pharm. , 53(6), 389-404 (1998); and references cited therein. 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 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 relates 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. 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 dose per day or a single dose per week. Generally, the dose for treating a pulmonary disorder will range from about 10 μg/day to about 500 μg/day.
In one of its method aspects, this invention relates to a method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient a therapeutically effective amount of a compound of formula I. Generally, the dose for treating COPD or asthma will range from about 10 μg/day to about 500 μg/day. The term "COPD" is understood by those of ordinary skill in the art to include a variety of respiratory conditions, including chronic obstructive bronchitis and emphysema, as exemplified by the teachings of Barnes, Chronic Obstructive Pulmonary Disease, N. Engl. J. Med., 2000: 343:269-78, and references cited therein. When administered by inhalation, the compounds of this invention typically have the effect of producing bronchodilation. Accordingly, in another of its method aspects, this invention relates to a method of producing bronchodilation in a mammal, the method comprising administering to a mammal a bronchodilation-producing amount of a compound of formula I. Generally, the dose for producing bronchodilation will range from about 10 μg/day to about 500 μg/day.
When used as a therapeutic agent, the compounds of this invention are optionally administered in combination with another therapeutic agent or agents. In particular, by administering the compounds of this invention with a steroidal anti-inflammatory agent, triple therapy, i.e., β2 adrenergic receptor agonist activity, muscarinic receptor antagonist activity and anti-inflammatory activity, can be achieved using only two therapeutic agents. Since pharmaceutical compositions (and combinations) containing two therapeutic agents are typically easier to formulate and/or administer compared to compositions containing three therapeutic agents, such two component compositions provide a significant advantage over compositions containing three therapeutic agents. Accordingly, in a particular embodiment, the pharmaceutical compositions, combinations and methods of this invention further comprise a steroidal anti-inflammatory agent.
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. Additionally, such compounds are useful in screening assays to discover, for example, 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. When used as a research tool, a biological system or sample comprising a β2 adrenergic receptor and/or a muscarinic receptor is typically contacted with a β2 adrenergic receptor-agonizing or muscarinic receptor-antagonizing amount of a compound of this invention. The effects on the biological system or sample caused by the compound are then determined or measured using conventional procedures and equipment, such as by measuring binding in a radioligand binding assays or ligand-mediated changes in a " functional assay or by determining the amount of bronchoprotection provided by the compound in a bronchoprotection assay in a mammal. . Representative ligand-mediated changes in a functional assay include ligand-mediated changes in intracellular cyclic adenosine monophosphate (cAMP); ligand-mediated changes in activity of the enzyme adenylyl cyclase (which synthesizes cAMP); ligand-mediated changes in incorporation of guanosine 5'-O-(thio)triphosρhate ([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 FLJPR® from Molecular Devices, Inc.); and the like. Compounds of this invention are expected to agonize or cause activation of a β2 adrenergic receptor and antagonize or decrease the activation of muscarinic receptors in the functional assays listed herein or in assays of a similar nature. The compounds of this invention will typically be used in these studies at a concentration ranging from about 0.1 nanomolar to about 100 nanomolar.
Additionally, the compounds of this invention can be used as research tools for evaluating other chemical compounds. In this aspect of the invention, a compound of formula I is used as a standard in an assay to allow comparison of the results obtained with a test compound and the compound of formula I. For example, β2 adrenergic receptor and/or muscarinic receptor binding data (as determined, for example, by in vitro radioligand displacement assays) for a test compound or a group of test compounds is compared to the β2 adrenergic receptor and/or muscarinic receptor binding data for a compound of formula I to identify those test compounds that have desirable binding, i.e. test compounds having binding about equal or superior to a compound of formula I, if any. Alternatively, for example, bronchoprotective effects can be determined for test compounds and a compound of formula I in a bronchoprotection assay in a mammal and this data compared to identify test compounds providing about equal or superior bronchoprotective effects. This aspect of the invention includes, as separate embodiments, both (i) the generation of comparison data (using the appropriate assays) and (ii) the analysis of the test data to identify test compounds of interest.
The properties and utility of the compounds of this invention can be demonstrated using various in vitro and in vivo assays well known to those of ordinary skill 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 and aspects of this invention. The illustration of specific embodiments and aspects, however, is not intended to limit the scope of this invention in any way unless specifically indicated.
All reagents, starting materials and solvents used in the following examples were purchased from commercial suppliers (such as Aldrich, Fluka, Sigma and the like) and were used without further purification unless otherwise indicated.
In the following examples, HPLC analysis was typically conducted using an Agilent (Palo Alto, CA) Series 1100 instrument with Zorbax Bonus RP 2.1 x 50 mm columns, supplied by Agilent, (a C 14 column), having a 3.5 micron particle size. Detection was by UV absorbance at 214 nm. Mobile phase "A" was 2 % acetonitrile, 97.9 % water, and 0.1% trifluoro acetic acid (v/v/v); and mobile phase "B" was 89.9 % acetonitrile, 10 % water, and 0.1 % trifluoroacetic acid (v/v/v). HPLC (10-70) data were obtained with a flow rate of 0.5 mL/minute of 10% to 70% mobile phase B gradient over a 6-minute period; HPLC (5-35) data were obtained with a flow rate of 0.5 mL/minute of 5% to 35% mobile phase B gradient over a 5-minute period; and HPLC (10-90) data were obtained with a flow rate of 0.5 mL/minute of 10% to 90% mobile phase B gradient over a 5-minute period.
Liquid chromatography mass spectrometry (LCMS) data typically were obtained with an Applied Biosystems (Foster City, CA) Model API-150EX instrument. LCMS 10- 90 data were obtained with a 10% to 90% mobile phase B gradient over a 5-minute period.
Small-scale purifications were typically conducted using an API 150EX Prep Workstation system from Applied Biosystems. The mobile phase "A" was water containing 0.05% trifluoroacetic acid (v/v); and mobile phase "B" was acetonitrile containing 0.05% trifluoroacetic acid (v/v). For small samples (about 3 to 50 mg recovered sample size), the following conditions were typically used: 20 mIVmin 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 samples (i.e., greater than about 100 mg crude sample), the following conditions were typically used: 60 mL/min flow rate; 30 min gradients and a 41.4 mm x 250 mm Microsorb BDS column with 10 micron particles (Varian, Palo Alto, CA).
Example 1 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 0C 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 500C 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 0C 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 (10N) 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 biphenyl-2-ylcarbamic acid piperidin-4-yl ester (155 g, 100%). HPLC (10-70) Rt
Figure imgf000037_0001
[M + H+] calc'd for C18H20N2O2 297.15; found 297.3.
Example 2 3-[4-(Biphenyl-2-ylcarbamoyIoxy)piperidin-l-yl]propionic Acid
To a solution of biphenyl-2-ylcarbamic acid piperidin-4-yl ester (50 g, 67.6 mmol) in dichloromethane (500 mL) was added acrylic acid (15.05 mL, 100 mmol). The resulting mixture was heated at 500C under reflux for 18 hours and then the solvent was removed. Methanol (600 mL) was added and this mixture was heated at 75 0C for 2 hours 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 3-[4-(biphenyl-2- ylcarbamoyloxy)piperidin-l-yl]propionic acid (6 Ig, 96% purity) as white powder. Example 3 iV-{5-[(ϋf)-2-Amino-l-(ter^-butyIdimethylsiIanyIoxy)ethyI]-2-hydroxyphenyl}- formamide Acetic Acid Salt
Step A - iV-{S-[(/?)-2-Benzylainino-l-(tert-butyIdiinethylsiIanyloxy)ethyI]-2- benzyloxyphenyl}formamide
To a 500 mL three-necked round-bottomed flask was added N-{2-benzyloxy-5- [(i?)-2-bromo-l-(terf-butyldimethylsilanyloxy)ethyl]phenyl}formamide (100 g, 215 mmol) and iV-methyl-2-pyrrolidone (300 mL). Benzylamine (69.4 mL, 648 mol) was added and the reaction mixture was flushed with nitrogen. The reaction mixture was then heated to 90 0C and stirred for about 8 hours. The reaction mixture was then cooled to room temperature and water (1.5 L) and ethyl acetate (1.5 L) were added. The layers were separated and the organic layer was washed with water (500 mL), a 1:1 mixture of water and saturated brine (500 mL total), and then again with water (500 mL). The organic layer was then dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to provide crude N-{5-[(i?)-2-benzylamino-l-(iert-butyldimethylsilanyloxy)ethyl]- 2-benzyloxyphenyl}formamide (100 g, 90% yield, 75-80% purity) as an orange-brown thick oil.
Step B — i\r-{5-[(JR)-2-Amino-l-(terι'-butyIdimethylsiIanyloxy)ethyl]-2-hydroxyphenyI}- formamide Acetic Acid Salt
Crude iV-{5-[(i?)-2-benzylamino-l-(tert-butyldimethylsilanyloxy)ethyl]-2- benzyloxyphenyl} formamide (100 g, 194 mmol) was dissolved in methanol (1 L) and acetic acid (25 mL, 291 mmol). The resulting mixture was purged with dry nitrogen and then palladium hydroxide on carbon (20 g, 20 wt. %, about 50% water) was added. Hydrogen was bubbled through the reaction mixture with stirring at room temperature for about 10 hours. The mixture was then purged with dry nitrogen and the mixture was filtered through Celite. The filtrate was concentrated on a rotary evaporator and ethyl acetate (600 mL) was added to the residue. This mixture was stirred for about 2 hours at which time a thick yellow slurry had developed: The slurry was filtered and the precipitate was air dried to provide N- {5-[(i.)-2-amino- 1 -(ter/-butyldimethylsilanyloxy)ethyl]-2- hydroxyphenyl} formamide acetic acid salt (48 g, 98% purity) as a yellow- white solid. LCMS (10-70) Rt= 3.62; [M + H4] found 311.3. Example 4
Biphenyl-2-yIcarbamic Acid l-[2-(4-{[(J?)-2-(3-FormyIamino-4-hydroxyphenyl)-2- hydroxyethyIamiπo]methyl}-2,5-dimethyIphenylcarbamoyl)ethyI]piperidin-4-yl Ester Step A - Methyl 2,5-Dimethyl-4-nitrobenzoate To a stirred solution of 2,5-diraethyl-4-nitrobenzoic acid (480 mg, 2.4 mmol) in dry methanol (8.2 mL) at 00C under dry nitrogen was added thionyl chloride (0.538 mL, 7.38 mmol). The resulting mixture as allowed to warm to room temperature and stirred for about 7 hours. Additional thionyl chloride (0.300 mL) was added and stirring was continued at room temperature overnight. The solvent was removed under reduced pressure and the residue was dissolved in ethyl acetate. This solution was washed with saturated aqueous sodium bicarbonate, dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide methyl 2,5-dimethyl-4-nitrobenzoate (578 mg) as a pale yellow solid. HPLC (10-70) R4 = 4.61; 1H NMR (300 MHz, CDCl3) δ 2.57 (3H, s), 2.61 (3H5 s), 3.94 (3H, s), 7.82 (IH, s), 7.87 (IH, s).
Step B - Methyl 4-Amino-2,5-dimethylbenzoate
To a stirred solution of methyl 2,5-dimethyl-4-nitrobenzoate (523 mg, 2.5 mmol) in a 9:1 mixture of methanol and water (25 mL total) at 00C was added ammonium chloride (401 mg, 7.5 mmol). Zinc (1.63 g, 25 mmol) was added portionwise and the resulting mixture was stirred at room temperature overnight. The reaction mixture was then filtered through Celite and the Celite pad was washed with methanol. The filtrate was concentrated under reduced pressure and the resulting residue was dissolved in ethyl acetate. This solution was washed with saturated aqueous sodium bicarbonate, dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide methyl 4- amino-2,5-dimethylbenzoate (450 mg) as a yellow oil. 1H NMR (300 MHz, CDCl3) δ 2.14 (3H, s), 2.53 (3H, s), 3.83 (3H, s), 3.85 (2H, br s), 6.48 (IH, s), 7.72 (IH, s).
