WO2010028011A1 - Amides of diazabicyclononanes and uses thereof - Google Patents

Amides of diazabicyclononanes and uses thereof Download PDF

Info

Publication number
WO2010028011A1
WO2010028011A1 PCT/US2009/055687 US2009055687W WO2010028011A1 WO 2010028011 A1 WO2010028011 A1 WO 2010028011A1 US 2009055687 W US2009055687 W US 2009055687W WO 2010028011 A1 WO2010028011 A1 WO 2010028011A1
Authority
WO
WIPO (PCT)
Prior art keywords
nonane
diazabicyclo
optionally substituted
compounds
ylcarbonyo
Prior art date
Application number
PCT/US2009/055687
Other languages
French (fr)
Inventor
Srinivisa Rao Akireddy
Anatoly Mazurov
Daniel Yohannes
Lan Miao
Jason Daniel Speake
Original Assignee
Targacept, 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 Targacept, Inc. filed Critical Targacept, Inc.
Publication of WO2010028011A1 publication Critical patent/WO2010028011A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the present invention relates to compounds that bind to and modulate the activity of neuronal nicotinic acetylcholine receptors, to processes for preparing these compounds, to pharmaceutical compositions containing these compounds, and to methods of using these compounds for treating a wide variety of conditions and disorders, including those associated with dysfunction of the central nervous system (CNS).
  • CNS central nervous system
  • NNRs neuronal nicotinic receptors
  • nAChRs nicotinic acetylcholine receptors
  • NNR ligands have been proposed as therapies are cognitive disorders, including Alzheimer's disease, attention deficit disorder, and schizophrenia (Newhouse et al., Curr. Opin. Pharmacol. 4: 36 (2004), Levin and Rezvani, Curr. Drug Targets: CNS Neurol. Disord. 1: 423 (2002), Graham et al., Curr. Drug Targets: CNS Neurol. Disord. 1: 387 (2002), Ripoll et al., Curr. Med. Res. Opin. 20(7): 1057 (2004), and McEvoy and Allen, Curr. Drug Targets: CNS Neurol. Disord.
  • nicotinic compounds are associated with various undesirable side effects due to non-specific binding to multiple nAChR subtypes.
  • binding to and stimulation of muscle and ganglionic nAChR subtypes can lead to side effects which can limit the utility of a particular nicotinic binding compound as a therapeutic agent.
  • the compounds of the present invention exhibit a high degree of binding to ⁇ 4 ⁇ 2 nAChR subtype and lower affinity for ⁇ 7, ganglionic and muscle nAChR subtypes.
  • these compounds can serve as therapeutic modulators of ⁇ 4 ⁇ 2 nAChRs in patients in need of such treatment, without producing side effects caused by non-specific nAChR subtype binding.
  • the present invention includes compounds of Formula I:
  • Z 1 is methylene and n is 0 or 1 ;
  • Z 2 is methylene and m is 0 or 1 ; when n is 1 , m is 0; when m is 1 , n is 0;;
  • X 1 is hydrogen, C 1-6 alkyl, or Ci -6 haloalkyl
  • X 2 is R 1 , OR 1 Or NRV; each of R 1 , R 1 ", and R ⁇ v are individually hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C 3-8 cycloalkyl, optionally substituted C 2-6 alkenyl, optionally substituted C ⁇ ecycloalkenyl, optionally substituted C ⁇ alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; or R 1 " and R ⁇ v can combine with the nitrogen atom to which they are attached to form an optionally substituted 3 to 8 membered ring that may contain one or more degrees of unsaturation and may contain one or more additional heteroatom selected from N, O, or S; and
  • the compounds of the present invention bind with high affinity to NNRs of the ⁇ 4 ⁇ 2 subtype and exhibit selectivity for this subtype over the ⁇ 7 NNR subtype, as well as ganglion and muscle subtypes.
  • the present invention also relates to pharmaceutically acceptable salts prepared from these compounds.
  • the present invention includes pharmaceutical compositions comprising a compound of the present invention or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical compositions of the present invention can be used for treating or preventing a wide variety of conditions or disorders, including those disorders characterized by dysfunction of nicotinic cholinergic neurotransmission or the degeneration of the nicotinic cholinergic neurons.
  • the present invention includes a method for treating or preventing disorders and dysfunctions, such as CNS disorders and dysfunctions, inflammation, inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders, or other disorders described in further detail herein.
  • the present invention includes a method for modulating neovascularization. The methods involve administering to a subject a therapeutically effective amount of a compound of the present invention, including a salt thereof, or a pharmaceutical composition that includes such compounds.
  • the present invention includes compounds that have utility as diagnostic agents and in receptor binding studies as described herein.
  • C x -C y alkyl refers to an alkyl group, as herein defined, containing the specified number of carbon atoms. Similar terminology will apply for other preferred terms and ranges as well.
  • One embodiment of the present invention includes so-called 'lower 1 alkyl chains of one to eight, preferably one to six carbon atoms.
  • CrC 6 alkyl represents a lower alkyl chain as hereinabove described.
  • alkyl refers to a straight or branched chain hydrocarbon having one to eight carbon atoms, preferably one to six carbon atoms, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
  • alkyl as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl, n-butyl, tert-butyl, isopentyl, and n-pentyl.
  • alkenyl refers to a straight or branched chain aliphatic hydrocarbon having two to twelve carbon atoms, preferably two to eight carbon atoms, and containing one or more carbon-to-carbon double bonds, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
  • alkenyl as used herein include, but are not limited to, vinyl, and allyl.
  • alkynyl refers to a straight or branched chain aliphatic hydrocarbon having two to twelve carbon atoms, preferably two to eight carbon atoms, and containing one or more carbon-to-carbon triple bonds, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
  • An example of “alkynyl” as used herein includes, but is not limited to, ethynyl.
  • cycloalkyl refers to a fully saturated optionally substituted three- to twelve-mem bered, preferably three- to eight-membered, monocyclic, bicyclic, or bridged hydrocarbon ring, with multiple degrees of substitution being allowed.
  • exemplary "cycloalkyl” groups as used herein include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • cycloalkenyl and “cycloalkynyl” refer to optionally substituted, partially saturated but non-aromatic, three-to-twelve membered, preferably either five- to eight-membered or seven- to ten-membered, monocyclic, bicyclic, or bridged hydrocarbon rings, with one or more degrees of unsaturation, and with multiple degrees of substitution being allowed.
  • heterocycle refers to an optionally substituted mono- or polycyclic ring system, optionally containing one or more degrees of unsaturation and also containing one or more heteroatoms, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
  • exemplary heteroatoms include nitrogen, oxygen, or sulfur atoms, including N-oxides, sulfur oxides, and dioxides.
  • the ring is three to twelve-membered, preferably three- to eight-membered and is either fully saturated or has one or more degrees of unsaturation.
  • Such rings may be optionally fused to one or more of another heterocyclic ring(s) or cycloalkyl ring(s).
  • heterocyclic groups as used herein include, but are not limited to, tetrahydrofuran, pyran, 1 ,4-dioxane, 1 ,3-dioxane, piperidine, pyrrolidine, morpholine, tetrahydrothiopyran, and tetrahydrothiophene.
  • aryl refers to a univalent benzene ring or fused benzene ring system, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
  • aryl groups as used include, but are not limited to, phenyl, 2-naphthyl, 1-naphthyl, anthracene, and phenanthrene.
  • Preferable aryl rings have five- to ten- members.
  • a fused benzene ring system encompassed within the term "aryl” includes fused polycyclic hydrocarbons, namely where a cyclic hydrocarbon with less than maximum number of noncumulative double bonds, for example where a saturated hydrocarbon ring (cycloalkyl, such as a cyclopentyl ring) is fused with an aromatic ring (aryl, such as a benzene ring) to form, for example, groups such as indanyl and acenaphthalenyl, and also includes such groups as, for non-limiting examples, dihydronaphthalene and hexahydrocyclopenta-cyclooctene.
  • aralkyl refers to an "aryl” group as herein defined attached through an alkylene linker.
  • heteroaryl refers to a monocyclic five to seven membered aromatic ring, or to a fused bicyclic aromatic ring system comprising two of such aromatic rings, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
  • heteroaryl rings contain five- to ten-members.
  • These heteroaryl rings contain one or more nitrogen, sulfur, and/or oxygen atoms, where N-oxides, sulfur oxides, and dioxides are permissible heteroatom substitutions.
  • heteroaryl groups as used herein include, but should not be limited to, furan, thiophene, pyrrole, imidazole, pyrazole, triazole, tetrazole, thiazole, oxazole, isoxazole, oxadiazole, thiadiazole, isothiazole, pyridine, pyridazine, pyrazine, pyrimidine, quinoline, isoquinoline, benzofuran, benzoxazole, benzothiophene, indole, indazole, benzimidazole, imidazopyridine, pyrazolopyridine, and pyrazolopyrimidine.
  • the term "heteroaragonal as used herein
  • halogen refers to fluorine, chlorine, bromine, or iodine.
  • haloalkyl refers to an alkyl group, as defined herein, that is substituted with at least one halogen.
  • branched or straight chained “haloalkyl” groups as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, and t-butyl substituted independently with one or more halogens, for example, fluoro, chloro, bromo, and iodo.
  • haloalkyl should be interpreted to include such substituents as perfluoroalkyl groups such as -CF 3 .
  • alkoxy refers to a group -OR a , where R a is alkyl as defined above.
  • nitro refers to a group -NO 2 .
  • cyano refers to a group -CN.
  • zido refers to a group -N 3 .
  • amino refers to a group -NR a R b , where each of R a and R b individually is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocylcyl, or heteroaryl.
  • R a or R b when either R a or R b is other than hydrogen, such a group may be referred to as a “substituted amino" or, for example if R a is H and R b is alkyl, as an "alkylamino.”
  • hydroxyl refers to a group -OH.
  • Z 1 is methylene and n is 0 or 1 ;
  • Z 2 is methylene and m is 0 or 1 ; when n is 1 , m is 0; when m is 1 , n is 0;
  • X 1 is hydrogen, C 1-6 alkyl, or Ci -6 haloalkyl;
  • X 2 is R 1 , OR 1 Or NR 111 R 17 ; each of R 1 , R 1 ", and R ⁇ v are individually hydrogen, optionally substituted C ⁇ alkyl, optionally substituted C 3-8 cycloalkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 3-8 cycloalkenyl, optionally substituted C 2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; or R 1 " and R ⁇ v can combine with the nitrogen atom to
  • R" is optionally substituted Ci -6 alkyl, optionally substituted C 3-8 cycloalkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 3-8 cycloalkenyl, optionally substituted C 2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; where the term "optionally substituted” refers to optional substitution of one or more hydrogen atoms by substituents independently selected from C 1-6 alkyl, C 3-8 cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, OR V , NR V R VI , Ci -6 haloalkyl, -CN, -
  • R v and R v ⁇ are individually hydrogen, Ci -6 alkyl, C 3-8 cycloalkyl, heterocyclyl, aryl, or arylalkyl; or a pharmaceutically acceptable salt thereof.
  • One embodiment of the present invention includes use of a compound of the present invention in the manufacture of a medicament.
  • One embodiment of the present invention includes a method for the treatment or prevention of a variety of disorders and dysfunctions, comprising administering to a mammal in need of such treatment, a therapeutically effective amount of the compound of the present invention.
  • the disorder or dysfunction may be selected from the group consisting of CNS disorders, inflammation, inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders or other disorders described in further detail herein.
  • One embodiment of the present invention includes a method for modulating neovascularization.
  • Another embodiment of the present invention includes compounds that have utility as diagnostic agents and in receptor binding studies as described herein. Additionally, these compounds may also have utility as diagnostic agents and in receptor binding studies as described herein.
  • One embodiment of the present invention includes a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and one or more pharmaceutically acceptable carriers.
  • One embodiment of the present invention includes the use of a pharmaceutical composition of the present invention in the manufacture of a medicament for treatment of central nervous system disorders and dysfunctions.
  • Another embodiment of the present invention includes a compound as herein described with reference to any one of the Examples.
  • Another embodiment of the present invention includes a compound of the present invention for use as an active therapeutic substance.
  • Another embodiment of the present invention includes a compound of the present invention for use to modulate an NNR in a subject in need thereof.
  • Another embodiment of the present invention includes a compound of the present invention for use in the treatment or prevention of conditions or disorders mediated by NNR.
  • Another embodiment of the present invention includes a use of a compound of the present invention in the manufacture of a medicament for use of modulating NNR in a subject in need thereof.
  • Another embodiment of the present invention includes a use of a compound of the present invention in the manufacture of a medicament for use in the treatment or prevention of conditions or disorders mediated by NNR.
  • Another embodiment of the present invention includes a method of modulating NNR in a subject in need thereof through the administration of a compound of the present invention.
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structure except for the replacement of a hydrogen atom by a deuterium or tritium, or the replacement of a carbon atom by a 13 C- or 14 C-enriched carbon are within the scope of the invention.
  • the compounds of the present invention may crystallize in more than one form, a characteristic known as polymorphism, and such polymorphic forms (“polymorphs") are within the scope of the present invention.
  • Polymorphism generally can occur as a response to changes in temperature, pressure, or both. Polymorphism can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility, and melting point. Certain of the compounds described herein contain one or more chiral centers, or may otherwise be capable of existing as multiple stereoisomers.
  • the scope of the present invention includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures.
  • the present invention also includes the individual isomers of the compounds represented by the formulas above as mixtures with isomers thereof in which one or more chiral centers are inverted.
  • the present invention includes a salt or solvate of the compounds herein described, including combinations thereof such as a solvate of a salt.
  • the compounds of the present invention may exist in solvated, for example hydrated, as well as unsolvated forms, and the present invention encompasses all such forms.
  • the salts of the present invention are pharmaceutically acceptable salts. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention.
  • Suitable pharmaceutically acceptable salts include inorganic acid addition salts such as chloride, bromide, sulfate, phosphate, and nitrate; organic acid addition salts such as acetate, galactarate, propionate, succinate, lactate, glycolate, malate, tartrate, citrate, maleate, fumarate, methanesulfonate, p-toluenesulfonate, and ascorbate; salts with acidic amino acid such as aspartate and glutamate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; ammonium salt; organic basic salts such as trimethylamine salt, triethylamine salt, pyridine salt, picoline salt, dicyclohexylamine salt, and N,N'-dibenzylethylenediamine salt; and salts with basic amino acid such as lysine salt and arginine salt.
  • inorganic acid addition salts such as chlor
  • the salts may be in some cases hydrates or ethanol solvates.
  • Representative salts are provided as described in U.S. Patent Nos. 5,597,919 to Dull et al., 5,616,716 to Dull et al. and 5,663,356 to Ruecroft et al, each of which is herein incorporated by reference with regard to such salts.
  • the present invention includes specific representative compounds, which are identified herein with particularity.
  • the compounds of this invention may be made by a variety of methods, including well-known standard synthetic methods. Illustrative general synthetic methods are set out below and then specific compounds of the invention are prepared in the working Examples. In all of the examples described below, protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles of synthetic chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts,
  • the present invention includes all possible stereoisomers and includes not only racemic compounds but the individual enantiomers as well.
  • a compound is desired as a single enantiomer, such may be obtained by stereospecif ic synthesis, by resolution of the final product or any convenient intermediate, or by chiral chromatographic methods as are known in the art. Resolution of the final product, an intermediate, or a starting material may be effected by any suitable method known in the art. See, for example, Stereochemistry of Organic Compounds (Wiley-lnterscience, 1994), incorporated by reference with regard to stereochemistry.
  • the present invention also provides a method for the synthesis of compounds useful as intermediates in the preparation of compounds of the present invention along with methods for their preparation.
  • the compounds can be prepared according to the following methods using readily available starting materials and reagents. In these reactions, variants may be employed which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail.
  • the 3,6-diazabicyclo[3.2.2]nonane scaffold was prepared as illustrated in Scheme 1. Pyridine-2,5-dicarboxylic acid (1), was converted to the methyl ester and hydrogenated over platinum oxide to yield compound 2. Protection of the amine moiety with with a t-butoxycarbonyl (Boc) protecting group, followed by hydride reduction of both esters and subsequent oxidative cleavage of the double bond, yields dialdehyde 3.
  • Boc t-butoxycarbonyl
  • Compound 4 can be selectively deprotected to yield 6-(tert- butoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane (5, Scheme 2), which can be subsequently used for the synthesis of 3-substituted 3,6- diazabicyclo[3.2.2]nonanes .
  • Compound 5 can also be treated sequentially with trifluoroacetic anhydride and trifluoroacetic acid to yield trifluoroacetyl (TFA) protected compound, 6, which can be subsequently used for the synthesis of 6-substituted 3,6-diazabicyclo[3.2.2]nonanes.
  • TFA trifluoroacetyl
