WO1994022868A1 - 7-azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents - Google Patents

7-azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents Download PDF

Info

Publication number
WO1994022868A1
WO1994022868A1 PCT/US1994/003573 US9403573W WO9422868A1 WO 1994022868 A1 WO1994022868 A1 WO 1994022868A1 US 9403573 W US9403573 W US 9403573W WO 9422868 A1 WO9422868 A1 WO 9422868A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
compound
pyridyl
exo
Prior art date
Application number
PCT/US1994/003573
Other languages
French (fr)
Inventor
T. Y. Shen
W. Dean Harman
Dao Fei Huang
Javier Gonzalez
Original Assignee
University Of Virginia
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 University Of Virginia filed Critical University Of Virginia
Priority to EP94912381A priority Critical patent/EP0691971A1/en
Priority to US08/532,584 priority patent/US6117889A/en
Priority to JP6522396A priority patent/JPH08511768A/en
Priority to AU64971/94A priority patent/AU695682B2/en
Publication of WO1994022868A1 publication Critical patent/WO1994022868A1/en
Priority to US09/023,844 priority patent/US6255490B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • This invention is in the area of
  • Opiates, and in particular, morphine are routinely administered for the treatment of
  • Agents that are less potent than morphine such as codeine, mixed agonist-antagonist opioids, and non-opiate
  • analgesics including non-steroidal
  • NSAIDS anti-inflammatory drugs
  • NSAtes anti-inflammatory drugs
  • Spande, et al. reported in 1992 that a potent nonopiate analgesic had been isolated from the skins of the Ecuadoran poison frog, Epipedobates tricolor.
  • Spande, et al. 1992 J. Am. Chem. Soc , 114 , 3475-3478.
  • the structure of the compound was determined by mass spectroscopy, infrared spectroscopy, and nuclear magnetic resonance as exo-2- (2-chloro-
  • epibatidine has a negligible affinity for opiate receptors (1/8000 times that of morphine). Based on this data, it appears that epibatidine is a very potent analgesic that acts via a non-opiate mechanism. Since epibatidine was isolated from the skins of the Ecuadoran poison frog, its identification and characterization was not based on the synthesis of the compound. In fact, compounds in the family of 7-azabicyclo [2.2.1]-heptanes and -heptenes (also referred to as 7-azanorbornanes and
  • analgesic activity or that can be derivatized to compounds with pharmacological activity.
  • R 1 and R 4 are independently hydrogen, alkyl, including CH 3 ; alkylhydroxy, including CH 2 OH;
  • alkyloxyalkyl including -CH 2 OCH 3 ; alkylthioalkyl, including -CH 2 SCH 3 ; alkylamino, including -CH 2 NH 2 ; alkylaminoalkyl or alkylaminodialkyl, including CH 2 NH(CH 3 ) and CH 2 N(CH 3 ) 2 ; oxyalkyl, including -OCH 3 ; carboalkoxy, including carbomethoxy; allyl, aryl and thioalkyl, including -SCH 3 ;
  • R 3 , R 5 and R 6 are independently hydrogen, alkyl, including -CH 3 ; alkylhydroxy, including -CH 2 OH;
  • alkyloxyalkyl including -CH 2 OCH 3 ; alkylthioalkyl, including -CH 2 SCH 3 ; alkylamino, including -CH 2 NH 2 ; alkylaminoalkyl or alkylaminodialkyl, including
  • -CO 2 H CO 2 alkyl, including -CO 2 CH 3 ; -C(O) alkyl, including -C(O)CH 3 ; -CN, -C(O)NH 2 , -C (O)NH (alkyl), -C(O)N (alkyl) 2 , including -C(O)N(CH 3 ) 2 ; allyl, -SO 2 (alkyl), -SO 2 aryl, including -SO 2 (C 6 H 5 ),
  • R 5 and R 6 together can be alkylidene or
  • haloalkylidene including -CH 2 - and -CF 2 -; epoxide (-O-); episulfide (-S-); imino (-N(alkyl)- or
  • C(O)Oheteroaryl COOaralkyl, -CN, Q, C(O)Q, - alkyl (Q), -alkenyl (Q), -alkynyl (Q), -O- (Q) -S-Q, - NH-Q or -N(alkyl)-Q;
  • R 2 and R 3 together can be -C (O) -NR 8 ) -C (O) or CH(OH) -N(R 8 ) -C(O) - wherein R 8 can be alkyl, aryl including phenyl, or heteroaryl;
  • alkylhydroxy including CH 2 CH 2 OH, alkylamino (alkyl) 2 , including CH 2 CH 2 N (CH 3 ) 2 alkyloxyalkyl,
  • alkylthioalkyl, aryl, dialkyl to form a quarternary ammonium including
  • R 9 is hydrogen or alkyl
  • Y' is CN, NO 2 , alkyl, OH, -O-alkyl; wherein Z is O or S;
  • R 10 and R 11 are each independently -O-, -OH, -O-alkyl, -O-aryl, -NH 2 , -NH(alkyl),
  • W is alkyl, including CH 3 ; halo, including Cl and F; aryl, heteroaryl, OH, oxyalkyl, including -OCH 3 ; SH, thioalkyl, including -SCH 3 ; -SO (alkyl) including -SOCH 3 ; -SO 2 alkyl, including -SO 2 CH 3 ;
  • -OCH 2 CH CH 2 , -OCH 2 (C 6 H 5 ), CF 3 , CN, alkylenedioxy, including -methylenedioxy-; -CO 2 H, -CO 2 alkyl, including -CO 2 CH 3 ; -OCH 2 CH 2 OH, -NO 2 , -NH 2 , -NH (alkyl), including -NHCH 3 ; -N (alkyl) 2 , including -N(CH 3 ) 2 ; -NHC(O) alkyl, including -NHC(O)CH 3 ; -SO 2 CF 3 , or -NHCH 2 aryl, including -NHCH 2 (C 6 H 5 ) ; and wherein
  • the - - - indicates an optional double bond.
  • a method for the treatment of pain includes administering an effective amount of the compound or its pharmaceutically acceptable salt or derivative, or mixtures thereof, to a host in need of analgesic therapy, optionally in a
  • the pharmaceutically active compounds, or their precursors are prepared by the cycloaddition reaction of pentaammineosmium(II) complexes of pyrroles with dipolarophiles, including 3 -vinyl pyridine derivatives.
  • Figure 1 is an illustration of the chemical structure of exo-2 -(2-chloro-5-pyridyl)-7- azabicyclo[2.2.1]heptane (epibatidine).
  • FIGS. 2a and 2b are schematic illustrations of processes for the preparation of active
  • alkyl refers to a saturated straight, branched, or cyclic (or a combination thereof) hydrocarbon of C 1 to C 10 , and specifically includes methyl, ethyl, propyl, isopropyl, cyclopropylmethyl, cyclobutylmethyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methylpentyl, 2,2-dimethylbutyl, 2,3-dimethyl- butyl, heptyl, octyl, nonyl, and decyl.
  • lower alkyl refers to a C 1 to C 6 saturated straight, branched, or cyclic (in the case of C 5-6 ) hydrocarbon, and specifically includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl,
  • alkylamino refers to an amino group that has an alkyl substituent.
  • alkynyl refers to a C 2 to C 10 straight or branched
  • hydrocarbon with at least one triple bond is hydrocarbon with at least one triple bond.
  • aryl refers to phenyl, or substituted phenyl, wherein the
  • substituent is halo, alkyl, alkoxy, alkylthio, haloalkyl, hydroxyalkyl, alkoxyalkyl,
  • aryl group can have up to 3
  • halo includes fluoro, chloro, bromo, and iodo.
  • aralkyl refers to an aryl group with an alkyl substituent.
  • alkaryl refers to an alkyl group that has an aryl substituent, including benzyl,
  • heteroaryl or heteroaromatic refers to an aromatic moiety that includes at least one sulfur, oxygen, or nitrogen in the aromatic ring.
  • Nonlimiting examples are furyl, pyridyl, pyrimidyl, thienyl, isothiazolyl,
  • organic or inorganic anion refers to an organic or inorganic moiety that carries a negative charge and can be used as the negative portion of a salt.
  • enantiomerically enriched composition or compound refers to a composition or compound that includes at least 95% by weight of a single enantiomer of the compound.
  • pharmaceutically active derivative refers to any compound that upon administration to the recipient, is capable of providing directly or indirectly, the compounds disclosed herein.
  • dipolarophile refers to a compound or moiety that reacts with a dipolar species to form a cycloaddition product.
  • dienophile refers to a compound or moiety that reacts with a diene to form a cycloaddition product.
  • refers to a pi-orbital complex of an unsaturated compound with a metal, and wherein the superscript after the ⁇ refers to the number of sp 2 carbon atoms bonded to the metal.
  • electron withdrawing substituent refers to a substituent that pulls electron density from the moiety to which it is attached through induction or resonance.
  • electron withdrawing substituents are well known to those skilled in organic synthesis.
  • anti-inflammatory activity can be administered to a mammal, including a human, to treat pain and inflammation.
  • a method for the treatment of pain is also presented that includes administering an effective amount of the compound or its pharmaceutically acceptable salt or
  • dipolarophile lie near the developing double bond in the diene. Exo configurations are formed when other unsaturated groups in the dienophile (or dipolarophile) lie away from the developing double bond in the diene. Depending on the substitution on the carbon atoms, the endo and exo orientations can yield different stereoisomers.
  • 7-azabicyclo[2.2.1]heptanes and carbon atoms 2 and 3 or 5 and 6 in 7-azabicyclo[2.2.1]heptanes are chiral when attached to different substituents. If at least one of the carbon in the molecule are chiral, the unsymmetrically substituted bicyclic compounds exist as one or more diastereomeric pairs.
  • the R groups in the active compounds described herein can also include chiral carbons, and thus, optically active centers.
  • enantiomers of a biologically active compound is more active, and perhaps less toxic, than other enantiomers of the same compound.
  • enantiomerically enriched compounds are preferred for pharmaceutical administration to humans or other hosts.
  • One of ordinary skill in the art can easily separate the enantiomers of the disclosed compounds using conventional processes, and can evaluate the biological activity of the isolated enantiomers using methods disclosed herein or otherwise known. Through the use of chiral NMR shift reagents, polarimetry, or chiral HPLC, the optical enrichment of the compound can be determined.
  • Classical methods of resolution include a variety of physical and chemical techniques. For example, since the compound has a basic amine (N 7 ) , it can be reacted with a chiral acid to form diastereomeric salts that may possess significantly different solubility properties.
  • chiral acids include malic acid, mandelic acid, dibenzoyl tartaric acid,
  • Any dipolarophile can be used in this reaction that reacts with the pentaammineosmium pyrrole complex to provide an optionally substituted
  • sulfones anhydrides, -CF 3 , pyridinium salts, and for example, CO(alkyl, aryl or heteroaryl), C(O)H, CO 2 (alkyl, aryl, or heteroaryl), SO 2 (alkyl, aryl, or heteroaryl), or wherein Z, and Z 2 are together
  • alpha-methylene- ⁇ -butyrolactone maleates, and fumarates.
  • any optionally substituted pyrrole can be used that on complexation with pentaammineosmium (II) will react with a
  • Suitable pyrroles include 2,5-dialkylpyrrole, 2-alkylpyrrole,
  • 3-alkylpyrrole 1-alkylpyrrole, 3,4-dialkylpyrrole, pyrrole, 1-silylated pyrrole, (1, 2, or 3)alkoxy or amino pyrrole, 2, 3-dialkoxypyrrole, 2,5- dialkoxypyrrole, and 3,4-dialkoxypyrrole.
  • osmium coordinates the heterocycle across C2 and C3. At 20°C, this species is in equilibrium with its linkage isomer in which the metal binds across C3 and C4. Although the 3,4- ⁇ species is only a minor component ( ⁇ G iso >3 kcal/mol), the metal coordination in this species renders the remaining portion of the pyrrole an azomethine ylide
  • Os (II) [Os(NH 3 )5] (OTf) 2 .
  • the resulting 7-azabicyclo[2.2.1]hept-5-ene ligand is unstable with respect to cycloreversion, but metal coordination greatly stabilizes the complex and thus provides the opportunity to carry out functional group transformations while keeping the bicyclic framework intact.
  • derivatization of electron-withdrawing groups in the 2- or 3-positions of the norbornene framework using conventional processes, provides a wide array of functionalized 7-azanorbornenes. Specifically, as shown in Scheme 2 below, the exo-carbonyl cycloadduct complex 2, prepared in a one-pot synthesis from 2,5-dimethylpyrrole, is reduced to the corresponding alcohol and oxidatively
  • This approach can be used to construct the epibatidine ring system if a 3 -vinyl pyridine is used as the dipolarophile.
  • bridgehead carbons C 1 and C 4 .
  • the reactivity of simple pentaammineosmium (II) - pyrrole complexes with dipolarophiles decreases in the order
  • Pentaammineosmium (II) is generated in situ by the reduction of pentaammineosmium (III) with a one electron reducing agent that has a reducing
  • pentaammineosmium (II) can be any anion that does not adversely affect the overall reaction.
  • Typical counteranions are CF 3 SO 3 - (Otf-), PF 6 , X-, and (alkyl or aryl)SO 3 -.
  • Any chemical or electrochemical reducing agent that can reduce the osmium complex from a III valence state to a II valence state and which does not cause or participate in undesired side
  • pentaammineosmium (III), and reducing agent are stirred at a temperature ranging between 0°C and 50°C until the desired organometallic complex is formed, typically between 0.1 and 1.0 hours.
  • the reaction can be carried out in a polar or nonpolar solvent, including but not limited to
  • reaction is carried out in the absence of 0 2 , and typically under nitrogen, at a pressure of 1 atm or greater.
  • the dipolarophile is added to the stirring solution of the pyrrole pentaammineosmium (II) complex to produce an optionally substituted 7- azabicyclo[2.2.1]hept-5-ene.
  • Any molar ratio of dipolarophile to pyrrole can be used that provides the desired results.