Step C - Methyl 4-{3-[4-(Biphenyl-2-ylcarbamoyloxy)piperidin-l- yl]propionylamino}-2,5-dimethylbenzoate To a stirred solution of 3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l-yl]propionic acid (670 mg, 1.82 mmol) and methyl 4-amino-2,5-dimethylbenzoate (390 mg, 2.18 mmol) in dichloromethane (3.6 mL) and diisopropylethylamine (0.413 mL) was added 0-(7- azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate (HATU) (829 mg, 2.18 mmol). The resulting mixture was stirred at room temperature overnight. The mixture was then washed with saturated aqueous sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with dichloromethane containing from 3% to 5% methanol to provide methyl 4- {3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l- ylJpropionylamino}-2,5-dimethylbenzoate (568 mg, 59% yield). LCMS (10-70) Rt = 4.55; [M + H+] found 530.4.
Step D - BiphenyI-2-yIcarbamic Acid l-[2-(4-HydroxymethyI-2,5- dimethyIphenylcarbamoyl)ethyl]piperidin-4-yl Ester
To a stirred solution of IM lithium aluminum hydride in THF (1.52 mL, 1.52 mmol) at 0 0C was added methyl 4-{3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l- yl]propionylamino}-2,5-dimethylbenzoate (400 mg, 0.76 mmol). The resulting mixture was stirred at 0 0C for 30 minutes and then a 1 : 1 mixture of IM aqueous sodium hydroxide (5 mL) and water (5 mL) was added and stirring was continued for 2 hours.
Dichloromethane was added and the organic layer was separated, dried over sodium sulfate and the solvent removed under reduced pressure. The residue was purified by silica gel chromatography eluting with dichloromethane containing 5% methanol to provide biphenyl-2-yIcarbamic acid l-[2-(4-hydroxymethyl-2,5-dimethylphenylcarbamoyl)- ethyl]piperidin-4-yl ester. LCMS (10-70) R1 = 3.94; [M + H+] found 502.5.
Step E - Biphenyl-2-ylcarbamic Acid l-[2-(4-Formyl-2,5- dimethylphenyIcarbamoyl)ethy]]pϊperidin-4-yI Ester
To a solution of biphenyl-2-ylcarbamic acid l-[2-(4-hydroxymethyl-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (151 mg, 0.3 mmol) in dichloromethane (3 mL) at 0 0C was added dimethyl sulfoxide (128 μL, 1.8 mmol) and diisopropylethylamine (157 μL, 0.9 mmol). After 15 minutes, sulfur trioxide pyridine complex (143 mg, 0.9 mmol) was added and stirring at 0 0C was continued for 1 hour. Water was added to quench the reaction and the layers were separated. The organic layer was dried over anhydrous sodium sulfate, filtered and the solvent removed under reduced pressure to give biphenyl-2-ylcarbamic acid l-[2-(4-formyl-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (150 mg, 100% yield), which was used without further purification. [M + H+] found 500.4. Step F - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(iϊ)-2-(tert-ButyIdimethylsilanyloxy)-2- (3-formylamino-4-hydroxyphenyl)ethylamino]methyl}-2,5- dimethyIphenylcarbamoyI)ethyl]piperidin-4-yl Ester A solution of biphenyl-2-ylcarbamic acid l-[2-(4-foπnyl-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (150 mg, 0.30 πunol) and N- {5-[(i?)- 2-amino-l-(tert-butyldimethylsilanyloxy)ethyl]-2-hydroxyphenyl}formamide (112 mg, 0.36 mmol) in a 1:1 mixture of dichloromethane and methanol (3.0 mL total) was stirred at room temperature for 30 minutes. Sodium triacetoxyborohydride (191 mg, 0.9 mmol) was added and the resulting mixture was stirred at room temperature overnight. Acetic acid was added to quench the reaction and the mixture was concentrated under reduced pressure to give biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(ter/-butyldimethylsilanyloxy)-2-(3- formylamino-4-hydroxyphenyl)ethylamino]methyl}-2,5-dimethylphenylcarbamoyl)- ethyl]piperidin-4-yl ester, which was used without further purification. LCMS (10-70) Rt = 4.55; [M + H+] found 794.6.
Step G - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(/?)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethyIphenylcarbamoyI)ethyl]piperidin-4-yl Ester To a suspension of biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-(tert- butyldimethylsilanyloxy)-2-(3-formylamino-4-hydroxyphenyl)ethylamino]methyl}-2,5- dimethylphenylcarbamoyl)-ethyl]piperidin-4-yl ester (238 mg, 0.30 mmol) in dichloromethane (3.0 mL) was added triethylamine trihydrofiuoride (147 μL, 0.90 mmol). This mixture was stirred at room temperature overnight and then the mixture was concentrated under reduced pressure. The residue 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 biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)- 2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester as the ditrifluoroacetate salt (50 mg, 97% purity). LPLC (2-90) Rt = 2.76; [M + H+] found 680.8. Example 5
BiphenyI-2-ylcarbamic Acid l-[2-(4-{[(-R)-2-(3-Formylamino-4-hydroxyphenyI)-2- hydroxyethylaminoJmethyl}-2,5-dimethyIphenyIcarbamoyl)ethyI]piperidin-4-yl Ester Step A - Dibenzyl-(4-iodo-2,5-dimethylphenyl)amine To a 2-liter round-bottomed flask equipped with an overhead stirrer, temperature control and an addition funnel was added 4-iodo-2,5-dimethylaniline (100.0 g, 0.405 mol) (from Spectra Group Limited, Inc., Millbury, OH). Ethanol (1 L) and solid potassium carbonate (160 g, 1.159 mol) were added and then neat benzyl bromide (140 niL, 1.179 mol) was added in one portion. The resulting mixture was stirred at 300C for about 18 hours at which time HPLC shows greater than 98 % conversion. The mixture was then cooled to room temperature and hexanes (1 L) were added. This mixture was stirred for 15 minutes and then filtered through a paper filter to removed solids and the filter cake was washed with hexanes (200 mL). Using a rotoevaporator, the volume of the filtrate was reduced to about 500 mL and concentrated hydrochloric acid (30 mL) was added. The remaining solvent was then removed using a rotoevaporator. To the resulting residue was added hexanes (500 mL) and this mixture was stirred for about 30 minutes as which time a free-flowing slurry had formed. The slurry was filtered and the filter cake was washed with hexanes (200 mL) and dried to provide dibenzyl-(4-iodo-2,5-dimethylphenyl)amine hydrochloride (115 g, 62% yield, 97.5% purity) as a greenish colored solid. The dibenzyl-(4-iodo-2,5-dimethylphenyl)amine hydrochloride was transferred to a 3 L flask and toluene (1 L) and 1 M aqueous sodium hydroxide (1 L) were added. The resulting mixture was stirred for 1 hour and then the layers were separated. The organic layer was washed with dilute brine (500 mL) and the solvent was removed by rotoevaporation to provide dibenzyl-(4-iodo-2,5-dimethylphenyl)amine (80 g) as a semi- solid thick oil. (Alternatively, dichloromethane can be used in place of toluene in this step). 1H NMR (300 MHz, DMSCW6) δ 2.05 (3H, s), 2.19 (3H, s), 3.90 (4H, s), 6.91 (IH, s), 7.05-7.20 (1OH, m), 7.42 (IH, s); MS [M + H+] found 428.
Step B - 4-Dibenzylamino-2,5-diniethylbenzaldehyde Hydrochloride To a 1 -liter 3 -necked round-bottomed flask equipped with an overhead stirrer, temperature control and an addition funnel was added dibenzyl-(4-iodo-2,5-dimethylphenyl)amine (15 g, 35 mmol). Toluene (300 mL) was added and the resulting mixture was stirred for about 15 minutes. The reaction flask was purged with dry nitrogen and cooled to about —200C and 1.6 M n-butyllithium in hexanes (33 mL, 53 mmol) was added dropwise via the addition funnel. During the addition, the internal temperature of the reaction mixture was maintained below —10 0C. When the addition was complete, the resulting mixture was stirred at about -15 0C for 15 minutes. ΛζN-Dimethylformamide (10 mL, 129 mmol) was then added dropwise while maintaining the internal reaction temperature below 0 0C. The resulting mixture was then stirred at —20 0C to 00C for about 1 hour. Aqueous 1 M hydrochloric acid (200 mL) was then added over a 5 minute period and the resulting mixture was stirred for 15 minutes. The layers were then separated and the organic layer was washed with dilute brine (100 mL). The organic layer was then dried over anhydrous sodium sulfate, filtered and the solvent removed under reduced pressure to provide 4- dibenzylamino-2,5-dimethylbenzaldehyde hydrochloride (11.5 g, 90% yield, 95% purity) as thick oil that solidified upon standing. The product contained about 3 to 5 % of the des- iodo by-product. 1H NMR (300 MHz, CDCl3) δ 2.42 (3H, s), 2.50 (3H, s), 4.25 (4H, s), 6.82 (IH, s), 7.10-7.30 (1OH, m), 7.62 (IH, s), 10.15 (IH, s); MS [M + H+] found 330.3.
Step C - 4-[l,3]Dioxolan-2-yI-2,5-dimethyIphenylamine
To a 500 mL round-bottomed flask was added 4-dibenzylamino-2,5- dimethylbenzaldehyde hydrochloride (11.5 g, 31.4 mmol) and toluene (150 mL) and the resulting mixture was stirred until the salt completely dissolved. The reaction flask was then purged with dry nitrogen for 5 minutes. Ethylene glycol (5.25 mL, 94.2 mmol) and p- toluenesulfonic acid (760 mg, 6.2 mmol) were added and the resulting mixture was heated at 60 0C to 80 0C for about 20 hours. The solvent was then removed slowly (over about 40 minutes) at 40 °C on a rotary evaporator. Toluene (100 mL) was added to the residue and the solvent was again removed slowly at 40 0C on a rotary evaporator. This process was repeated using another aliquot of toluene (100 mL) and the mixture was evaporated to dryness. Ethyl acetate (150 mL) and saturated aqueous sodium bicarbonate (100 mL) were added to the residue and the layers were separated. The organic layer was washed with brine (50 mL) and then dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to provide crude dibenzyl-(4-[l,3]dioxolan-2-yl-2,5-dimethyl- phenyl)amine (11.4 g).
The crude dibenzyl-(4-[l,3]dioxolan-2-yl-2,5-dimethyl-ρhenyl)amine was dissolved in a 2:1 mixture of ethanol and water (150 mL total) and the resulting mixture was purged with dry nitrogen for 5 minutes. Palladium on carbon (2.3 g, 10 Wt. % containing about 50% water) and solid sodium bicarbonate (1.0 g) were added and the resulting mixture was hydrogenated at about 1 atm of hydrogen at 25 0C to 30 0C for about 8 hours. The mixture was then filtered through Celite and the filtrate was concentrated on a rotary evaporator to provide crude 4-[l,3]dioxolan-2-yl-2,5-dimethylphenylamine (5.6 g, 92% yield) as a thick oil. 1H NMR (300 MHz, DMSO-^6) δ 2.05 (3H, s), 2.22 (3H, s), 3.7- 3.9 (4H, m), 3.95 (4H, s), 5.59 (IH, s), 6.72 (IH, s), 7.0-7.25 (HH, m).
Step D -7V-(4-[l,3]Dioxolan-2-yl-2,5-dimethylphenyl)acrylaini(Ie
To a 500 mL round-bottom flask was added crude 4-[l,3]dioxolan-2-yl-2,5- dimethylphenylarnine (5.6 g, 29 mmol), dichloromethane (100 mL) and diisopropylethylamine (7.6 mL, 43.5 mmol). The resulting mixture was stirred at room temperature until the ingredients dissolved and then the mixture was cooled to 0 0C.
Acryloyl chloride (2.35 mL, 29 mmol) was then added dropwise over a 5 minute period.
The reaction mixture was stirred at 0 0C to 5 0C for 1 hour and then water (50 mL) was added and stirring was continued for about 30 minutes at which time fine solids had formed. The mixture was filtered to collect the solids. The layers of the filtrate were then separated and the organic layer was concentrated under reduced pressure to dryness.
Dichloromethane (50 mL) was added to the residue and this mixture was stirred until a free- flowing slurry developed. The slurry was filtered (using the same runnel used to collect the fine solids above) and the filter cake was washed with dichloromethane (10 mL) and dried to provide N-(4-[l,3]dioxolan-2-yl-2,5-dimethylphenyl)acrylamide (3.1 g, 97% purity) as a white to off-white solid.
The filtrate from above was then evaporated to dryness and methanol (10 mL) was added to the residue. This mixture was stirred for 15 minutes and then the precipitate was collected by filtration, washed with methanol (5 mL) and dried to give a second crop of N-
(4-[l,3]dioxolan-2-yl-2,5-dimethylρhenyl)acrylamide (0.8 g, 95% purity). 1H NMR (300
MHz, CD3OD) δ 2.10 (3H, s), 2.23 (3H, s), 3.85-4.10 (4H, m), 5.60-6.40 (3H, m), 5.59
(IH, s), 7.18 (IH, s), 7.23 (IH, s).
Step E - Biphenyl-2-yIcarbamic Acid 1 -[2-(4-Formy 1-2,5- dimethylphenylcarbamoyI)ethyl]piperidin-4-yI Ester Hydrochloride