  • Amide compounds of the present invention can be prepared via the coupling of mono-protected diazabicycle (5 or 6) with a suitably functionalized acid chloride or other reactive carboxylic acid derivative.
  • a suitably functionalized acid chloride or other reactive carboxylic acid derivative Such a compound may be available commercially or prepared by treatment of the appropriate carboxylic acid with, among other reagents, thionyl chloride or oxalyl chloride. After amide formation, removal of the protecting group will afford the compounds of the present invention.
  • carbamate and urea compounds of the present invention can be prepared by the coupling 5 or 6 with a suitably functionalized chloroformate or isocyanate derivative, respectively.
  • Other compounds of the present invention can be synthesized by alkylation of the remaining basic (and nucleophilic) nitrogen with an activated alkyl compound such as an alkyl halide or alkyl sulfate.
  • compositions of the present invention include the salts described herein, in the pure state or in the form of a composition in which the compounds are combined with any other pharmaceutically compatible product, which can be inert or physiologically active.
  • the resulting pharmaceutical compositions can be used to prevent a condition or disorder in a subject susceptible to such a condition or disorder, and/or to treat a subject suffering from the condition or disorder.
  • the pharmaceutical compositions described herein include one or more compounds of Formula I and/or pharmaceutically acceptable salts thereof.
  • compositions are preferably administered orally (e.g., in liquid form within a solvent such as an aqueous or non-aqueous liquid, or within a solid carrier).
  • Preferred compositions for oral administration include pills, tablets, capsules, caplets, syrups, and solutions, including hard gelatin capsules and time-release capsules.
  • Standard excipients include binders, fillers, colorants, solubilizers and the like.
  • Compositions can be formulated in unit dose form, or in multiple or subunit doses.
  • Preferred compositions are in liquid or semisolid form.
  • Compositions including a liquid pharmaceutically inert carrier such as water or other pharmaceutically compatible liquids or semisolids can be used.
  • compositions can also be administered via injection, i.e., intravenously, intramuscularly, subcutaneously, intraperitoneally, intraarterially, intrathecally; and intracerebroventricularly.
  • Intravenous administration is the preferred method of injection.
  • Suitable carriers for injection are well known to those of skill in the art and include 5% dextrose solutions, saline, and phosphate- buffered saline.
  • the compounds can also be administered as an infusion or injection (e.g., as a suspension or as an emulsion in a pharmaceutically acceptable liquid or mixture of liquids).
  • the formulations can also be administered using other means, for example, rectal administration.
  • Formulations useful for rectal administration are well known to those of skill in the art.
  • the compounds can also be administered by inhalation (e.g., in the form of an aerosol either nasally or using delivery articles of the type set forth in U.S. Patent No. 4,922,901 to Brooks et al., the disclosure of which is incorporated herein in its entirety); topically (e.g., in lotion form); transdermal ⁇ (e.g., using a transdermal patch) or iontophoretically; or by sublingual or buccal administration.
  • inhalation e.g., in the form of an aerosol either nasally or using delivery articles of the type set forth in U.S. Patent No. 4,922,901 to Brooks et al., the disclosure of which is incorporated herein in its entirety
  • topically e.g., in lotion form
  • transdermal ⁇ e.g., using a transdermal patch
  • iontophoretically iontophoretically
  • compositions can contain a liquid carrier that can be oily, aqueous, emulsified or contain certain solvents suitable to the mode of administration.
  • the compositions can be administered intermittently or at a gradual, continuous, constant or controlled rate to a warm-blooded animal (e.g., a mammal such as a mouse, rat, cat, rabbit, dog, pig, cow, or monkey), but advantageously are administered to a human being.
  • a warm-blooded animal e.g., a mammal such as a mouse, rat, cat, rabbit, dog, pig, cow, or monkey
  • time of day and the number of times per day that the pharmaceutical formulation is administered can vary.
  • Other suitable methods for administering the compounds of the present invention are described in U.S. Patent No. 5,604,231 to Smith et al., the contents of which are hereby incorporated by reference.
  • the compound of the present invention may be administered in combination with other therapeutic compounds.
  • a compound of this invention can be used in combination with other NNR ligands (such as varenicline), antioxidants (such as free radical scavenging agents), antibacterial agents (such as penicillin antibiotics), antiviral agents (such as nucleoside analogs, like zidovudine and acyclovir), anticoagulants (such as warfarin), anti-inflammatory agents (such as NSAIDs), anti-pyretics, analgesics, anesthetics (such as used in surgery), acetylcholinesterase inhibitors (such as donepezil and galantamine), antipsychotics (such as haloperidol, clozapine, olanzapine, and quetiapine), immuno-suppressants (such as cyclosporin and methotrexate), neuroprotective agents, steroids (such as steroid hormones
  • steroids such as steroid hormones
  • the compounds of the present invention may be employed alone or in combination with other therapeutic agents, including other compounds of the present invention.
  • Such a combination of pharmaceutically active agents may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order.
  • the amounts of the compounds or agents and the relative timings of administration will be selected in order to achieve the desired therapeutic effect.
  • the administration in combination of a compound of the formulae of the present invention including salts or solvates thereof with other treatment agents may be in combination by administration concomitantly in: (1) a unitary pharmaceutical composition including both compounds; or (2) separate pharmaceutical compositions each including one of the compounds.
  • the combination may be administered separately in a sequential manner wherein one treatment agent is administered first and the other second or vice versa.
  • the compounds of the present invention may be used in the treatment of a variety of disorders and conditions and, as such, the compounds of the present invention may be used in combination with a variety of other suitable therapeutic agents useful in the treatment or prophylaxis of those disorders or conditions.
  • the appropriate dose of the compound is that amount effective to prevent occurrence of the symptoms of the disorder or to treat some symptoms of the disorder from which the patient suffers.
  • effective amount By “effective amount”, “therapeutic amount” or “effective dose” is meant that amount sufficient to elicit the desired pharmacological or therapeutic effects, thus resulting in effective prevention or treatment of the disorder.
  • an effective amount of compound is an amount sufficient to pass across the blood-brain barrier of the subject, to bind to relevant receptor sites in the brain of the subject and to modulate the activity of relevant NNR subtypes (e.g., provide neurotransmitter secretion, thus resulting in effective prevention or treatment of the disorder).
  • Prevention of the disorder is manifested by delaying the onset of the symptoms of the disorder.
  • Treatment of the disorder is manifested by a decrease in the symptoms associated with the disorder or an amelioration of the recurrence of the symptoms of the disorder.
  • the effective amount is sufficient to obtain the desired result, but insufficient to cause appreciable side effects.
  • the effective dose can vary, depending upon factors such as the condition of the patient, the severity of the symptoms of the disorder, and the manner in which the pharmaceutical composition is administered.
  • the effective dose of typical compounds generally requires administering the compound in an amount sufficient to modulate the activity of relevant NNRs, but the amount should be insufficient to induce effects on skeletal muscles and ganglia to any significant degree.
  • the effective dose of compounds will of course differ from patient to patient, but in general includes amounts starting where CNS effects or other desired therapeutic effects occur but below the amount where muscular effects are observed.
  • the compounds described herein when employed in effective amounts in accordance with the methods described herein, can provide some degree of prevention of the progression of, ameliorate symptoms of, and ameliorate to some degree of the recurrence of CNS or other disorders.
  • the effective amounts of those compounds are typically below the threshold concentration required to elicit any appreciable side effects, for example those effects relating to skeletal muscle or ganglia.
  • the compounds can be administered in a therapeutic window in which certain CNS and other disorders are treated and certain side effects are avoided.
  • the effective dose of the compounds described herein is sufficient to provide the desired effects upon the disorder but is insufficient (i.e., is not at a high enough level) to provide undesirable side effects.
  • the compounds are administered at a dosage effective for treating the CNS or other disorders but less than 1/5, and often less than 1/10, the amount required to elicit certain side effects to any significant degree.
  • effective doses are at very low concentrations, where maximal effects are observed to occur, with a minimum of side effects.
  • An effective dose of such compounds may require administering the compound in an amount of less than 5 mg/kg of patient weight.
  • the compounds of the present invention may be administered in an amount from less than about 1 mg/kg patent weight and usually less than about 100 ⁇ g/kg of patient weight, but may be between about 10 ⁇ g to less than 100 ⁇ g/kg of patient weight.
  • the foregoing doses typically represent that amount administered as a single dose, or as one or more doses administered over a 24-hour period.
  • an effective dose of typical compounds generally requires administering the compound in an amount of at least about 1 , often at least about 10, and frequently at least about 100 mg/ 24 hr/ patient.
  • an effective dose of typical compounds requires administering the compound which generally does not exceed about 500, often does not exceed about 400, and frequently does not exceed about 300 mg/ 24 hr/ patient.
  • the compositions may be advantageously administered at an effective dose such that the concentration of the compound within the plasma of the patient normally does not exceed 50 ng/mL, often does not exceed 30 ng/mL, and frequently does not exceed 10 ng/mL.
  • the compounds of the present invention can be used for the prevention or treatment of various conditions or disorders for which other types of nicotinic compounds have been proposed or are shown to be useful as therapeutics, such as CNS disorders, inflammation, inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders, addictions, obesity or other disorders described in further detail herein.
  • This compound can also be used as a diagnostic agent in receptor binding studies (in vitro and in vivo).
  • Such therapeutic and other teachings are described, for example, in references previously listed herein, including Williams et al., Drug News Perspec. 7(4): 205 (1994), Arneric et al., CNS Drug Rev. 1(1): 1-26 (1995), Arneric et al., Exp. Opin.
  • CNS Disorders The compounds and their pharmaceutical compositions are useful in the treatment or prevention of a variety of CNS disorders, including neurodegenerative disorders, neuropsychiatric disorders, neurologic disorders, and addictions.
  • the compounds and their pharmaceutical compositions can be used to treat or prevent cognitive deficits and dysfunctions, age-related and otherwise; attentional disorders and dementias, including those due to infectious agents or metabolic disturbances; to provide neuroprotection; to treat convulsions and multiple cerebral infarcts; to treat mood disorders, compulsions and addictive behaviors; to provide analgesia; to control inflammation, such as mediated by cytokines and nuclear factor kappa B; to treat inflammatory disorders; to provide pain relief; and to treat infections, as anti-infectious agents for treating bacterial, fungal, and viral infections.
  • AAMI age-associated memory impairment
  • MCI mild cognitive impairment
  • ARCD age-related cognitive decline
  • pre-senile dementia early onset Alzheimer's disease
  • senile dementia dementia of the
  • Alzheimer's type Alzheimer's disease, cognitive impairment no dementia (CIND), Lewy body dementia, HIV-dementia, AIDS dementia complex, vascular dementia, Down syndrome, head trauma, traumatic brain injury (TBI), dementia pugilistica, Creutzfeld-Jacob Disease and prion diseases, stroke, central ischemia, peripheral ischemia, attention deficit disorder, attention deficit hyperactivity disorder, dyslexia, schizophrenia, schizophreniform disorder, schizoaffective disorder, cognitive dysfunction in schizophrenia, cognitive deficits in schizophrenia, Parkinsonism including Parkinson's disease, postencephalitic parkinsonism, parkinsonism-dementia of Gaum, frontotemporal dementia Parkinson's Type (FTDP), Pick's disease, Niemann-Pick's Disease, Huntington's Disease, Huntington's chorea, tardive dyskinesia, hyperkinesia, progressive supranuclear palsy, progressive supranuclear paresis, restless leg syndrome, Creutzfeld-Jakob disease, multiple
  • Cognitive impairments or dysfunctions may be associated with psychiatric disorders or conditions, such as schizophrenia and other psychotic disorders, including but not limited to psychotic disorder, schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic disorder, and psychotic disorders due to a general medical conditions, dementias and other cognitive disorders, including but not limited to mild cognitive impairment, pre-senile dementia, Alzheimer's disease, senile dementia, dementia of the Alzheimer's type, age-related memory impairment, Lewy body dementia, vascular dementia, AIDS dementia complex, dyslexia, Parkinsonism including Parkinson's disease, cognitive impairment and dementia of Parkinson's Disease, cognitive impairment of multiple sclerosis, cognitive impairment caused by traumatic brain injury, dementias due to other general medical conditions, anxiety disorders, including but not limited to panic disorder without agoraphobia, panic disorder with agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia, obsessive-compulsive disorder, post-traumatic stress disorder, acute
  • Cognitive performance may be assessed with a validated cognitive scale, such as, for example, the cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog).
  • ADAS-cog Alzheimer's Disease Assessment Scale
  • One measure of the effectiveness of the compounds of the present invention in improving cognition may include measuring a patient's degree of change according to such a scale.
  • the compounds of the present invention may be used as a therapy for nicotine addiction and for other brain-reward disorders, such as substance abuse including alcohol addiction, illicit and prescription drug addiction, eating disorders, including obesity, and behavioral addictions, such as gambling, or other similar behavioral manifestations of addiction.
  • substance abuse including alcohol addiction, illicit and prescription drug addiction
  • eating disorders including obesity
  • behavioral addictions such as gambling, or other similar behavioral manifestations of addiction.
  • the treatment or prevention of diseases, disorders and conditions occurs without appreciable adverse side effects, including, for example, significant increases in blood pressure and heart rate, significant negative effects upon the gastro-intestinal tract, and significant effects upon skeletal muscle.
  • the compounds of the present invention when employed in effective amounts, are believed to modulate the activity of the ⁇ 4 ⁇ 2 and ⁇ 7 NNRs without appreciable interaction with the nicotinic subtypes that characterize the human ganglia, as demonstrated by a lack of the ability to elicit nicotinic function in adrenal chromaffin tissue, or skeletal muscle, further demonstrated by a lack of the ability to elicit nicotinic function in cell preparations expressing muscle-type nicotinic receptors.
  • these compounds are believed capable of treating or preventing diseases, disorders and conditions without eliciting significant side effects associated activity at ganglionic and neuromuscular sites.
  • administering is believed to provide a therapeutic window in which treatment of certain diseases, disorders and conditions is provided, and certain side effects are avoided. That is, an effective dose of the compound is believed sufficient to provide the desired effects upon the disease, disorder or condition, but is believed insufficient, namely is not at a high enough level, to provide undesirable side effects.
  • the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, for use in therapy, such as a therapy described above.
  • the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a CNS disorder, such as a disorder, disease or condition described hereinabove.
  • the nervous system primarily through the vagus nerve, is known to regulate the magnitude of the innate immune response by inhibiting the release of macrophage tumor necrosis factor (TNF).
  • TNF macrophage tumor necrosis factor
  • This physiological mechanism is known as the "cholinergic anti-inflammatory pathway" (see, for example, Tracey, "The Inflammatory Reflex,” Nature 420: 853-9 (2002)).
  • Excessive inflammation and tumor necrosis factor synthesis cause morbidity and even mortality in a variety of diseases. These diseases include, but are not limited to, endotoxemia, rheumatoid arthritis, osteoarthritis, psoriasis, asthma, atherosclerosis, idiopathic pulmonary fibrosis, and inflammatory bowel disease.
  • Inflammatory conditions that can be treated or prevented by administering the compounds described herein include, but are not limited to, chronic and acute inflammation, psoriasis, endotoxemia, gout, acute pseudogout, acute gouty arthritis, arthritis, rheumatoid arthritis, osteoarthritis, allograft rejection, chronic transplant rejection, asthma, atherosclerosis, mononuclear-phagocyte dependent lung injury, idiopathic pulmonary fibrosis, atopic dermatitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, acute chest syndrome in sickle cell disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis, acute cholangitis, aphteous stomatitis, pouchitis, glomerulonephritis, lupus nephritis, thrombosis, and graft vs. host reaction.
  • bacterial and/or viral infections are associated with side effects brought on by the formation of toxins, and the body's natural response to the bacteria or virus and/or the toxins.
  • the body's response to infection often involves generating a significant amount of TNF and/or other cytokines.