  • a molar ratio of dipolarophile to pyrrole ranging between
  • reaction solution is stirred at a temperature ranging between 10 and 50°C until the product is formed, typically between 1 and 24 hours.
  • bicyclic ring system In an optional step after the bicyclic ring system is formed, and while pentaammineosmium is still complexed to the pi-orbital of the heptene moiety, functional groups on the bicyclic ring can be derivatized using conventional processes. For example, esters can be reduced to alcohols, nitriles to amines, sulfones to sulfides, nitro groups to amines, and amides to amines. Sulfones and carboxylates can be reductively eliminated using the Barton decarboxylation procedure. High temperatures and strong bases should be avoided in the functionalization procedures to avoid ring disruption and unwanted side reactions.
  • 7-azabicyclo[2.2.1]hept-5-ene can be treated with one equivalent of cerium reagent at 20°C in a polar solvent such as acetonitrile.
  • a polar solvent such as acetonitrile.
  • Appropriate reagents include Ce (NO 3 ) 6 (NH 4 ) 2 ,DDQ, and other inorganic or organic oxidants with E > +.70 volts versus
  • the osmium reagent can be removed by heating the complex as necessary, usually between approximately 50°C and 100°C.
  • acetonitrile was protonated with excess triflic acid (250 mg, 1.67 mmol) and treated at -10°C with a likewise-cooled solution of eerie ammonium nitrate (560 mg, 1.02 mmol) and triflic acid (560 mg, 3.73 mmol) in 2 grams acetonitrile. Water (1-2 ml) was added to dissolve the precipitated salts, the mixture made basic with 40 ml 10% aqueous sodium carbonate and the product extracted with 5 X 20 ml methylene chloride. The extract was dried over MgSO 4 and the solvent evaporated, yielding 147 mg of brown oil.
  • the crude aminoalcohol 8 obtained from 1.0 mmol of the osmium complex as described above was suspended in a solution of aqueous Na 2 CO 3 (0.38 grams in 2 grams water), and the mixture chilled to 0°C.
  • Benzyl chloroformate 510 mg, 3 mmol was added, and the mixture allowed to warm to room temperature with vigorous stirring. After 20 hours at 25°C the mixture was extracted with methylene chloride, and the extracts dried and
  • CDCI 3 d 176.5 (CO), 67.7, 63.4, 53.0, 51.3, 44.0,
  • the 2,5-dimethylpyrrole complex (669 mg, 1.0 mmol) was suspended in 2 grams methyl acrylate and the slurry stirred for 1 hour.
  • Acetonitrile (c. 1 ml) was added to dissolve the solids and the resulting solution added dropwise to 50 ml of ether while stirring. The precipitate was filtered, washed with ether and dried, yielding 730 mg (97%) of an off-white powder.
  • the yield of this reaction is typically 90-95% of a yellow-orange solid containing approximately 8% of a binuclear impurity.
  • the pentaammineosmium-pyrrole complex obtained from Example 11 was treated with an excess (3-30 eq) of a dipolarophile in either acetonitrile or N,N-dimethylacetamide solution. After 1-10 hours, the solution was added to ether or methylene chloride with stirring (20 ml of ether per gram of acetonitrile or 75 ml methylene chloride per gram of N,N-dimethylacetamide). The resulting
  • a dipolarophile e.g., methyl acrylate was added directly to the reaction mixture in the synthesis of the parent pyrrole complex as
  • Example 11 After a suitable reaction time (e.g., 1-10 hours), the mixture was filtered to remove the magnesium, and the filtrate was added to 1:1 methylene-chloride/ether (100 ml for every gram of N,N-dimethylacetamide used in the
  • Example 14 One-Pot Process for the Synthesis of 7-Azanorbornanes from the
  • the cycloadduct complex (1.0 mmol) prepared in Example 13 was dissolved in acetonitrile (4 g), protonated with triflic acid (3-5 eq), and treated with DDQ (1 eq).
  • the dark solution was transferred to a 50-ml round-bottomed flask with the aid of an additional 20 ml of acetonitrile, treated with 10% palladium-on-carbon (approximately 0.5 g, 40 mole%), and hydrogenated under 1 atm H 2 (balloon) for a suitable period of time (2-20 hours)
  • the picrate salt (both isomers combined) was crystallized from wet ethanol (m.p. 102-108 °C); Anal. Calcd. for C 15 H 18 N 4 O 9 ; C, 45.23; H, 4.55; N, 14.07. Found: C, 45.42; H, 4.59; N, 14.10.
  • This compound was prepared in 42% overall yield from pyrrole and dimethyl fumarate using the procedures set forth in Examples 11, 12 (using acetonitrile as a solvent), and 14 (hydrogenation solvent - methanol; reaction time - 2 h; workup A).
  • This compound was obtained as a 4:1 mixture of exo and endo isomers, respectively, in 39% overall yield from pyrrole and N-phenylmaleimide using the procedures set forth in Examples 11, 12 (using acetonitrile as a solvent), and 14 (hydrogenation solvent - methanol; reaction time - 2 hours; workup A).
  • This compound was formed when the synthesis of hexahydro-2-phenyl-4,7-imino-1H-isoindole-1, 3 (2H- dione was carried out using acetonitrile in the hydrogenation step of the method set forth in
  • Example 16 The nitrile formed in Example 16 (55 mg, 0.4 mmol) was treated with excess lithium aluminum hydride (30 mg, 0.79 mmol) in 10 ml ether with stirring. After 5 minutes (a white suspension formed), the reaction was quenched with methanol (0.1 g), then water (0.1 g), acidified with 1 M HCl, then basified with cone, NH 4 OH, and extracted with methylene chloride. Drying and evaporation of the extract yielded the corresponding primary amine as an oil (17 mg, 30%).
  • 1 H NMR (CDCl 3 ) ⁇ 3.18 (t, J
  • Example 21 The primary amine formed in Example 21 (17 mg, 0.121 mmol) was treated with 2,5- dimethoxytetrahydrofuran (25 mg, 0.189 mmol) in acetic acid (0.1 g) at 150°C for 5 minutes in an oil bath. Extraction of the basified (10% aqueous Na 2 CO 3 ) reaction mixture with methylene chloride yielded a mixture of products from which was obtained 8 mg (-30%) of crude exo-2-(1- pyrrolylmethyl) product by preparative thin layer chromatography using 1:1:8
  • Example 15 The aminoester formed in Example 15 (41 mg, 0.243 mmol) was treated with lithium aluminum hydride (10 mg, 0.264 mmol) in 5 ml ether. After 5 minutes, the reaction mixture was quenched with methanol, acidified with 1 M HCl, basified with cone. NH 4 OH, and extracted with methylene chloride. Evaporation of the extract yielded the desired product (11 mg, 32%).
  • active compounds are prepared through the Diels-Alder reaction of an N-(electron withdrawing- substituted) pyrrole with an arylsulfonyl (optionally substituted aryl or heterocyclic) acetylene.
  • the electron withdrawing group at the N 7 -position decreases the aromaticity of the pyrrole ring and activates the ring in favor of the cycloaddition reaction.
  • heterocyclic) acetylene is a 7-(electron withdrawing substituted)-2-(optionally substituted aryl or heteroaromatic)-3-arylsulfonyl-7-azabicyclo[2.2.1 ]-hepta-2,5-diene (compounds 23 and 32, Fig. 2).
  • This diene can be derivatized using conventional methods to a wide variety of 7-azabicyclo[2.2.1]- heptanes and -heptenes.
  • an R 3 alkyl or aralkyl group can be added by reacting the
  • Nonlimiting examples of addition reactions include hydrogenation, hydroboration, hydrohalogenation, hydroxylation, halohydrination, alkylation, carbene and dihalo carbene addition and epoxidation followed by ring opening reactions with nucleophiles such as alkoxide, amines,
  • alkylsulfide halide, and hydroxide.
  • the reactive chloro in compounds 24 and 25 ( Figure 2) is easily displaced by nucleophiles such as alkoxy, including methoxy, alkylthio, hydroxy, amino, cyano, azide, bromide, iodide, and
  • arylsulfonyl (optionally substituted aryl or heterocyclic) acetylene is carried out in excess N- (electron withdrawing substituted) -pyrrole or in a solvent, for example, toluene, tetrahydrofuran, dimethylformamide, diethoxyethane or other inert solvents.
  • a solvent for example, toluene, tetrahydrofuran, dimethylformamide, diethoxyethane or other inert solvents.
  • Any molar ratio of pyrrole to dienophile can be used that provides an acceptable yield of product, and typically ranges between 0.5:1 to 50:1, preferable (1-5):1.
  • the reaction is conducted at any temperature that produces the desired product, and typically, between room temperature and 150°C, until the reaction is completed, for typically between 1 hour and 72 hours at 1 atm. or elevated pressure in a sealed reactor.
  • Compound 25 is successfully deblocked by treatment with hydrobromic acid in acetic acid for 24 hours at room temperature.
  • the recovered starting material is essentially the pure endo isomer of 25, indicating some
  • 3- (thioalkyl) pyrrole including 3-(SCH 3 )pyrrole; 2,5-dialkylpyrrole, including 2,5-dimethylpyrrole; 3,4- dihaloalkylpyrrole, including 3,4- bis (trifluoromethyl) pyrrole, 2-alkylpyrrole, including 2-methylpyrrole; 2-alkoxyalkylpyrrole, including 2-methoxymethylpyrrole; 2- alkylthioalkylpyrrole, including 2- methylthiomethylpyrrole; 2- dialkylaminoalkylpyrrole, including 2- dimethylaminomethylpyrrole; alkyl pyrrole 2- acetate, including dimethylaminomethylpyrrole;
  • alkyl pyrrole 2-acetate including methyl pyrrole 2-acetate; 2-alkoxyalkoxyalkylpyrrole, including 2- methoxymethoxyethylpyrrole; 3-aryloxyalkylpyrrole, including 3-benzyloxymethylpyrrole; 2- alkoxypyrrole, including 2-methoxypyrrole; 3- alkoxypyrrole, including 3-methoxypyrrole; 3- aryloxypyrrole, including 3-benzyloxypyrrole; 3,4- dialkylpyrrole, and 3-alkylpyrrole, including 3- methylpyrrole and 3,4-dimethylpyrrole; 1,6 and 4,5- alkylidene pyrrole, including 4,5,6,7- tetrahydroindole and 2-methyl-4,5,6,7- tetrahydroindole.
  • the N-substituent on the pyrrole ring is any moiety that is electron withdrawing and that activates the ring toward cycloaddition with a dienophile.
  • the N-substituent is preferably carbomethoxy, however, other electron withdrawing moieties, including carbobenzyloxy, tert- butoxycarbonyl and optically active alkoxycarbonyl, including (+) and (-)-menthyloxycarbonyl can also be used.
  • Arylsulfonyl (optionally
  • heteroaromatic is reacted with the N-(electron withdrawing-substituted) pyrrole or its derivative.
  • the arylsulfonyl-(optionally substituted aryl or heteroaromatic)-acetylene can be prepared by methods known to those of skill in the art. In one embodiment, described in detail in the Example 26 below, the compound is prepared by reacting the lithium salt of methyl (aryl) sulfone with the desired optionally substituted aryl or
  • heteroaromatic acid chloride to produce a 1-(aryl or heteroaromatic)-2-arylsulfonylethanone, that is converted to the corresponding acetylene via an enolphosphate intermediate as described in Example 27 below.
  • Any optionally substituted aryl or heteroaromatic acid chloride can be used, including without limitation, the acid chloride of nicotinic acid, isonicotinic acid, 5-chloronicotinic acid, 6- methylnicotinic acid, 6-methoxynicotinic acid, 6- phenylnicotinic acid, 6-methylthionicotinic acid, 2-chloropyridine-4-carboxylic acid, 2,6- dimethylpyridine-4-carboxylic acid, l-methyl-2 (1H) - pyridone-3-carboxylic acid, 6-methylthionicotinic acid, 3-quinolinic acid, 4-quinolinic acid, 7- chloro-3-quinolinic acid, 6-methoxy-3-quino
  • Substituents that can be positioned on the aromatic or heteroaromatic group include, but are not limited to, alkyl, halo, aryl, alkoxy,
  • the aryl group attached to the sulfone can be any group that sufficiently activates the
  • acetylenic group to act as a dienophile toward the activated pyrrole and which does not interfere with the cycloaddition reaction.
  • Nonlimiting examples are phenyl, p-alkylphenyl, including p- methylphenyl; halophenyl, and including p- chlorophenyl, p-fluorophenyl, and p-nitrophenyl.
  • Fluoroalkanesulfonyl including CF 3 SO 2 and C 4 F 9 SO 2 , can also be used to activate an aryl- or
  • arylsulfonyl-(aryl or heteroaromatic)-acetylenes are described in Bhattacharya, S.N., et al,
  • arylsulfonyl (optionally substituted aryl or
  • heterocyclic) acetylene is set out in detail in the working examples below. These examples are merely illustrative, and not intended to limit the scope of the process or the compounds that can be made according to the process. As discussed above, this is a general method that can be combined with conventional synthetic techniques to provide a wide variety of products, all of which are considered to fall within the scope of the invention.
  • the compounds are numbered as illustrated in Figure 2.
  • Example 26 Preparation of 1-(2-chloro-5- pyridyl)-2-phenylsulfony1ethanone (9) To a cold solution (-30°C) of 20 g methyl phenyl sulfone in 400 ml dried tetrahydrofuran was added 128 ml 2.5M n-butyllithium (2.4 eq) slowly. The resulting solution was stirred at -30°C for 30 minutes. A solution of 26 g 6-chloronicotinyl chloride in 100 ml tetrahydrofuran was then added during a 20 minute period. After stirring at the same temperature for 30 minutes, the mixture was quenched by addition of sat. ammonium chloride (ca.
  • ketosulfones are obtained.
  • chlorophosphate was added in one portion. The mixture was stirred at room temperature overnight, then cooled to -78°C, and 1.35 g potassium t- butoxide is added in portions. The brown solution was stirred at -78°C for another 10 minutes and allowed to warm to ca. -30°C. Water was added and the aqueous layer extracted with methylene
  • Example 30 are hydrogenated to the corresponding substituted 7-aza-bicyclo[2.2.1]heptane analogs.