To a 50 mL round-bottom flask was added biphenyl-2-ylcarbamic acid piperidin-4- yl ester (1.2 g, 4.04 mmol) and iV-(4-[l,3]dioxolan-2-yl-255-dimethylphenyl)acrylamide (1.0 g, 4.04 mmol). Ethanol (10 mL) and dichloromethane (10 mL) were added to form a slurry. The reaction mixture was heated at 45 0C to 50 0C for about 18 hours and then cooled to room temperature. Aqueous IM hydrochloric acid (10 mL) was added and the resulting mixture was stirred vigorously for about 3 hours. Dichloromethane (10 mL) was added and the resulting mixture was stirred for about 5 minutes. The layers were then separated and the organic layer was dried over anhydrous sodium sulfate, filtered and concentrated on a rotary evaporator to provide crude biphenyl-2-ylcarbamic acid l-[2-(4- formyl-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester hydrochloride (1.9 g). 1H NMR (300 MHz, DMSO-^6) δ 1.2-1.4 (2H, m), 1.58-1.75 (2H, m), 2.0-2.17 (2H5 m), 2.19 (3H, s), 2.38 (3H, s), 2.41-2.50 (4H, m), 2.5-2.75 (2H, m), 4.31-4.42 (IH, m), 7.10-7.35 (9H, m), 7.55 (IH5 s), 7.75 (IH, s), 8.59 (IH, s), 9.82 (IH, s), 9.98 (IH5 s); MS [M + H+] found 500.2.
Step F - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(jR)-2-(ter/-ButyldimethylsiIanyloxy)-2- (3-formylamino-4-hydroxyphenyl)ethylamino]raethyl}-2,5-dimethyIphenyI- carbamoyl)ethyl]piperidin-4-yl Ester
To a 2 L three-necked round-bottom flask was added biphenyl-2-ylcarbamic acid 1- [2-(4-formyI-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester hydrochloride (38 g, 70 mmol) and N-{5-[(i?)-2-amino-l-(rer/-butyldimethylsilanyloxy)ethyl]-2- hydroxyphenyl}formamide acetic acid salt (33.6 g, 91 mmol). Dichloromethane (500 mL) and methanol (500 mL) were added and the resulting mixture was stirred at room temperature under dry nitrogen for about 3 hours. The reaction mixture was then cooled to 0 0C to 5 0C and solid sodium triacetoxyborohydride (44.5 g, 381 mmol) was added in portions over a 10 minute period. The reaction mixture was slowly warmed from 0 °C to room temperature over a period of about 2 hours and then cooled to 0 0C. Saturated aqueous sodium bicarbonate (500 mL) and dichloromethane (500 mL) were added. This mixture was stirred thoroughly and then the layers were separated. The organic layer was washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude biphenyl-2-ylcarbamic acid l-[2-(4- {[(i?)-2-(rert-butyldimethylsilanyloxy)-2-(3-formylamino-4- hydroxyphenyl)ethylamino]methyl}-2,5-dimethylphenylcarbamoyl)-ethyl]piperidin-4-yl ester (55 g, 86% purity) as a yellow solid. The crude product (30 g) was dissolved in dichloromethaπe containing 2% methanol (150 mL total) and loaded onto a silica gel column (300 g) that had been packed and equilibrated with dichloromethane containing 2% methanol and 0.5% ammonium hydroxide. The product was eluted from the column using dichloromethane containing 2% methanol and 0.5% ammonium hydroxide (1 L); dichloromethane containing 4% methanol and 0.5% ammonium hydroxide (1 L) and dichloromethane containing 5% methanol and 0.5% ammonium hydroxide (about 3 L). Fractions (200 mL) were collected and those fractions having a purity greater than 90% were combined and concentrated under reduced pressure to provide biphenyl-2-ylcarbamic acid \-[2-(4-{[(R)-2-(tert- butyldimethylsilanyloxy)-2-(3-formylamino-4-hydroxyphenyl)ethylamino]methyl} -2,5- dimethylphenylcarbamoyl)-ethyl]piperidin-4-yl ester (21.6 g, 96.5% purity) as a yellowish solid. MS [M + H+] found 794.6.
Step G - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(if)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylaminolmethyl}-2,5-dimethylphenyl- carbamoyI)ethyl]piperidin-4-yl Ester Hydrofluoride Salt
To a 1 L round-bottom flask was added biphenyl-2-ylcarbamic acid l-[2-(4-{[(iϋ)- 2-(/er^-butyldimethylsilanyloxy)-2-(3-formylamino-4-hydroxyphenyl)ethylamino]methyl}- 2,5-dimethylphenylcarbamoyI)ethyl]piperidin-4-yl ester (21.5 g, 27.1 mmol) and dichloromethane (200 mL). The resulting mixture was stirred at room temperature until the ingredients dissolved and then triethylamine trihydrofluoride (8.85 mL, 54.2 mmol) was added and the resulting mixture was stirred at 25 0C for about 48 hours. The solvent was removed on a rotary evaporator to provide a thick paste. Dichloromethane (100 mL) and ethyl acetate (200 mL) were added to the paste and the resulting mixture was stirred for 30 minutes. The resulting slurry was slowly filtered under dry nitrogen and the filter cake was washed with a 1 :2 mixture of dichloromethane and ethyl acetate (100 mL total), dried under nitrogen for 2 hours and then dried under vacuum overnight to provide biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(/?)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl}-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester as a hydrofluoride salt (25 g, 96.9% purity) which was a hard clay-like solid. MS [M + H+] . found 680.8. Step H ~ BiphenyI-2-ylcarbamic Acid l-[2-(4-{[(iϊ)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5-dimelhyIphenylcarbamoyl)- ethyl]piperidin-4-yl Ester
Biphenyl-2-ylcarbamic acid 1 -[2-(4- {[(Λ)-2-(3-formylamino-4-hydroxyphenyl)-2- hydroxyethylamino]methyl} -2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester hydrofluoride salt (25 g) was purified on a 6-inch reverse-phase column (Microsorb solid phase) in three equal batches using a 10 % to 50 % mixture of acetonitrile in water containing 1% trifluoroacetic acid as the mobile phase. Fractions with greater than 99% purity were combined and then diluted with one volume of water. The resulting mixture was cooled to 00C and solid sodium bicarbonate was added until the pH of the mixture was about 7.5 to 8.0. Within about 5 minutes, a white slurry developed. The slurry was stirred for 30 minutes and then filtered. The filter cake was washed with water (500 mL), air dried for about 4 hours and then dried in vacuum overnight to provide biphenyl-2- ylcarbamic acid l-[2-(4-{[(Λ)-2-(3-foπnylamino-4-hydroxyphenyl)-2- hydroxyethylarnino]methyl}-2,5-dimethylphenyl-carbamoyl)ethyl]piperidin-4-yl ester (12 g,, 99+% purity), as a semi-crystalline free base.
Example 6
Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(/?)-2-(3-Formylamino-4-hydroxyphenyl)-2- hydroxyethylammo]methyl}-2,5-dimethyIphenylcarbamoyl)ethyl]piperidin-4-yl Ester Step A - Biphenyl-2-ylcarbamic Acid l~[2-(4-[l,3]DioxoIan-2-yl-2,5- dimethylphenyIcarbamoyl)ethyl]piperidin-4-yl Ester
To a 500 mL round-bottom flask was added biphenyl-2-ylcarbamic acid piperidin- 4-yl ester (17.0 g, 58 mmol) andiV-(4-[l,3]dioxolan-2-yl-2,5-dimethylphenyl)acrylamide (13.1 g, 52.9 mmol).' Ethanol (150 mL) and dicloromethane (150 mL) were added to form a slurry. The reaction mixture was heated at 50 0C to 55 0C for about 24 hours and then cooled to room temperature. Most of the solvent was removed on a rotary evaporator, resulting in a thick slurry. Ethanol (reagent grade) was added to form a total volume of about 200 mL and the resulting mixture was heated to 80 0C and then cooled slowly to room temperature. The resulting thick white slurry was filtered, washed with ethanol (20 mL) and dried in vacuum to provide biphenyl-2-ylcarbamic acid l-[2-(4-[l,3]dioxolan-2- yl-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (23.8 g, about 98% purity) as a white solid. Step B - Biphenyl-2-ylcarbamic Acid l-[2-(4-Formyl-2,5-dimethyIphenyl- carbamoyl)ethyl]piperidin-4-yl Ester
To a 500 mL round-bottom flask was added biphenyl-2-ylcarbamic acid l-[2-(4- [l,3]dioxolan-2-yl-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (15 g, 27.6 mmol) and acetonitrile (150 mL) to form a slurry. Aqueous 2 M hydrochloric acid (75 mL) was added and the resulting mixture was stirred at 30 0C for 1 hour. The mixture was then cooled to room temperature and ethyl acetate (150 mL) was added. Aqueous 2 M sodium hydroxide (75 mL) was added, the pH was checked and then additional 2 M sodium hydroxide was added until the pH of the solution was in the range of 9 to 10. The layers were separated and the organic layer was washed with diluted brine (75 mL; 1:1 brine/water), dried over anhydrous sodium sulfate and the solvent removed on a rotary evaporator to give biphenyl-2-ylcarbamic acid l-[2-(4-formyl-2,5-dimethylphenyl- carbamoyl)ethyl]piperidin-4-yl ester (12.5 g, about 98% purity). If desired, the purity of this intermediate can be increased by forming a slurry with ethanol (3 volumes of ethanol), heating the slurry to 80 0C, and then cooling slowly to room temperature and isolating by filtration.
Step C - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(i?)-2-(te/t-ButyldimethylsilanyIoxy)-2- (3-formyIamino-4-hydroxyphenyl)ethylimino]methyI}-2,5-dimethyl- phenylcarbamoyl)ethyl]piperidin-4-yl Ester
To a 250 mL round-bottom flask was added biphenyl-2-ylcarbamic acid l-[2-(4- formyl-235-dimethylphenylcarbamoyl)ethyl]piperidin-4-yi ester (7.1 g, 14.2 mmol) and JV- {5-[(JR)-2-amino-l-(/ert-butyldimethylsilanyloxy)ethyl]-2-hydroxyphenyl}formamide acetic acid salt (5.8 g. 15.6 mmol). Methanol (100 mL) was added to form a slurry and this mixture was stirred at 45 0C to 50 0C under nitrogen for 1 hour. The mixture was then cooled to room temperature and toluene (50 mL) was added and the solvent was removed on a rotary evaporator at temperature ranging from 35 0C to 45 0C. Toluene (50 mL) was added to the residue and the solvent was removed to provide biphenyl-2-ylcarbamic acid 1- [2-(4- { [(i?)-2-(tert-butyldimethylsilanyloxy)-2-(3-formylamino-4- hydroxyphenyl)ethylimino]methyl}-2,5-dimethylρhenylcarbamoyl)ethyl]piperidin-4-yl ester (12 g) as a yellow-orange solid. Step D -Biphenyl-2-yIcarbamic Acid l-[2-(4-{[(i?)-2-(ϊert-ButyldimethylsiIanyloxy)-2- (3-formylamino-4-hydroxyphenyl)ethyIamino]methyl}-2,5-dimethylphenyl- carbamoyl)ethyI]piperidin-4-yI Ester
To a hydrogenation flask was added biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2- (?er?-butyldiraethylsilanyloxy)-2-(3-foiτnylamino-4-hydroxyphenyl)ethylimino]niethyl}- 2,5-dimethylphenylcarbamoyl)ethyl]piρeridin-4-yl ester (4.6 g) and 2- methyltetrahydrofuran (50 mL). The resulting mixture was stirred until the solid dissolved (about 5 min.) and then the mixture was purged with nitrogen. Platinum on carbon (920 mg, 5 wt. %, support activated carbon) was added and the mixture was hydrogenated at 50 psi (Parr shaker) for 6 hours. The mixture was then filtered through Celite and the Celite was washed with 2-methyltetrahyrofuran (10 mL). To the filtrate was added a thiopropyl- modified silica gel (20% of weight of solution, Silicycle) and this mixture was stirred at 25 0C to 30 0C for 3 hours. The mixture was then filtered through Celite and concentrated to remove the solvent. The residue was dissolved in methanol (5 mL per gram of residue) and then the resulting solution was added slowly to a vigorously stirred 1 :1 mixture of saturated aqueous sodium bicarbonate and water (40 mL per gram of residue). The resulting off-white slurry was stirred for 20 minutes and then filtered. The filter cake was washed with water (20 volumes), air dried for 3 hours and then dried in vacuum at room temperature overnight to provide biphenyl-2-ylcarbamic acid l-[2-(4-{[(R)-2-(tert- butyldimethylsilanyloxy)-2-(3-formylamino-4-hydroxyphenyl)ethylamino]methyl}-2,5- dimethylρhenylcarbamoyl)ethyl]piperidin-4-yl ester (80% recovery, about 96% purity).
Step E - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[CR)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethyIamino]methyI}-2,5-dimetbyIphenyI- carbamoyl)ethyl]piperidin-4-yI Ester L-Tartrate Salt
To a 200 mL round-bottom flask was added biphenyl-2-ylcarbamic acid l-[2-(4- {[(i?)-2-(/er?-butyldimethylsilanyloxy)-2-(3-formylamino-4-hydroxyphenyl)ethylamino]- methyl} -2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (3.8 g, 4.8 mmol) and 2- methyltetrahydrofuran (40 mL). The resulting mixture was stirred at room temperature until the ingredients dissolved (about 15 min.) and then triethylamine trihydrofluoride (0.94 mL, 5.76 mmol) was added and the resulting mixture was stirred at 25 0C for about 24 hours. To this mixture was added a 1 :1 mixture of saturated aqueous sodium bicarbonate and water (40 mL) and 2-methyltetrahydrofuran and the resulting mixture was stirred until the solid dissolved (solution pH about 8). The layers were separated and the organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated on a rotary evaporator. The residue was dissolved in 2- methyltetrahydrofuran (50 mL) and solid L-tartaric acid (650 mg) was added. The resulting mixture was stirred at 25 0C to 30 0C for 18 hours and then filtered through filter paper. The filter cake was washed with 2-methyltetxahydrofuran (10 mL), isopropanol (10 mL) and immediately put under vacuum to provide biphenyl-2-ylcarbamic acid l-[2-(4- {[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenyl-carbamoyl)ethyl]piperidin-4-yl ester L-tartaric acid salt (3.7 g, >97 % purity).
Step F - Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(Λ)-2-(3-Formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5-dimethyIphenylcarbamoyI)- ethyl]piperidin-4-yI Ester To a 250 mL round-bottom flask was added biphenyl-2-ylcarbamic acid l-[2-(4-