  • the over-expression of these cytokines can result in significant injury, such as septic shock (when the bacteria is sepsis), endotoxic shock, urosepsis and toxic shock syndrome.
  • Cytokine expression is mediated by NNRs, and can be inhibited by administering agonists or partial agonists of these receptors.
  • Those compounds described herein that are agonists or partial agonists of these receptors can therefore be used to minimize the inflammatory response associated with bacterial infection, as well as viral and fungal infections. Examples of such bacterial infections include anthrax, botulism, and sepsis. Some of these compounds may also have antimicrobial properties.
  • Antitoxins can also be used as adjunct therapy in combination with existing therapies to manage bacterial, viral and fungal infections, such as antibiotics, antivirals and antifungals.
  • Antitoxins can also be used to bind to toxins produced by the infectious agents and allow the bound toxins to pass through the body without generating an inflammatory response. Examples of antitoxins are disclosed, for example, in U.S. Patent No. 6,310,043 to Bundle et al. Other agents effective against bacterial and other toxins can be effective and their therapeutic effect can be complemented by co-administration with the compounds described herein.
  • the compounds can be administered to treat and/or prevent pain, including acute, neurologic, inflammatory, neuropathic and chronic pain.
  • the compounds can be used in conjunction with opiates to minimize the likelihood of opiate addiction (e.g., morphine sparing therapy).
  • opiate addiction e.g., morphine sparing therapy.
  • the analgesic activity of compounds described herein can be demonstrated in models of persistent inflammatory pain and of neuropathic pain, performed as described in U.S. Published Patent Application No. 20010056084 A1 (Allgeier et at.) (e.g., mechanical hyperalgesia in the complete Freund's adjuvant rat model of inflammatory pain and mechanical hyperalgesia in the mouse partial sciatic nerve ligation model of neuropathic pain).
  • the analgesic effect is suitable for treating pain of various genesis or etiology, in particular in treating inflammatory pain and associated hyperalgesia, neuropathic pain and associated hyperalgesia, chronic pain (e.g., severe chronic pain, post-operative pain and pain associated with various conditions including cancer, angina, renal or biliary colic, menstruation, migraine, and gout).
  • chronic pain e.g., severe chronic pain, post-operative pain and pain associated with various conditions including cancer, angina, renal or biliary colic, menstruation, migraine, and gout.
  • Inflammatory pain may be of diverse genesis, including arthritis and rheumatoid disease, teno-synovitis and vasculitis.
  • Neuropathic pain includes trigeminal or herpetic neuralgia, diabetic neuropathy pain, causalgia, low back pain and deafferentation syndromes such as brachial plexus avulsion.
  • the ⁇ 7 NNR is associated with neovascularization.
  • Inhibition of neovascularization for example, by administering antagonists (or at certain dosages, partial agonists) of the ⁇ 7 NNR can treat or prevent conditions characterized by undesirable neovascularization or angiogenesis.
  • Such conditions can include those characterized by inflammatory angiogenesis and/or ischemia-induced angiogenesis.
  • Neovascularization associated with tumor growth can also be inhibited by administering those compounds described herein that function as antagonists or partial agonists of ⁇ 7 NNR.
  • Specific antagonism of ⁇ 7 NNR-specific activity reduces the angiogenic response to inflammation, ischemia, and neoplasia.
  • Representative tumor types that can be treated using the compounds described herein include NSCLC, ovarian cancer, pancreatic cancer, breast carcinoma, colon carcinoma, rectum carcinoma, lung carcinoma, oropharynx carcinoma, hypopharynx carcinoma, esophagus carcinoma, stomach carcinoma, pancreas carcinoma, liver carcinoma, gallbladder carcinoma, bile duct carcinoma, small intestine carcinoma, urinary tract carcinoma, kidney carcinoma, bladder carcinoma, urothelium carcinoma, female genital tract carcinoma, cervix carcinoma, uterus carcinoma, ovarian carcinoma, choriocarcinoma, gestational trophoblastic disease, male genital tract carcinoma, prostate carcinoma, seminal vesicles carcinoma, testes carcinoma, germ cell tumors, endocrine gland carcinoma, thyroid carcinoma, adrenal carcinoma, pituitary gland carcinoma, skin carcinoma, hemangiomas, melanomas, sarcomas, bone and soft tissue sarcoma, Kaposi's sarcoma, tumors of the brain, tumors
  • the compounds can also be administered in conjunction with other forms of anti-cancer treatment, including co-administration with antineoplastic antitumor agents such as cis-platin, adriamycin, daunomycin, and the like, and/or anti-VEGF (vascular endothelial growth factor) agents, as such are known in the art.
  • antineoplastic antitumor agents such as cis-platin, adriamycin, daunomycin, and the like
  • anti-VEGF vascular endothelial growth factor
  • the compounds can be administered in such a manner that they are targeted to the tumor site.
  • the compounds can be administered in microspheres, microparticles or liposomes conjugated to various antibodies that direct the microparticles to the tumor.
  • the compounds can be present in microspheres, microparticles or liposomes that are appropriately sized to pass through the arteries and veins, but lodge in capillary beds surrounding tumors and administer the compounds locally to the tumor.
  • Such drug delivery devices are known in the art.
  • the compounds of the present invention can be also used to prevent or treat certain other conditions, diseases, and disorders in which NNRs play a role.
  • autoimmune disorders such as Lupus, disorders associated with cytokine release, cachexia secondary to infection (e.g., as occurs in AIDS, AIDS related complex and neoplasia), obesity, pemphitis, urinary incontinence, retinal diseases, infenctious diseases, myasthenia, Eaton- Lambert syndrome, hypertension, preeclampsia, osteoporosis, vasoconstriction, vasodilatation, cardiac arrhythmias, type I diabetes, bulimia, anorexia as well as those indications set forth in published PCT application WO 98/25619.
  • the compounds of this invention can also be administered to treat convulsions such as those that are symptomatic of epilepsy, and to treat conditions such as syphill
  • the compounds can be used in diagnostic compositions, such as probes, particularly when they are modified to include appropriate labels.
  • the probes can be used, for example, to determine the relative number and/or function of specific receptors, particularly the ⁇ 4 ⁇ 2 and ⁇ 7 receptor subtypes.
  • the compounds of the present invention most preferably are labeled with a radioactive isotopic moiety such as 11 C, 18 F, 76 Br, 123 I or 125 I.
  • the administered compounds can be detected using known detection methods appropriate for the label used. Examples of detection methods include position emission topography (PET) and single-photon emission computed tomography (SPECT).
  • PET position emission topography
  • SPECT single-photon emission computed tomography
  • the radiolabels described above are useful in PET (e.g., 11 C, 18 F or 76 Br) and SPECT (e.g., 123 I) imaging, with half-lives of about 20.4 minutes for 11 C, about 109 minutes for 18 F, about 13 hours for 123 I, and about 16 hours for 76 Br.
  • a high specific activity is desired to visualize the selected receptor subtypes at non-saturating concentrations.
  • the administered doses typically are below the toxic range and provide high contrast images.
  • the compounds are expected to be capable of administration in non-toxic levels. Determination of dose is carried out in a manner known to one skilled in the art of radiolabel imaging. See, for example, U.S. Patent No. 5,969,144 to London
  • the compounds can be administered using known techniques. See, for example, U.S. Patent No. 5,969,144 to London et al., as noted.
  • the compounds can be administered in formulation compositions that incorporate other ingredients, such as those types of ingredients that are useful in formulating a diagnostic composition.
  • Compounds useful in accordance with carrying out the present invention most preferably are employed in forms of high purity. See, U.S. Patent No. 5,853,696 to Elmalch etal.
  • a subject e.g., a human subject
  • the presence of that compound within the subject can be imaged and quantified by appropriate techniques in order to indicate the presence, quantity, and functionality of selected NNR subtypes.
  • the compounds can also be administered to animals, such as mice, rats, dogs, and monkeys.
  • SPECT and PET imaging can be carried out using any appropriate technique and apparatus. See Villemagne et al., In: Arneric et al. (Eds.) Neuronal Nicotinic Receptors: Pharmacology and Therapeutic Opportunities, 235-250 (1998) and U.S. Patent No. 5,853,696 to Elmalch et al., each herein incporated by reference, for a disclosure of representative imaging techniques.
  • the radiolabeled compounds bind with high affinity to selective NNR subtypes (e.g., ⁇ 4 ⁇ 2, ⁇ 7) and preferably exhibit negligible non-specific binding to other nicotinic cholinergic receptor subtypes (e.g., those receptor subtypes associated with muscle and ganglia).
  • the compounds can be used as agents for noninvasive imaging of nicotinic cholinergic receptor subtypes within the body of a subject, particularly within the brain for diagnosis associated with a variety of CNS diseases and disorders.
  • the diagnostic compositions can be used in a method to diagnose disease in a subject, such as a human patient.
  • the method involves administering to that patient a detectably labeled compound as described herein, and detecting the binding of that compound to selected NNR subtypes (e.g., ⁇ 4 ⁇ 2 and ⁇ 7 receptor subtypes).
  • selected NNR subtypes e.g., ⁇ 4 ⁇ 2 and ⁇ 7 receptor subtypes.
  • Those skilled in the art of using diagnostic tools, such as PET and SPECT can use the radiolabeled compounds described herein to diagnose a wide variety of conditions and disorders, including conditions and disorders associated with dysfunction of the central and autonomic nervous systems.
  • Such disorders include a wide variety of CNS diseases and disorders, including Alzheimer's disease, Parkinson's disease, and schizophrenia.
  • These and other representative diseases and disorders that can be evaluated include those that are set forth in U.S. Patent No. 5,952,339 to Bencherif et al.
  • the diagnostic compositions can be used in a method to monitor selective nicotinic receptor subtypes of a subject, such as a human patient.
  • the method involves administering a detectably labeled compound as described herein to that patient and detecting the binding of that compound to selected nicotinic receptor subtypes namely, the ⁇ 4 ⁇ 2 and ⁇ 7 receptor subtypes.
  • the compounds of this invention can be used as reference ligands in binding assays for compounds which bind to NNR subtypes, particularly the ⁇ 4 ⁇ 2 and ⁇ 7 receptor subtypes.
  • the compounds of this invention are preferably labeled with a radioactive isotopic moiety such as 3 H, or 14 C. Examples of such binding assays are described in detail below.
  • Example 2 Synthesis of 1-(tert-butoxycarbonyl)-2,5- bis(hydroxymethyl)piperidine To a solution of 1 -(tert-butoxycarbonyl)-2,5-bis(methoxycarbonyl)piperidine (46 g, 0.15 mol) in anhydrous THF (500 mL) at 0 0 C was added lithium aluminum hydride (700 ml. of 1 M solution in THF, 0.70 mol) over 10 minutes. The reaction was stirred at 0 0 C for 30 min. The reaction was carefully quenched by addition of powdered sodium sulfate decahydrate (100 g) and the mixture was stirred for 1 h at ambient temperature.
  • lithium aluminum hydride 700 ml. of 1 M solution in THF, 0.70 mol
  • Example 3 1-(tert-butoxycarbonyl)piperidine-2,5-dialdehyde To a mixture of 1 -(tert-butoxycarbonyl)-2,5-bis(hydroxymethyl)piperidine (23 g,
  • Example 4 6-(tert-butoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane
  • 1 -(tert-butoxycarbonyl)piperidine-2,5-dialdehyde 7.O g, 29.5 mmol
  • benzylamine 3.21 mL, 29.5 mmol
  • dichloromethane methanol
  • sodium triaceoxyborohydride 31.5 g, 148 mmol
  • Certain amide derivatives can be prepared by reaction of ⁇ -ftert-butoxycarbonyl ⁇ e-diazabicyclo ⁇ Jnonane with acid chlorides and similar reagents.
  • Certain other amide derivatives can be made by reaction of 3-(trifluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane with acid chlorides and similar reagents. The following procedures are exemplary.
  • the crude amide was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 3-(trifluoroacetyl)-6-(N-propylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane.
  • the relevant fractions were combined, and the solvent evaporated.
  • the residue was dissolved in methanol (3 mL), and 20% aqueous potassium carbonate solution (0.5 mL) was added. The reaction mixture was stirred for 16 h at ambient temperature. The solvent was evaporated and the residue was triturated with dichloromethane (2 x 3 mL).
  • Example 9 6-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane
  • 2-fluoroethyl chloroformate (0.13 g, 1.1 mmol) followed by triethylamine (1 mL) at 0 0 C.
  • the solution was stirred for 2 h at ambient temperature.
  • the reaction solution was washed with water (2 mL) and concentrated.
  • the crude amide was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 3-(trifluoroacetyl)-6-(2-fluoroethoxycarbonyl)-3,6- diazabicyclo[3.2.2]nonane.
  • the relevant fractions were combined, and the solvent evaporated.
  • the residue was dissolved in methanol (3 mL), and 20% aqueous potassium carbonate solution (0.5 mL) was added. The reaction mixture was stirred for 16 h at ambient temperature. The solvent was evaporated and the residue was triturated with dichloromethane (2 x 3 mL).
  • Rats Male, Sprague- Dawley
  • Rats weighing 150-250 g
  • Animals were anesthetized with 70% CO 2 , and then decapitated. Brains were removed and placed on an ice-cold platform.
  • the cerebral cortex was removed and placed in 20 volumes (weight:volume) of ice-cold preparative buffer (137 mM NaCI, 10.7 mM KCI, 5.8 mM KH 2 PO 4 , 8 mM Na 2 HPO 4 , 20 mM HEPES (free acid), 5 mM iodoacetamide, 1.6 mM EDTA, pH 7.4); PMSF, dissolved in methanol to a final concentration of 100 ⁇ M, was added and the suspension was homogenized by Polytron. The homogenate was centrifuged at 18,000 x g for 20 min at 4 0 C and the resulting pellet was re-suspended in 20 volumes of ice-cold water. After 60 min incubation on ice, a new pellet was collected by centrifugation at 18,000 x g for 20 min at 4 0 C. The final pellet was re-suspended in 10 volumes of buffer and stored at -20 0 C.
  • ice-cold preparative buffer
  • the frozen membranes were thawed and spun at 48,000 x g for 20 min. The supernatant was decanted and discarded. The pellet was resuspended in Dulbecco's phosphate buffered saline (PBS, Life Technologies) pH 7.4 and homogenized with the Polytron for 6 seconds. Protein concentrations were determined using a Pierce BCA Protein Assay Kit, with bovine serum albumin as the standard (Pierce Chemical Company, Rockford, IL).
  • Membrane preparations (approximately 50 ⁇ g for human and 200-300 ⁇ g protein for rat ⁇ 4 ⁇ 2) were incubated in PBS (50 ⁇ L and 100 ⁇ L respectively) in the presence of competitor compound (0.01 nM to 100 ⁇ M) and 5 nM [ 3 H]nicotine for 2-3 hours on ice. Incubation was terminated by rapid filtration on a multi-manifold tissue harvester (Brandel, Gaithersburg, MD) using GF/B filters presoaked in 0.33% polyethyleneimine (w/v) to reduce non-specific binding. Tissue was rinsed 3 times in PBS, pH 7.4. Scintillation fluid was added to filters containing the washed tissue and allowed to equilibrate. Filters were then counted to determine radioactivity bound to the membranes by liquid scintillation counting (2200CA Tri-Carb LSC, Packard Instruments, 50% efficiency or Wallac Trilux 1450 MicroBeta, 40% efficiency, Perkin Elmer).
  • Ki IC 50 / (1 + N/Kd) where N is the concentration of [ 3 H]nicotine and Kd is the affinity of nicotine (3 nM, determined in a separate experiment). ⁇ 7 nAChR Subtype
  • Rats male, Sprague-Dawley
  • weighing 150-250 g were maintained on a 12 h light/dark cycle and were allowed free access to water and food supplied by PMI Nutrition International, Inc.
  • Animals were anesthetized with 70% CO 2 , then decapitated. Brains were removed and placed on an ice-cold platform.
  • the hippocampus was removed and placed in 10 volumes (weight:volume) of ice- cold preparative buffer (137 mM NaCI, 10.7 mM KCI, 5.8 mM KH 2 PO 4 , 8 mM Na 2 HPO 4 , 20 mM HEPES (free acid), 5 mM iodoacetamide, 1.6 mM EDTA, pH 7.4); PMSF, dissolved in methanol to a final concentration of 100 ⁇ M, was added and the tissue suspension was homogenized by Polytron. The homogenate was centrifuged at 18,000 x g for 20 min at 4 0 C and the resulting pellet was re-suspended in 10 volumes of ice-cold water. After 60 min incubation on ice, a new pellet was collected by centrifugation at 18,000 x g for 20 min at 4 0 C. The final pellet was re-suspended in 10 volumes of buffer and stored at -20 0 C.
  • ice- cold preparative buffer
  • tissue was thawed, centrifuged at 18,000 x g for 20 min, and then re-suspended in ice-cold PBS (Dulbecco's Phosphate Buffered Saline, 138 mM NaCI, 2.67 mM KCI, 1.47 mM KH 2 PO 4, 8.1 mM
  • PBS Dulbecco's Phosphate Buffered Saline
  • the binding of [ 3 H]MLA was determined using a 2 h incubation at 21 0 C. Incubations were conducted in 48-weil micro-titre plates and contained about 200 ⁇ g of protein per well in a final incubation volume of 300 ⁇ L. The incubation buffer was PBS and the final concentration of [ 3 H]MLA was 5 nM.
  • the binding reaction was terminated by filtration of the protein containing bound ligand onto glass fiber filters (GF/B, Brandel) using a Brandel Tissue Harvester at room temperature. Filters were soaked in de- ionized water containing 0.33% polyethyleneimine to reduce non-specific binding. Each filter was washed with PBS (3 x 1 mL) at room temperature. Non-specific binding was determined by inclusion of 50 ⁇ M non-radioactive MLA in selected wells.
  • the inhibition of [ 3 H]MLA binding by test compounds was determined by including seven different concentrations of the test compound in selected wells. Each concentration was replicated in triplicate. IC 50 values were estimated as the concentration of compound that inhibited 50 percent of specific [ 3 H]MLA binding. Inhibition constants (Ki values), reported in nM, were calculated from the IC 50 values using the method of Cheng et al., Biochem. Pharmacol. 22: 3099-3108 (1973), herein incorporated by reference.
  • Receptor binding data for compounds of the present invention are shown in Table 1.
  • Ki values inhibition constants
  • ND inhibition constants
  • failing to bind sufficiently in HTS means, for the ⁇ 4 ⁇ 2 subtype, that the compound failed to inhibit, at 5 ⁇ M concentration, the binding of 5 nM 3 H- nicotine by at least 50%, and for the ⁇ 7 subtype, that the compound failed to inhibit, at 5 ⁇ M concentration, the binding of 5 nM 3 H-MLA (methyllycaconitine) by at least 50%.
  • the specific pharmacological responses observed may vary according to and depending on the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with practice of the present invention.