  • Example 32 Preparation of racemic epibatidine (19) and endo-epibatidine (19')
  • N-acetyl derivatives of epibatidine can be prepared from epibatidine and acetic anhydride in the presence of triethylamine.
  • other N-substituted 7-azabicyclo[2.2.1]heptanes described in Example 31 are deprotected to the corresponding free amine.
  • the amines are readily acylated to the amide, alkylated to the tertiary amine and
  • amines also form stable and water-soluble salts with organic and inorganic acids as preferred in the pharmaceutical
  • N-carbomethoxy-5-(2-chloro-5-pyridyl)- 7-aza-bicyclo[2.2.1]hept-2-ene 25 (16 mg) was dissolved in 3 ml methanol containing 7 mg 10% palladium on carbon. The mixture was hydrogenated under a slightly elevated pressure of hydrogen for one hour. After removal of catalyst and solvent, the residue was partitioned between ether and aqueous sodium bicarbonate. The aqueous layer was extracted with ether and the combined organic layer was dried over magnesium sulfate. Removal of solvent gave 10 mg of 7 -carbomethoxy-2-(3-pyridyl)- 7-azanorbornane (12) . MS (Cl) m/z 233 (M+1), H 1 -NMR 3.72, 3.66 (2s, N-COOCH 3 ).
  • 4-methylthiobenzoic acid 4- methylsulfinylbenzoic acid, 4-methylsulfonylbenzoic acid, 3, 5-difluorobenzoic acid, 2-naphthoic acid,
  • ketosulfones described in Example 37 are converted to the corresponding substituted aryl and aralkyl acetylenic derivatives.
  • Phenyl phenylsulfonyl acetylene 34 (84.3 mg, 0.35 mmol) was mixed with 0.42 g of N-carbomethoxy pyrrole (21). The mixture was heated to and maintained at 85°C for 48 hours. After removal of the excess pyrrole, the residue was chromatographed on silica gel column and eluted with 25-33% ethyl acetate in petroleum ether to give 30 mg (23%) of the adduct as a colorless oil. MS(CI) m/z
  • the bicyclic adduct 35 was reductively
  • Example 39 the other bicyclic adducts in Example 39 are converted to the corresponding 2-substituted aryl-7-aza-bicyclo[2.2.1]heptanes.
  • the bicyclic adduct 35 is reductively
  • Example 39 the other bicyclic adducts in Example 39 are converted to the corresponding 2-substituted aryl-7-aza- bicyclo[2.2.1]hept-5-enes.
  • alkylsulfide halide, hydroxide, etc.
  • Epibatidine 19 prepared in Example 32 is alkylated with methyl iodide (1.1 eq) in dry tetrahydrofuran at room temperature, followed by the usual isolation procedure, to give the
  • alkylation with ethyl iodide, isopropyl bromide, allyl bromide, cyclopropylmethyl bromide, benzyl chloride, 4-methoxybenzyl chloride, 3,4-dimethoxybenzyl chloride, phenethyl bromide, propargyl bromide, hydroxyethyl chloride and methoxyethyl iodide yield the corresponding
  • alkylated to their 7-N-alkyl is derivatives in the same manner.
  • Example 7 The N-acetyl derivative of epibatidine in Example 7 is reduced to the N-ethyl derivative by the treatment of lithium aluminum hydride in dry tetrahydrofuran at room temperature. Similarly, the 7-N-propionyl, N-benzoyl, N-phenylacetyl and N-2-furoyl derivative of epibatidine are reduced to the corresponding 7-propyl, 7-benzyl, 7-phenethyl and 7-(2-furfuryl) derivatives.
  • Example 45 Resolution of racemic compounds
  • the substituted 7-aza-bicyclo[2.2.1]heptane derivatives are resolved to their optical isomers by conventional methods including chromatography on a chiral column, fractional crystallization of diastereomeric salts of chiral acids and separation of the chiral ester or amide derivatives followed by regeneration of the optically pure enantiomers.
  • N-carbo-(-)-menthyloxy pyrrole is prepared from pyrrole and (-)-menthyl chloroformate by the method described above.
  • the chiral pyrrole is treated with the sulfonyl acetylene 22 or 34 as in Example 29 to give a diastereoisomeric mixture of the chiral cycloadduct 7-aza-bicyclo[2.2.1]hepta -2,5-diene derivative.
  • sodium amalgam as in Example 30
  • hepta-5-ene derivatives is obtained. These diastereomers are separated by chromatography to give the d and 1 enantiomers .
  • the optically active intermediates are each reduced and treated with HBr/AcOH to yield optically active epibatidine enantiomers. Similarly, other substituted
  • Scheme 5 illustrates a method for the
  • N-methyl isoindole was prepared according to the method set forth in B. Zeeh and K. H. Konig, Synthesis 1972, 45. b) Preparation of 2-(6-chloro-3-pyridyl)-3- phenylsulfonyl-1,4-dihydronaphthalene- 1,4-imine (44)
  • N-methyl isoindole (91 mg, 0.7 mmol) was mixed with 1-(6-chloro-3-pyridyl)-2-phenylsulfonyl acetylene 22 (139 mg, 0.5 mmol) in ethyl ether.
  • Racemic-epibatidine 19 (42 mg, 0.2 mmol) was mixed with 77 mg (0.7 mmol) freshly prepared ethyl formamidinate hydrochloride and 129 mg (1.0 mmol) diisopropyl ethylamine in 1 ml acetonitrile. After stirring at room temperature for 48 hours, the mixture was acidified with 1.0 M hydrogen chloride in ether. After evaporation in vacuo, the residue was separated on silica gel preparative thin layer chromatography, using a solvent system of 25% methanol in chloroform, to give 25 mg of the compound 45 as a hygroscopic solid. Yield 36%. MS (CI), 236, 238 (free base M+1). H 1 -NMR (CD 3 OD). ⁇ 3.40 (M, 1H, H 2 ). Example 51
  • Example 50 The process of Example 50 was repeated with the replacement of ethyl formamidinate by S-methyl pseudothiourea, S-methyl-N-methyl pseudothiourea, S-methyl-N-nitro pseudothiourea, or methyl
  • N-Formamidinyl epibatidine (12 mg, 0.04 mmol) 45 was dissolved in 2 ml methanol containing 5 mg 10% palladium on carbon. After hydrogenation under 1 atm hydrogen for 3 hours, the catalyst was removed by filtration. The filtrate was
  • Adduct 73 (445 mg) was dissolved in a mixture of 20 ml methanol and 10 ml tetrahydrofuran
  • Scheme 8 shows the preparation of compound 79.
  • Compound 80 was prepared according to the procedure set forth in J. Leroy et al, Synthesis, 1982 313.
  • Compound 82 was hydrogenated under high pressure of hydrogen, providing compound 79.
  • Humans, equine, canine, bovine and other animals, and in particular, mammals, suffering from pain can be treated by administering to the patient an effective amount of one or more of the
  • the active materials can be administered by any appropriate route, for example, orally,
  • salts or complexes refers to salts or complexes that retain the desired biological activity of the above-identified compounds and exhibit minimal undesired toxicological effects.
  • Nonlimiting examples of such salts are (a) acid addition salts formed with inorganic acids (for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, naphthalenedisulfonic acid, and polygalacturonic acid; (b) base addition salts formed with metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, sodium, potassium, and the like, or
  • the active compound is included in the
  • a preferred dose of the active compound for all of the above-mentioned conditions is in the range from about 0.0001 to 20 mg/kg, preferably 0.001 to 2 mg/kg per day, more generally 0.05 to about 0.5 mg per kilogram body weight of the recipient per day.
  • a typical topical dosage will range from 0.001% to 0.5% wt/wt in a suitable carrier.
  • the effective dosage range of the pharmaceutically acceptable derivatives can be calculated based on the weight of the parent compound to be delivered. If the derivative exhibits activity in itself, the
  • effective dosage can be estimated as above using the weight of the derivative, or by other means known to those skilled in the art.
  • the compound is conveniently administered in any suitable unit dosage form, including but not limited to one containing 0.001 to 1000 mg, preferably 0.01 to 500 mg of active ingredient per unit dosage form.
  • a oral dosage of 0.1 to 200 mg is usually convenient.
  • the active ingredient can be administered by the intravenous injection of a solution or
  • formulation of the active ingredient optionally in saline, or an aqueous medium or administered as a bolus of the active ingredient.
  • the concentration of active compound in the drug composition will depend on absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the
  • compositions and that the concentration ranges set forth herein are exemplary only and are not
  • the active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets.
  • the active compound can be any active compound.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid,
  • Primogel, or corn starch a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange
  • dosage unit form When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or other enteric agents.
  • the active compound or pharmaceutically acceptable salt or derivative thereof can be administered as a component of an elixir,
  • a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the active compound or pharmaceutically acceptable derivatives or salts thereof can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antibiotics, antifungals, antiinflammatories, or antiviral compounds.
  • Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline
  • the parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS).
  • PBS physiological saline or phosphate buffered saline
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art . The materials can also be obtained commercially from Alza Corporation.
  • a wide variety of biological assays have been used to evaluate the ability of a compound to act as an analgesic. Any of these known assays can be used to evaluate the analgesic ability of the compounds disclosed herein.
  • the Straub-tail reaction which is characteristic of opiate alkaloids, has been used as an assay for opiate agonists and antagonists. The assay is described in detail in Br. J. of Pharmacol . 1969, 36, 225.
  • Another accepted assay for analgesic activity is the hot plate analgesia assay, described in J. of Pharmacol . Exp. Therap. 1953, 107, 385.
  • An assay for the evaluation of the ability of a compound to bind to an opiate receptor is described in Mol . Pharmacol . 1974, 10 , 868.
  • peripheral anti-inflammatory activities also possess varying degrees of peripheral anti-inflammatory and analgesic effects which are useful for therapeutic applications.
  • the following assays for the evaluation peripheral anti-inflammatory activities are described in
  • Table 4 provides the analgesic activity measured as ED 50 ( ⁇ g/Kg) for selected compounds disclosed herein, as determined using the Straub- Tail assay, as describe by J. Daly et al. J. Am.

Abstract

7-Azabicyclo[2.2.1]-heptane and -heptene derivatives with analgesic or anti-inflammatory activity are disclosed that can be administered to a mammal, including a human, to treat pain and inflammatory disorders. A method for the treatment of pain or inflammatory disorders is also presented that includes administering an effective amount of the compound or its pharmaceutically acceptable salt or derivative, or mixtures thereof, to a host in need of analgesic anti-inflammatory therapy, optionally in a pharmaceutically acceptable carrier or diluent.

Description

7-AZABICYCLO-[2.2.1]-HEPTANE AND
-HEPTENE DERIVATIVES AS ANALGESICS
AND ANTI-INFLAMMATORY AGENTS
This invention is in the area of
7-azabicyclo [2.2.1] heptane and -heptene derivatives and their method of manufacture and pharmaceutical use.
BACKGROUND OF THE INVENTION
Opiates, and in particular, morphine, are routinely administered for the treatment of
moderate to severe pain. Agents that are less potent than morphine, such as codeine, mixed agonist-antagonist opioids, and non-opiate
analgesics, including non-steroidal
anti-inflammatory drugs (NSAIDS) are often used to relieve mild to moderate pain. Because of the well-known side effects of opiates, including chemical dependence and respiratory depression, there is a strong need for a non-opiate based analgesic for moderate to severe pain that would equal or exceed the potency of opiate analgesics, yet lack the serious side effects associated with the administration of opiates. Spande, et al., reported in 1992 that a potent nonopiate analgesic had been isolated from the skins of the Ecuadoran poison frog, Epipedobates tricolor. Spande, et al., 1992 J. Am. Chem. Soc , 114 , 3475-3478. The structure of the compound was determined by mass spectroscopy, infrared spectroscopy, and nuclear magnetic resonance as exo-2- (2-chloro-
5-pyridyl)-7-azabicyclo [2.2.1] heptane (see Figure 1). The compound, which was named epibatidine, is the first member of the class of
7-azabicyclo [2.2.1] heptane compounds to be found in nature. Limited pharmacological evaluation of epibatidine indicated that it is approximately 500 times more potent than morphine in eliciting the Straub-tail response, and that this effect is not reversed by the opiate antagonist naloxone. In the hot plate analgesia assay, epibatidine is
approximately 200 times as potent as morphine. It has also been determined that epibatidine has a negligible affinity for opiate receptors (1/8000 times that of morphine). Based on this data, it appears that epibatidine is a very potent analgesic that acts via a non-opiate mechanism. Since epibatidine was isolated from the skins of the Ecuadoran poison frog, its identification and characterization was not based on the synthesis of the compound. In fact, compounds in the family of 7-azabicyclo [2.2.1]-heptanes and -heptenes (also referred to as 7-azanorbornanes and
7-azanorbornenes, respectively) have historically been difficult to synthesize.
A possible entry into the 7-azabicyclo[2.2. 1] -heptane and -heptene ring systems involves the construction of the bicyclic ring through the 2 + 4 cycloaddition of appropriately substituted 3-vinyl pyridine and pyrrole fragments, as indicated below.
Figure imgf000004_0001
Unfortunately, pyrrole and its derivatives readily undergo substitution reactions upon treatment with dienophiles (Diels, 0. and Alder, K., Ann. 1932 498, 1.), with only a few exceptions reported
(Wittig, G., Angrew Chem. 1957 69, 245). The partial aromatic character of pyrrole limits its reactivity as a diene and Michael -type addition products usually dominate. (Jones, R.A., The
Chemistry of Heterocyclic Compounds, v.48 Pyrroles Wiley & Sons: New York. 1990.) Rate and yield enhancement of the Diels-Alder reaction between pyrroles and olefins have been obtained using Lewis acids (Donnini, G.P.; Just, G. J. Heterocycl . Chew. 1977, 14, 1423; Bansal, R.C.; McCulloch,
A.W.;McInnes, A.G. Can . J. Chem. 1969, 47, 2391), and high pressures (Kotsuki, H.; Mori, Y; Nishizawa, H.; Masamitsu, 0.; Matsμoka, K. Heterocycles, 1982, 19, 1915; Drew, M.G.B.; George, A.V.; Isaacs, is N.S.; Rzepa, H.S. T.C.S. Perkin Trans 1 , 1985, 1277), but thus far these approaches have been limited in scope. The limitation in these
reactions can often be traced to the inherent instability of the 7-azabicyclo[2-2.1]hept-2-ene products with respect to either retro-cycloaddition or retro-Mannich and re-aromatization pathways.