{[(i?)-2-(3-foππylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester L-tartaric acid salt (3.5 g) and methanol (35 mL) and the resulting mixture was stirred for 15 min. A 1:1 mixture of saturated aqueous sodium bicarbonate and water (70 mL) was added over a 5 min. period and stirring was continued for 2 hours. The resulting off-white slurry was filtered and the filter cake was washed with water (20 mL), air dried for 2 hours and then dried in a vacuum overnight to provide biphenyl-2-ylcarbamic acid l-[2-(4-{[(i.)-2-(3-formylamino- 4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5-dimethylphenylcarbamoyl)- ethyl]piperidin-4-yl ester'(2.3 g) as a semi-crystalline free base.
1H and 13C NMR spectra were obtained for a sample of biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-255- dimethylphenylcarbamoyl)-ethyl]piperidin-4-yl ester (22.2 mg in about 0.75 mL of DMSO-cfe) at ambient temperature using a JEOL ECX-400 NMR spectrometer: 1H NMR (400 MHz, DMSO-^6), major isomer, δ 9.64 (br, IH), 9.54 (br s, IH),
9.43 (s, IH), 8.67 (s, IH), 8.26 (s, IH), 8.03 (d, J = 1.9, IH), 7.25-7.45 (m, 9H), -7.3 (nd, IH), 7.07 (s, IH), 6.88 (dd, J = 8.2, 1.9, IH), 6.79 (d, J = 8.2, IH), 5.15 (br, IH), 4.53 (dd, J = 7.3, 4.7, IH), 4.47 (m, IH), -3.65 and -3.60 (AB pair, 2H), 2.68 (br m, 2H), -2.59 (nd, 4H), 2.44 (br t, J = 6.5, 2H), 2.20 (s, 3H), -2.17 (br m, 2H), 2.14 (s, 3H), 1.73 (br, 2H), 1.44 (br q, J = -9.0, 2H).
1H NMR (400 MHz, DMSCW6), minor isomer, δ 9.64 (br, IH), 9.43 (s IH), 9.26 (br d, J = -7.0, IH), 8.67 (s, IH), 8.50 (br d, J = -7.0, IH), 7.25-7 '.45 (m, 9H), -7.3 (nd, IH), 7.07 (s, IH), -7.07 (nd, IH), 6.95 (dd, J = 8.3, 1.8, IH), 6.83 (d, J =8.3, IH), 5.15 (br, IH), 4.47 (m, IH), -3.65 and -3.60 (AB pair, 2H), 2.68 (br m, 2H), -2.59 (nd, 2H), 2.44 (br t, J = 6.5, 2H), 2.20 (s, 3H), -2.17 (br m, 2H), 2.14 (s, 3H), 1.73 (br, 2H), 1.44 (br q, J = -9.0, 2H).
13C NMR (100 MHz, DMSO-d6), major isomer, δ 170.0, 159.9, 153.9, 145.5, 139.3, 137.6, 135.2, 135.0, 134.8, 133.4, 133.4, 130.2, 130.2, 128.6, 128.2, 127.8, 127.4,
127.2, 127.0, 126.1, 125.7, 125.6, 121.7, 118.6, 114.5, 71.4, 70.0, 57.4, 53.9, 50.3, 50.1, 33.7, 30.7, 18.2, 17.5.
13C NMR (100 MHz, DMSCW6), minor isomer, δ 170.0, 163.4, 153.9, 147.8,
139.3, 137,6, 135.7, 135.2, 135.0, 134.8, 133.4, 133.4, 130.2, 130.2, 128.6, 128.2, 127.8, 127.4, 127.2, 127.0, 126.1, 125.7, 123.0, 119.6, 115.6, 71.0, 70.0, 57.3, 53.9, 50.3, 50.1,
33.7, 30.7, 18.2, 17.5.
The 1H and 13C NMR spectra showed the presence of a major isomer (about 82 mole percent) and a minor isomer (about 18 mole percent) believed to be rotational isomers resulting from hindered rotation about the -NH-C(O)H bond. The phenyl group is believed to be syn to the carbonyl oxygen in the major isomer and anti in the minor isomer.
Example 7
Seed Crystals of Form II of Biphenyl-2-ylcarbamic Acid l-[2-(4-{[(2?)-2-(3- Formylamino-4-hydroxyphenyI)-2-hydroxyethylamino]methyl}-2,5- dimethyIphenylcarbamoyl)ethyl]piperidin-4-yI Ester
Semi-crystalline biphenyl-2-ylcarbamic acid l-[2-(4-{[(i2)-2-(3-formylamino-4- hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5-dimethylphenyl- carbamoyl)ethyl]piperidin-4-yl ester (500 mg) was dissolved in methanol (50 mL) and water was added until the cloud point was reached. The resulting mixture was stirred at 25 0C for 3 hours and the resulting crystalline material was isolated by filtration to provide crystalline biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3-formylamino-4-hydroxyphenyl)- 2-hydroxyethylamino]methyl}-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (420 mg). This crystalline free base was determined to have a differential scanning calorimetry (DSC) trace that exhibit a peak in endothermic heat flow at about 142 CC to about 150 0C; and a powder x-ray diffraction (PXRD) pattern having significant diffraction peaks, among other peaks, at 2Θ values of about 20.7±0.3, 21.6±0.3, 22.5±0.3 and 23.2±0.3. This crystalline free base form is designated as Form II. Additional information on Form II and other crystalline free base forms of this compound is disclosed in commonly-assigned U.S. Application No. , filed on even date herewith (Attorney
Docket No. P-222-US 1) and U.S. Provisional Application No. 60/794,709, filed April 25, 2006; the disclosures of which are incorporated herein by reference in their entirety.
Example 8
Crystallization of Form II of Biphenyl-2-ylcarbamic acid l-[2-(4-{[(Λ)-2-(3- FormyIamino-4-hydroxyphenyl)-2-hydroxyethyIamino]methyl}-2,5- dimethyIphenylcarbamoyI)ethyI]piperidin-4-yI Ester
To a 3 L three-necked round-bottomed flask equipped with an over-head stirrer, temperature control and addition funnel was added semi-crystalline biphenyl-2-ylcarbamic acid l-[2-(4-{[(i2)-2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}- 2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (14 g) and methanol (1.4 L). Water (500 mL) was added in one portion and then additional water (200 mL) was added slowly until the cloud point was reached. Seed crystals of biphenyl-2-ylcarbamic acid 1- [2-(4-{[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenyl-carbamoyl)ethyl]piperidin-4-yl ester Form II (50 mg) were added and the resulting mixture was stirred at 25 0C for 3 hours at which time a free-flowing slurry had developed. Water (300 mL) was added over a 15-minute period and the resulting mixture was stirred at 25 0C overnight. The mixture was then filtered and the filter cake was washed with water (100 mL), air dried for about 2 hours and then dried under vacuum at room temperature for 48 hours to provide crystalline biphenyl-2-ylcarbamic acid l-[2-(4- {[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2)5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (12.5 g, 99.6% purity). This crystalline salt was determined to be Form II. Example 9
Biphenyl-2-ylcarbamic Acid l-[2-(4-Formyl-2,5-dimethyIphenyIcarbamoyl)ethyll- piperidin-4-yl Ester
Step A - 4-Iodo-2,5-diraethylphenylamine To a solution of 2,5-dimethylaniline (2Og, 165 ramol) in a 1:1 mixture of dichloromethane and methanol (400 mL) was added sodium bicarbonate (20.8g, 250 mmol) and tetramethylammonium dichloroiodate(I) (44.7g, 165 mmol). The resulting mixture was stirred at room temperature for 1 hour and then water was added (500 mL). The organic layer was removed and washed with 5% aqueous sodium thiosulfate (500 mL) and brine (500 mL). The organic layer was then dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide 4-iodo-2,5-dimethylphenylamine (39.6 g, 98% yield). The product was used without further purification.
Step B -iV-(4-Iodo-2,5-dimethylphenyl)acrylamide To a solution of 4-iodo-2,5-dimethylphenylamine (37.2 g, 151 mmol) in dichloromethane (500 mL) was added sodium bicarbonate (25.4 g, 302 mmol). The resulting mixture was cooled to 00C and acryolyl chloride (12.3 mL, 151 mmol) was added slowly over a period of 25 minutes. The resulting mixture was stirred at room temperature overnight and then filtered. The volume of the filtrate was reduced to about 100 mL and a precipitate formed. The precipitate was filtered, dried, washed with water (1 L) and then dried again to afford Λ^-(4-iodo-2,5-dimethylphenyl)acrylam.ide (42.98g, 95% purity, 90 % yield). The product was used without further purification.
Step C - BiphenyI-2-ylcarbaπuc Acid l-[2-(4-Iodo-2,5-dimethyIpheuyIcarbamoyl)- ethyl]piperidin-4-yl Ester
To a solution of Λr-(4-iodo-2,5-dimethylphenyl)acrylamide (32.2 g, 107 mmol) in a 6:1 v/v mixture of N^-dimethylformarnide and isopropanol (700 mL) was added biphenyl- 2-ylcarbamic acid piperidin-4-yl ester (36.3 g, 123 mmol). The resulting mixture was heated at 500C for 24 hours and then at 80 0C for 24 hours. The reaction mixture was then cooled to room temperature and concentrated under vacuum. The residue was dissolved in dichloromethane (1 L) and this solution was washed with IN aqueous hydrochloric acid (500 mL), water (500 mL), brine (500 mL) and saturated aqueous sodium bicarbonate solution (500 mL). The organic layer was then dried over anhydrous magnesium sulfate and filtered. Ethanol (400 mL) was added and the resulting mixture was concentrated under vacuum to a volume of about 400 mL, at which time a precipitate had formed. The precipitate was filtered and dried to afford biphenyl-2-ylcarbamic acid l-[2-(4-iodo-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (59.6g, 84% purity, 79% yield), m/z: [IvH- H+] calcd for C29H32DST3O3 598.49; found 598.5.
Step D - 4-{3-[4-(BiphenyI-2-ylcarbamoyloxy)piperidiπ-l-yI]propionylamino}-2,5- dimethylbenzoic Acid Methyl Ester
To a solution of biphenyl-2-ylcarbamic acid l-[2-(4-iodo-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (56 g, 94 mmol) in a 5:1 v/v mixture of N,N-dimethylformarnide and methanol (600 mL) were added diispropylethylamine (49 rnL, 281 mmol), l,3-bis(diphenylphosphino)propane (3.9 g, 9.4 mmol) and palladium(II) acetate (2.1 g, 9.4 mmol). The resulting mixture was purged with carbon monoxide and then stirred overnight at 70 0C to 80 0C under a carbon monoxide atmosphere (balloon pressure). The reaction mixture was concentrated under vacuum and the residue was dissolved in dichloromethane (500 mL). This mixture was washed with IN aqueous hydrochloric acid (500 mL), water (500 mL) and then brine (500 mL). The organic layer was then dried over anhydrous magnesium sulfate, filtered and then concentrated under vacuum. The residue was mixed with ethanol (about 5:1 v/w ethanol to residue) and the mixture was heated until all solid material dissolved. This solution was allowed to slowly cool to room temperature and the resulting precipitate was isolated by filtration to afford 4- {3 -[4-(biphenyl-2-ylcarbamoyloxy)piperidin- 1 -yl]propionylamino } -2,5-dimethylbenzoic acid methyl ester (47.3 g, 97% purity, 92% yield), m/z: [M + H+] calcd for C31H35N3O5 530.63; found 530.4.
Step E - Biphenyl-2-ylcarbamic Acid l-[2-(4-Hydroxymethyl-2,5-dimethyl- phenyIcarbamoyl)ethyl]piperidin-4-yl Ester
To a solution of 4-{3-[4-(biphenyl-2-ylcarbamoyloxy)piperidin-l- yl]propionylamino}-2,5-dimethylbenzoic acid methyl ester (49.8 g, 93.9 mmol) in tetrahydrofuran (200 mL) was cooled to 0 0C and lithium aluminum hydride (10.7 g, 281.7 mmol) added portion-wise (10 x 1.07 g). The resulting mixture was stirred for 3 hours and then water (10.7 mL) was added, followed by IN aqueous sodium hydroxide (10.7 mL) and additional water (32.1 mL). This mixture was stirred overnight and then filtered. The organic layer was concentrated under vacuum and the residue was mixed with ethyl acetate (about 5:1 v/w ethyl acetate to residue). This mixture was heated until all the solid material dissolved and then the solution was allowed to cool to room temperature. The resulting precipitate was filtered and dried to afford biphenyl-2-ylcarbamic acid l-[2-(4- hydroxymethyl-2,5-dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (24.6 g, 95% purity, 47.5% yield). This material was used without further purification, m/z: [M + H+] calcd for C30H35N3O4 502.62; found 502.5.
Step F - Biphenyl-2-ylcarbamic Acid l-[2-(4-Formyl-2,5- dimethyIphenyIcarbamoyl)ethyl]piperidin-4-yl Ester
To a solution of biphenyl-2-ylcarbamic acid l-[2-(4-hydroxymethyl-2,5- dimethylphenylcarbamoyl)ethyl]ρiperidin-4-yl ester (5.0 g, 10 mmol) in dichloromethane (200 mL) was added diisopropylethylamine (8.7 mL, 50 mmol) and dimethylsulfoxide (5.6 mL, 100 mmol). The resulting mixture was cooled to 0 0C and sulfur trioxide pyridine complex (8.0 g, 50 mmol) was added. The reaction mixture was stirred for 1 hour at 0 0C and then water (300 mL) was added. The organic layer was removed and washed with IN aqueous hydrochloric acid (300 mL) and brine (300 mL). The organic layer was then dried over anhydrous magnesium sulfate and filtered. The resulting solution containing biphenyl-2-ylcarbamic acid 1 -[2-(4-formyl-2,5-dimethylphenylcarbamoyl)ethyl]piperidin- 4-yl ester was used without further purification, m/z: [M + H+] calcd for C3QH33N3O4 500.60; found 500.4.
Example 10
Cell Culture and Membrane Preparation From Cells Expressing Human Mi, M25 M3 and M4 Muscarinic Receptors