Abstract

The present invention relates to compounds of the following formula (I) that bind to and modulate the activity of neuronal nicotinic acetylcholine receptors, to processes for preparing these compounds, to pharmaceutical compositions containing these compounds, and to methods of using these compounds for treating a wide variety of conditions and disorders, including those associated with dysfunction of the central nervous system (CNS).

Description

AMIDES OF DIAZABICYCLONONANES AND USES THEREOF
Field of the Invention
The present invention relates to compounds that bind to and modulate the activity of neuronal nicotinic acetylcholine receptors, to processes for preparing these compounds, to pharmaceutical compositions containing these compounds, and to methods of using these compounds for treating a wide variety of conditions and disorders, including those associated with dysfunction of the central nervous system (CNS).
Background of the Invention
The therapeutic potential of compounds that target neuronal nicotinic receptors (NNRs), also known as nicotinic acetylcholine receptors (nAChRs), has been the subject of several reviews. See, for example, Breining et al., Ann. Rep. Med. Chem. 40: 3 (2005), Hogg and Bertrand, Curr. Drug Targets: CNS Neurol. Disord. 3: 123 (2004), Suto and Zacharias, Expert Opin. Ther. Targets 8: 61 (2004), Dani et al., Bioorg. Med. Chem. Lett. 14: 1837 (2004), Bencherif and Schmitt, Curr. Drug Targets: CNS Neurol. Disord. 1: 349 (2002), each incorporated by reference with regard to such teaching. Among the kinds of indications for which NNR ligands have been proposed as therapies are cognitive disorders, including Alzheimer's disease, attention deficit disorder, and schizophrenia (Newhouse et al., Curr. Opin. Pharmacol. 4: 36 (2004), Levin and Rezvani, Curr. Drug Targets: CNS Neurol. Disord. 1: 423 (2002), Graham et al., Curr. Drug Targets: CNS Neurol. Disord. 1: 387 (2002), Ripoll et al., Curr. Med. Res. Opin. 20(7): 1057 (2004), and McEvoy and Allen, Curr. Drug Targets: CNS Neurol. Disord. 1: 433 (2002)); pain and inflammation (Decker et al., Curr. Top. Med. Chem. 4(3): 369 (2004), Vincler, Expert Opin. Invest. Drugs 14(10): 1191 (2005), Jain, Curr. Opin. Inv. Drugs 5: 76 (2004), Miao et al., Neuroscience 123: 777 (2004)); depression and anxiety (Shytle et al., MoI. Psychiatry 7: 525 (2002), Damaj et al., MoI. Pharmacol. 66: 675 (2004), Shytle et al., Depress. Anxiety 16: 89 (2002)); neurodegeneration (O'Neill et al., Curr. Drug Targets: CNS Neurol. Disord. 1: 399 (2002), Takata et al., J. Pharmacol. Exp. Ther. 306: 772 (2003), Marrero et al., J. Pharmacol. Exp. Ther. 309: 16 (2004)); Parkinson's disease (Jonnala and Buccafusco, J. Neurosci. Res. 66: 565 (2001)); addiction (Dwoskin and Crooks, Biochem. Pharmacol. 63: 89 (2002), Coe et al., Bioorg. Med. Chem. Lett. 15(22): 4889 (2005)); obesity (Li et al., Curr. Top. Med. Chem. 3: 899 (2003)); and Tourette's syndrome (Sacco et al., J. Psychopharmacol. 18(4): 457 (2004), Young et al., Clin. Ther. 23(4): 532 (2001 )), each of these references incorporated by reference with regard to the nexus of the receptor and the named indication(s).
A limitation of some nicotinic compounds is that they are associated with various undesirable side effects due to non-specific binding to multiple nAChR subtypes. For example, binding to and stimulation of muscle and ganglionic nAChR subtypes can lead to side effects which can limit the utility of a particular nicotinic binding compound as a therapeutic agent. The compounds of the present invention exhibit a high degree of binding to α4β2 nAChR subtype and lower affinity for α7, ganglionic and muscle nAChR subtypes. Thus, these compounds can serve as therapeutic modulators of α4β2 nAChRs in patients in need of such treatment, without producing side effects caused by non-specific nAChR subtype binding.
Summary of the Invention
The present invention includes compounds of Formula I:
Figure imgf000003_0001
Formula I wherein:
Z1 is methylene and n is 0 or 1 ;
Z2 is methylene and m is 0 or 1 ; when n is 1 , m is 0; when m is 1 , n is 0;;
X1 is hydrogen, C1-6 alkyl, or Ci-6 haloalkyl;
X2 is R1, OR1Or NRV; each of R1, R1", and Rιv are individually hydrogen, optionally substituted C1-6 alkyl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-6alkenyl, optionally substituted C^ecycloalkenyl, optionally substituted C^alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; or R1" and Rιv can combine with the nitrogen atom to which they are attached to form an optionally substituted 3 to 8 membered ring that may contain one or more degrees of unsaturation and may contain one or more additional heteroatom selected from N, O, or S; and
R" is optionally substituted C1^ alkyl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-6alkenyl, optionally substituted C3-8 cycloalkenyl, optionally substituted C2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; where the term "optionally substituted" refers to optional substitution of one or more hydrogen atoms by substituents independently selected from Ci-s alkyl, C3-8 cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, ORV, NRVRVI, C1-6 haloalkyl, -CN, - NO2, -C2RV, -SRV, -N3, -C(=O)NRVRVI, -NRvC(=O)Rvl, -OC(=O)NRVRVI, -NRVC(=O)ORVI, -SO2RV, -SO2NRV, and -NRVSO2RVI, where Rv and R are individually hydrogen, Ci-6 alkyl, C3-8 cycloalkyl, heterocyclyl, aryl, or arylalkyl; or a pharmaceutically acceptable salt thereof.
The compounds of the present invention bind with high affinity to NNRs of the α4β2 subtype and exhibit selectivity for this subtype over the α7 NNR subtype, as well as ganglion and muscle subtypes. The present invention also relates to pharmaceutically acceptable salts prepared from these compounds. The present invention includes pharmaceutical compositions comprising a compound of the present invention or a pharmaceutically acceptable salt thereof. The pharmaceutical compositions of the present invention can be used for treating or preventing a wide variety of conditions or disorders, including those disorders characterized by dysfunction of nicotinic cholinergic neurotransmission or the degeneration of the nicotinic cholinergic neurons.
The present invention includes a method for treating or preventing disorders and dysfunctions, such as CNS disorders and dysfunctions, inflammation, inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders, or other disorders described in further detail herein. The present invention includes a method for modulating neovascularization. The methods involve administering to a subject a therapeutically effective amount of a compound of the present invention, including a salt thereof, or a pharmaceutical composition that includes such compounds. Additionally, the present invention includes compounds that have utility as diagnostic agents and in receptor binding studies as described herein. The foregoing and other aspects of the present invention are explained in further detail in the detailed description and examples set forth below.
Detailed Description I. Compounds The following definitions are meant to clarify, but not limit, the terms defined. If a particular term used herein is not specifically defined, such term should not be considered indefinite. Rather, terms are used within their accepted meanings.
As used throughout this specification, the preferred number of atoms, such as carbon atoms, will be represented by, for example, the phrase "Cx-Cy alkyl," which refers to an alkyl group, as herein defined, containing the specified number of carbon atoms. Similar terminology will apply for other preferred terms and ranges as well. One embodiment of the present invention includes so-called 'lower1 alkyl chains of one to eight, preferably one to six carbon atoms. Thus, for example, CrC6 alkyl represents a lower alkyl chain as hereinabove described.
As used herein the term "alkyl" refers to a straight or branched chain hydrocarbon having one to eight carbon atoms, preferably one to six carbon atoms, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed. Examples of "alkyl" as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl, n-butyl, tert-butyl, isopentyl, and n-pentyl.
As used herein the term "alkenyl" refers to a straight or branched chain aliphatic hydrocarbon having two to twelve carbon atoms, preferably two to eight carbon atoms, and containing one or more carbon-to-carbon double bonds, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed. Examples of "alkenyl" as used herein include, but are not limited to, vinyl, and allyl.
As used herein the term "alkynyl" refers to a straight or branched chain aliphatic hydrocarbon having two to twelve carbon atoms, preferably two to eight carbon atoms, and containing one or more carbon-to-carbon triple bonds, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed. An example of "alkynyl" as used herein includes, but is not limited to, ethynyl.
As used herein, the term "cycloalkyl" refers to a fully saturated optionally substituted three- to twelve-mem bered, preferably three- to eight-membered, monocyclic, bicyclic, or bridged hydrocarbon ring, with multiple degrees of substitution being allowed. Exemplary "cycloalkyl" groups as used herein include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
Similarly, as used herein, the terms "cycloalkenyl" and "cycloalkynyl" refer to optionally substituted, partially saturated but non-aromatic, three-to-twelve membered, preferably either five- to eight-membered or seven- to ten-membered, monocyclic, bicyclic, or bridged hydrocarbon rings, with one or more degrees of unsaturation, and with multiple degrees of substitution being allowed.
As used herein, the term "heterocycle" or "heterocyclyl" refers to an optionally substituted mono- or polycyclic ring system, optionally containing one or more degrees of unsaturation and also containing one or more heteroatoms, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed. Exemplary heteroatoms include nitrogen, oxygen, or sulfur atoms, including N-oxides, sulfur oxides, and dioxides. Preferably, the ring is three to twelve-membered, preferably three- to eight-membered and is either fully saturated or has one or more degrees of unsaturation. Such rings may be optionally fused to one or more of another heterocyclic ring(s) or cycloalkyl ring(s). Examples of "heterocyclic" groups as used herein include, but are not limited to, tetrahydrofuran, pyran, 1 ,4-dioxane, 1 ,3-dioxane, piperidine, pyrrolidine, morpholine, tetrahydrothiopyran, and tetrahydrothiophene.
As used herein, the term "aryl" refers to a univalent benzene ring or fused benzene ring system, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed. Examples of "aryl" groups as used include, but are not limited to, phenyl, 2-naphthyl, 1-naphthyl, anthracene, and phenanthrene. Preferable aryl rings have five- to ten- members.
As used herein, a fused benzene ring system encompassed within the term "aryl" includes fused polycyclic hydrocarbons, namely where a cyclic hydrocarbon with less than maximum number of noncumulative double bonds, for example where a saturated hydrocarbon ring (cycloalkyl, such as a cyclopentyl ring) is fused with an aromatic ring (aryl, such as a benzene ring) to form, for example, groups such as indanyl and acenaphthalenyl, and also includes such groups as, for non-limiting examples, dihydronaphthalene and hexahydrocyclopenta-cyclooctene. As used herein, the term "aralkyl" refers to an "aryl" group as herein defined attached through an alkylene linker.
As used herein, the term "heteroaryl" refers to a monocyclic five to seven membered aromatic ring, or to a fused bicyclic aromatic ring system comprising two of such aromatic rings, which may be optionally substituted as herein further described, with multiple degrees of substitution being allowed.
Preferably, such rings contain five- to ten-members. These heteroaryl rings contain one or more nitrogen, sulfur, and/or oxygen atoms, where N-oxides, sulfur oxides, and dioxides are permissible heteroatom substitutions. Examples of "heteroaryl" groups as used herein include, but should not be limited to, furan, thiophene, pyrrole, imidazole, pyrazole, triazole, tetrazole, thiazole, oxazole, isoxazole, oxadiazole, thiadiazole, isothiazole, pyridine, pyridazine, pyrazine, pyrimidine, quinoline, isoquinoline, benzofuran, benzoxazole, benzothiophene, indole, indazole, benzimidazole, imidazopyridine, pyrazolopyridine, and pyrazolopyrimidine. As used herein, the term "heteroaralkyl" refers to an "heteroaryl" group as herein defined attached through an alkylene linker.
As used herein the term "halogen" refers to fluorine, chlorine, bromine, or iodine.
As used herein the term "haloalkyl" refers to an alkyl group, as defined herein, that is substituted with at least one halogen. Examples of branched or straight chained "haloalkyl" groups as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, and t-butyl substituted independently with one or more halogens, for example, fluoro, chloro, bromo, and iodo. The term "haloalkyl" should be interpreted to include such substituents as perfluoroalkyl groups such as -CF3.
As used herein the term "alkoxy" refers to a group -ORa, where Ra is alkyl as defined above.
As used herein the term "nitro" refers to a group -NO2.
As used herein the term "cyano" refers to a group -CN. As used herein the term "azido" refers to a group -N3.
As used herein "amino" refers to a group -NRaRb, where each of Ra and Rb individually is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocylcyl, or heteroaryl. As used herein, when either Ra or Rb is other than hydrogen, such a group may be referred to as a "substituted amino" or, for example if Ra is H and Rb is alkyl, as an "alkylamino."
As used herein, the term "hydroxyl" refers to a group -OH.
One embodiment of the present invention includes a compound as represented by Formula I:
Figure imgf000008_0001
Formula I wherein:
Z1 is methylene and n is 0 or 1 ; Z2 is methylene and m is 0 or 1 ; when n is 1 , m is 0; when m is 1 , n is 0; X1 is hydrogen, C1-6 alkyl, or Ci-6 haloalkyl; X2 is R1, OR1Or NR111R17; each of R1, R1", and Rιv are individually hydrogen, optionally substituted C^ alkyl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-6 alkenyl, optionally substituted C3-8cycloalkenyl, optionally substituted C2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; or R1" and Rιv can combine with the nitrogen atom to which they are attached to form an optionally substituted 3 to 8 membered ring that may contain one or more degrees of unsaturation and may contain one or more additional heteroatom selected from N, O, or S; and
R" is optionally substituted Ci-6 alkyl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-6 alkenyl, optionally substituted C3-8cycloalkenyl, optionally substituted C2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; where the term "optionally substituted" refers to optional substitution of one or more hydrogen atoms by substituents independently selected from C1-6 alkyl, C3-8 cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, ORV, NRVRVI, Ci-6 haloalkyl, -CN, -
NO2, -C2RV, -SRV, -N3, -C(=O)NRVRVI, -NRVC(=O)RVI, -OC(=O)NRVRVI,
-NRVC(=O)ORVI, -SO2RV, -SO2NRVRVI, and -NRVSO2RVI, where Rv and R are individually hydrogen, Ci-6 alkyl, C3-8 cycloalkyl, heterocyclyl, aryl, or arylalkyl; or a pharmaceutically acceptable salt thereof.
One embodiment of the present invention includes a compound selected from the group consisting of:
3-(acetyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(methoxyacetyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(fluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(propanoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(butanoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(cyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1 R,2R)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1 R,2S)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1S,2R)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1S,2S)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(2,2,3,3-tetramethylcyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(cyclobutanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(cyclopentanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(cyclopent-3-enecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(N-methylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(N-ethylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(N-propylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(N-butylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(N-cyclopentylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(methoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(ethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(propoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(isopropoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(2-methoxyethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(phenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(4-fluorophenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, S-lfuran^-ylcarbonyO-S.Θ-diazabicycloβ^^lnonane, S-^-methylfuran^-ylcarbonyO-S.e-diazabicycloβ^^nonane, 3-(4-methylfuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, S-^-methylfuran^-ylcarbonyO-S.e-diazabicycloβ^^lnonane, 3-(3-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(4-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(5-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(4-methyloxazol-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(5-methyloxazol-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(5-methyloxazol-4-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(oxazol-5-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(3-methylisoxazol-5-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, S-^-methylisoxazol-S-ylcarbonyO-S.β-diazabicycloβ^^nonane, S-φenzofuran^-ylcarbonyiy-S.β-diazabicyclofS^^nonane, 3-(cyclopropanecarbonyl)-6-methyl-3,6-diazabicyclo[3.2.2]nonane, S-^uran^-ylcarbonyO-θ-methyl-S.δ-diazabicyclofS^^nonane, 6-(acetyl)-3,6-diazabicyclo[3.2.2]nonane,
6-(fluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(methoxyacetyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(propanoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(butanoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1R,2R)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1 R,2S)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1S,2R)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1S,2S)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(2,2,3,3-tetramethylcyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclobutanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclopentanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclopent-3-enecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(N-methylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(N-ethylcarbamoyi)-3,6-diazabicyclo[3.2.2]nonane, 6-(N-propylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(N-butylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(N-cyclopentylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(methoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(ethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(propoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, β-OsopropoxycarbonylJ-S.β-diazabicycloβ^^jnonane, 6-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(2-methoxyethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(phenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, β-^-fluorophenoxycarbonyO-S.Θ-diazabicycloβ^^nonane, 6-(furan-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(3-methylfuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, θ-^-methylfuran^-ylcarbonyO-S.Θ-diazabicycloβ^^nonane, 6-(5-methylfuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(3-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(4-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(5-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(4-methyloxazol-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, β^δ-methyloxazol^-ylcarbonyO-S.Θ-diazabicycloβ^^nonane, β^S-methyloxazol^-ylcarbonylJ-S^-diazabicyclotS^^Jnonane, θ-faxazol-δ-ylcarbonylJ-S.Θ-diazabicycloβ^^Jnonane, 6-(3-methylisoxazol-5-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(4-methylisoxazol-5-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, θ^benzofuran^-ylcarbonyO-S.β-diazabicycloβ^^lnonane, 6-(cyclopropanecarbonyl)-3-methyl-3,6-diazabicyclo[3.2.2]nonane, and β^furan^-ylcarbonylJ-S-methyl-S.δ-diazabicyclofS^^Jnonane.or a pharmaceutically acceptable salt thereof.
One embodiment of the present invention includes use of a compound of the present invention in the manufacture of a medicament. One embodiment of the present invention includes a method for the treatment or prevention of a variety of disorders and dysfunctions, comprising administering to a mammal in need of such treatment, a therapeutically effective amount of the compound of the present invention. More specifically, the disorder or dysfunction may be selected from the group consisting of CNS disorders, inflammation, inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders or other disorders described in further detail herein. One embodiment of the present invention includes a method for modulating neovascularization. Another embodiment of the present invention includes compounds that have utility as diagnostic agents and in receptor binding studies as described herein. Additionally, these compounds may also have utility as diagnostic agents and in receptor binding studies as described herein.