This is especially true for cases where the
resulting olefin is functionalized by
electron-withdrawing groups. (Altenbach, H.J.;
constant, D.; Martin, H.D.; Mayer, B.; Muller, M.; Vogel, E. Chem. Ber. 1991, 124, 791.) An
alternative method for the synthesis of the
7-azanorbornane system was reported by Fraser, et al. ( Can . J. Chem. 1970, 48, 2065) but the long synthetic route and drastic reaction conditions involved are unsuitable for the synthesis of epibatidine. For a review of 7-azanorbornene chemistry see: Kricka, L.J.; Vernon, J.M. Adv. in Heterocycl . Chem. 1974, 16, 87.
N-Carboalkoxypyrrole has been used in
Diels-Alder reactions with several acetylenic dienophiles to prepare norbornane derivatives.
None of these reported schemes, however, place an aromatic or heteroaromatic group in the important is 2 -position of the heptane or heptene ring.
(Altenbach, H-J., et al., Chem . Ber. 1991 , 124 ,
791; Altenbach, H-J., et al., Angew. Chem . Int . Ed.
Engl . 1992 21(10), 778; Gabel, N.W., J. Org. Chem. 1962, 27, 301; Toube, T.P. (1992) in: Pyrroles, Part
2 (Jones, R.A., ed.) John Wiley, New York, pp
92-95.)
In light of the analgesic potency of
epibatidine as well as the strong need for new potent, non-opiate analgesics, it would be useful to provide methods for the synthesis of 7- azabicyclo [2.2.1]-heptane and -heptene derivatives that have pharmacological activity, and in
particular, analgesic activity, or that can be derivatized to compounds with pharmacological activity.
Therefore, it is an object of the present invention to provide new 7-azabicyclo[2.2.1]- heptane and -heptene derivatives with analgesic activity.
It is another object of the present invention to provide methods for the synthesis of
7-azabicyclo[2.2.1]-heptane and -heptene
derivatives with analgesic activity.
It is still another object of the present invention to provide new methods for the treatment of pain.
SUMMARY OF THE INVENTION
7-Azabicyclo[2.2.1]-heptane and -heptene compounds are disclosed of Formula (I):
Figure imgf000007_0001
wherein:
R1 and R4 are independently hydrogen, alkyl, including CH3; alkylhydroxy, including CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; carboalkoxy, including carbomethoxy; allyl, aryl and thioalkyl, including -SCH3;
R3, R5 and R6 are independently hydrogen, alkyl, including -CH3; alkylhydroxy, including -CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including
CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; thioalkyl, including -SCH3; halo, including Cl, F; haloalkyl, including CF3; NH2, alkylamino or dialkylamino, including -N(CH3)2 and -NHCH3; cyclic dialkylamino, including
Figure imgf000008_0001
amidine, cyclic amidine including
Figure imgf000008_0002
and their N-alkyl derivatives;
Figure imgf000008_0003
-CO2H; CO2alkyl, including -CO2CH3; -C(O) alkyl, including -C(O)CH3; -CN, -C(O)NH2, -C (O)NH (alkyl), -C(O)N (alkyl) 2, including -C(O)N(CH3)2; allyl, -SO2 (alkyl), -SO2aryl, including -SO2(C6H5),
-S(O) alkyl, -S(O)aryl, aryl, heteroaryl;
Figure imgf000008_0004
R5 and R6 together can be alkylidene or
haloalkylidene, including -CH2- and -CF2-; epoxide (-O-); episulfide (-S-); imino (-N(alkyl)- or
-N(H)-) or a fused aryl or heteroaryl ring
including a fused phenyl ring;
R2 is independently hydrogen, alkyl, including CH3; alkenyl including -CH2-HC=CH2; alkylhydroxy, including -CH2-OH; alkyloxyalkyl including -CH2-O- (alkyl) , alkylamine, including -CH2NH2; carboxylate, C(O)Oalkyl, including CO2Me; C(O)Oaryl,
C(O)Oheteroaryl, COOaralkyl, -CN, Q, C(O)Q, - alkyl (Q), -alkenyl (Q), -alkynyl (Q), -O- (Q) -S-Q, - NH-Q or -N(alkyl)-Q;
R2 and R3 together can be -C (O) -NR8) -C (O) or CH(OH) -N(R8) -C(O) - wherein R8 can be alkyl, aryl including phenyl, or heteroaryl;
R7 is hydrogen, alkyl, including CH3, or CH2CH3; alkyl substituted with one or more halogens, including CH2CH2Cl; -CH2-(cycloalkyl), including -CH2-(eyelopropyl); -CH2CH=CH2, -CH2CH2 (C6H5),
alkylhydroxy, including CH2CH2OH, alkylamino (alkyl)2, including CH2CH2N (CH3)2 alkyloxyalkyl,
alkylthioalkyl, aryl, dialkyl to form a quarternary ammonium including
c
Figure imgf000009_0001
or
Figure imgf000010_0001
wherein R9 is hydrogen or alkyl;
wherein Y' is CN, NO2, alkyl, OH, -O-alkyl; wherein Z is O or S;
wherein R10 and R11 are each independently -O-, -OH, -O-alkyl, -O-aryl, -NH2, -NH(alkyl),
-N(alkyl)2, -NH(aryl) and -N(aryl)2;
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
and wherein the Q moiety can be optionally substituted with 1 to 3 W substituents; and
W is alkyl, including CH3; halo, including Cl and F; aryl, heteroaryl, OH, oxyalkyl, including -OCH3; SH, thioalkyl, including -SCH3; -SO (alkyl) including -SOCH3; -SO2alkyl, including -SO2CH3;
-OCH2CH=CH2, -OCH2(C6H5), CF3, CN, alkylenedioxy, including -methylenedioxy-; -CO2H, -CO2alkyl, including -CO2CH3; -OCH2CH2OH, -NO2, -NH2, -NH (alkyl), including -NHCH3; -N (alkyl)2, including -N(CH3)2; -NHC(O) alkyl, including -NHC(O)CH3; -SO2CF3, or -NHCH2aryl, including -NHCH2 (C6H5) ; and wherein
the - - - indicates an optional double bond.
These compounds have central and peripheral analgesic activity, and, or alternatively,
anti-inflammatory activity, and thus can be
administered to a mammal, including a human, to treat pain and inflammatory disorders. A method for the treatment of pain is also presented that includes administering an effective amount of the compound or its pharmaceutically acceptable salt or derivative, or mixtures thereof, to a host in need of analgesic therapy, optionally in a
pharmaceutically acceptable carrier or diluent.
Facile processes for the preparation of the disclosed 7-azabicyclo[2.2.1]-heptane and -heptene derivatives are also provided. In one embodiment, the pharmaceutically active compounds, or their precursors, are prepared by the cycloaddition reaction of pentaammineosmium(II) complexes of pyrroles with dipolarophiles, including 3 -vinyl pyridine derivatives. In another embodiment, pharmaceutically active compounds or their
precursors, are prepared by the Diels-Alder
reaction of an N- (electron
withdrawing-substituted) pyrrole with an arylsulfonyl (optionally substituted aryl, alkyl, heterocyclic or heteroaryl) acetylene.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is an illustration of the chemical structure of exo-2 -(2-chloro-5-pyridyl)-7- azabicyclo[2.2.1]heptane (epibatidine).
Figures 2a and 2b are schematic illustrations of processes for the preparation of active
compounds through the Diels-Alder reaction of an N- (electron withdrawing substituted) pyrrole with an arylsulfonyl (optionally substituted aryl or heterocyclic) acetylene.
DETAILED DESCRIPTION OF THE INVENTION
I. Definitions The term alkyl, as used herein, refers to a saturated straight, branched, or cyclic (or a combination thereof) hydrocarbon of C1 to C10, and specifically includes methyl, ethyl, propyl, isopropyl, cyclopropylmethyl, cyclobutylmethyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methylpentyl, 2,2-dimethylbutyl, 2,3-dimethyl- butyl, heptyl, octyl, nonyl, and decyl.
The term lower alkyl, as used herein, refers to a C1 to C6 saturated straight, branched, or cyclic (in the case of C5-6) hydrocarbon, and specifically includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl,
cyclopropylmethyl, pentyl, cyclopentyl,
cyclobutylmethyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methylpentyl,
2,2-dimethylbutyl, and 2,3-dimethylbutyl. The term alkylamino refers to an amino group that has an alkyl substituent.
The term alkynyl, as referred to herein, refers to a C2 to C10 straight or branched
hydrocarbon with at least one triple bond.
The term lower alkynyl, as referred to
herein, refers to a C2 to C6 alkynyl group,
specifically including acetylenyl and propynyl.
The term aryl, as used herein, refers to phenyl, or substituted phenyl, wherein the
substituent is halo, alkyl, alkoxy, alkylthio, haloalkyl, hydroxyalkyl, alkoxyalkyl,
methylenedioxy, cyano, C(O) (lower alkyl), carboxy, CO2alkyl, amide, amino, alkylamino and dialkylamino, and wherein the aryl group can have up to 3
substituents.
The term halo, as used herein, includes fluoro, chloro, bromo, and iodo.
The term aralkyl refers to an aryl group with an alkyl substituent.
The term alkaryl refers to an alkyl group that has an aryl substituent, including benzyl,
substituted benzyl, phenethyl or substituted phenethyl, wherein the substituents are as defined for aryl groups.
The term heteroaryl or heteroaromatic, as used herein, refers to an aromatic moiety that includes at least one sulfur, oxygen, or nitrogen in the aromatic ring. Nonlimiting examples are furyl, pyridyl, pyrimidyl, thienyl, isothiazolyl,
imidazolyl, pyrazinyl, benzofuranyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl,
pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl,
isothiazolyl, 1,2,5-thiadiazolyl, isooxazolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cinnolinyl, phthalazinyl, quinoxalinyl, xanthinyl, hypoxanthinyl, pteridinyl, 5- azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolopyrimidinyl.
The term organic or inorganic anion refers to an organic or inorganic moiety that carries a negative charge and can be used as the negative portion of a salt.
The term "pharmaceutically acceptable
application" refers to an organic or inorganic moiety that carries a positive charge and that can be administered in association with a
pharmaceutical agent, for example, as a
counteraction in a salt.
The term enantiomerically enriched composition or compound" refers to a composition or compound that includes at least 95% by weight of a single enantiomer of the compound.
The term pharmaceutically active derivative refers to any compound that upon administration to the recipient, is capable of providing directly or indirectly, the compounds disclosed herein.
As used herein, the term dipolarophile refers to a compound or moiety that reacts with a dipolar species to form a cycloaddition product.
As used herein, the term dienophile refers to a compound or moiety that reacts with a diene to form a cycloaddition product.
As used herein, the term η refers to a pi-orbital complex of an unsaturated compound with a metal, and wherein the superscript after the η refers to the number of sp2 carbon atoms bonded to the metal.
The term electron withdrawing substituent as used herein refers to a substituent that pulls electron density from the moiety to which it is attached through induction or resonance. A wide variety of electron withdrawing substituents are well known to those skilled in organic synthesis.
II. Examples of Active Compounds
7-Azabicyclo[2.2.1]-heptane and -heptene derivatives of Formula (I) are provided that have central and peripheral analgesic and, or
alternatively, anti-inflammatory activity, and thus can be administered to a mammal, including a human, to treat pain and inflammation. A method for the treatment of pain is also presented that includes administering an effective amount of the compound or its pharmaceutically acceptable salt or
derivative, or mixtures thereof, to a host in need of analgesic therapy, optionally in a
pharmaceutically acceptable carrier or diluent.
The numbering scheme for 7-azabicyclo
[2.2.1]-heptane and -heptene derivatives is as illustrated below.
Figure imgf000018_0001
The 7-azabicyclo[2.2.1]-heptanes and -heptenes disclosed herein can exhibit a number of
stereochemical configurations. As discussed above, the compounds are prepared in a Diels-Alder
cycloaddition reaction of a dienophile with a pyrrole, or a modification of the Diels Alder reaction involving the reaction of a dipolarophile with a pentaammineosmium(II) activated pyrrole. In the transition state of the cycloaddition reaction, there are two possible relative orientations of the diene or dienophile, referred to as endo and exo. Endo configurations are formed when other
unsaturated groups in the dienophile (or
dipolarophile) lie near the developing double bond in the diene. Exo configurations are formed when other unsaturated groups in the dienophile (or dipolarophile) lie away from the developing double bond in the diene. Depending on the substitution on the carbon atoms, the endo and exo orientations can yield different stereoisomers.
Carbon atoms 2, 3, 5 and 6 in
7-azabicyclo[2.2.1]heptanes and carbon atoms 2 and 3 or 5 and 6 in 7-azabicyclo[2.2.1]heptanes are chiral when attached to different substituents. If at least one of the carbon in the molecule are chiral, the unsymmetrically substituted bicyclic compounds exist as one or more diastereomeric pairs. The R groups in the active compounds described herein can also include chiral carbons, and thus, optically active centers.
It is sometimes found that one or more
enantiomers of a biologically active compound is more active, and perhaps less toxic, than other enantiomers of the same compound. Such
enantiomerically enriched compounds are preferred for pharmaceutical administration to humans or other hosts.
One of ordinary skill in the art can easily separate the enantiomers of the disclosed compounds using conventional processes, and can evaluate the biological activity of the isolated enantiomers using methods disclosed herein or otherwise known. Through the use of chiral NMR shift reagents, polarimetry, or chiral HPLC, the optical enrichment of the compound can be determined.