CHO cell lines stably expressing cloned human hMi, IiM2, bM3 and I1M4 muscarinic receptor subtypes, respectively, were grown to near confluency in Hams F-12 media supplemented with 10% FBS and 250 μg/mL Geneticin. The cells were grown in a 5% CO2, 37 0C 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 EC 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 (Kinematica AG; 20 seconds x 2 bursts). Crude membranes were centrifuged at 40,000 x g for 15 minutes at 4 0C. The membrane pellet was then resuspended with re- suspension buffer and homogenized again with the Polytron tissue disrupter. The protein concentration of the membrane suspension was determined by the method described in Lowry et al<5 1951, Journal of Biochemistry, 193, 265. All membranes were stored frozen in aliquots at —800C or used immediately. Aliquots of prepared I1M5 receptor membranes were purchased directly from PerkinElmer, Inc. (Wellesley, MA) and stored at -80 0C until use.
Example 11 Radioligand Binding Assay for Muscarinic Receptors
Radioligand binding assays for cloned 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, I1M2, hM3, hJVLj or I1M5 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 TaM4, 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 K^ values of the radioligand were performed using L-|7V-methyl- 3H] scopolamine methyl chloride ([3H]-NMS) (TRK666, 84.0 Ci/mmol, Amersham Pharmacia Biotech, Buckinghamshire, England) at concentrations ranging from 0.001 nM to 20 nM. Displacement assays for determination of K,- values of test compounds were performed with [3H]-NMS at 1 nM and eleven different test compound concentrations. The test compounds were initially dissolved to a concentration of 400 μM in dilution buffer and then serially diluted 5x with dilution buffer to final concentrations ranging from 10 pM to 100 μM. The addition order and volumes to the assay plates were as follows: 25 μL radioligand, 25 μL diluted test compound, and 50 μL membranes. Assay plates were incubated for 60 minutes at 37 0C. Binding reactions were terminated by rapid filtration over GF/B glass fiber filter plates (PerkinElmer, Inc.) 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.) were added to each well. The plates were then counted in a PerkinElmer Topcount liquid scintillation counter (PerkinElmer, Inc.). 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 Ko value of the radioligand using the Cheng-Prusoff equation (Cheng Y; Prusoff WH. (1973) Biochemical Pharmacology, 22(23):3099-l 08). Kj values were converted to pKi values to determine the geometric mean and 95% confidence intervals. These summary statistics were then converted back to Ki values for data reporting.
In this assay, a lower K,- value indicates that the test compound has a higher binding affinity for the receptor tested. Biphenyl-2-yicarbamic acid l-[2-(4-{[(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl} -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ha) was found to have a Kj value of less than 10 nM for the Mi, M2, M3, M4 and M5 muscarinic receptor subtypes.
Example 12 Cell Culture and Membrane Preparation From Cells Expressing Human βi, P2 or βa Adrenergic Receptors
Human embryonic kidney (HEK-293) cell lines stably expressing cloned human βi and β2 adrenergic receptors or Chinese hamster ovarian (CHO) cell lines stably expressing cloned human β3 adrenergic receptors were grown to near confiuency in DMEM or Hams F- 12 media with 10% FBS in the presence of 500 μg/mL Geneticin. The cell monolayer was lifted with 2 mM 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 βi and β2 receptor expressing membranes, cell pellets were re-suspended in lysis buffer (10 mM HEPES/HC1, 10 mM EDTA, pH 7.4 at 4 0C) 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 (10 mM Tris/HCl, pH 7.4) supplemented with one tablet of "Complete Protease Inhibitor Cocktail Tablets with 2 mM EDTA" per 50 mL buffer (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, 12.5 mM MgCl2, 1 mM EDTA). The protein concentration of the 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 0C or used immediately.
Example 13
Radioligand Binding Assay for Human βj, β2 and β3 Adrenergic Receptors
Binding assays were performed in 96-well microliter plates in a total assay volume of 100 μL with 10-15 μg of membrane protein containing the human βi, β2 or β3 adrenergic receptors in assay buffer (75 mM Tris/HCl pH 7.4 at 25 0C, 12.5 mM MgCl2, 1 mM EDTA, 0.2% BSA). Saturation binding studies for determining K^ values of the radioligand were done using [3H]-dihydroalprenolol (NET-720, 100 Ci/mmol, PerkinElmer Life Sciences Inc., Boston, MA) for the βt 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 K1- 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 0C, and then binding reactions were terminated by rapid filtration over GF/B for the βi 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 0C, 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 Kd value of the 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. Biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3- formylarnino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ila) was found to have a Kj value of less than 10 nM for the β2 adrenergic receptor and Ki values greater than 1000 nM for the βi and β3 adrenergic receptors.
Example 14
Functional Assays of Antagonism for Muscarinic Receptor Subtypes Assay A - Blockade of Agonist-Mediated Inhibition of cAMP Accumulation
In this assay, the functional potency of a test compound as an antagonist for the hM2 receptor was determined by measuring the ability of the test compound to block oxotremorine-inhibition of forskolin-mediated cAMP accumulation in CHO-Kl cells expressing the hM2 receptor. cAMP assays were performed in a radioimmunoassay format using the Flashplate Adenylyl Cyclase Activation Assay System with l25I-cAMP (NEN SMP004B, PerkinElmer Life Sciences Inc., Boston, MA), according to the manufacturer's instructions. Cells were 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 were washed twice by centrifugation at 650 x g for five minutes in 50 mL dPBS. The cell pellet was then re-suspended in 10 mL dPBS, and the cells were counted with a Coulter Zl Dual Particle Counter (Beckman Coulter, Fullerton, CA). The cells were centrifuged again at 650 x g for five minutes and re-suspended in stimulation buffer to an assay concentration of 1.6 x 106 to 2.8 x 106 cells/mL. The test compound was 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 was diluted in a similar manner.
To measure oxotremorine inhibition of adenylyl cyclase activity, 25 μL forskolin (25 μM final concentration diluted in dPBS), 25 μL diluted oxotremorine, and 50 μL cells were added to agonist assay wells. To measure the ability of a test compound to block oxotremorine-inhibited adenylyl cyclase 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 were added to remaining assay wells.
Reactions were incubated for 10 minutes at 37 0C and stopped by addition of 100 μL ice-cold detection buffer. Plates were 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) was calculated based on the counts observed for the samples and cAMP standards, as described in the manufacturer's user manual. Data was 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 K^s, using the EC50 of the oxotremorine concentration- response curve and the oxotremorine assay concentration as the KD and [L], respectively.
In this assay, a lower Kobs value indicates that the test compound has a higher functional activity at the receptor tested. Biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ila) was found to have a Kobs value of less than about 10 nM for blockade of oxotremorine-inhibition of forskolin- mediated cAMP accumulation in CHO-Kl cells expressing the hM2 receptor.
Assay B - Blockade of Agonist-Mediated [35S]GTPyS Binding
In this functional assay, the functional potency of a test compound as an antagonist of the hM2 receptor was determined by measuring the ability of the test compound to block oxotremorine-stimulated [35S]GTPyS 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 to 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 EC90 value (effective concentration for 90% maximal response) for stimulation of [35S]GTPyS binding by the agonist oxotremorine was determined in each experiment.
To determine the ability of a test compound to inhibit oxotremorine-stimulated [35S]GTPyS binding, the following was added to each well of 96 well plates: 25 μL of assay buffer with [35S]GTPyS (0.4nM), 25 μL of oxotremorine(EC9o) 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 0C 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 Kobs. using the IC50 values of the concentration-response curve for the test compound and the oxotremorine concentration in the assay as the KD and [L], ligand concentration, respectively.
In this assay, a lower Kobs value indicates that the test compound has a higher functional activity at the receptor tested. Biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl} -2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound Ha) was found to have a Kobs value of less than about 10 nM for blockade of oxotremorine-stimulated [35S]GTPyS binding in CHO-Kl cells expressing the I1M2 receptor.
Assay C - Blockade of Agonist-Mediated Calcium Release via FLIPR Assays
In this functional assay, the functional potency of a test compound as an antagonist of ItM1, 11M3 and cMs receptors was determined by measuring the ability of the test compound to inhibit agonist-mediated increases in intracellular calcium.
CHO cells stably expressing the receptors were seeded into 96-well FLIPR plates the night before the assay was done. Seeded cells were washed twice 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) using Cellwash (MTX Labsystems, Inc.) to remove growth media. After washing, each well contained 50 μL of FLIPR buffer. The cells were then incubated with 50 μL/well of 4 μM FLUO-4AM (a 2X solution was made) for 40 minutes at 37 0C, 5% carbon dioxide. Following the dye incubation period, cells were washed two times with FLIPR buffer, leaving a final volume of 50 μL in each well. The dose-dependent stimulation of intracellular Ca2+ release for oxotremorine was determined so that the test compound could be measured against oxotremorine stimulation at an EC90 concentration. Cells were first incubated with compound dilution buffer for 20 minutes and then oxotremorine was added. An ECg0 value for oxotremorine was generated according to the method detailed in the FLEPR measurement and data reduction section below, in conjunction with the formula ECF = ((F/100-F)Al/H) * EC50 • An oxotremorine concentration of 3 x ECF was prepared in stimulation plates such that an EC90 concentration of oxotremorine was added to each well in test assay plates.
The parameters used for the FLIPR were: exposure length of 0.4 seconds, laser strength of 0.5 watts, excitation wavelength of 488 nm, and emission wavelength of
550 nm. Baseline was determined by measuring the change in fluorescence for 10 seconds prior to addition of oxotremorine. Following oxotremorine 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 was expressed as maximum fluorescence minus baseline fluorescence for each well. The raw data was analyzed against the logarithm of test compound concentration by nonlinear regression with GraphPad Prism (GraphPad Software, Inc., San Diego, CA) using the built-in model for sigmoidal dose-response. Antagonist KobS values were determined by Prism using the oxotremorine EC50 value as the Ko and the oxotremorine EC90 for the ligand concentration according to the Cheng-Prusoff equation (Cheng & Prusoff, 1973).
In this assay, a lower Kobs value indicates that the test compound has a higher functional activity at the receptor tested. Biphenyl-2-ylcarbamic acid l-[2-(4-{[(i.)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (compound EIa) was found to have a Kobs value of less than about 10 nM for blockade of agonist-mediated calcium release in CHO cells stably expressing the hMi, I1M3 and cMs receptors. Example 15
Whole-cell cAMP Flashplate Assay in HEK-293 and 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 βi and β2 receptor agonist potency (EC50), HEK-293 cell lines stably expressing cloned human P1 and β2 receptors were grown to near confluency in DMEM supplemented with 10% FBS and Geneticin (500 μg/mL). For the determination β3 receptor agonist potency (EC50), CHO-Kl cell line stably expressing cloned human or β3 adrenergic receptors was grown to near confluency in Hams 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% trypsin/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 IBMX (PerkinElmer Kit) pre-warmed to room temperature to a concentration of 1.6 x 106 to 2.8 x 106 cells/mL. About 40,000 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 0C 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. Biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5-dimethylphenyl- carbamoyl)ethyl]piperidin-4-yl ester (compound Ila) was found to have an EC50 value less than about 10 nM for the β2 adrenergic receptor; an EC50 value of about 30 nM for the P1 adrenergic receptor; and an EC50 value greater than 700 nM for the β3 adrenergic receptor.