One embodiment of the present invention includes a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and one or more pharmaceutically acceptable carriers. One embodiment of the present invention includes the use of a pharmaceutical composition of the present invention in the manufacture of a medicament for treatment of central nervous system disorders and dysfunctions.
Another embodiment of the present invention includes a compound as herein described with reference to any one of the Examples. Another embodiment of the present invention includes a compound of the present invention for use as an active therapeutic substance.
Another embodiment of the present invention includes a compound of the present invention for use to modulate an NNR in a subject in need thereof.
Another embodiment of the present invention includes a compound of the present invention for use in the treatment or prevention of conditions or disorders mediated by NNR.
Another embodiment of the present invention includes a use of a compound of the present invention in the manufacture of a medicament for use of modulating NNR in a subject in need thereof. Another embodiment of the present invention includes a use of a compound of the present invention in the manufacture of a medicament for use in the treatment or prevention of conditions or disorders mediated by NNR.
Another embodiment of the present invention includes a method of modulating NNR in a subject in need thereof through the administration of a compound of the present invention.
The scope of the present invention includes combinations of embodiments.
Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of a hydrogen atom by a deuterium or tritium, or the replacement of a carbon atom by a 13C- or 14C-enriched carbon are within the scope of the invention.
The compounds of the present invention may crystallize in more than one form, a characteristic known as polymorphism, and such polymorphic forms ("polymorphs") are within the scope of the present invention. Polymorphism generally can occur as a response to changes in temperature, pressure, or both. Polymorphism can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility, and melting point. Certain of the compounds described herein contain one or more chiral centers, or may otherwise be capable of existing as multiple stereoisomers. The scope of the present invention includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures. Also included within the scope of the invention are the individual isomers of the compounds represented by the formulae of the present invention, as well as any wholly or partially equilibrated mixtures thereof. The present invention also includes the individual isomers of the compounds represented by the formulas above as mixtures with isomers thereof in which one or more chiral centers are inverted. The present invention includes a salt or solvate of the compounds herein described, including combinations thereof such as a solvate of a salt. The compounds of the present invention may exist in solvated, for example hydrated, as well as unsolvated forms, and the present invention encompasses all such forms. Typically, but not absolutely, the salts of the present invention are pharmaceutically acceptable salts. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention.
Examples of suitable pharmaceutically acceptable salts include inorganic acid addition salts such as chloride, bromide, sulfate, phosphate, and nitrate; organic acid addition salts such as acetate, galactarate, propionate, succinate, lactate, glycolate, malate, tartrate, citrate, maleate, fumarate, methanesulfonate, p-toluenesulfonate, and ascorbate; salts with acidic amino acid such as aspartate and glutamate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; ammonium salt; organic basic salts such as trimethylamine salt, triethylamine salt, pyridine salt, picoline salt, dicyclohexylamine salt, and N,N'-dibenzylethylenediamine salt; and salts with basic amino acid such as lysine salt and arginine salt. The salts may be in some cases hydrates or ethanol solvates. Representative salts are provided as described in U.S. Patent Nos. 5,597,919 to Dull et al., 5,616,716 to Dull et al. and 5,663,356 to Ruecroft et al, each of which is herein incorporated by reference with regard to such salts.
As noted herein, the present invention includes specific representative compounds, which are identified herein with particularity. The compounds of this invention may be made by a variety of methods, including well-known standard synthetic methods. Illustrative general synthetic methods are set out below and then specific compounds of the invention are prepared in the working Examples. In all of the examples described below, protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles of synthetic chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts,
Protecting Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, New York (1999), incorporated by reference with regard to protecting groups). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of the present invention.
Those skilled in the art will recognize if a stereocenter exists. As noted hereinabove, the present invention includes all possible stereoisomers and includes not only racemic compounds but the individual enantiomers as well. When a compound is desired as a single enantiomer, such may be obtained by stereospecif ic synthesis, by resolution of the final product or any convenient intermediate, or by chiral chromatographic methods as are known in the art. Resolution of the final product, an intermediate, or a starting material may be effected by any suitable method known in the art. See, for example, Stereochemistry of Organic Compounds (Wiley-lnterscience, 1994), incorporated by reference with regard to stereochemistry.
The present invention also provides a method for the synthesis of compounds useful as intermediates in the preparation of compounds of the present invention along with methods for their preparation. The compounds can be prepared according to the following methods using readily available starting materials and reagents. In these reactions, variants may be employed which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail.
II. General Synthetic Methods Several methods for the preparation of 3,6-diazabicyclo[3.2.2]nonanes have been published. One such method described by Fray et al. in J. Org. Chem. 53: 896 (1988) involves the Diels-Alder condensation of 1 ,3-cyclohexadiene and methylene bis-urethane to give 2-carbethoxy-2-azabocyclo[2.2.2]oct-5-ene. Ozonolysis of this material followed by reaction with excess benzylamine hydrochloride and reduction with sodium cyanoborohydride yielded 3-benzyl-6- carbethoxy-3,6-diazabicyclo[3.2.2]nonane. This methodology is also described by Fox et al. in EP 0297858. Van den Branden et al. in Tetrahedron 48 (44): 9753 (1992) describes an alternative pathway into the diazabicyclo[3.2.2]nonane system through an intramolecular substitution reaction, exemplified by the reaction of cis-N- ((1 -benzyl-5-(p-toluenesulfonyloxymethyl)-2-piperidinyl)methyl)-4-fluorobenzamide with potassium iodide and sodium hydride in THF to give 6-benzyl-3-(4-fluorobenzoyl)- 3,6-diazabicyclo[3.2.2.]nonane. Coleman et al. in WO 08008517 describes the synthesis of compounds containing the 3,6-diazabicyclo[3.2.2]nonane skeleton. Thus, the reaction of p-anisidine, 2-cyclohexenone, and formaldehyde in the presence of L- proline yielded (1 S,4S)-2-(4-methoxyphenyl)-2-azabicyclo[2.2.2]octan-5-one.
Reaction of this compound with sodium azide in concentrated sulfuric acid resulted in the production of two ring expansion products, one of which was (1S,5S)-6-(4- methoxyphenyl)-3,6-diazabicyclo[3.2.2]nonan-2-one, thus providing a route to the single enantiomers of various 3,6-diazabicyclo[3.2.2]nonane derivatives. Any of these synthetic schemes could be modified using standard synthetic techniques, known to those skilled in the art, to give a route to the compounds of the present invention.
For compounds of the present invention, the 3,6-diazabicyclo[3.2.2]nonane scaffold was prepared as illustrated in Scheme 1. Pyridine-2,5-dicarboxylic acid (1), was converted to the methyl ester and hydrogenated over platinum oxide to yield compound 2. Protection of the amine moiety with with a t-butoxycarbonyl (Boc) protecting group, followed by hydride reduction of both esters and subsequent oxidative cleavage of the double bond, yields dialdehyde 3. Reaction of this compound with benzylamine in the presence of sodium triacetoxyborohydride yields 3- benzyl-e^tert-butoxycarbonyO-S.e-diazabicycloP^^lnonane (4) (Bn = benzyl).
Figure imgf000016_0001
Figure imgf000016_0002
Scheme 1
Compound 4 can be selectively deprotected to yield 6-(tert- butoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane (5, Scheme 2), which can be subsequently used for the synthesis of 3-substituted 3,6- diazabicyclo[3.2.2]nonanes . Compound 5 can also be treated sequentially with trifluoroacetic anhydride and trifluoroacetic acid to yield trifluoroacetyl (TFA) protected compound, 6, which can be subsequently used for the synthesis of 6-substituted 3,6-diazabicyclo[3.2.2]nonanes. Such methods for installation and removal of the benzyl, tert-butoxycarbonyl, and trifluoroacetyl amine protecting groups which are well known by those skilled in the art and are described in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, New York (1999), herein incorporated by reference for such synthetic teaching.
Figure imgf000016_0003
4 5 6
Scheme 2
Amide compounds of the present invention can be prepared via the coupling of mono-protected diazabicycle (5 or 6) with a suitably functionalized acid chloride or other reactive carboxylic acid derivative. Such a compound may be available commercially or prepared by treatment of the appropriate carboxylic acid with, among other reagents, thionyl chloride or oxalyl chloride. After amide formation, removal of the protecting group will afford the compounds of the present invention. Similarly, carbamate and urea compounds of the present invention can be prepared by the coupling 5 or 6 with a suitably functionalized chloroformate or isocyanate derivative, respectively. Other compounds of the present invention can be synthesized by alkylation of the remaining basic (and nucleophilic) nitrogen with an activated alkyl compound such as an alkyl halide or alkyl sulfate.
Those skilled in the art of organic synthesis will appreciate that there exist multiple means of producing compounds of the present invention which are labeled with a radioisotope appropriate to various diagnostic uses. Thus, condensation of a 11C- or 18F-labeled carboxylic acid with either compound 5 or compound 6 followed by removal of the protecting group as described above will produce a compound suitable for use in positron emission tomography. Further derivatization of this compound is possible as described above by alkylation of the remaining basic nitrogen with an activated alkyl compound.
III. Pharmaceutical Compositions
The pharmaceutical compositions of the present invention include the salts described herein, in the pure state or in the form of a composition in which the compounds are combined with any other pharmaceutically compatible product, which can be inert or physiologically active. The resulting pharmaceutical compositions can be used to prevent a condition or disorder in a subject susceptible to such a condition or disorder, and/or to treat a subject suffering from the condition or disorder. The pharmaceutical compositions described herein include one or more compounds of Formula I and/or pharmaceutically acceptable salts thereof.
The manner in which the compounds are administered can vary. The compositions are preferably administered orally (e.g., in liquid form within a solvent such as an aqueous or non-aqueous liquid, or within a solid carrier). Preferred compositions for oral administration include pills, tablets, capsules, caplets, syrups, and solutions, including hard gelatin capsules and time-release capsules. Standard excipients include binders, fillers, colorants, solubilizers and the like. Compositions can be formulated in unit dose form, or in multiple or subunit doses. Preferred compositions are in liquid or semisolid form. Compositions including a liquid pharmaceutically inert carrier such as water or other pharmaceutically compatible liquids or semisolids can be used. The use of such liquids and semisolids is well known to those of skill in the art. The compositions can also be administered via injection, i.e., intravenously, intramuscularly, subcutaneously, intraperitoneally, intraarterially, intrathecally; and intracerebroventricularly. Intravenous administration is the preferred method of injection. Suitable carriers for injection are well known to those of skill in the art and include 5% dextrose solutions, saline, and phosphate- buffered saline. The compounds can also be administered as an infusion or injection (e.g., as a suspension or as an emulsion in a pharmaceutically acceptable liquid or mixture of liquids). The formulations can also be administered using other means, for example, rectal administration. Formulations useful for rectal administration, such as suppositories, are well known to those of skill in the art. The compounds can also be administered by inhalation (e.g., in the form of an aerosol either nasally or using delivery articles of the type set forth in U.S. Patent No. 4,922,901 to Brooks et al., the disclosure of which is incorporated herein in its entirety); topically (e.g., in lotion form); transdermal^ (e.g., using a transdermal patch) or iontophoretically; or by sublingual or buccal administration. Although it is possible to administer the compounds in the form of a bulk active chemical, it is preferred to present each compound in the form of a pharmaceutical composition or formulation for efficient and effective administration.
Exemplary methods for administering such compounds will be apparent to the skilled artisan. The usefulness of these formulations can depend on the particular composition used and the particular subject receiving the treatment. These formulations can contain a liquid carrier that can be oily, aqueous, emulsified or contain certain solvents suitable to the mode of administration. The compositions can be administered intermittently or at a gradual, continuous, constant or controlled rate to a warm-blooded animal (e.g., a mammal such as a mouse, rat, cat, rabbit, dog, pig, cow, or monkey), but advantageously are administered to a human being. In addition, the time of day and the number of times per day that the pharmaceutical formulation is administered can vary. Other suitable methods for administering the compounds of the present invention are described in U.S. Patent No. 5,604,231 to Smith et al., the contents of which are hereby incorporated by reference.
In an embodiment of the present invention and as will be appreciated by those skilled in the art, the compound of the present invention may be administered in combination with other therapeutic compounds. For example, a compound of this invention can be used in combination with other NNR ligands (such as varenicline), antioxidants (such as free radical scavenging agents), antibacterial agents (such as penicillin antibiotics), antiviral agents (such as nucleoside analogs, like zidovudine and acyclovir), anticoagulants (such as warfarin), anti-inflammatory agents (such as NSAIDs), anti-pyretics, analgesics, anesthetics (such as used in surgery), acetylcholinesterase inhibitors (such as donepezil and galantamine), antipsychotics (such as haloperidol, clozapine, olanzapine, and quetiapine), immuno-suppressants (such as cyclosporin and methotrexate), neuroprotective agents, steroids (such as steroid hormones), corticosteroids (such as dexamethasone, predisone, and hydrocortisone), vitamins, minerals, nutraceuticals, anti-depressants (such as imipramine, fluoxetine, paroxetine, escitalopram, sertraline, venlafaxine, and duloxetine), anxiolytics (such as alprazolam and buspirone), anticonvulsants (such as phenytoin and gabapentin), vasodilators (such as prazosin and sildenafil), mood stabilizers (such as valproate and aripiprazole), anti-cancer drugs (such as antiproliferatives), antihypertensive agents (such as atenolol, clonidine, amlopidine, verapamil, and olmesartan), laxatives, stool softeners, diuretics (such as furosemide), anti-spasmotics (such as dicyclomine), anti-dyskinetic agents, and anti-ulcer medications (such as esomeprazole). The compounds of the present invention may be employed alone or in combination with other therapeutic agents, including other compounds of the present invention. Such a combination of pharmaceutically active agents may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order. The amounts of the compounds or agents and the relative timings of administration will be selected in order to achieve the desired therapeutic effect. The administration in combination of a compound of the formulae of the present invention including salts or solvates thereof with other treatment agents may be in combination by administration concomitantly in: (1) a unitary pharmaceutical composition including both compounds; or (2) separate pharmaceutical compositions each including one of the compounds. Alternatively, the combination may be administered separately in a sequential manner wherein one treatment agent is administered first and the other second or vice versa. Such sequential administration may be close in time or remote in time. The compounds of the present invention may be used in the treatment of a variety of disorders and conditions and, as such, the compounds of the present invention may be used in combination with a variety of other suitable therapeutic agents useful in the treatment or prophylaxis of those disorders or conditions.
The following examples are provided to illustrate the present invention, and should not be construed as limiting thereof. In these examples, all parts and percentages are by weight, unless otherwise noted.
The appropriate dose of the compound is that amount effective to prevent occurrence of the symptoms of the disorder or to treat some symptoms of the disorder from which the patient suffers. By "effective amount", "therapeutic amount" or "effective dose" is meant that amount sufficient to elicit the desired pharmacological or therapeutic effects, thus resulting in effective prevention or treatment of the disorder.
When treating a CNS disorder, an effective amount of compound is an amount sufficient to pass across the blood-brain barrier of the subject, to bind to relevant receptor sites in the brain of the subject and to modulate the activity of relevant NNR subtypes (e.g., provide neurotransmitter secretion, thus resulting in effective prevention or treatment of the disorder). Prevention of the disorder is manifested by delaying the onset of the symptoms of the disorder. Treatment of the disorder is manifested by a decrease in the symptoms associated with the disorder or an amelioration of the recurrence of the symptoms of the disorder. Preferably, the effective amount is sufficient to obtain the desired result, but insufficient to cause appreciable side effects.
The effective dose can vary, depending upon factors such as the condition of the patient, the severity of the symptoms of the disorder, and the manner in which the pharmaceutical composition is administered. For human patients, the effective dose of typical compounds generally requires administering the compound in an amount sufficient to modulate the activity of relevant NNRs, but the amount should be insufficient to induce effects on skeletal muscles and ganglia to any significant degree. The effective dose of compounds will of course differ from patient to patient, but in general includes amounts starting where CNS effects or other desired therapeutic effects occur but below the amount where muscular effects are observed.
The compounds described herein, when employed in effective amounts in accordance with the methods described herein, can provide some degree of prevention of the progression of, ameliorate symptoms of, and ameliorate to some degree of the recurrence of CNS or other disorders. The effective amounts of those compounds are typically below the threshold concentration required to elicit any appreciable side effects, for example those effects relating to skeletal muscle or ganglia. The compounds can be administered in a therapeutic window in which certain CNS and other disorders are treated and certain side effects are avoided. Ideally, the effective dose of the compounds described herein is sufficient to provide the desired effects upon the disorder but is insufficient (i.e., is not at a high enough level) to provide undesirable side effects. Preferably, the compounds are administered at a dosage effective for treating the CNS or other disorders but less than 1/5, and often less than 1/10, the amount required to elicit certain side effects to any significant degree. Most preferably, effective doses are at very low concentrations, where maximal effects are observed to occur, with a minimum of side effects. An effective dose of such compounds may require administering the compound in an amount of less than 5 mg/kg of patient weight. The compounds of the present invention may be administered in an amount from less than about 1 mg/kg patent weight and usually less than about 100 μg/kg of patient weight, but may be between about 10 μg to less than 100 μg/kg of patient weight. The foregoing doses typically represent that amount administered as a single dose, or as one or more doses administered over a 24-hour period.