Classical methods of resolution include a variety of physical and chemical techniques. For example, since the compound has a basic amine (N7) , it can be reacted with a chiral acid to form diastereomeric salts that may possess significantly different solubility properties. Nonlimiting examples of chiral acids include malic acid, mandelic acid, dibenzoyl tartaric acid,
3-bromocamphor-B-sulfonic acid, 10 -camphorsulfonic acid, and di-p-toluoyltartaric acid, and
(-)-menthyl chloroformate. Similarly, acylation of a free amine or hydroxyl group in the molecule with a chiral acid also results in the formation of a diastereomeric amide or ester whose physical properties may differ sufficiently to permit separation. Enantiomerically pure or enriched compounds can be also obtained by passing the racemic mixture through a chromatographic column that has been designed for chiral separations, including cyclodextrin bonded columns marketed by Rainin Corporation.
The following are nonlimiting examples of specific compounds that fall within the scope of the invention. These examples are merely
exemplary, and are not intended to limit the scope of the invention.
(A) Epibatidine isomers:
1-7-aza-2-exo-(2-chloro-5-pyridyl)-bicyclo[2.2.1] heptane and its pharmaceutically acceptable salts, including the hydrochloride salt; 1 -7 -aza-2 - exo- (2-chloro-5-pyridyl)-bicyclo[2.2.1] heptane and its pharmaceutically acceptable salts, including the hydrochloride salt;
d and 1-7-aza-endo-(2-chloro- 5-pyridyl)- bicyclo[2.2.1] heptane and its
pharmaceutically acceptable salts, including the hydrochloride salts;
(B) d and 1 enantiomers of the 7-aza-bicyclo[2.2.1]heptane derivatives containing the following substituents:
A combination of 7-methyl, 7-allyl-,
7-cyclopropylmethyl, 7-cyclobutylmethyl,
7-phenethyl, 7-hydroxyethyl, 7-methoxyethyl,
7-methylthioethyl, 7-dimethylaminopropyl, 7- formamidinyl, 7-(2-chloroethyl), 7-disodium
phosphate and 7-(4-methoxybenzyl) substituents with a 2-exo-(2-chloro-5-pyridyl) substituent;
2-exo-(3-pyridyl);
2 - endo-(3-pyridyl); 7-methyl-2-exo-(3-pyridyl);
7-cyclopropylmethyl-2-exo-(3-pyridyl); 7- phenethyl-2-exo-(3-pyridyl);
2-exo-(4-pyridyl); 7-methyl-2-exo-
(4-pyridyl); 7- allyl-2-exo-(4-pyridyl);
7-cyclopropylmethyl-2-exo-(4-pyridyl);
2-exo-(3-chloro-4-pyridyl);
7-cyclopropylmethyl-2- exo-(3-chloro-4-pyridyl);
7-phenethyl-2-exo-(3-chloro-4-pyridyl) 2-exo-(2 chloro-3-pyridyl); 2-exo-(2-chloro-4-pyridyl);
2-exo-(2-fluoro-5-pyridyl);
2-exo-(2-methoxy-5-pyridyl); 2-exo-(2-methylthio-
5-pyridyl); 2-exo-(2-methyl-5-pyridyl);
2-exo-(2-dimethylamino-5-pyridyl); 2-exo-(2- hydroxy-5-pyridyl) and their 7-cyclopropylmethyl derivatives;
The exo and endo isomers of:
2-phenyl; 2-(3-chlorophenyl);
2-(3-dimethylaminophenyl); 2-(3- trifluoromethylphenyl);
2-(3,4-methylenedioxyphenyl); 2-(3,4- dimethoxyphenyl); 2-(4-fluorophenyl);
2-(4-hydroxyphenyl); 2-(4-methylthiophenyl); 2-(4-methylsulfonylphenyl), 2-(3,5- difluorophenyl); 2-(2-chlorophenyl);
2-(2-naphthyl); 2-(7-methoxy-2-naphthyl);
2-(5-chloro-2-thienyl); 2-(chloro-5-thiazolyl); 2-(4-pyrimidyl); 2-(2-chloro-5-pyrimidyl); 2-(5- chloro-2-pyridazinyl); 2-(1,2,5-thiadiazol-3-yl);
2-(5-dimethylamino-2-furyl); 2-(5-indolyl);
2-(5-fluoro-3-indolyl); 2-(5-methoxy-3-indolyl);
2-(4-chlorobenzyl); 2-(5-chloro-3-pyridylmethyl); 2-(4-pyridylmethyl); 2-nicotinyl; 2-(6- chloronicotinyl); 2-isonicotinyl;
2-(3-chloro-isonicotinyl); 2(4-chlorobenzoyl);
2-(4-dimethylaminobenzoyl); 2- (3,4- dimethoxybenzoyl) and their 7-methyl,
7-cyclopropylmethyl, 7-allyl and 7-phenethyl derivatives.
(C) The exo and endo isomers of
7-aza-2-(2-chloro-5-pyridyl)-bicyclo[2.2.1]heptane containing the following substituents at the 1, 2, 3, 4, 5 or 6 positions:
1 or 4-methyl; 1 or 4-hydroxymethyl;
1 or 4- methoxymethyl; 1 or 4-carbomethoxy; 1 or
4-allyl; 1 or 4-benzyl; 1 or 4-(4-fluorobenzyl); 1 or 4-(4-methoxybenzyl); 1,4-dimethyl;
1, 4-bis(hydroxymethyl); 1,4-bis(methoxymethyl); 1,6 or 4, 5-butylidene;
Endo or exo-3-methyl;
3 -hydroxymethyl; 3 -methoxymethyl; 3-carbomethoxy;
3-carboxy; 3-carbamyl; 3-cyano; 3-acetyl;
3-aminomethyl; 3-dimethylaminomethyl,
3-methylthiomethyl; 3-phenylsulfonyl,
3-methanesulfonyl; 3 -benzyl; 3-allyl; 3-cyano-
1,4-dimethyl; 3 -hydroxymethyl-1,4-dimethyl,
3-methoxymethyl-1, 4-dimethyl;
3-methylthiomethyl-1, 4-dimethyl; 5,6- bis (trifluoromethyl); 5 or 6-methoxy; 5 or
6-methyl; 5,6-dimethyl; 5,6-dicarbomethoxy; 5,6-bis (hydroxymethyl); 5,6-bis(methoxymethyl); 5 or 6-chloro; 5 or 6-hydroxy; 5,6-dehydro;
5,6-dehydro-1,4-dimethyl; 3,3-dimethyl; 2-methyl;
2,3-dimethyl, 5,6-methylene;
and their corresponding 7-methyl, 7- cyclopropylmethyl, 7-allyl, 7-phenethyl and
7-(4-fluorobenzyl) derivatives.
(D) 7-Aza-2-(2-chloro-5-pyridyl)- bicyclo[2.2.1]hept-2-ene and its 7-methyl, 7-allyl, 7-cyclopropylmethyl, 7-phenethyl and
7-(4-methoxyphenethyl) derivatives; and
the corresponding 1,4-dimethyl; 1 or
4-methyl; 5,6-dimethyl and 5, 6-bis (trifluoromethyl) analogs.
(E) Benzo [5a, 6a] epibatidine and its N-methyl derivative; 2,3-dehydroepibatidine; 5,6- bis (trifluoromethyl) deschloroepibatidine; 2- carbomethoxy-7-methyl-7-azabicyclo[2.2.1]heptane;
2-cyano-7-methyl-7-azabicyclo[2.2.1]heptane; trans- 2,3-bis-carbomethoxy-7-azabicyclo[2.2.1]- heptane; exo-2-amino-7-methyl-7-azabicyclo[2.2.1]- heptane; exo-2-(1-pyrrolylmethyl)-7-methyl-7-azabicyclo
[2.2.1] heptane; exo-2-hydroxymethyl-7-methyl-7- azabicyclo[2.2.1]heptane; exo-2-hydroxymethyl-7- methyl-2-azabicyclo[2.2.1]heptane.
III. Methods for the Synthesis of Optionally
Substituted 7-Azabicyclo[2.2.1]-heptanes and -heptenes
A. SYNTHESIS OF THE 7-AZABICYCLO[2.2.1]-HEPTANE OR -HEPTENE RING SYSTEM FROM PYRROLES VIA PENTAAMMINEOSMIUM(II) COMPLEXES
It has been discovered that
7-azabicyclo[2.2.1]-heptane and -heptene
derivatives can be prepared by combining a
dipolarophile with an optionally substituted pyrrole that has been complexed with
pentaammineosmium (II).
Any dipolarophile can be used in this reaction that reacts with the pentaammineosmium pyrrole complex to provide an optionally substituted
7-azabicyclo[2.2.1]-heptene, which is easily converted to the corresponding 7-azabicyclo [2.2.1] - heptane. Examples of dipolarophiles include compounds of the is structure Z1-C=C-Z2, wherein Z1 and Z2 are independently electron withdrawing groups, including without limitation, esters, nitriles, ketones, aldehydes, amides, -NO2,
sulfones, anhydrides, -CF3, pyridinium salts, and for example, CO(alkyl, aryl or heteroaryl), C(O)H, CO2 (alkyl, aryl, or heteroaryl), SO2 (alkyl, aryl, or heteroaryl), or wherein Z, and Z2 are together
(CO)2O, or (CO)2N. Specific compounds include
N-methylated and 6-carboxylated pyridyl acrylates, alkyl acrylate, alkyl methacrylate, pyridyl
substituted vinyl sulfones, acrylonitriles, anhydrides, maleimides,
alpha-methylene-δ-butyrolactone, maleates, and fumarates.
Analogously, any optionally substituted pyrrole can be used that on complexation with pentaammineosmium (II) will react with a
dipolarophile. Examples of suitable pyrroles include 2,5-dialkylpyrrole, 2-alkylpyrrole,
3-alkylpyrrole, 1-alkylpyrrole, 3,4-dialkylpyrrole, pyrrole, 1-silylated pyrrole, (1, 2, or 3)alkoxy or amino pyrrole, 2, 3-dialkoxypyrrole, 2,5- dialkoxypyrrole, and 3,4-dialkoxypyrrole.
As shown below in Scheme 1, a complex is readily formed between pyrrole and the 7r-base pentaammineosmium (II) in which the
osmium coordinates the heterocycle across C2 and C3. At 20°C, this species is in equilibrium with its linkage isomer in which the metal binds across C3 and C4. Although the 3,4-η species is only a minor component (ΔGiso>3 kcal/mol), the metal coordination in this species renders the remaining portion of the pyrrole an azomethine ylide
(R2C+-N(R) -C-R2«R2C=N+ (R) -CR2), and thereby
dramatically enhances the tendency of the ligand to undergo a 1,3-dipolar cycloaddition with suitable dipolarophiles.
Figure imgf000025_0001
Scheme 1. Dipolar cycloaddition of η2-pyrrole complex with dipolarophile.
Os (II) = [Os(NH3)5] (OTf)2.
The resulting 7-azabicyclo[2.2.1]hept-5-ene ligand is unstable with respect to cycloreversion, but metal coordination greatly stabilizes the complex and thus provides the opportunity to carry out functional group transformations while keeping the bicyclic framework intact. For example, derivatization of electron-withdrawing groups in the 2- or 3-positions of the norbornene framework, using conventional processes, provides a wide array of functionalized 7-azanorbornenes. Specifically, as shown in Scheme 2 below, the exo-carbonyl cycloadduct complex 2, prepared in a one-pot synthesis from 2,5-dimethylpyrrole, is reduced to the corresponding alcohol and oxidatively
decomplexed to yield the relatively inaccessible 5-hydroxymethyl-7-azanorbornene 3.
Figure imgf000026_0001
71%
overall
Scheme 2 Synthesis of a 5-Substituted
7-azanorbornene (Os (II) = [Os (NH3)5] (Otf)2); DMAc = N,N- dimethylacetamide; Otf- = CF3SO3
This approach can be used to construct the epibatidine ring system if a 3 -vinyl pyridine is used as the dipolarophile. The use of
methyl-trans-3-(3-pyridyl)-acrylate in the above reaction sequence (using the 2,5-dimethylpyrrole complex shown in Scheme 2), yields compound 4, shown below, which contains the carbon skeleton of the natural product.
Figure imgf000026_0002
Epibatidine has no substitution at the
bridgehead carbons (C1 and C4) . The reactivity of simple pentaammineosmium (II) - pyrrole complexes with dipolarophiles decreases in the order
2,5-dimethylpyrrole >N-methylpyrrole>pyrrole. Generally, additional activation of the
dipolarophile, by careful selection of the electron withdrawing group attached to the olefin, or high pressure is required to obtain cycloadducts without substitution at the bridgehead positions. Although the parent pyrrole complex gives complex mixtures, the N-methyl pyrrole reacts with the N-methylated and 6-carboxylated pyridyl acrylates to yield cycloadducts 5 and 6 as single diastereomers.
Figure imgf000027_0001
An alternative method for stabilization of the azabicyclo[2.2.1]heptane nucleus involves
protonation of the secondary amine (and pyridyl group) followed by oxidative removal of the metal and in si tu hydrogenation of the azanorbornene. An example of this method is shown in Scheme 3 below for the synthesis of the 1,4-dimethyl-exo- carbomethoxy-norchloroepibatidine 7.
(
Figure imgf000028_0001
Scheme 3. Decomplexation and hydrogenation to generate a 7-azanorbornane.
([Os]2+=[Os(NH3)5] (Otf)2)
The process for preparing optionally
substituted 7-azabicyclo[2.2.1]heptanes and
7-azabicyclo[2.2.1]hept-5-enes via
pentaammineosmium (II) complexes proceeds in three steps. In the first step, the optionally
substituted pyrrole is treated with
pentaammineosmium (II). An excess of the pyrrole complex is usually preferred.