Example 16 Whole-cell cAMP Flashplate Assay With a Lung Epithelial Cell Line Endogenously Expressing Human βz Adrenergic Receptor
In this assay, the agonist potency and intrinsic activity of a test compound were determined using a cell line expressing endogenous levels of the β2 adrenergic receptor. Cells from a human lung epithelial cell line (BEAS-2B) (ATCC CRL-9609, American Type Culture Collection, Manassas, VA) (January B, et al., British Journal of
Pharmacology, 1998, 123, 4, 701-11) were grown to 75-90% confluency in complete, serum-free medium (LHC-9 medium containing epinephrine and retinoic acid, Biosource International, Camarillo, CA). The day before the assay, the medium was switched to LHC-8 (no epinephrine or retinoic acid, Biosource International, Camarillo, CA). 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.
On the day of the 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 0C, 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 0C and stopped by addition of 100 μL of iceLcold detection buffer. Plates were sealed, incubated over night at 4 0C 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. Biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3- formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5-dimethylphenyl- carbamoyl)ethyl]piρeridin-4-yl ester (compound Ila) was found to have an ECs0 value of less than 10 nM with intrinsic activity value of greater than 0.3 compared with a full β2 agonist isoproterenol (1.0),
Example 17
Einthoven Assay for Determining Bronchoprotective Efficacy and Duration In this assay, the bronchoprotective efficacy and duration of test compounds were determined using guinea pigs. This assay was derived from the procedures described in Einthoven (1892) Pfugers Arch. 51: 367 - 445; and Mohammed et al. (2000) PuIm Pharmacol Ther. 13 (6)..-287 '-92. In this assay, changes in ventilation pressure serve as a surrogate measure of airway resistance. Following pre-treatment with a test compound, muscarinic antagonist potency was determined using bronchoconstrictor dose-response curves to intravenous methacholine in the presence of propranolol. Similarly, β2 agonist bronchoprotective potency was determined using histamine. Combined bronchoprotective potency was determined using methacholine in the absence of propranolol.
The assay was conducted using male Duncan-Hartley guinea pigs (Harlan, Indianapolis, IN), weighing between 250 and 400 g. A 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 generated from an LC Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA) driven by Bioblend (a mixture of 5% CO2; 21% O2; and 74% N2) at a pressure of 22 psi. Pulmonary function was evaluated at various time points after inhalation dosing.
Seventy- five minutes prior to the start of the assay, the guinea pigs were anesthetized with an intramuscular (IM) injection of a mixture of ketamine (43.7 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 niethacholine or histamine. 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 connected to a Biopac (TSD 137C) pre-amplifier. Body temperature was maintained at 37 0C using a heating pad. Prior to initiating data collection, pentobarbital (25 mg/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 of the bronchoconstrictor (methacholine or histamine). When methacholine was used as the bronchoconstrictor agent, animals were pre-treated with propranolol (5 mg/kg, IV) to isolate the antimuscarinic effects of the test compound. The propranolol was administered 30 minutes prior to construction of the dose-response curve to methacholine or histamine. 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 methacholine or histamine was fitted to a four parameter logistic equation using GraphPad Prism, version 3.00 for Windows (GraphPad Software, San Diego, California). The following equation was used:
Y = Min + (Max-Min)/(1 + 10 ((log '∞0"^ Hillslope) )
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 of the bronchoconstrictor response to a submaximal dose of methacholine or histamine was calculated at each dose of the 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) x 100). Inhibition curves were fitted using the four parameter logistic equation from GraphPad software. ID50 (dose required to produce 50% inhibition of the bronchoconstrictor response) and Emax (maximal inhibition) were also estimated wherever appropriate. The magnitude of bronchoprotection at different time-points after inhalation of the test compound was used to estimate the pharmacodynamic half-life (PD T1Z2). PD Tj/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 Ty2 = 0.69/K. Plateau was constrained to 0.
At 1.5 hours post-dose, biphenyl-2-ylcarbamic acid l-[2-(4-{[(Λ)-2-(3- ' formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-2,5- dimethylphenylcarbamoyl)ethyl]piperidin-4-yl ester (Ha) was found to have an ID50 of less than about 50 μg/mL for both methacholine-induced bronchoconstriction and histamine- induced bronchoconstriction.
Additionally, this compound produced significant bronchoprotection for up to about 72 hours when administered as a single sub-maximal dose (100 μg/mL). In this assay, salmeterol (3 μg/mL) (a β2 adrenergic receptor agonist) exhibited significant bronchoprotection for 6 to 14 hours; and tiotropium (10 μg/mL) (a muscarinic receptor antagonist) exhibited significant bronchoprotection for greater than 72 hours.
Example 18
Plethysmograph Guinea Pig Assay for Determining Bronchoprotective Efficacy and
Duration In this assay, the bronchoprotective efficacy and duration of test compounds was determined using a guinea pig assay.
Groups of 6 male guinea pigs (Duncan-Hartley (HsdPoctDH) 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 arranged 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). The aerosols were generated from aqueous solutions using an LC Star Nebulizer Set (Model 22F51, PARI Respiratory Equipment, Inc. Midlothian, VA) driven by a mixture of gases (CO2 = 5%, O2= 21% and N2 = 74%) at a pressure of 22 psi. The gas flow through the nebulizer at this operating pressure was approximately 3 L per 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 of the filled nebulizer.
The bronchoprotective effects of test compounds administered via inhalation were evaluated using whole body plethysmography at 1.5, 24, 48 and 72 hours post-dose. Forty-five minutes prior to the start of the pulmonary evaluation, each guinea pig was anesthetized with an intramuscular injection of ketamine (43.75 mg/kg), xylazine (3.50 mg/kg) and acepromazine (1.05 mg/kg). The surgical site was shaved and cleaned with 70% alcohol and a 2 to 3 cm midline incision of the ventral aspect of the neck was made. 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 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 of the anesthetic mixture. The depth of anesthesia was monitored and adjusted if the animal responded to pinching of its paw or if the respiration rate was greater than 100 breaths per minute.
Once the cannulations were completed, the animal was placed into a plethysmo graph (#PLY3114, Buxco Electronics, Inc., Sharon, CT) and an esophageal pressure cannula (PE- 160, Becton Dickinson, Sparks, MD) was inserted to measure pulmonary driving pressure. The teflon tracheal tube was attached to the opening of the 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 three 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 had not collapsed and that the animal did not suffer from hyperventilation.
The pulmonary evaluation was initiated after determining that baseline values were within the range of 0.3 to 0.9 mL per cm H2O for compliance and within the range of 0.1 to 0.199 cm H2O per mL per second for resistance. A Buxco pulmonary measurement computer pro gam was used for the collection and derivation of pulmonary values. Starting the program initiated the experimental protocol and data collection. The changes in volume over time that occurred 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 breth. This signal and the pulmonary driving pressure changes, collected using a Sensym pressure transducer (TRD4100), were connected via a Buxco (MAX 2270) preamplifier to a data collection interface (SFT3400 and SFT3813). AU other pulmonary parameters are derived from these two inputs. Baseline values were collected for 5 minutes, after which time the guinea pigs were challenged with either acetylcholine or histamine. When evaluating the muscarinic < antagonist effects of a test compound, propanolol (5 mg/Kg, iv) (Sigma- Aldrich, St. Louis, MO) was administered 15 minutes prior to challenge with acetylcholine. Acetylcholine (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 of the experiment: 1.9 μg/minute at 5 minutes, 3.8 μg/minute at 10 minutes, 7.5 μg/minute at 15 minutes, 15.0 μg/minute at 20 minutes, 30 μg/minute at 25 minutes and 60 μg/minute at 30 minutes. Alternatively, bronchoprotective effects of the test compound was assessed in the acetylcholine challenge model without pretreatment with propanolol.
When evaluating the β2 adrenergic receptor agonist effects of the test compound, 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 did not returned to baseline values at 3 minutes following each acetylcholine or histamine dose, the guinea pig's lungs were inflated 3 times with 4 mL of air from a 10 mL calibration syringe. The pulmonary parameters recorded included respiration frequency (breaths per minute), compliance (mL per cm H2O) and pulmonary resistance (cm H2O per 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 per mL per second) was calculated from the ratio of change in pressure to the change in flow. The RL response to acetylcholine (60 μg/min, IH) was computed for the vehicle and the test compound. The mean acetylcholine response in vehicle-treated animals, at each pre-treatment time, was calculated and used to compute percent inhibition of acetylcholine response, at the corresponding 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 BD50 (dose required to inhibit the acetylcholine (60 μg/min) bronchoconstriction response by 50%). The following equation was used:
Y = Min + (Max-Mrn)/(1 + 10 ((Iog m5^r Hillslope))
where X is the logarithm of dose, Y is the response (percent inhibition of acetylcholine- 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 acetylcholine or histamine needed to cause a doubling of the baseline pulmonary resistance, was calculated using the pulmonary resistance values derived from the flow and the pressure over a range of acetylcholine or histamine challenges using the following equation (which was derived from an equation used to calculate PC20 values described in American Thoracic Society. Guidelines for methacholine and exercise challenge testing - 1999. Am J Respir Crit Care Med. 2000; 161: 309-329):
(log C2 - log 0(2R0 - Ri) PD2 = antilog [ log C,+ J
R2 - R1 where:
Ci — concentration of acetylcholine or histamine preceding C2 C2 = concentration of acetylcholine or histamine resulting in at least a 2-fold increase in pulmonary resistance (R1)
R0 = Baseline RL value R1 = RL value after C1 R2 = RL value after C2
Statistical analysis of the data was performed using a two tailed Students t-test. A P-vahie <0.05 was considered significant.
Compound 50 described in U.S. Patent Publication No. US 2004/0167167A1, published August 24, 2004, was tested in this assay. The chemical structure of compound 50 is as follows:
Figure imgf000071_0001
This compound lacks the alkyl groups present on phenyl ring of the compounds of the present invention. In this assay, compound 50 showed no significant bronchoprotection at 24 hours post-dose for doses ranging from 3 μg/mL to 300 μg/mL. The PD2x valves for compound 50 at 24 hours were similar to the vehicle (water) group. In this assay, salmeterol (100 μg/mL) (a β2 adrenergic receptor agonist) exhibited significant bronchoprotection for at least 24 hours; and tiotropium (10 μg/mL) (a muscarinic receptor antagonist) exhibited significant bronchoprotection for at least 24 hours.
While the present invention has been described with reference to specific aspects or embodiments thereof, it will be understood by those of ordinary skilled in the art that various changes can be made or equivalents can be substituted without departing from the true spirit and scope of the invention. Additionally, to the extent permitted by applicable patent statutes 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