For human patients, an effective dose of typical compounds generally requires administering the compound in an amount of at least about 1 , often at least about 10, and frequently at least about 100 mg/ 24 hr/ patient. For human patients, an effective dose of typical compounds requires administering the compound which generally does not exceed about 500, often does not exceed about 400, and frequently does not exceed about 300 mg/ 24 hr/ patient. In addition, the compositions may be advantageously administered at an effective dose such that the concentration of the compound within the plasma of the patient normally does not exceed 50 ng/mL, often does not exceed 30 ng/mL, and frequently does not exceed 10 ng/mL.
IV. Method of Using Pharmaceutical Compositions
The compounds of the present invention can be used for the prevention or treatment of various conditions or disorders for which other types of nicotinic compounds have been proposed or are shown to be useful as therapeutics, such as CNS disorders, inflammation, inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders, addictions, obesity or other disorders described in further detail herein. This compound can also be used as a diagnostic agent in receptor binding studies (in vitro and in vivo). Such therapeutic and other teachings are described, for example, in references previously listed herein, including Williams et al., Drug News Perspec. 7(4): 205 (1994), Arneric et al., CNS Drug Rev. 1(1): 1-26 (1995), Arneric et al., Exp. Opin. Invest. Drugs 5(1): 79-100 (1996), Bencherif et al., J. Pharmacol. Exp. Ther. 279: 1413 (1996), Lippiello et al., J. Pharmacol. Exp. Ther. 279: 1422 (1996), Damaj et al., J. Pharmacol. Exp. Ther. 291: 390 (1999); Chiari et al., Anesthesiology 91 : 1447 (1999), Lavand'homme and Eisenbach, Anesthesiology 91: 1455 (1999), Holladay et al., J. Med. Chem. 40(28): 4169-94 (1997), Bannon et al., Science 279: 77 (1998), PCT WO 94/08992, PCT WO
96/31475, PCT WO 96/40682, and U.S. Patent Nos. 5,583,140 to Bencherif et al., 5,597,919 to Dull et al., 5,604,231 to Smith et al. and 5,852,041 to Cosford et al.
CNS Disorders The compounds and their pharmaceutical compositions are useful in the treatment or prevention of a variety of CNS disorders, including neurodegenerative disorders, neuropsychiatric disorders, neurologic disorders, and addictions. The compounds and their pharmaceutical compositions can be used to treat or prevent cognitive deficits and dysfunctions, age-related and otherwise; attentional disorders and dementias, including those due to infectious agents or metabolic disturbances; to provide neuroprotection; to treat convulsions and multiple cerebral infarcts; to treat mood disorders, compulsions and addictive behaviors; to provide analgesia; to control inflammation, such as mediated by cytokines and nuclear factor kappa B; to treat inflammatory disorders; to provide pain relief; and to treat infections, as anti-infectious agents for treating bacterial, fungal, and viral infections. Among the disorders, diseases and conditions that the compounds and pharmaceutical compositions of the present invention can be used to treat or prevent are: age-associated memory impairment (AAMI), mild cognitive impairment (MCI), age-related cognitive decline (ARCD), pre-senile dementia, early onset Alzheimer's disease, senile dementia, dementia of the
Alzheimer's type, Alzheimer's disease, cognitive impairment no dementia (CIND), Lewy body dementia, HIV-dementia, AIDS dementia complex, vascular dementia, Down syndrome, head trauma, traumatic brain injury (TBI), dementia pugilistica, Creutzfeld-Jacob Disease and prion diseases, stroke, central ischemia, peripheral ischemia, attention deficit disorder, attention deficit hyperactivity disorder, dyslexia, schizophrenia, schizophreniform disorder, schizoaffective disorder, cognitive dysfunction in schizophrenia, cognitive deficits in schizophrenia, Parkinsonism including Parkinson's disease, postencephalitic parkinsonism, parkinsonism-dementia of Gaum, frontotemporal dementia Parkinson's Type (FTDP), Pick's disease, Niemann-Pick's Disease, Huntington's Disease, Huntington's chorea, tardive dyskinesia, hyperkinesia, progressive supranuclear palsy, progressive supranuclear paresis, restless leg syndrome, Creutzfeld-Jakob disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS), motor neuron diseases (MND), multiple system atrophy (MSA), corticobasal degeneration, Guillain-Barre Syndrome (GBS), and chronic inflammatory demyelinating polyneuropathy (CIDP), epilepsy, autosomal dominant nocturnal frontal lobe epilepsy, mania, anxiety, depression, premenstrual dysphoria, panic disorders, bulimia, anorexia, narcolepsy, excessive daytime sleepiness, bipolar disorders, generalized anxiety disorder, obsessive compulsive disorder, rage outbursts, conduct disorder, oppositional defiant disorder, Tourette's syndrome, autism, drug and alcohol addiction, tobacco addiction, obesity, cachexia, psoriasis, lupus, acute cholangitis, aphthous stomatitis, ulcers, asthma, ulcerative colitis, inflammatory bowel disease, Crohn's disease, irritable bowel syndrome, spastic dystonia, diarrhea, constipation, pouchitis, viral pneumonitis, arthritis, including, rheumatoid arthritis and osteoarthritis, endotoxaemia, sepsis, atherosclerosis, idiopathic pulmonary fibrosis, acute pain, chronic pain, neuropathies, urinary incontinence, diabetes, sexual dysfunction, neoplasias, and preeclampsia.
Cognitive impairments or dysfunctions may be associated with psychiatric disorders or conditions, such as schizophrenia and other psychotic disorders, including but not limited to psychotic disorder, schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic disorder, and psychotic disorders due to a general medical conditions, dementias and other cognitive disorders, including but not limited to mild cognitive impairment, pre-senile dementia, Alzheimer's disease, senile dementia, dementia of the Alzheimer's type, age-related memory impairment, Lewy body dementia, vascular dementia, AIDS dementia complex, dyslexia, Parkinsonism including Parkinson's disease, cognitive impairment and dementia of Parkinson's Disease, cognitive impairment of multiple sclerosis, cognitive impairment caused by traumatic brain injury, dementias due to other general medical conditions, anxiety disorders, including but not limited to panic disorder without agoraphobia, panic disorder with agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia, obsessive-compulsive disorder, post-traumatic stress disorder, acute stress disorder, generalized anxiety disorder and generalized anxiety disorder due to a general medical condition, mood disorders, including but not limited to major depressive disorder, dysthymic disorder, bipolar depression, bipolar mania, bipolar I disorder, depression associated with manic, depressive or mixed episodes, bipolar Il disorder, cyclothymic disorder, and mood disorders due to general medical conditions, sleep disorders, including but not limited to dyssomnia disorders, primary insomnia, primary hypersomnia, narcolepsy, parasomnia disorders, nightmare disorder, sleep terror disorder and sleepwalking disorder, mental retardation, learning disorders, motor skills disorders, communication disorders, pervasive developmental disorders, attention-deficit and disruptive behavior disorders, attention deficit disorder, attention deficit hyperactivity disorder, feeding and eating disorders of infancy, childhood, or adults, tic disorders, elimination disorders, substance-related disorders, including but not limited to substance dependence, substance abuse, substance intoxication, substance withdrawal, alcohol-related disorders, amphetamine or amphetamine-like-related disorders, caffeine-related disorders, cannabis-related disorders, cocaine-related disorders, hallucinogen-related disorders, inhalant- related disorders, nicotine-related disorders, opioid-related disorders, phencyclidine or phencyclidine-like-related disorders, and sedative-, hypnotic- or anxiolytic-related disorders, personality disorders, including but not limited to obsessive-compulsive personality disorder and impulse-control disorders.
Cognitive performance may be assessed with a validated cognitive scale, such as, for example, the cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog). One measure of the effectiveness of the compounds of the present invention in improving cognition may include measuring a patient's degree of change according to such a scale.
Regarding compulsions and addictive behaviors, the compounds of the present invention may be used as a therapy for nicotine addiction and for other brain-reward disorders, such as substance abuse including alcohol addiction, illicit and prescription drug addiction, eating disorders, including obesity, and behavioral addictions, such as gambling, or other similar behavioral manifestations of addiction.
The above conditions and disorders are discussed in further detail, for example, in the American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision, Washington, DC,
American Psychiatric Association, 2000. This Manual may also be referred to for greater detail on the symptoms and diagnostic features associated with substance use, abuse, and dependence.
Preferably, the treatment or prevention of diseases, disorders and conditions occurs without appreciable adverse side effects, including, for example, significant increases in blood pressure and heart rate, significant negative effects upon the gastro-intestinal tract, and significant effects upon skeletal muscle.
The compounds of the present invention, when employed in effective amounts, are believed to modulate the activity of the α4β2 and α7 NNRs without appreciable interaction with the nicotinic subtypes that characterize the human ganglia, as demonstrated by a lack of the ability to elicit nicotinic function in adrenal chromaffin tissue, or skeletal muscle, further demonstrated by a lack of the ability to elicit nicotinic function in cell preparations expressing muscle-type nicotinic receptors. Thus, these compounds are believed capable of treating or preventing diseases, disorders and conditions without eliciting significant side effects associated activity at ganglionic and neuromuscular sites. Thus, administration of the compounds is believed to provide a therapeutic window in which treatment of certain diseases, disorders and conditions is provided, and certain side effects are avoided. That is, an effective dose of the compound is believed sufficient to provide the desired effects upon the disease, disorder or condition, but is believed insufficient, namely is not at a high enough level, to provide undesirable side effects.
Thus, the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, for use in therapy, such as a therapy described above.
In yet another aspect the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a CNS disorder, such as a disorder, disease or condition described hereinabove.
Inflammation
The nervous system, primarily through the vagus nerve, is known to regulate the magnitude of the innate immune response by inhibiting the release of macrophage tumor necrosis factor (TNF). This physiological mechanism is known as the "cholinergic anti-inflammatory pathway" (see, for example, Tracey, "The Inflammatory Reflex," Nature 420: 853-9 (2002)). Excessive inflammation and tumor necrosis factor synthesis cause morbidity and even mortality in a variety of diseases. These diseases include, but are not limited to, endotoxemia, rheumatoid arthritis, osteoarthritis, psoriasis, asthma, atherosclerosis, idiopathic pulmonary fibrosis, and inflammatory bowel disease. Inflammatory conditions that can be treated or prevented by administering the compounds described herein include, but are not limited to, chronic and acute inflammation, psoriasis, endotoxemia, gout, acute pseudogout, acute gouty arthritis, arthritis, rheumatoid arthritis, osteoarthritis, allograft rejection, chronic transplant rejection, asthma, atherosclerosis, mononuclear-phagocyte dependent lung injury, idiopathic pulmonary fibrosis, atopic dermatitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, acute chest syndrome in sickle cell disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis, acute cholangitis, aphteous stomatitis, pouchitis, glomerulonephritis, lupus nephritis, thrombosis, and graft vs. host reaction.
Inflammatory Response Associated with Bacterial and/or Viral Infection
Many bacterial and/or viral infections are associated with side effects brought on by the formation of toxins, and the body's natural response to the bacteria or virus and/or the toxins. As discussed above, the body's response to infection often involves generating a significant amount of TNF and/or other cytokines. The over-expression of these cytokines can result in significant injury, such as septic shock (when the bacteria is sepsis), endotoxic shock, urosepsis and toxic shock syndrome.
Cytokine expression is mediated by NNRs, and can be inhibited by administering agonists or partial agonists of these receptors. Those compounds described herein that are agonists or partial agonists of these receptors can therefore be used to minimize the inflammatory response associated with bacterial infection, as well as viral and fungal infections. Examples of such bacterial infections include anthrax, botulism, and sepsis. Some of these compounds may also have antimicrobial properties.
These compounds can also be used as adjunct therapy in combination with existing therapies to manage bacterial, viral and fungal infections, such as antibiotics, antivirals and antifungals. Antitoxins can also be used to bind to toxins produced by the infectious agents and allow the bound toxins to pass through the body without generating an inflammatory response. Examples of antitoxins are disclosed, for example, in U.S. Patent No. 6,310,043 to Bundle et al. Other agents effective against bacterial and other toxins can be effective and their therapeutic effect can be complemented by co-administration with the compounds described herein.
Pain
The compounds can be administered to treat and/or prevent pain, including acute, neurologic, inflammatory, neuropathic and chronic pain. The compounds can be used in conjunction with opiates to minimize the likelihood of opiate addiction (e.g., morphine sparing therapy). The analgesic activity of compounds described herein can be demonstrated in models of persistent inflammatory pain and of neuropathic pain, performed as described in U.S. Published Patent Application No. 20010056084 A1 (Allgeier et at.) (e.g., mechanical hyperalgesia in the complete Freund's adjuvant rat model of inflammatory pain and mechanical hyperalgesia in the mouse partial sciatic nerve ligation model of neuropathic pain).
The analgesic effect is suitable for treating pain of various genesis or etiology, in particular in treating inflammatory pain and associated hyperalgesia, neuropathic pain and associated hyperalgesia, chronic pain (e.g., severe chronic pain, post-operative pain and pain associated with various conditions including cancer, angina, renal or biliary colic, menstruation, migraine, and gout).
Inflammatory pain may be of diverse genesis, including arthritis and rheumatoid disease, teno-synovitis and vasculitis. Neuropathic pain includes trigeminal or herpetic neuralgia, diabetic neuropathy pain, causalgia, low back pain and deafferentation syndromes such as brachial plexus avulsion.
Neovascularization
The α7 NNR is associated with neovascularization. Inhibition of neovascularization, for example, by administering antagonists (or at certain dosages, partial agonists) of the α7 NNR can treat or prevent conditions characterized by undesirable neovascularization or angiogenesis. Such conditions can include those characterized by inflammatory angiogenesis and/or ischemia-induced angiogenesis. Neovascularization associated with tumor growth can also be inhibited by administering those compounds described herein that function as antagonists or partial agonists of α7 NNR. Specific antagonism of α7 NNR-specific activity reduces the angiogenic response to inflammation, ischemia, and neoplasia. Guidance regarding appropriate animal model systems for evaluating the compounds described herein can be found, for example, in Heeschen, C. et a/., "A novel angiogenic pathway mediated by non-neuronal nicotinic acetylcholine receptors," J. CHn. Invest. 110(4):527-36 (2002). Representative tumor types that can be treated using the compounds described herein include NSCLC, ovarian cancer, pancreatic cancer, breast carcinoma, colon carcinoma, rectum carcinoma, lung carcinoma, oropharynx carcinoma, hypopharynx carcinoma, esophagus carcinoma, stomach carcinoma, pancreas carcinoma, liver carcinoma, gallbladder carcinoma, bile duct carcinoma, small intestine carcinoma, urinary tract carcinoma, kidney carcinoma, bladder carcinoma, urothelium carcinoma, female genital tract carcinoma, cervix carcinoma, uterus carcinoma, ovarian carcinoma, choriocarcinoma, gestational trophoblastic disease, male genital tract carcinoma, prostate carcinoma, seminal vesicles carcinoma, testes carcinoma, germ cell tumors, endocrine gland carcinoma, thyroid carcinoma, adrenal carcinoma, pituitary gland carcinoma, skin carcinoma, hemangiomas, melanomas, sarcomas, bone and soft tissue sarcoma, Kaposi's sarcoma, tumors of the brain, tumors of the nerves, tumors of the eyes, tumors of the meninges, astrocytomas, gliomas, glioblastomas, retinoblastomas, neuromas, neuroblastomas, Schwannomas, meningiomas, solid tumors arising from hematopoietic malignancies (such as leukemias, chloromas, plasmacytomas and the plaques and tumors of mycosis fungoides and cutaneous T-cell lymphoma/leukemia), and solid tumors arising from lymphomas.
The compounds can also be administered in conjunction with other forms of anti-cancer treatment, including co-administration with antineoplastic antitumor agents such as cis-platin, adriamycin, daunomycin, and the like, and/or anti-VEGF (vascular endothelial growth factor) agents, as such are known in the art.
The compounds can be administered in such a manner that they are targeted to the tumor site. For example, the compounds can be administered in microspheres, microparticles or liposomes conjugated to various antibodies that direct the microparticles to the tumor. Additionally, the compounds can be present in microspheres, microparticles or liposomes that are appropriately sized to pass through the arteries and veins, but lodge in capillary beds surrounding tumors and administer the compounds locally to the tumor. Such drug delivery devices are known in the art.
Other Disorders In addition to treating CNS disorders, inflammation, and neovascularization, and pain, the compounds of the present invention can be also used to prevent or treat certain other conditions, diseases, and disorders in which NNRs play a role. Examples include autoimmune disorders such as Lupus, disorders associated with cytokine release, cachexia secondary to infection (e.g., as occurs in AIDS, AIDS related complex and neoplasia), obesity, pemphitis, urinary incontinence, retinal diseases, infenctious diseases, myasthenia, Eaton- Lambert syndrome, hypertension, preeclampsia, osteoporosis, vasoconstriction, vasodilatation, cardiac arrhythmias, type I diabetes, bulimia, anorexia as well as those indications set forth in published PCT application WO 98/25619. The compounds of this invention can also be administered to treat convulsions such as those that are symptomatic of epilepsy, and to treat conditions such as syphillis and Creutzfeld-Jakob disease.
Diagnostic Uses
The compounds can be used in diagnostic compositions, such as probes, particularly when they are modified to include appropriate labels. The probes can be used, for example, to determine the relative number and/or function of specific receptors, particularly the α4β2 and α7 receptor subtypes. For this purpose the compounds of the present invention most preferably are labeled with a radioactive isotopic moiety such as 11C, 18F, 76Br, 123I or 125I.
The administered compounds can be detected using known detection methods appropriate for the label used. Examples of detection methods include position emission topography (PET) and single-photon emission computed tomography (SPECT). The radiolabels described above are useful in PET (e.g., 11C, 18F or 76Br) and SPECT (e.g., 123I) imaging, with half-lives of about 20.4 minutes for 11C, about 109 minutes for 18F, about 13 hours for 123I, and about 16 hours for 76Br. A high specific activity is desired to visualize the selected receptor subtypes at non-saturating concentrations. The administered doses typically are below the toxic range and provide high contrast images. The compounds are expected to be capable of administration in non-toxic levels. Determination of dose is carried out in a manner known to one skilled in the art of radiolabel imaging. See, for example, U.S. Patent No. 5,969,144 to London et al.