Pentaammineosmium (II) is generated in situ by the reduction of pentaammineosmium (III) with a one electron reducing agent that has a reducing
potential of less than -0.75 volts versus hydrogen. The counteranion of pentaammineosmium (II) can be any anion that does not adversely affect the overall reaction. Typical counteranions are CF3SO3- (Otf-), PF6, X-, and (alkyl or aryl)SO3-.
Any chemical or electrochemical reducing agent that can reduce the osmium complex from a III valence state to a II valence state and which does not cause or participate in undesired side
reactions is suitable. Examples of appropriate reducing agents include magnesium, zinc, aluminum, sodium, cobaltocene and electrochemical reduction. In a preferred embodiment, activated magnesium powder is used. The optionally substituted pyrrole,
pentaammineosmium (III), and reducing agent are stirred at a temperature ranging between 0°C and 50°C until the desired organometallic complex is formed, typically between 0.1 and 1.0 hours. The reaction can be carried out in a polar or nonpolar solvent, including but not limited to
N,N-dimethylacetamide, N,N-dimethylformamide, water, methanol, acetonitrile, acetone,
dimethylsulfoxide, CH2Cl2, or dimethoxyethane. The reaction is carried out in the absence of 02, and typically under nitrogen, at a pressure of 1 atm or greater.
In the second step of the process, the dipolarophile is added to the stirring solution of the pyrrole pentaammineosmium (II) complex to produce an optionally substituted 7- azabicyclo[2.2.1]hept-5-ene. Any molar ratio of dipolarophile to pyrrole can be used that provides the desired results. Typically, a molar ratio of dipolarophile to pyrrole ranging between
approximately 1 and 10 provides a suitable yield of product. The reaction solution is stirred at a temperature ranging between 10 and 50°C until the product is formed, typically between 1 and 24 hours.
In an optional step after the bicyclic ring system is formed, and while pentaammineosmium is still complexed to the pi-orbital of the heptene moiety, functional groups on the bicyclic ring can be derivatized using conventional processes. For example, esters can be reduced to alcohols, nitriles to amines, sulfones to sulfides, nitro groups to amines, and amides to amines. Sulfones and carboxylates can be reductively eliminated using the Barton decarboxylation procedure. High temperatures and strong bases should be avoided in the functionalization procedures to avoid ring disruption and unwanted side reactions.
In the third step of the reaction, the
pentaammineosmium (II) complex is removed from the optionally substituted
7-azabicyclo [2.2.1] hept-5-ene by, for example, treatment with cerium (IV) or oxygen in acidic solution. For example, the
7-azabicyclo[2.2.1]hept-5-ene can be treated with one equivalent of cerium reagent at 20°C in a polar solvent such as acetonitrile. Appropriate reagents include Ce (NO3)6 (NH4)2,DDQ, and other inorganic or organic oxidants with E > +.70 volts versus
hydrogen. Alternatively, the osmium reagent can be removed by heating the complex as necessary, usually between approximately 50°C and 100°C.
Using the method of synthesis described above, a wide variety of substituted 7-azanorbornanes and 7-azanorbornenes can be prepared. Examples of representative compounds are summarized in Tables 1 and 2.
Some of them are useful as intermediates for the synthesis of desired compounds containing complex heteroaryl or polar substituents as R2 and/or R3.
Figure imgf000031_0001
Figure imgf000032_0001
Methods for preparing compounds of Formula (I) via derivatization of a 5,6-η2-7- aza-bicyclo[2.2.1]hept-5-ene are set out below.
These examples are merely illustrative, and are not intended to limit the scope of the invention.
Example 1 Preparation of
1,4-Dimethyl-2-exo-(hydroxymethyl)- 7-azabicyclo[2.2.1]hept-5-ene (8)
A solution of the 5 , 6 -η2 osmium complex of compound 8 (727 mg, 1.0 mmol) in 2 grams
acetonitrile was protonated with excess triflic acid (250 mg, 1.67 mmol) and treated at -10°C with a likewise-cooled solution of eerie ammonium nitrate (560 mg, 1.02 mmol) and triflic acid (560 mg, 3.73 mmol) in 2 grams acetonitrile. Water (1-2 ml) was added to dissolve the precipitated salts, the mixture made basic with 40 ml 10% aqueous sodium carbonate and the product extracted with 5 X 20 ml methylene chloride. The extract was dried over MgSO4 and the solvent evaporated, yielding 147 mg of brown oil. The crude product was purified by silica gel column chromatography using 1:10 of 15 wt % NH3 in methanol/methylene chloride, yielding 62 mg (41%) of pure 8. (oil, Rf = 0.5). 1H NMR (300 MHz, CDCl3) d 6.31 (d, J = 5.3 Hz, 1H), 6.09 (d, J = 5.3 Hz, 1H), 3.99 (dd, J = 10.3, 2.1 Hz, 1H), 3.67 (dd, J = 10.3, 2.1, 1H), 3.6-2.8 (v br, ~2H, OH and NH), 1.4-1.8 (m, 3H), 1.48 (s, 3H), 1.47 (s, 3H); 13C NMR (75 MHz, CDCl3) d 145.2 (CH), 141.5 (CH), 69.9 (C), 67.0 (C), 61.5 (CH2), 41.7 (CH), 37.0 (CH2), 18.9 (CH3), 15.7 (CH3); This material was further characterized by conversion to the picrate salt. m.p. 186-188°C; Anal. Calcd. for C15H18N4O8: C, 47.12; H, 4.75; N, 14.65. Found: C, 46.96; H, 4.52; N, 14.66. Example 2 Preparation of N-CBZ-1,4-Dimethyl-2- exo-(hydroxymethyl)-7- azabicyclo[2.2.1]hept-5-ene (9) and N,O-Bis-CBZ-1,4-Dimethyl-2-exo- ( hydroxymethyl)-7- azabicyclo[2.2.1]hept-5-ene (10)
The crude aminoalcohol 8 obtained from 1.0 mmol of the osmium complex as described above was suspended in a solution of aqueous Na2CO3 (0.38 grams in 2 grams water), and the mixture chilled to 0°C. Benzyl chloroformate (510 mg, 3 mmol) was added, and the mixture allowed to warm to room temperature with vigorous stirring. After 20 hours at 25°C the mixture was extracted with methylene chloride, and the extracts dried and
rotoevaporated, yielding 0.4 grams of brown oil. The crude material was chromatographed twice using 1:8 ethyl acetate/petroleum ether, yielding 43 mg (10%) of 9 and 64 mg (22%) of 10 (Rf = 0.5 and 0.1, respectively) For 9: 1H NMR (300 MHz, CDCl3) d
7.32 (m, 5H, Phenyl), 6.06 (ABq, J = 5.7 Hz, 2H, H5 and H6), 5.04 (s, 2H, OCH2Ph), 3.69 (m, 2H, CH2OH), 2.18 (br s, 1H, OH), 1.75 (2Xs, 6H, CH3), 1.7 (m, overlap, 1H), 1.55 (m, 2H); 13C NMR (75 MHz, CDCl3) d 155.2 (CO), 140.5 (CH, C5 or C6),
140.2 (CH, C6 or C5), 136.4 (C, ipso), 128.3 (CH),127.9 (CH), 127.8 (CH), 71.1 (C), 69.0 (C), 66.4 (CH2OH), 63.0 (CH2), 45.6 (CH), 37.7 (CH2), 19.4 (CH3), 16.8 (CH3). For 10: 1H NMR(300 MHz, CDCl3) d 7.37 (m, 5H, Phenyl), 7.32 (m, 5H,
Phenyl) , 6.07 (ABq, J = 5.5 Hz, 2H, H5 and H6), 5.16 (s, 2H, OCH2Ph) , 5.05 (ABq, J = 13.5 Hz,2H, OCH2Ph), 4.33 (dd, J = 10.5, 7 Hz, 1H, 1/2
CH2OCBZ) , 4.06 (dd, J = 10.5, 7.5 Hz, 1H,
1/2CH2OCBZ) 1.94 (m, 1H, H2) , 1.79 (s, 3H, CH3)
1.75 (s, 3H, CH3), 1.60 (dd, J = 11.4, 9 Hz, 1H, H3endo), 1.4 (dd, J = 11.4, 3.6 Hz, H3exo). 13C NMR (75 MHz, CDCl3) d 155.0 (CO), 154.9 (CO) , 140.5 (CH, C5 or C6), 140.5 (CH, C6 or C5), 136.4 (C, ipso), 135.2 (C, ipso), 128.5 (ove rlap of 2 X CH), 128.4 (CH), 128.3 (CH), 128.0 (CH), 127.8 (CH), 70.8 (C), 69.6 (overlap of 2X CH2), 68.9 (C), 66.3 (CH2O), 43.2 (CH, C5), 38.7 (CH2, C6), 19.3 (CH3), 17.0 (CH3).
Example 3 Preparation of 1,4-Dimethyl-2-endo-
(3'-pyridyl)-3-exo-(hydroxymethyl)- 7-azabicyclo[2.2.1]hept-5-ene (11) The corresponding 5,6-η2 osmium complex was treated as described above for compound 8.
Diagnostic ΗH NMR information: 6.43 (d, J = 6H, 1H, H5 or H6), 6.0 (d, J = 6 Hz, 1H, H6 or H5), 4.0 (dd, J = 10,2.5 Hz, 1H, 1/2 CH2OH), 3.75 (dd, J = 10, 2.5 Hz, 1/2 CH2OH), 1.55 (s, CH3), 1.38 (s, CH3).
Example 4 Preparation of 1,4-Dimethyl-2-exo-
(hydroxymethyl)-7- azabicyclo[2.2.1]heptane (12)
A sample of crude compound 8 (85 mg, 0.56 mmol) was stirred with 30 mg 10% Pd-on-C and 0.5 g methanol in a 5-ml round-bottomed flask under 1 atmosphere of H2 for 30 minutes. The reaction mixture was filtered through celite and evaporated, yielding 78 mg of oil. Purification by preparative thin layer chromatography (0.25 mm, 20 X 20 cm;
Eluent = 1:6 15% NH3 in MeOH, CH2Cl2), yielded 14 mg
(16%) of pure 12 (Rf = 0.5) 1H NMR (300 MHz, CDCl3) d 3.89 (br, 2H, NH and OH), 3.82 (d J = 10.6 Hz,
1/2 CH2OH) , 3.38 (d, J = 10.6 Hz, 1/2 CH2OH) , 1.7- 1.5 (m, 7H, 3 X CH2 + CH) , 1.41 (s, 3H, CH3) , 1.37
(s, 3H, CH3) ; 13C NMR (75 MHz, CDCl3) d 66.8, 64.0, 63.8, 45.5, 40.0, 39.1, 39.07, 20.6, 17.8 Example 5 Preparation of 1,4-Dimethyl-2-exo- carboxymethyl-7- azabicyclo[2.2.1]heptane (13)
The corresponding 2,3-772-osmium complex 18 was protonated and decomplexed with Ce(IV) as described for 8. The acetonitrile was evaporated and the unstable, protonated 7-azanorbornene hydrogenated in methanol as described for 12. Compound 13 was obtained as an oil following an aqueous workup (e.g., see procedure for 8) and preparative thin layer chromatography purification. 1H NMR (300 MHz, CDCI3) d 3.60 (s, 3H, CH3O) , 2.63 (dd, J = 8.1, 5.1 Hz, 1H, H2), 2.49 (br s, 1H, NH), 1.82 (dd, J =
12.2, 8.1 Hz, 1H, H3endo), 1.75-1.2 (m, overlap, 5H), 1.32 (s, CH3), 1.2 (s, 3H, CH3); 13C NMR (75 MHz,
CDCI3) d 176.5 (CO), 67.7, 63.4, 53.0, 51.3, 44.0,
38.3, 36.7, 20.5, 18.3.
Example 6 Preparation of 1,4-Dimethyl-2-endo-
(3'-pyridyl)-3-exo-carboxymethyl-7- azabicyclo[2.2.1]heptane (14a) and its exo-pyridyl-endo-carboxyl isomer (14b)
These isomers were obtained as an inseparable 94:6 mixture from the corresponding mixture of osmium complexes following the procedure for 13.
For 14a, 1H NMR (300 MHz, CDCl3) d 8.45 (m, 2H, H2' and H6' overlap), 7.49 (dt, J = 7.8, 1.5 Hz, 1H, H4'), 7.23 (dd, J = 7.8, 4.8 Hz, 1H, H5'), 3.64 (s, 3H, CH3O), 3.29 (dd, J = 5.9, 2.1 Hz, 1H, H2), 2.95 (d, J = 5.9 Hz, 1H, H3), 2.62 (br s, 1H, NH), 1.85- 1.6 (m, 2H, CH2's), 1.5 (m, 1H), 1.35 (m, 1H), 1.29 (s, 3H, CH3), 1.26 (s, 3H, CH3); 13C NMR (75 MHz, CDCI3) d 175.7 (CO), 149.8 (CH), 148.2 (CH), 135.3 (CH), 134.1 (C), 123.1 (CH), 67.6 (2XC overlap), 58.7 (CH), 58.3 (CH), 51.7 (CH3O), 38.6 (CH2), 30.3
(CH2), 19.3 (CH3), 18.7 (CH3). Diagnostic features of 14b : d 3 . 3 6 (d , J = 6 Hz , H2 ) , 2 . 8 (dd , J = 6 , 2 Hz , H3 )
Example 7 Preparation of 1,4-Dimethyl-2-endo-
(3'-pyridyl)-3-exo-(hydroxymethyl)- 7-azabicyclo[2.2.1]heptane (15)
Compound 14 was reduced with lithium aluminum hydride in ether, yielding a clear resin after an aqueous workup. Diagnostic 1H NMR resonances: 3.87 (dd, J = 10.6, 2.8 Hz, 1H, 1/2 CH2OH), 3.46 (dd, J = 10.6, 3.0 Hz, 1H, 1/2 CH2OH), 3.16 (dd, J = 5.0,
1.9 Hz, 1H, H2), 1.5 (s, 3H, CH3), 1.25 (s, 3H, CH3)
Example 8 Preparation of 1,4-Dimethyl-2-endo-
(3'-pyridyl)-3- exo-phenylsulfonyl-7- azabicyclo[2.2.1]heptane (16a) and its exo-pyridyl, endo-phenylsulfonyl isomer (16b)
The procedure for compounds 13 and 14 was followed yielding a mixture of isomeric 7- azanorbornanes. Diagnostic 1H NMR peaks for major isomer: 3.6 (d, J = 7 Hz, 1H, CHendo), 2.95 (dd, J = 7, 1.5 Hz, 1H, CHexo), 1.85 (s, 3H, CH3), 1.25 (s, 3H, CH3)
Example 9 Preparation of [Os (NH3)5(2,3-η2-2,5- dimethylpyrrole)] (OTf)2 (17) To a solution of [Os (NH3)5OTf] OTf2 (1.445 grams, 2.00 mmol) in 1.5 grams N,N-dimethylacetamide was added 2, 5-dimethylpyrrole (1.5 g, 16 mmol) and activated Mg° (1.0 g, 41 mmol) and the slurry stirred for 45-60 minutes. The slurry was filtered through a medium-porosity frit into 150 ml CH2Cl2, giving a light yellow precipitate, which was filtered, washed with CH2Cl2 and ether, then dried. The yield of a light-yellow powder was 1.23-1.31 g (92-98%).