CLAIMS:
1. A compound of formula I:
Figure imgf000072_0001
wherein
R1 is methyl or ethyl;
R2 is methyl or ethyl; or a pharmaceutically acceptable salt or solvate or stereoisomer thereof.
2. A compound of Claim 1 having formula II:
Figure imgf000072_0002
wherein
R1 is methyl or ethyl;
R2 is methyl or ethyl; or a pharmaceutically acceptable salt thereof.
3. A compound of Claim 1 having formula Ha:
Figure imgf000073_0001
4. A compound of Claim 1, wherein the compound is a pharmaceutically acceptable salt of a compound of formula Ha:
Figure imgf000073_0002
5. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of any one of Claims 1 to 4.
6. A pharmaceutical composition comprising:
(a) a compound of any one of Claims 1 to 4;
(b) a steroidal anti-inflammatory agent; and
(c) a pharmaceutically acceptable carrier.
7. A combination of therapeutic agents, comprising:
(a) a compound of any one of Claims 1 to 4; and
(b) a steroidal anti-inflammatory agent.
8. A kit comprising:
(a) a first pharmaceutical composition comprising a compound of any one of Claims 1 to 4 and a first pharmaceutically acceptable carrier; and (b) a second pharmaceutical composition comprising a steroidal antiinflammatory agent and a second pharmaceutically acceptable carrier; wherein the first and second pharmaceutical compositions are separate pharmaceutical compositions.
9. A method of 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 4.
10. A method of treating chronic obstructive pulmonary disease or asthma, the method comprising administering to a patient a therapeutically effective amount of a compound of any one of Claims 1 to 4.
11. A method of producing bronchodilation in a mammal, the method comprising administering to a mammal a bronchodilation-producing amount of a compound of any one of Claims 1 to 4.
12. A method of antagonizing a muscarinic receptor and agonizing a β2 adrenergic receptor in a mammal, the method comprising administering to the mammal a compound of any one of Claims 1 to 4.
13. A method of using a compound of any one of Claims 1 to 4 as a research tool, the method comprising conducting a biological assay using a compound of any one of Claims 1 to 4.
14. A method of evaluating a test compound in a biological assay, the method comprising:
(a) conducting a biological assay with a test compound to provide a first assay value; (b) conducting the biological assay with a compound of any one of Claims 1 to 4 to provide a second assay value; wherein step (a) is conducted either before, after or concurrently with step (b); and
(c) comparing the first assay value from step (a) with the second assay value from step (b).
15. The method of Claim 14, wherein the biological assay is a muscarine receptor binding assay or a β2 adrenergic receptor binding assay.
16. The method of Claim 14, wherein the biological assay is a bronchoprotection assay in a mammal.
17. A process of preparing the compound of Claim 1, the process comprising deprotecting a compound of formula 6:
Figure imgf000075_0001
wherein P1 is a hydroxyl-protecting group, to provide a compound of formula I.
18. A process of preparing the compound of Claim 1, the process comprising deprotecting a compound of formula 6a:
Figure imgf000076_0001
wherein Ra, Rb and Rc are independently selected from CM alkyl, phenyl, -C1-4 alkyl-(phenyl), or one of Rla, Rlb and Rlc is -0-(CM alkyl); to provide a compound of formula I.
19. A process of preparing the compound of Claim 1, the process comprising: (a) reacting a compound of formula 4:
Figure imgf000076_0002
with a compound of formula 5 :
Figure imgf000076_0003
wherein P! is a hydroxyl-protecting group, in the presence of a reducing agent to provide a compound of formula 6:
Figure imgf000077_0001
and (b) deprotecting the compound of formula 6 to provide a compound of formula I.
20. A process of preparing a pharmaceutically acceptable salt of the compound of Claim 1, the process comprising contacting a compound of formula I in free base form with a pharmaceutically acceptable acid.
21. An intermediate for preparing a compound of any one of Claims 1 to 4, wherein the intermediate is a compound of formula III:
Figure imgf000077_0002
wherein
Y1 is selected from -CHO, -CN, -CH2OH5 -CH(OR3a)OR3b, -C(O)OH, -C(O)OR3c, bromo and iodo, where R »3a a „n„d, R > 3b are selected independently from C1-6 alkyl, or R 3a and R3b are joined to form C2-6 alkylene, R3c is selected from Ci-β alkyl; R1 is methyl or ethyl; R2 is methyl or ethyl; or a salt or stereoisomer thereof.
22. The compound of Claim 21 , wherein R1 and R2 are methyl.
23. The compound of Claim 21 , wherein Y1 is -CHO.
24. The compound of Claim 21 , wherein Y1 is -CHO; and R1 and R2 are methyl.
25. A compound of Claim 4, wherein the compound is biphenyl-2-ylcarbamic acid l-[2-(4-{[(i?)-2-(3-formylamino-4-hydroxyphenyl)-2-hydroxyethylamino]methyl}-
2,5-dimethylphenyl-carbamoyl)ethyl]piperidin-4-yl ester L-tartaric acid salt.
26. A compound of any one of Claims 1 to 4 for use in therapy.
27. Use of a compound of any one of Claims 1 to 4 for the manufacture of a medicament for treating a pulmonary disorder.
PCT/US2007/009925 2006-04-25 2007-04-24 DIALKYLPHENYL COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY WO2007127196A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
MX2008013555A MX2008013555A (en) 2006-04-25 2007-04-24 Dialkylphenyl compounds having î²2 adrenergic receptor agonist and muscarinic receptor antagonist activity.
JP2009507757A JP2009541209A (en) 2006-04-25 2007-04-24 Dialkylphenyl compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity
ES07776093T ES2391584T3 (en) 2006-04-25 2007-04-24 Dialkylphenyl compounds that have beta2-adrenergic receptor agonist activity and muscarinic receptor antagonist
EP07776093A EP2010489B1 (en) 2006-04-25 2007-04-24 Dialkylphenyl compounds having 2 adrenergic receptor agonist and muscarinic receptor antagonist activity
CA002650530A CA2650530A1 (en) 2006-04-25 2007-04-24 Dialkylphenyl compounds having .beta.2 adrenergic receptor agonist and muscarinic receptor antagonist activity
BRPI0710767-6A BRPI0710767A2 (en) 2006-04-25 2007-04-24 dialkylphenyl compounds having b2-adrenergic receptor agonist and muscarinic receptor antagonist activity
AU2007243481A AU2007243481A1 (en) 2006-04-25 2007-04-24 Dialkylphenyl compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
IL194334A IL194334A0 (en) 2006-04-25 2008-09-25 Dialkylphenyl compounds having ??2 adrenergic receptor agonist and muscarinic receptor antagonist activity
NO20084603A NO20084603L (en) 2006-04-25 2008-10-30 Dialkylphenyl compounds having beta2 adrenergic receptor agonist and muscarinic receptor agonist activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79470206P 2006-04-25 2006-04-25
US60/794,702 2006-04-25