The compounds can be administered using known techniques. See, for example, U.S. Patent No. 5,969,144 to London et al., as noted. The compounds can be administered in formulation compositions that incorporate other ingredients, such as those types of ingredients that are useful in formulating a diagnostic composition. Compounds useful in accordance with carrying out the present invention most preferably are employed in forms of high purity. See, U.S. Patent No. 5,853,696 to Elmalch etal. After the compounds are administered to a subject (e.g., a human subject), the presence of that compound within the subject can be imaged and quantified by appropriate techniques in order to indicate the presence, quantity, and functionality of selected NNR subtypes. In addition to humans, the compounds can also be administered to animals, such as mice, rats, dogs, and monkeys. SPECT and PET imaging can be carried out using any appropriate technique and apparatus. See Villemagne et al., In: Arneric et al. (Eds.) Neuronal Nicotinic Receptors: Pharmacology and Therapeutic Opportunities, 235-250 (1998) and U.S. Patent No. 5,853,696 to Elmalch et al., each herein incporated by reference, for a disclosure of representative imaging techniques. The radiolabeled compounds bind with high affinity to selective NNR subtypes (e.g., α4β2, α7) and preferably exhibit negligible non-specific binding to other nicotinic cholinergic receptor subtypes (e.g., those receptor subtypes associated with muscle and ganglia). As such, the compounds can be used as agents for noninvasive imaging of nicotinic cholinergic receptor subtypes within the body of a subject, particularly within the brain for diagnosis associated with a variety of CNS diseases and disorders.
In one aspect, the diagnostic compositions can be used in a method to diagnose disease in a subject, such as a human patient. The method involves administering to that patient a detectably labeled compound as described herein, and detecting the binding of that compound to selected NNR subtypes (e.g., α4β2 and α7 receptor subtypes). Those skilled in the art of using diagnostic tools, such as PET and SPECT, can use the radiolabeled compounds described herein to diagnose a wide variety of conditions and disorders, including conditions and disorders associated with dysfunction of the central and autonomic nervous systems. Such disorders include a wide variety of CNS diseases and disorders, including Alzheimer's disease, Parkinson's disease, and schizophrenia. These and other representative diseases and disorders that can be evaluated include those that are set forth in U.S. Patent No. 5,952,339 to Bencherif et al.
In another aspect, the diagnostic compositions can be used in a method to monitor selective nicotinic receptor subtypes of a subject, such as a human patient. The method involves administering a detectably labeled compound as described herein to that patient and detecting the binding of that compound to selected nicotinic receptor subtypes namely, the α4β2 and α7 receptor subtypes.
Receptor Binding The compounds of this invention can be used as reference ligands in binding assays for compounds which bind to NNR subtypes, particularly the α4β2 and α7 receptor subtypes. For this purpose the compounds of this invention are preferably labeled with a radioactive isotopic moiety such as 3H, or 14C. Examples of such binding assays are described in detail below.
Synthetic Examples
The following examples are provided to illustrate the present invention, and should not be construed as limiting thereof. In these examples, all parts and percentages are by weight, unless otherwise noted.
Example 1: Synthesis of 1-(tert-butoxycarbonyl)-2,5- bis(methoxycarbonyl)piperidine
To a solution of pyridine-2,5-dicarboxylic acid (35 g, 0.21 mol) in anhydrous methanol (1L) was added thionyl chloride (35 mL, 0.48 mol) at 0 0C over 15 min. The cooling source was removed, and the solution was stirred at room temperature for 18 h. The solvent was evaporated by rotary evaporation, and the crude product was dissolved in methanol (300 mL) and subjected to hydrogenation over platinum dioxide (1.0 g) at 55 psi for 16 h. The reaction mixture was filtered, and the filtrate was concentrated by rotary evaporation. The resulting crude product was dissolved in water (300 mL), and the pH was adjusted to pH 8 with solid sodium bicarbonate. A solution of di-tert-butyldicarbonate (50 g) in THF (200 mL) was added, and the mixture was stirred at room temperature for 1 h. The reaction mixture was extracted with ethyl acetate (500 mL). The organic layer was dried over anhydrous sodium sulfate, concentrated and purified by silica gel column chromatography (hexane/ethyl acetate) to give 1-(tert-butoxycarbonyl)-2,5-bis(methoxycarbonyl)piperidine (47 g, 74% yield). MS (m/z): 302 (M+1), 246 (M+1-56).
Example 2: Synthesis of 1-(tert-butoxycarbonyl)-2,5- bis(hydroxymethyl)piperidine To a solution of 1 -(tert-butoxycarbonyl)-2,5-bis(methoxycarbonyl)piperidine (46 g, 0.15 mol) in anhydrous THF (500 mL) at 0 0C was added lithium aluminum hydride (700 ml. of 1 M solution in THF, 0.70 mol) over 10 minutes. The reaction was stirred at 0 0C for 30 min. The reaction was carefully quenched by addition of powdered sodium sulfate decahydrate (100 g) and the mixture was stirred for 1 h at ambient temperature. The mixture was filtered, and the filtrate was concentrated by rotary evaporation and dried in vacuo to obtain 1-(tert-butoxycarbonyl)-2,5- bis(hydroxymethyl)piperidine (23.7 g; 64% yield) as an oil. MS (m/z): 246 (M+1), 190 (M+1-56).
Example 3: 1-(tert-butoxycarbonyl)piperidine-2,5-dialdehyde To a mixture of 1 -(tert-butoxycarbonyl)-2,5-bis(hydroxymethyl)piperidine (23 g,
94 mmol) and powdered Molecular Sieves (4 A, 60 g) in dichloromethane (600 mL) was added pyridinium chlorochromate (51 g, 235 mmol) at 0 0C. The cooling source was removed and the solution was stirred at ambient temperature for 3 h. The reaction mixture was filtered through diatomaceous earth, and the filtrate was concentrated by rotary evaporation and immediately purified by silica gel chromatography (1 :1 hexane/ethyl acetate) to give 1-(tert-butoxycarbonyl)piperidine- 2,5-dialdehyde (8.26 g; 36% yield) as an oil.
Example 4: 6-(tert-butoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane To a solution of 1 -(tert-butoxycarbonyl)piperidine-2,5-dialdehyde (7.1O g, 29.5 mmol) and benzylamine (3.21 mL, 29.5 mmol) in 4:1 dichloromethane:methanol (250 mL) was added sodium triaceoxyborohydride (31.5 g, 148 mmol). The solution was stirred for 6 h at ambient temperature. The solvent was evaporated by rotary evaporation, diluted with ethyl acetate (200 mL) and washed with saturated aqueous sodium bicarbonate (2 x 100 mL). The organic layer was dried with anhydrous sodium sulfate, concentrated by rotary evaporation and purified by silica gel chromatography (hexane/ethyl acetate) to give 3-benzyl-6-(tert-butoxycarbonyl)-3,6- diazabicyclo[3.2.2]nonane (3.3 g), which was dissolved in methanol (50 mL) and hydrogenated over 10% palladium on carbon (0.5 g) at 50 psi. The reaction was filtered and filtrate was concentrated to give 6-(tert-butoxycarbonyl)-3,6- diazabicyclo[3.2.2]nonane (2.3 g; 34% yield). 1H NMR (CDCI3, 300 MHz): δ 4.28^.22 and 4.12-4.06 (m, 1 H), 3.56-3.43 (m, 1 H), 3.29-2.83 (m, 5 H), 2.20-2.06 (m, 1 H), 1.92-1.70 (m, 4 H), 1.46 and 1.45 (s, 9 H); MS (m/z): 227 (M+1 ).
General procedure for making amides of 3,6-diaza-bicyclo[3.2.2]nonane
Certain amide derivatives (those at the 3-position) can be prepared by reaction of θ-ftert-butoxycarbonyl^e-diazabicycloβ^^Jnonane with acid chlorides and similar reagents. Certain other amide derivatives (those at the 6-position) can be made by reaction of 3-(trifluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane with acid chlorides and similar reagents. The following procedures are exemplary.
Example 5: 3-(furan-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane
To a solution of θ-^ert-butoxycarbonyO-S.e-diazabicycloIS^^jnonane (0.070 g, 0.31 mmol) in acetonitrile (5 mL) was added triethylamine (0.17 ml_, 1.2 mmol) and 2-furoyl chloride (0.050 g, 0.38 mmol). The solution was stirred at ambient temperature for 1 h. The solvent was evaporated, and the crude amide was purified by reverse phase HPLC (acetonitrile/0.05% aqueous trifluoroacetic acid), to obtain 6- (tert-butoxycarbonyl)-3-(furan-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane as an oil. This material was dissolved in dichloromethane (3 mL), combined with trifluoroacetic acid (2 mL), and stirred at ambient temperature for 1 h. The solvent was evaporated and the residue purified by HPLC (acetonitrile/0.05% aqueous trifluoroacetic acid) to obtain 0.023 g (32% yield) of 3-(furan-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane as an oil. 1H NMR (CD3OD, 300 MHz): δ 7.71 (dd, J = 1.83 and 0.85 Hz, 1 H), 7.10 (dd, J = 3.54 and 0.73 Hz, 1 H), 6.61 (dd, J = 3.54 and 1.83 Hz, 1 H), 4.85^.75 (m, 1 H), 4.63-4.57 (m, 1 H), 3.94-3.88 (m, 1 H), 3.65-3.42 (m, 4 H), 2.46-2.39 (m, 1 H), 2.11- 1.78 (m, 4 H) ; MS (m/z): 221 (M+1 ).
Example 6: 3-(trifluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane
To a solution of eKtert-butoxycarbonyO-S.β-diazabicycloP^^Jnonane (4.00 g, 17.7 mmol) and triethylamine (3.72 mL, 26.6 mmol, 1.5 eq.) in dichloromethane (60 mL) at -78 0C was slowly added trifluoroacetic anhydride (3.7 mL, 26.55 mmol). The solution was allowed to reach ambient temperature over 3 h. The reaction solution was diluted with dichloromethane (100 mL) and washed with water (100 mL). The organic layer was dried over sodium sulfate and concentrated. The residue was dissolved in dichloromethane (50 mL) and trifluoroacetic acid (50 mL) was added. The solution was stirred for 1 h at ambient temperature. The solution was concentrated to obtain 3-(trifluoroacetyl)- 3,6-diazabicyclo[3.2.2]nonane as the trifluoroacetate salt. The crude product was dissolved in dichloromethane (50 mL) and used without further purification. Example 7: 6-(propanoyl)-3,6-diazabicyclo[3.2.2]nonane
To a solution of 3-(trifluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane in dichloromethane from Example 6 (2 mL) was added propionyl chloride (0.10 g, 1.1 mmol) followed by triethylamine (1 mL) at 0 0C. The solution was stirred for 2 h at ambient temperature. The reaction solution was washed with water (2 mL) and concentrated. The crude amide was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain S^trifluoroacetylHHpropanoyO-S.Θ-diazabicycloβ^^nonane. The relevant fractions were combined, and the solvent evaporated. The residue was dissolved in methanol (3 mL), and 2M aqueous sodium carbonate solution (1 mL) was added. The reaction mixture was stirred for 24 h at ambient temperature. The solvent was evaporated and the residue was triturated with dichloromethane (2 x 3 mL). The solvent was evaporated, and the product was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 6-(propanoyl)-3,6-diazabicyclo[3.2.2]nonane trifluoroacetate as gum (0.033 g). 1H NMR (CD3OD, 300 MHz): δ 4.81-4.77 and 4.43-4.39 (m, 1 H), 3.74-3.16 (m, 6 H), 2.57-2.54 (m, 1H), 2.52-2.37 (m, 2H), 2.12-1.82 (m, 4 H), 1.18-1.09 (m, 3 H); MS (m/z): 183 (M+1).
Example 8: 6-(N-propylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane
To a solution of S^trifluoroacetyO-S.Θ-diazabicyclofS^^jnonane in dichloromethane from Example 6 (2 mL) was added propyl isocyanate (0.91 g, 1.1 mmol) followed by triethylamine (1 mL) at 0 0C. The solution was stirred for 2 h at ambient temperature. The reaction solution was washed with water (2 mL) and concentrated. The crude amide was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 3-(trifluoroacetyl)-6-(N-propylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane. The relevant fractions were combined, and the solvent evaporated. The residue was dissolved in methanol (3 mL), and 20% aqueous potassium carbonate solution (0.5 mL) was added. The reaction mixture was stirred for 16 h at ambient temperature. The solvent was evaporated and the residue was triturated with dichloromethane (2 x 3 mL). The solvent was evaporated, and the product was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 6-(N-propylcarbamoyl)-3,6- diazabicyclo[3.2.2]nonane trifluoroacetate as white powder (0.161 g). 1H NMR (CD3OD, 300 MHz): δ 4.58-4.55 (m, 1 H), 3.3.59-3.10 (m, 8 H), 2.57-2.48 (m, 1 H), 2.06-1.84 (m, 4 H), 1.52 (sextet, J = 7.4 Hz, 2 H), 0.91 (t, J = 7.4 Hz, 3 H); MS (m/z): 212 (M+1).
Example 9: 6-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane To a solution of 3-(trifluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane in dichloromethane from Example 6 (2 mL) was added 2-fluoroethyl chloroformate (0.13 g, 1.1 mmol) followed by triethylamine (1 mL) at 0 0C. The solution was stirred for 2 h at ambient temperature. The reaction solution was washed with water (2 mL) and concentrated. The crude amide was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 3-(trifluoroacetyl)-6-(2-fluoroethoxycarbonyl)-3,6- diazabicyclo[3.2.2]nonane. The relevant fractions were combined, and the solvent evaporated. The residue was dissolved in methanol (3 mL), and 20% aqueous potassium carbonate solution (0.5 mL) was added. The reaction mixture was stirred for 16 h at ambient temperature. The solvent was evaporated and the residue was triturated with dichloromethane (2 x 3 mL). The solvent was evaporated, and the product was purified by reverse phase HPLC using acetonitrile and 0.05% aqueous trifluoroacetic acid as the mobile phase, to obtain 6-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane trifluoroacetate as gum (0.119 g). 1H NMR (CD3OD, 300 MHz): 54.68-4.65 (m, 1 H), 4.56-4.53 (m, 1 H), 4.47-4.44 (m, 1 H), 4.394.37 (m, 1 H), 4.32-4.30 (m, 1 H), 3.71-3.24, (m, 6 H), 2.56-2.45 (m, 1 H), 2.17-1.84 (m, 4 H); MS (m/z): 217 (M+1).
The above illustrated amide coupling procedures were used as a basis to make the compounds shown in Table 1. Reagents and conditions will be readily apparent to those skilled in the art. In some cases, compounds were characterized by nuclear magnetic resonance (NMR) data. In other cases, compounds were structurally characterized by LCMS.
VIII. Biological Assays
Example 10: Radioligand Binding at CNS nAChRs α4β2 nAChR Subtype
Preparation of membranes from rat cortex: Rats (female, Sprague- Dawley), weighing 150-250 g, were maintained on a 12 h light/dark cycle and were allowed free access to water and food supplied by PMI Nutrition International, Inc. Animals were anesthetized with 70% CO2, and then decapitated. Brains were removed and placed on an ice-cold platform. The cerebral cortex was removed and placed in 20 volumes (weight:volume) of ice-cold preparative buffer (137 mM NaCI, 10.7 mM KCI, 5.8 mM KH2PO4, 8 mM Na2HPO4, 20 mM HEPES (free acid), 5 mM iodoacetamide, 1.6 mM EDTA, pH 7.4); PMSF, dissolved in methanol to a final concentration of 100 μM, was added and the suspension was homogenized by Polytron. The homogenate was centrifuged at 18,000 x g for 20 min at 4 0C and the resulting pellet was re-suspended in 20 volumes of ice-cold water. After 60 min incubation on ice, a new pellet was collected by centrifugation at 18,000 x g for 20 min at 4 0C. The final pellet was re-suspended in 10 volumes of buffer and stored at -20 0C.
Preparation of membranes from SH-EP1 /human α4β2 clonal cells: Cell pellets from 40 150 mm culture dishes were pooled, and homogenized by Polytron (Kinematica GmbH, Switzerland) in 20 milliliters of ice-cold preparative buffer. The homogenate was centrifuged at 48,000 g for 20 minutes at 40C. The resulting pellet was re-suspended in 20 mL of ice-cold preparative buffer and stored at -2O0C.
On the day of the assay, the frozen membranes were thawed and spun at 48,000 x g for 20 min. The supernatant was decanted and discarded. The pellet was resuspended in Dulbecco's phosphate buffered saline (PBS, Life Technologies) pH 7.4 and homogenized with the Polytron for 6 seconds. Protein concentrations were determined using a Pierce BCA Protein Assay Kit, with bovine serum albumin as the standard (Pierce Chemical Company, Rockford, IL).
Assay: Membrane preparations (approximately 50 μg for human and 200-300 μg protein for rat α4β2) were incubated in PBS (50 μL and 100 μL respectively) in the presence of competitor compound (0.01 nM to 100 μM) and 5 nM [3H]nicotine for 2-3 hours on ice. Incubation was terminated by rapid filtration on a multi-manifold tissue harvester (Brandel, Gaithersburg, MD) using GF/B filters presoaked in 0.33% polyethyleneimine (w/v) to reduce non-specific binding. Tissue was rinsed 3 times in PBS, pH 7.4. Scintillation fluid was added to filters containing the washed tissue and allowed to equilibrate. Filters were then counted to determine radioactivity bound to the membranes by liquid scintillation counting (2200CA Tri-Carb LSC, Packard Instruments, 50% efficiency or Wallac Trilux 1450 MicroBeta, 40% efficiency, Perkin Elmer).
Data were expressed as disintegrations per minute (DPMs). Within each assay, each point had 2-3 replicates. The replicates for each point were averaged and plotted against the log of the drug concentration. IC50, which is the concentration of the compound that produces 50% inhibition of binding, was determined by least squares non-linear regression. Ki values were calculated using the Cheng-Prussof equation (1973):
Ki = IC50/ (1 + N/Kd) where N is the concentration of [3H]nicotine and Kd is the affinity of nicotine (3 nM, determined in a separate experiment). α7 nAChR Subtype
Preparation of membranes from rat hippocampus: Rats (female, Sprague-Dawley), weighing 150-250 g, were maintained on a 12 h light/dark cycle and were allowed free access to water and food supplied by PMI Nutrition International, Inc. Animals were anesthetized with 70% CO2, then decapitated. Brains were removed and placed on an ice-cold platform. The hippocampus was removed and placed in 10 volumes (weight:volume) of ice- cold preparative buffer (137 mM NaCI, 10.7 mM KCI, 5.8 mM KH2PO4, 8 mM Na2HPO4, 20 mM HEPES (free acid), 5 mM iodoacetamide, 1.6 mM EDTA, pH 7.4); PMSF, dissolved in methanol to a final concentration of 100 μM, was added and the tissue suspension was homogenized by Polytron. The homogenate was centrifuged at 18,000 x g for 20 min at 4 0C and the resulting pellet was re-suspended in 10 volumes of ice-cold water. After 60 min incubation on ice, a new pellet was collected by centrifugation at 18,000 x g for 20 min at 4 0C. The final pellet was re-suspended in 10 volumes of buffer and stored at -20 0C.
On the day of the assay, tissue was thawed, centrifuged at 18,000 x g for 20 min, and then re-suspended in ice-cold PBS (Dulbecco's Phosphate Buffered Saline, 138 mM NaCI, 2.67 mM KCI, 1.47 mM KH2PO4, 8.1 mM
Na2HPO4, 0.9 mM CaCI2, 0.5 mM MgCI2, Invitrogen/Gibco, pH 7.4) to a final concentration of approximately 2 mg protein/mL. Protein was determined by the method of Lowry et al., J. Biol. Chem. 193: 265 (1951 ), herein incorporated by reference, using bovine serum albumin as the standard. Assay: The binding of [3H]MLA was measured using a modification of the methods of Davies et al., Neuropharmacol. 38: 679 (1999), herein incorporated by reference. [3H]MLA (Specific Activity = 25-35 Ci/mmol) was obtained from Tocris. The binding of [3H]MLA was determined using a 2 h incubation at 21 0C. Incubations were conducted in 48-weil micro-titre plates and contained about 200 μg of protein per well in a final incubation volume of 300 μL. The incubation buffer was PBS and the final concentration of [3H]MLA was 5 nM. The binding reaction was terminated by filtration of the protein containing bound ligand onto glass fiber filters (GF/B, Brandel) using a Brandel Tissue Harvester at room temperature. Filters were soaked in de- ionized water containing 0.33% polyethyleneimine to reduce non-specific binding. Each filter was washed with PBS (3 x 1 mL) at room temperature. Non-specific binding was determined by inclusion of 50 μM non-radioactive MLA in selected wells.
The inhibition of [3H]MLA binding by test compounds was determined by including seven different concentrations of the test compound in selected wells. Each concentration was replicated in triplicate. IC50 values were estimated as the concentration of compound that inhibited 50 percent of specific [3H]MLA binding. Inhibition constants (Ki values), reported in nM, were calculated from the IC50 values using the method of Cheng et al., Biochem. Pharmacol. 22: 3099-3108 (1973), herein incorporated by reference.
Example 11 : Tabular Receptor Binding Data
Receptor binding data for compounds of the present invention are shown in Table 1.
Table 1
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Summary of Biological Data
Compounds of Table 1 , representative of the present invention, exhibited inhibition constants (Ki values) at the rat and human α4β2 subtypes in the ranges of 1 nM to > 10,000 nM. Ki values at the human α7 subtype vary within the range of 850 nM to >10,000 nM. The notation "ND" means that the Ki value was not determined. In some cases, this was a result of the assay being unavailable for a period of time, and in other cases, this was because the compounds failed to bind sufficiently in high through-put screening (HTS) to warrant Ki determination. This latter situation was much more common for binding at the α7 subtype, as compared to the α4β2 subtype. In this regard, failing to bind sufficiently in HTS means, for the α4β2 subtype, that the compound failed to inhibit, at 5 μM concentration, the binding of 5 nM 3H- nicotine by at least 50%, and for the α7 subtype, that the compound failed to inhibit, at 5 μM concentration, the binding of 5 nM 3H-MLA (methyllycaconitine) by at least 50%. The specific pharmacological responses observed may vary according to and depending on the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with practice of the present invention.
Although specific embodiments of the present invention are herein illustrated and described in detail, the invention is not limited thereto. The above detailed descriptions are provided as exemplary of the present invention and should not be construed as constituting any limitation of the invention. Modifications will be obvious to those skilled in the art, and all modifications that do not depart from the spirit of the invention are intended to be included with the scope of the appended claims.