Example 10 Preparation of 5,6 - exo- η2-OB(NH3)5-1,4- dimethyl-2-exo-carbomethoxy-7- azabicyclo-[2.2.1]hept-5-ene) (OTf)2 (18)
The 2,5-dimethylpyrrole complex (669 mg, 1.0 mmol) was suspended in 2 grams methyl acrylate and the slurry stirred for 1 hour. Acetonitrile (c. 1 ml) was added to dissolve the solids and the resulting solution added dropwise to 50 ml of ether while stirring. The precipitate was filtered, washed with ether and dried, yielding 730 mg (97%) of an off-white powder. 1H NMR (300 MHz, CD3CN) d 3.97 (br s, 3H, trans -NH3), 3.65 (s, 3H, CH3O), 3.34 (br s, 12H, cis-NH3), 3.17 (d, J = 6.3 Hz, 2H, H5 or H6), 3.13 (d, J = 6.3 Hz, 1H, H6 or H5), 2.77 (dd, J = 8.1, 4.2 Hz, 1H, H2), 2.14 (br s, 1H, NH), 2.05 (dd, J = 11.6, 8.1 Hz, 1H, 112endo) , 1.63 (dd J = 11.6, 4.2 Hz, H3exo), 1.39 (s, 3H, CH3), 1.24 (s, 3H, CH3); 13C NMR (75 MHz, CD3CN) d 176.4 (CO), 75.7 (C), 71.0 (C), 59.1 (CH), 58.0 (CH), 55.3 (CH), 51.6 (OCH3), 47.1 (CH2), 18.3 (CH3), 15.9 (CH3);
Anal. Calcd. for C12H30N6O8S2F6Os :
C, 19.10; H, 4.01; N, 11.14. Found: C, 18.57; H, 3.96; N, 11.02.
Example 11 Preparation of Pentaammineosmium-
Pyrrole Complexes: 2,3-η2-[Os(NH3)5]- Ligand] (OTf)2, where Ligand is pyrrole or N-methyl pyrrole
A mixture of [Os (NH3)5OTf] (OTf2) (723 mg, 1.0 mmol), N,N-dimethylacetamide (1 g), DME (3 g), pyrrole or N-methyl pyrrole (1 g) and magnesium (0.5 g) was stirred for 1 hour. The solution was filtered through a 60-ml medium fritted glass funnel with the aid of 10-15 ml of DME, and the filtrate added dropwise to methylene chloride (150 ml). The resulting precipitate was filtered, and washed with portions of methylene chloride (20 ml) and ether (2 X 20 ml), and dried under nitrogen.
The yield of this reaction is typically 90-95% of a yellow-orange solid containing approximately 8% of a binuclear impurity.
Example 12 Preparation of Pentaammineosmium- Cycloadduct Complexes
The pentaammineosmium-pyrrole complex obtained from Example 11 was treated with an excess (3-30 eq) of a dipolarophile in either acetonitrile or N,N-dimethylacetamide solution. After 1-10 hours, the solution was added to ether or methylene chloride with stirring (20 ml of ether per gram of acetonitrile or 75 ml methylene chloride per gram of N,N-dimethylacetamide). The resulting
precipitate was worked up as described in Example 11 providing a yield of 85-95%.
Example 13 One-Pot Process for the Synthesis of
Pentaammineosmium-Cycloadduct
Complexes
A dipolarophile (e.g., methyl acrylate) was added directly to the reaction mixture in the synthesis of the parent pyrrole complex as
described in Example 11. After a suitable reaction time (e.g., 1-10 hours), the mixture was filtered to remove the magnesium, and the filtrate was added to 1:1 methylene-chloride/ether (100 ml for every gram of N,N-dimethylacetamide used in the
synthesis) with stirring. The solid was isolated as described in Example 11 yielding the cycloadduct complex as mono-N,N-dimethylacetamide solvate in -95% yield. Example 14 One-Pot Process for the Synthesis of 7-Azanorbornanes from the
Pentaammineosmium-Cycloadduct Complex
The cycloadduct complex (1.0 mmol) prepared in Example 13 was dissolved in acetonitrile (4 g), protonated with triflic acid (3-5 eq), and treated with DDQ (1 eq). The dark solution was transferred to a 50-ml round-bottomed flask with the aid of an additional 20 ml of acetonitrile, treated with 10% palladium-on-carbon (approximately 0.5 g, 40 mole%), and hydrogenated under 1 atm H2 (balloon) for a suitable period of time (2-20 hours) (The pyrrole-derived complexes, lacking a substituent on nitrogen, underwent reductive amination to N-ethyl derivatives in acetonitrile. In these cases the solvent was evaporated and the reduction carried out in methanol). Workup A: The reaction mixture was filtered through celite to remove the Pd/C, the cake washed with acetonitrile (or methanol), and the filtrate evaporated. The residue was dissolved in water (approximately 10-15 ml), transferred to a separatory funnel, rendered basic with 10% aqueous Na2CO3 (20 ml) and extracted with methylene chloride (3 x 40 ml). The extract was dried over MgSO4 and evaporated, yielding the crude 7-azanorbornanes. Workup B: The hydrogenation reaction mixture was treated with 1 ml NH4OH, diluted with an equal volume of methylene chloride (about 30 ml), then filtered directly through 20 cc of silica gel in a 30-ml medium fritted glass funnel. The flask and silica were washed with an additional 2 X 30 ml of 1:1 methylene chloride/acetonitrile (or methanol) containing -3-5% NH4OH, and the combined eluent evaporated, yielding the crude 7-azanorbornanes. Example 15 Preparation of 2-Carbomethoxy-7- methyl-7-azabicyclo[2.2.1]heptanes
These compounds, obtained as a 1:1 mixture of isomers, were prepared in 66% overall yield from N- methyl pyrrole and methyl acrylate using the method set forth in Examples 13 and 14 (workup B). The isomers were separated by preparative thin layer chromatography using 1:1:5 HMDS/Methanol/methylene chloride: Exo isomer (1): Rf = 0.76; 1H NMR (CDCl3) δ 3.66 (s, 3H, CH3O), 3.62 (d, J = 4.2 Hz, 1H, H4), 3.30 (t, J = 4.0 Hz, 1H, H4), 2.40 (dd, J = 9.6, 5.4 Hz, 1H, H2), 2.21 (s, 3H, CH3N), 2.18 (m, 1H), 1.86 (m, 2H), 1.57 (dd, J = 12.6, 9.6 Hz, 1H,
H3endo), 1.33 (m, 2H); 13C NMR (CDCl3) δ 174.6 (C, CO), 64.2 (CH, C1 or C4), 61.1 (CH, C4 or Cl), 51.9 (CH3, CH3O), 47.4 (CH, C2), 34.5 (CH3, CH3N), 33.3 (CH2), 26.7 (CH2), 26.2 (CH2); Endo isomer (2): Rf = 0.62; 1H NMR (CDCl3) δ 3.65 (s, 3H, CH3O), 3.44 (t, J = 4.5 Hz, 1H, H1 or H4), 3.21 (t, J = 4.5 Hz, 1H, H4 or H1), 3.08 (m, 1H, H2), 2.26 (s, 3H, CH3N), 1.95 (m, 1H), 1.75 (m, overlap, 3H), 1.36 (m, 2H); 13C NMR (CDCl3, 50°C) δ 174.3 (C, CO), 64.1 (CH, C1 or C4), 62.1 (CH, C4 or Cl), 51.4 (CH3, CH3O), 45.2 (CH, C2), 34.4 (CH3, CH3N), 30.6 (CH2), 28.0 (CH2), 24.2 (CH2). The picrate salt (both isomers combined) was crystallized from wet ethanol (m.p. 102-108 °C); Anal. Calcd. for C15H18N4O9; C, 45.23; H, 4.55; N, 14.07. Found: C, 45.42; H, 4.59; N, 14.10.
Example 16 Preparation of 2-Cyano-7-methyl-7- azabicyclo[2.2.1] heptanes
These compounds, obtained as a 1:1 mixture of isomers, were prepared in 57% overall yield from N- methyl pyrrole and acrylonitrile using the method set forth in Examples 13 and 14 (workup B). The isomers were separated by preparative thin layer chromatography, using 1:1:8 HMDS/methanol/methylene chloride. Exo isomer (3) : Rf = 0.71; 1H NMR (CDCl3) δ 3.53 (d, J = 3.3 Hz, 1H, H1) , 3.37 (t, J = 3.8 Hz, 1H, H4) , 2.44 (dd, J = 9.3, 5.1 Hz, 1H, H2) , 2.36 (s, 3H, CH3N) , 2.1 (m, 1H) , 1.83 (m, 2H) , 1.75 (dd, J = 12.6, 9.3 Hz, 1H, H3endo) , 1.3 (m, 2H) ; 13C
NMR (CDCl3) δ 122.7 (C, CN) , 65.5 (CH, Cl or C4) , 60.8 (CH, C4 or C1) , 35.7 (CH2) , 35.3 (CH3) , 31.9 (CH) , 27.5 (CH2) , 26.9 (CH2) ; Endo isomer (4) : Rf = 0.55; 1H NMR (CDCl3) δ 3.44 (t, J = 4.5 Hz , 1H, H1 or H4) , 3.29 (t, J = 4.5 Hz, 1H, H4 or H1) , 2.92 (dtd [11 line pattern] , J = 12, -4.8, 1.8 Hz, 1H, H2) , 2.26 (s, m overlap, 4H, CH3N and H3exo), 2.0-1.8 (m, 3H) , 1.57 (dd, J = 12.3, 5.1 Hz, 1H, H3endo) , 1.45 (m, 1H) ; 13C NMR (CDCl3, 50°C) δ 121.7 (C, CN) , 63.8 (CH, Cl or C4) , 61.6 (CH, C4 or Cl) , 34.6
(CH2), 34.4 (CH3, CH3N), 29.2 (CH, C2), 27.9 (CH2), 24.1 (CH2). The picrate salt (both isomers
combined) was crystallized from ethanol (mp 218-224 °C) : Anal. Calcd. for C14H15N5O7: C, 46.03; H, 4.14; N, 19.17. Found: C, 45.85; H, 4.08; N, 18.88.
Example 17 Preparation of trans-2, 3-Bis- carbomethoxy-7- azabicyclo[2.2.1]heptane
This compound was prepared in 42% overall yield from pyrrole and dimethyl fumarate using the procedures set forth in Examples 11, 12 (using acetonitrile as a solvent), and 14 (hydrogenation solvent - methanol; reaction time - 2 h; workup A). 1H NMR (CDCl3) δ 3.95 (t, J = 4.5 Hz, 1H, H4), 3.84 (d, J = 4.8 Hz, 1H, H1), 3.70 (s, 3H, CH3O), 3.695 (s, 3H, CH3O) , 3.22 (td, J = 4.8, 1.8 Hz, 1H, H3), 3.03 (d, J = 4.8 Hz, 1H, H2), 2.55 (br s, 1H, NH), 1.8-1.3 (overlapping m, 4H); 13C NMR (CDCl3) δ 174.8 (C, CO), 172.1 (C, CO), 61.8 (CH, Cl or C4), 59.1 (CH, C4 or Cl), 52.3 (CH), 52.1 (CH3, CH3O), 52.0 (CH3, CH3O), 50.1 (CH), 28.7 (CH2), 24.9 (CH2). Example 18 Preparation of Hexahydro-2-phenyl-4, 7-imino-1H-isoindole-1,3 (2H)-dione
This compound was obtained as a 4:1 mixture of exo and endo isomers, respectively, in 39% overall yield from pyrrole and N-phenylmaleimide using the procedures set forth in Examples 11, 12 (using acetonitrile as a solvent), and 14 (hydrogenation solvent - methanol; reaction time - 2 hours; workup A). The crude material was chromatographed on a preparative thin layer chromatography plate (20 X 20 cm, 2 mm) using a gradient elution of ether containing -4% cone. NH4OH and 5, 10, and 20% methanol. Two bands were extracted with ether- methanol: Fl (Rf = 0.75, ether containing 3% NH4OH and 10% methanol). This material was
recrystallized from ethyl acetate-petroleum ether, yielding colorless crystals (mp 206-209°C); exo isomer. 1H NMR (CDCl3) δ 7.5-7.3 (m, 5H, Ph), 4.15 (t, J = 2 Hz, 2H, H1, H4), 2.86 (s, 2H, H2, H3), 1.7 (m, 4H, 2 X CH2), 1.54 (br s, 1H, NH); 13C NMR (CDCl3) δ 177.3 (CO), 132.1 (C), 129.0 (CH), 128.5 (CH), 126.5 (CH), 59.9 (CH, Cl, C4), 49.0 (CH, C2 , C3), 29.5 (CH2). The second fraction (Rf = 0.21) yielded the endo isomer: 1H NMR δ 7.6-7.2 (m, 5H, Ph), 4.18 (br s, 2H, H1 and H4), 3.64 (br s, 1H, NH), 3.41 (br s, 2H, H2 and H3), 1.8-1.6 (m, 4H); 13C NMR δ 175.9 (C), 132.0 (C), 129.7 (CH), 129.3 (CH), 126.9 (CH), 59.6 (CH), 51.5 (CH), 26.5 (CH2).