Publications (2)

Publication Number Publication Date
WO2007127196A2 true WO2007127196A2 (en) 2007-11-08
WO2007127196A3 WO2007127196A3 (en) 2007-12-21

Family

ID=38577470

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/009925 WO2007127196A2 (en) 2006-04-25 2007-04-24 DIALKYLPHENYL COMPOUNDS HAVING β2 ADRENERGIC RECEPTOR AGONIST AND MUSCARINIC RECEPTOR ANTAGONIST ACTIVITY

Country Status (20)

Country Link
US (3) US7524965B2 (en)
EP (1) EP2010489B1 (en)
JP (1) JP2009541209A (en)
KR (1) KR20090005389A (en)
CN (1) CN101426764A (en)
AR (1) AR060647A1 (en)
AU (1) AU2007243481A1 (en)
BR (1) BRPI0710767A2 (en)
CA (1) CA2650530A1 (en)
CO (1) CO6140054A2 (en)
ES (1) ES2391584T3 (en)
IL (1) IL194334A0 (en)
MA (1) MA30556B1 (en)
MX (1) MX2008013555A (en)
NO (1) NO20084603L (en)
PE (1) PE20080094A1 (en)
RU (1) RU2008146383A (en)
TW (1) TW200811105A (en)
WO (1) WO2007127196A2 (en)
ZA (1) ZA200808817B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011512331A (en) * 2008-02-06 2011-04-21 アストラゼネカ・アクチエボラーグ Compound
WO2011081937A1 (en) 2009-12-15 2011-07-07 Gilead Sciences, Inc. Corticosteroid-beta-agonist-muscarinic antagonist compounds for use in therapy
EP3061821A1 (en) 2009-07-22 2016-08-31 Puretech Ventures Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10265311B2 (en) 2009-07-22 2019-04-23 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10925832B2 (en) 2018-09-28 2021-02-23 Karuna Therapeutics, Inc. Compositions and methods for treatment of disorders ameliorated by muscarinic receptor activation

Families Citing this family (9)

* 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
PE20040950A1 (en) 2003-02-14 2005-01-01 Theravance Inc BIPHENYL DERIVATIVES AS AGONISTS OF ß2-ADRENERGIC RECEPTORS AND AS ANTAGONISTS OF MUSCARINAL RECEPTORS
TWI341836B (en) 2004-03-11 2011-05-11 Theravance Inc Biphenyl compounds useful as muscarinic receptor antagonists
EP1778626A1 (en) * 2004-08-16 2007-05-02 Theravance, Inc. 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
TW200811105A (en) * 2006-04-25 2008-03-01 Theravance Inc Dialkylphenyl compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
SI2599778T1 (en) 2009-04-23 2017-08-31 Theravance Respiratory Company, Llc Diamide Compounds having Muscarinic Receptor Antagonist and Beta 2 Adrenergic Receptor Agonist Activity
GB201009801D0 (en) 2010-06-11 2010-07-21 Astrazeneca Ab Compounds 950
CN114409556A (en) * 2022-01-28 2022-04-29 兰州康鹏威耳化工有限公司 Preparation method of 3, 4-disubstituted-2-aminobenzaldehyde

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6433027B1 (en) 1999-05-12 2002-08-13 Boehringer Ingelheim Pharma Kg Medicament compositions based on tiotropium bromide and formoterol fumarate
US20040167167A1 (en) 2003-02-14 2004-08-26 Mathai Mammen Biphenyl derivatives

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995006635A1 (en) 1993-09-02 1995-03-09 Yamanouchi Pharmaceutical Co., Ltd. Carbamate derivative and medicine containing the same
KR970701174A (en) 1994-02-10 1997-03-17 오노다 마사요시 Novel carbamate derivative and medicinal composition containing the same
US6040344A (en) * 1996-11-11 2000-03-21 Sepracor Inc. Formoterol process
US6362371B1 (en) * 1998-06-08 2002-03-26 Advanced Medicine, Inc. β2- adrenergic receptor agonists
US6693202B1 (en) * 1999-02-16 2004-02-17 Theravance, Inc. Muscarinic receptor antagonists
UA73965C2 (en) * 1999-12-08 2005-10-17 Theravance Inc b2 ADRENERGIC RECEPTOR ANTAGONISTS
ATE381537T1 (en) 2001-03-22 2008-01-15 Glaxo Group Ltd FORMANILIDE DERIVATIVES AS BETA2 ADRENORECEPTOR AGONISTS
US20030018019A1 (en) * 2001-06-23 2003-01-23 Boehringer Ingelheim Pharma Kg Pharmaceutical compositions based on anticholinergics, corticosteroids and betamimetics
TWI249515B (en) * 2001-11-13 2006-02-21 Theravance Inc Aryl aniline beta2 adrenergic receptor agonists
CN100569760C (en) * 2003-11-21 2009-12-16 施万制药 Has β 2The active compound of 3 adrenergic receptor agonists and muscarinic receptor antagonist
US7456199B2 (en) 2004-03-11 2008-11-25 Theravance, Inc. Biphenyl compounds useful as muscarinic receptor antagonists
EP1778626A1 (en) * 2004-08-16 2007-05-02 Theravance, Inc. Compounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
TWI374883B (en) 2004-08-16 2012-10-21 Theravance Inc Crystalline form of a biphenyl compound
EP1833822A2 (en) * 2004-08-16 2007-09-19 Theravance, Inc. Compounds having beta2 adrenergic receptor agonist 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

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6433027B1 (en) 1999-05-12 2002-08-13 Boehringer Ingelheim Pharma Kg Medicament compositions based on tiotropium bromide and formoterol fumarate
US20040167167A1 (en) 2003-02-14 2004-08-26 Mathai Mammen Biphenyl derivatives
US7141671B2 (en) 2003-02-14 2006-11-28 Theravance, Inc. Biphenyl derivatives

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011512331A (en) * 2008-02-06 2011-04-21 アストラゼネカ・アクチエボラーグ Compound
US10369143B2 (en) 2009-07-22 2019-08-06 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
EP3061821A1 (en) 2009-07-22 2016-08-31 Puretech Ventures Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10238643B2 (en) 2009-07-22 2019-03-26 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10265311B2 (en) 2009-07-22 2019-04-23 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10369144B2 (en) 2009-07-22 2019-08-06 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
EP3646870A1 (en) 2009-07-22 2020-05-06 Puretech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10695339B2 (en) 2009-07-22 2020-06-30 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
WO2011081937A1 (en) 2009-12-15 2011-07-07 Gilead Sciences, Inc. Corticosteroid-beta-agonist-muscarinic antagonist compounds for use in therapy
US10925832B2 (en) 2018-09-28 2021-02-23 Karuna Therapeutics, Inc. Compositions and methods for treatment of disorders ameliorated by muscarinic receptor activation
US10933020B2 (en) 2018-09-28 2021-03-02 Karuna Therapeutics, Inc. Compositions and methods for treating disorders ameliorated by muscarinic receptor activation
US11452692B2 (en) 2018-09-28 2022-09-27 Karuna Therapeutics, Inc. Compositions and methods for treating disorders ameliorated by muscarinic receptor activation
US11471413B2 (en) 2018-09-28 2022-10-18 Karuna Therapeutics, Inc. Compositions and methods for treating disorders ameliorated by muscarinic receptor activation
US11890378B2 (en) 2018-09-28 2024-02-06 Karuna Therapeutics, Inc. Compositions and methods for treating disorders ameliorated by muscarinic receptor activation

Also Published As

Publication number Publication date
WO2007127196A3 (en) 2007-12-21
NO20084603L (en) 2008-11-19
US8134006B2 (en) 2012-03-13
AU2007243481A1 (en) 2007-11-08
EP2010489A2 (en) 2009-01-07
ZA200808817B (en) 2010-03-31
JP2009541209A (en) 2009-11-26
US20100137603A1 (en) 2010-06-03
RU2008146383A (en) 2010-05-27
PE20080094A1 (en) 2008-02-17
BRPI0710767A2 (en) 2011-06-07
ES2391584T3 (en) 2012-11-28
CN101426764A (en) 2009-05-06
TW200811105A (en) 2008-03-01
IL194334A0 (en) 2009-08-03
MX2008013555A (en) 2008-11-04
EP2010489B1 (en) 2012-08-22
AR060647A1 (en) 2008-07-02
KR20090005389A (en) 2009-01-13
US20070249675A1 (en) 2007-10-25
CO6140054A2 (en) 2010-03-19
US7524965B2 (en) 2009-04-28
US7687521B2 (en) 2010-03-30
MA30556B1 (en) 2009-07-01
US20090176833A1 (en) 2009-07-09
CA2650530A1 (en) 2007-11-08

Similar Documents

Publication Publication Date Title
EP2010489B1 (en) Dialkylphenyl compounds having 2 adrenergic receptor agonist and muscarinic receptor antagonist activity
EP2010490B1 (en) Crystalline forms of a dimethylphenyl compound
CA2557479C (en) Biphenyl compounds useful as muscarinic receptor antagonists
US8501776B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
US7858797B2 (en) Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087732A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
WO2005087735A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723112A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723109A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists
EP1723115A1 (en) Biphenyl compounds useful as muscarinic receptor antagonists

Legal Events

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

Ref document number: 07776093

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 571478

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 194334

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 8157/DELNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007776093

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007243481

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200780014098.4

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2650530

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/013555

Country of ref document: MX

Ref document number: 2008101742

Country of ref document: EG

WWE Wipo information: entry into national phase

Ref document number: 2009507757

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 12008502361

Country of ref document: PH

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 08115666

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2007243481

Country of ref document: AU

Date of ref document: 20070424

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020087028589

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2008146383

Country of ref document: RU

ENP Entry into the national phase

Ref document number: PI0710767

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20081024