Claims

ClaimsWe Claim:
1. A compound as represented by Formula I:
Figure imgf000050_0001
Formula I wherein:
Z1 is methylene and n is 0 or 1 ; Z2 is methylene and m is 0 or 1 ; when n is 1 , m is 0; when m is 1 , n is O;;
X1 is hydrogen, C1-6 alkyl, or C1-6 haloalkyl; X2 is R', OR" or NRRlv; each of R', R1", and Rιv are individually hydrogen, optionally substituted C1-6 alkyl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-6alkenyl, optionally substituted C3-8cycloalkenyl, optionally substituted C2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; or R"' and Rιv can combine with the nitrogen atom to which they are attached to form an optionally substituted 3 to 8 membered ring that may contain one or more degrees of unsaturation and may contain one or more additional heteroatom selected from N, O, or S; and
R" is optionally substituted C1-6 alkyl, optionally substituted C3-8 cycloalkyl, optionally substituted C2-6alkenyl, optionally substituted C^cycloalkenyl, optionally substituted C2-6 alkynyl, optionally substituted heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl; where the term "optionally substituted" refers to optional substitution of one or more hydrogen atoms by substituents independently selected from C1-6 alkyl, C34, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, ORV, NRVRVI, C1-6 haloalkyl, -CN, -NO2, -C2RV, -SRV, -N3, -Cf=O)NfW, -NRVC(=O)RVI, -OC(=O)NRVRVI, -NRVC(=O)ORVI, -SO2RV, -SO2NRVRVI, and -NRVSO2RVI, where Rv and R are individually hydrogen, C1-6 alkyl, C3-8cycloalkyl, heterocyclyl, aryl, or arylalkyl; or a pharmaceutically acceptable salt thereof.
2. A compound selected from the group consisting of:
3-(acetyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(methoxyacetyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(fluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(propanoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(butanoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(cyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1 R,2R)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1R,2S)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1S,2R)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
(1S,2S)-3-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(2,213,3-tetramethylcyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(cyclobutanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(cyclopentanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(cyclopent-3-enecarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
S^N-methylcarbamoyO-S.δ-diazabicyclotS^^Jnonane,
3-(N-ethylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(N-propylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(N-butylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(N-cyclopentylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(methoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(ethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(propoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(isopropoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(2-methoxyethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(phenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(4-fluorophenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(furan-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(3-methylfuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
3-(4-methylfuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane,
S^δ-methylfuran^-ylcarbonyO-S.Θ-diazabicyclotS^^lnonane, 3-(3-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(4-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 3-(5-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, S-^-methyloxazol^-ylcarbonyO-S.Θ-diazabicycloβ^^lnonane, S-^-methyloxazol^-ylcarbonyO-S.δ-diazabicycloβ^^nonane, 3-(5-methyloxazol-4-ylcarbonyf)-3,6-diazabicyclo[3.2.2]nonane, S^oxazol-δ-ylcarbonyO-S.e-diazabicycloβ^^nonane, S-^-methylisoxazol-δ-ylcarbonyO-S.e-diazabicycloIS^^Jnonane, S-^-methylisoxazol-S-ylcarbonylJ-S.Θ-diazabicycloIS^^jnonane, S-fbenzofuran^-ylcarbonyO-S.δ-diazabicyclolS^^nonane,
3-(cyclopropanecarbonyl)-6-methyl-3,6-diazabicyclo[3.2.2]nonane, S^furan^-ylcarbonyO-β-methyl-S.β-diazabicyclop^^Jnonane, 6-(acetyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(fluoroacetyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(methoxyacetyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(propanoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(butanoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1R,2R)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1R,2S)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1S,2R)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, (1S,2S)-6-(2-fluorocyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(2,2,3,3-tetramethylcyclopropanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclobutanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclopentanecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(cyclopent-3-enecarbonyl)-3,6-diazabicyclo[3.2.2]nonane, δ^N-methylcarbamoyO-S.e-diazabicyclotS^^Jnonane, 6-(N-ethylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, β^N-propylcarbamoyO-S.Θ-diazabicycloIS^^lnonane, β^N-butylcarbamoyO-S.β-diazabicycloβ^^Jnonane,
6-(N-cyclopentylcarbamoyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(methoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(ethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(propoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(isopropoxycarbonyl)-3,6-diazabicyclof3.2.2]nonane, 6-(2-fluoroethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(2-methoxyethoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(phenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(4-fluorophenoxycarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(furan-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, θ-β-methylfuran^-ylcarbonyO-S.δ-diazabicycloβ^^nonane, β-^-methylfuran^-ylcarbonyO-S.Θ-diazabicyclofS^^Jnonane, 6-(5-methylfuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(3-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(4-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, 6-(5-chlorofuran-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, β-^-methyloxazol^-ylcarbonyO-S.Θ-diazabicycloβ^^nonane, 6-(5-methyloxazol-2-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, θ-^-methyloxazoM-ylcarbonyO-S.Θ-diazabicyclofS^^Jnonane, β-tøxazol-δ-ylcarbonyO-S.Θ-diazabicycloβ^^nonane, 6-(3-methylisoxazol-5-ylcarbonyl)-3,6-diazabicyclo[3.2.2]nonane, β-^-methylisoxazol-δ-ylcarbonyO-S.Θ-diazabicycloβ^^Jnonane, β-φenzofuran^-ylcarbonyO-S.β-diazabicycloβ^^nonane, 6-(cyclopropanecarbonyl)-3-methyl-3,6-diazabicyclo[3.2.2]nonane, and θ^furan^-ylcarbonyO-S-methyl-S.Θ-diazabicyclop^^Jnonane; or a pharmaceutically acceptable salt thereof.
3. Use of the compound according to any one of claims 1 or 2, in the manufacture of a medicament.
4. A method for treatment of central nervous system disorders and dysfunctions, comprising administering to a mammal in need of such treatment, a therapeutically effective amount of the compound according to any one of claims 1 or 2.
5. The method of claim 4, wherein the disorder is selected from the group consisting of age-associated memory impairment, mild cognitive impairment, presenile dementia, early onset Alzheimer's disease, senile dementia, dementia of the Alzheimer's type, Lewy body dementia, vascular dementia, Alzheimer's disease, stroke, AIDS dementia complex, attention deficit disorder, attention deficit hyperactivity disorder, dyslexia, schizophrenia, schizophreniform disorder, schizoaffective disorder, cognitive deficits in schizophrenia, and cognitive dysfunction in schizophrenia.
6. The method of claim 4, wherein the disorder is selected from the group consisting of mild to moderate dementia of the Alzheimer's type, attention deficit disorder, attention deficit hyperactivity disorder, mild cognitive impairment, age- associated memory impairment, cognitive deficits in schizophrenia, and cognitive dysfunction in schizophrenia.
7. A pharmaceutical composition comprising a compound according to any one of claims 1 or 2, and one or more pharmaceutically acceptable carrier.
8. Use of the pharmaceutical composition according to claim 7 in the manufacture of a medicament for treatment of central nervous system disorders and dysfunctions.
9. A method of treating inflammation, the inflammatory response associated with bacterial and/or viral infection, pain, metabolic syndrome, autoimmune disorders, addictions, and obesity, comprising administering to a mammal in need of such treatment, a therapeutically effective amount of the compound according to any one of claims 1 or 2.
PCT/US2009/055687 2008-09-05 2009-09-02 Amides of diazabicyclononanes and uses thereof WO2010028011A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US9463108P 2008-09-05 2008-09-05
US61/094,631 2008-09-05

Publications (1)

Publication Number Publication Date
WO2010028011A1 true WO2010028011A1 (en) 2010-03-11

Family

ID=41256099

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/055687 WO2010028011A1 (en) 2008-09-05 2009-09-02 Amides of diazabicyclononanes and uses thereof

Country Status (1)

Country Link
WO (1) WO2010028011A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104030998A (en) * 2014-06-05 2014-09-10 华东师范大学 4- perfluoroalkyl-4,5-disubstituted isoxazole derivative and preparation method thereof
CN104098565A (en) * 2013-04-02 2014-10-15 上海药明康德新药开发有限公司 Preparation method for 3, 6-diazabicyclo[3.2.2]nonane derivative
WO2016064759A1 (en) 2014-10-20 2016-04-28 Oyster Point Pharma, Inc. Methods of treating ocular conditions
US10709707B2 (en) 2016-04-07 2020-07-14 Oyster Point Pharma, Inc. Methods of treating ocular conditions
US11325915B2 (en) 2017-10-13 2022-05-10 The Institute Of Cancer Research: Royal Cancer Hospital Lysyl oxidase inhibitors
CN115626925A (en) * 2022-12-08 2023-01-20 山东汇海医药化工有限公司 Synthesis method of bispidine

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0885888A1 (en) * 1996-02-02 1998-12-23 Nippon Shinyaku Company, Limited Isoquinoline derivatives and drugs
WO2002096911A1 (en) * 2001-06-01 2002-12-05 Neurosearch A/S Novel heteroaryl-diazabicyclo-alkanes as cns-modulators
US20030225268A1 (en) * 1999-01-29 2003-12-04 Bunnelle William H. Diazabicyclic CNS active agents
US20050101602A1 (en) * 2003-09-19 2005-05-12 Anwer Basha Substituted diazabicycloalkane derivatives
WO2008008517A2 (en) * 2006-07-14 2008-01-17 Merck & Co., Inc. Bridged diazepan orexin receptor antagonists
WO2008057938A1 (en) * 2006-11-02 2008-05-15 Targacept, Inc. Nicotinic acetylcholine receptorsub-type selective amides of diazabicycloalkanes

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0885888A1 (en) * 1996-02-02 1998-12-23 Nippon Shinyaku Company, Limited Isoquinoline derivatives and drugs
US20030225268A1 (en) * 1999-01-29 2003-12-04 Bunnelle William H. Diazabicyclic CNS active agents
WO2002096911A1 (en) * 2001-06-01 2002-12-05 Neurosearch A/S Novel heteroaryl-diazabicyclo-alkanes as cns-modulators
US20050101602A1 (en) * 2003-09-19 2005-05-12 Anwer Basha Substituted diazabicycloalkane derivatives
WO2008008517A2 (en) * 2006-07-14 2008-01-17 Merck & Co., Inc. Bridged diazepan orexin receptor antagonists
WO2008057938A1 (en) * 2006-11-02 2008-05-15 Targacept, Inc. Nicotinic acetylcholine receptorsub-type selective amides of diazabicycloalkanes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
W. H. BUNNELLE: "Structure-Activity studies and analgesic efficayc of N-(3-pyridinyl)-bridged bicyclic diamines, exceptionally potent agonists at nicotinic acetylcholine receptors", J. MED. CHEM., vol. 50, no. 15, 2007, pages 3627 - 3644, XP002554553 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104098565A (en) * 2013-04-02 2014-10-15 上海药明康德新药开发有限公司 Preparation method for 3, 6-diazabicyclo[3.2.2]nonane derivative
CN104030998A (en) * 2014-06-05 2014-09-10 华东师范大学 4- perfluoroalkyl-4,5-disubstituted isoxazole derivative and preparation method thereof
CN104030998B (en) * 2014-06-05 2015-11-18 华东师范大学 4-Polyfluoroalkyl-4,5-bis-substituted isoxazoles derivative and preparation method thereof
US10456396B2 (en) 2014-10-20 2019-10-29 Oyster Point Pharma, Inc. Dry eye treatments
EP3848028A1 (en) 2014-10-20 2021-07-14 Oyster Point Pharma, Inc. Methods of treating ocular conditions
US9504645B2 (en) 2014-10-20 2016-11-29 Oyster Point Pharma, Inc. Pharmaceutical formulations for treating ocular conditions
US9532944B2 (en) 2014-10-20 2017-01-03 Oyster Point Pharma, Inc. Methods of improving ocular discomfort
US9597284B2 (en) 2014-10-20 2017-03-21 Oyster Point Pharma, Inc. Dry eye treatments
WO2016064759A1 (en) 2014-10-20 2016-04-28 Oyster Point Pharma, Inc. Methods of treating ocular conditions
US11911380B2 (en) 2014-10-20 2024-02-27 Oyster Point Pharma, Inc. Compositions and use of varenicline for treating dry eye
US9504644B2 (en) 2014-10-20 2016-11-29 Oyster Point Pharma, Inc. Methods of increasing tear production
US11224598B2 (en) 2014-10-20 2022-01-18 Oyster Point Pharma, Inc. Methods of increasing lacrimal proteins
US11903942B2 (en) 2014-10-20 2024-02-20 Oyster Point Pharma, Inc. Compositions and use of varenicline for treating dry eye
US11903943B2 (en) 2014-10-20 2024-02-20 Oyster Point Pharma, Inc. Compositions and use of varenicline for treating dry eye
US11903941B2 (en) 2014-10-20 2024-02-20 Oyster Point Pharma, Inc. Compositions and use of varenicline for treating dry eye
US10709707B2 (en) 2016-04-07 2020-07-14 Oyster Point Pharma, Inc. Methods of treating ocular conditions
US11325915B2 (en) 2017-10-13 2022-05-10 The Institute Of Cancer Research: Royal Cancer Hospital Lysyl oxidase inhibitors
CN115626925A (en) * 2022-12-08 2023-01-20 山东汇海医药化工有限公司 Synthesis method of bispidine

Similar Documents

Publication Publication Date Title
JP5739398B2 (en) Nicotinic Acetylcholine Receptor Subtype Selective Diazabicycloalkane Amides
US20120053168A1 (en) Fused benzoazepines as neuronal nicotinic acetylcholine receptor ligands
US20110071180A1 (en) Sub-type selective amides of diazabicycloalkanes
AU2010206638B2 (en) Preparation and therapeutic applications of (2S,3R)-N-2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)-3,5-difluorobenzamide
US8802694B2 (en) 3,6-diazabicyclo[3.1.1]heptanes as neuronal nicotinic acetycholine receptor ligands
WO2010028011A1 (en) Amides of diazabicyclononanes and uses thereof
CA2618700A1 (en) Heteroaryl-substituted diazatricycloalkanes, methods for its preparation and use thereof
US20100144700A1 (en) Heterocyclic-carbonyl-diazabicycloalkanes as modulators of the neuronal nicotinic acetylcholine alpha 4 beta 2, subtype receptor for the treatment of cns related disorders
US20110263629A1 (en) Amides of diazabicyclooctanes and uses thereof
US20140249141A1 (en) 1,4-diazabicyclo[3.2.2]nonanes as neuronal nicotinic acetylcholine receptor ligands
WO2013116413A1 (en) Diazabicyclo[3.3.1]nonanes, methods of synthesis, and uses thereof
US20110257168A1 (en) Derivatives of oxabispidine as neuronal nicotinic acetylcholine receptor ligands
US20150119378A1 (en) Azetidinyloxy-, pyrrolidinyloxy-, and piperidinyloxy-substituted metanicotines as neuronal nicotinic acetylcholine receptor ligands

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: 09792155

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09792155

Country of ref document: EP

Kind code of ref document: A1