Example 19 Preparation of 8-Ethylhexahydro-2- phenyl-exo-4,7-imino-1H-isoindole-
1,3(2H)-dione
This compound was formed when the synthesis of hexahydro-2-phenyl-4,7-imino-1H-isoindole-1, 3 (2H- dione was carried out using acetonitrile in the hydrogenation step of the method set forth in
Example 14 (reaction time - 18 h, workup A). The crude material was chromatographed on silica gel (3.5 x 13 cm column). Elution with ether yielded 56 mg (21%) of the title product (Rf = 0.8; ether containing NH4OH). Further elution with ether containing 10% methanol and 3% cone. NH4OH yielded a second fraction containing 69 mg of crude
hexahydro-2-phenyl-4,7-imino-1H-isoindole-1,3(2H)- dione (Rf = 0.2; ether containing NH4OH). The first fraction was treated with decolorizing charcoal, filtered, evaporated, and the residue
recrystallized from ethyl acetate/petroleum ether. Yield = 21 mg of lustrous colorless crystals mp 126-128°C. 1H NMR (CDCl3) δ 7.5-7.25 (m, 5H, Ph), 3.82 (t, J = 2.2 Hz, 2H, H1, H4), 2.80 (s, 2H, H2, H3), 2.37 (q, J = 7.2 Hz, 2H, NCH2), 1.93 (m, 2H, H5exo, H6exo), 1.51 (m, 2H, H5endo, H6endo), 1.04 (t, J = 7.2 Hz, 3H, CH3); 13C NMR (CDCl3) δ 177.8 (CO), 132.4 (C, C1'), 129.1 (CH), 128.5 (CH), 126.7 (CH), 62.6 (CH, C1, C4), 49.5 (CH, C2, C3), 40.4 (CH2N), 25.0 (CH2), 14.5 (CH3).
Example 20 Preparation of Hexahydro-1-hydroxy-2- phenyl-4,7-imino-1H-isoindole-3(2H)- one
The exo imide formed in Example 18 (25 mg.
~0.1 mmol) was treated with excess sodium
borohydride (40 mg, -1.0 mmol) in 5 ml ethanol and the mixture refluxed for 20 minutes. The ethanol was evaporated, the residue acidified with 1 M HCl, and treated with Na2CO3 and methylene chloride.
Evaporation of the extract yielded 20 mg of crude material. Preparative thin layer chromatography (gradient elution; ether containing 5% NH4OH and 10- 20% methanol) yielded the product (Rf = 0.25, ether with 3% NH4OH and 10% methanol), still contaminated with a minor product. 1H NMR (CDCl3) δ 7.55-7.2 (m, 5H, Ph), 5.22 (s, 1H, NCH(OH)), 3.82 (d, J = 2 Hz, 1H), 2.60 (d, J = 2H, 1H), 2.71 (d, J = 10 Hz, 1H),
2.08 (d, J = 10 Hz, 1H), 1.63-1.3 (m, overlap, 6H, 2 X CH2, NH, OH).
Example 21 Preparation of exo-2-aminomethyl-7- methyl-7-azabicyclo[2.2.1]heptane
The nitrile formed in Example 16 (55 mg, 0.4 mmol) was treated with excess lithium aluminum hydride (30 mg, 0.79 mmol) in 10 ml ether with stirring. After 5 minutes (a white suspension formed), the reaction was quenched with methanol (0.1 g), then water (0.1 g), acidified with 1 M HCl, then basified with cone, NH4OH, and extracted with methylene chloride. Drying and evaporation of the extract yielded the corresponding primary amine as an oil (17 mg, 30%). 1H NMR (CDCl3) δ 3.18 (t, J =
3.9 Hz, 1H, H4), 3.03 (d, J= 3.9 Hz, 1H, H1), 2.70 (dd, J = 12, 7.8 Hz, 1H, 1/2 CH2N), 2.51 (dd, J = 12, 6 Hz, 1H, 1/2 CH2N), 2.22 (s, 3H, CH3N), 1.86 (m, 2H), 1.6-1.2 (m, 7H, CH2 + NH2 overlap). Example 22 Preparation of exo-2-(1-
Pyrrolylmethyl)-7-methyl-7- azabicyclo[2.2.1]heptane
The primary amine formed in Example 21 (17 mg, 0.121 mmol) was treated with 2,5- dimethoxytetrahydrofuran (25 mg, 0.189 mmol) in acetic acid (0.1 g) at 150°C for 5 minutes in an oil bath. Extraction of the basified (10% aqueous Na2CO3) reaction mixture with methylene chloride yielded a mixture of products from which was obtained 8 mg (-30%) of crude exo-2-(1- pyrrolylmethyl) product by preparative thin layer chromatography using 1:1:8
hexamethyldisilazane/methanol/methylene chloride. 1H NMR (CDCl3) δ 6.68 (s, 2H), 6.18 (s, 2H), 3.92 (dd, J = 15, 12 Hz, 1H, l/2CH2N), 3.72 (dd, J = 15, 7 Hz, 1H, 1/2CH2N), 3.22 (m, 1H), 2.96 (m, 1H), 2.26 (s, 3H, CH3N), 1.98 (m, 1H), 1.83 (m, 2H), 1.5-1.22 (m, 4H).
Example 23 Preparation of exo-2-Hydroxymethyl-7- methyl-7-azabicyclo[2.2.1]heptane
The aminoester formed in Example 15 (41 mg, 0.243 mmol) was treated with lithium aluminum hydride (10 mg, 0.264 mmol) in 5 ml ether. After 5 minutes, the reaction mixture was quenched with methanol, acidified with 1 M HCl, basified with cone. NH4OH, and extracted with methylene chloride. Evaporation of the extract yielded the desired product (11 mg, 32%). 1H NMR (CDCl3) δ 3.80 (dd, J = 9, 1 Hz, 1H, 1/2 CH2O), 3.39 (dd, J = 9, 2 Hz, 1H, 1/2 CH2O), 3.21 (t, J= 5 Hz, 1H, H4), 3.19 (d, J = 4 Hz, 1H, H1), 2.18 (s, 3H, CH3N), 1.82 (m, 3H), 1.7 (m, 1H), 1.5-1.2 (m, 4H).
Example 24 Preparation of exo-2- benzoyloxymethyl-7-methyl-7- azabicyclo[2.2.1]heptane
The alcohol formed in Example 23 (11 mg, 0.078 mmol) was treated with benzoic anhydride (34 mg, 0.15 mmol) and DMAP (10 mg) in methylene chloride. The product was purified by preparative thin layer chromatography (20 X 20 cm X 0.25 mm) using 1:3:80 NH4OH/methanol/ether (Rf = 0.6). Yield: 10 mg
(52%). 1H NMR (CDCl3) δ 8.05 (d, J = 7.2 Hz, 2H, ortho-H), 7.55 (t, J = 7.2 Hz, 1H, para-H) , 7.44 (t, J =7.2 Hz, 2H, meta-H), 4.18 (m, 2H, CH2O), 3.22 (t, J = 3.9 Hz, 1H, H4 ), 3.18 (d, J = 3.6 Hz, 1H, H1), 2.25 (s, 3H, CH3N), 2.05-1.85 (m, overlap, 3H), 1.48 (dd, J = 12, 9 Hz, 1H, H3endo), 1.34 (m, 3H). Example 25 Preparation of Norbornane Analog of Epibatidine using Reductive Heck Methodology: exo-2-(3-pyridyl) bicyclo[2.2.1]heptane This procedure is based on that described by R. Larock et al. ( J. Chem . Soc . Chem . Comm . 1989, 1368). A mixture of norbornene (101 mg, 1.07 mmol), 3-iodopyridine (205 mg, 1.0 mmol), tetra-n- butylammonium chloride (287 mg. 1.03 mmol), potassium formate (255 mg, 3.03 mmol), and
palladium acetate (28 mg, 0.125 mmol) was stirred in DMF (1.2 g) at room temperature for 72 hours. The mixture was diluted with 10 ml of 10% Na2CO3 (aq) and 10 ml of ether and the aqueous phase extracted again with ether. The combined extracts were dried over MgSO4, filtered and evaporated, and the residue purified by preparative thin layer chromatography (20 X 20 cm, 2.0 mm, 1:1 petroleum ether/ethyl acetate, Rf = 0.5), yielding the title product as an oil (73 mg, 42%). 1H NMR (CDCl3) δ
8.42 (s, 1H, H2'), 8.33 (d, J= 4.5 Hz, 1H, H6'),
7.43 (d, J = 7.8 Hz, 1H, H4'), 7.11 (dd, J = 7.8, 4.5 Hz, 1H, H5'), 2.67 (dd, J= 8.7, 5.7 Hz, 1H, H2), 2.30 (m, 2H, 1H, H1 and H4), 1.8-1.2 (m, overlap, 8H, 4 X CH2, 13C NMR (CDCl3) δ 149.1 (CH),
146.3 (CH), 142.3 (C), 134.0 (CH), 122.9 (CH), 44.7 (CH), 42.5 (CH), 38.7 (CH2), 36.7 (CH), 35.9 (CH2), 30.3 (CH2), 28.6 (CH2).
B. SYNTHESIS OF THE 7-AZABICYCLO [2.2.1]
-HEPTANE OR -HEPTENE RING SYSTEM USING
DIELS-ALDER APPROACH
In an alternative embodiment, as illustrated in Figures 2a and 2b, active compounds, or their precursors, are prepared through the Diels-Alder reaction of an N-(electron withdrawing- substituted) pyrrole with an arylsulfonyl (optionally substituted aryl or heterocyclic) acetylene. The electron withdrawing group at the N7-position decreases the aromaticity of the pyrrole ring and activates the ring in favor of the cycloaddition reaction.
The product of the reaction between the N- (electron withdrawing-substituted) pyrrole with the arylsulfonyl (optionally substituted aryl or
heterocyclic) acetylene is a 7-(electron withdrawing substituted)-2-(optionally substituted aryl or heteroaromatic)-3-arylsulfonyl-7-azabicyclo[2.2.1 ]-hepta-2,5-diene (compounds 23 and 32, Fig. 2). This diene can be derivatized using conventional methods to a wide variety of 7-azabicyclo[2.2.1]- heptanes and -heptenes. For example, an R3 alkyl or aralkyl group can be added by reacting the
saturated bicycloheptane derivative of compound 23 or 32 with n-butyl lithium and R3I, followed by treatment with a reducing agent to remove the 3- arylsulfonyl moiety. (Julia, M. and Paris, J-M., Tetrahedron Letters, 49, 4833 (1973).) R5 and R6 groups can be added to compound 24 (Figure 2) by appropriate and conventional reactions of the double bond. (See Advanced Organic Chemistry F.A. Carey and R.J. Sundberg (1990) pp. 167-218 Plenum Publishing Co.) Nonlimiting examples of addition reactions include hydrogenation, hydroboration, hydrohalogenation, hydroxylation, halohydrination, alkylation, carbene and dihalo carbene addition and epoxidation followed by ring opening reactions with nucleophiles such as alkoxide, amines,
alkylsulfide, halide, and hydroxide.
The reactive chloro in compounds 24 and 25 (Figure 2) is easily displaced by nucleophiles such as alkoxy, including methoxy, alkylthio, hydroxy, amino, cyano, azide, bromide, iodide, and
dimethylamino. The reaction between the N-(electron
withdrawing-substituted) pyrrole with the
arylsulfonyl (optionally substituted aryl or heterocyclic) acetylene is carried out in excess N- (electron withdrawing substituted) -pyrrole or in a solvent, for example, toluene, tetrahydrofuran, dimethylformamide, diethoxyethane or other inert solvents. Any molar ratio of pyrrole to dienophile can be used that provides an acceptable yield of product, and typically ranges between 0.5:1 to 50:1, preferable (1-5):1.
The reaction is conducted at any temperature that produces the desired product, and typically, between room temperature and 150°C, until the reaction is completed, for typically between 1 hour and 72 hours at 1 atm. or elevated pressure in a sealed reactor.
Several methods have been investigated for the removal of the N-electron withdrawing group, and specifically, the N-carbomethoxy protecting group, after synthesis of the desired 7-azabicyclo[2.2.1] -heptane or -heptene framework. Hydrolysis of compound 25 (Figure 2) with potassium hydroxide in methanol results in substitution of the moderately reactive chlorine in the pyridine ring by a methoxy group. Treatment of 25 with methyllithium stopped at the formation of N-acetyl epibatidine (identical with an authentic sample from acetylation of rac- epibatidine as described below), which resisted further cleavage by methyllithium even after a prolonged treatment. This is in accordance with the known stability of N-acetyl epibatidine.
Compound 25 is successfully deblocked by treatment with hydrobromic acid in acetic acid for 24 hours at room temperature. The products isolated from silica gel chromatography, with a mixed solvent system of ethyl acetate, methylene chloride and ammonia in methanol as the eluent, were rac- epibatidine (19, 25%), rac-endo-epibatidine (19', 28.4%) and unchanged carbamate (25, 20%). Notably, the recovered starting material is essentially the pure endo isomer of 25, indicating some
stereoselectivity in the cleavage of the N- carbomethoxy group with hydrobromic acid. The exo- isomer was apparently cleaved at a higher rate than the endo-isomer, presumably influenced by the proximity of the pyridyl group and the carbamate group. The rac-epibatidine thus obtained, m.p. 50- 51°, is very pure, as evidenced by its spectral data. i). N-(electron withdrawing- substituted)pyrrole
Many substituted pyrroles are known and are easily converted to N-(electron withdrawing- substituted) -pyrroles for use in the Diels-Alder process to prepare 7-azabicyclo[2.2.1]heptanes and -heptenes. For example, 3- (thioalkyl) pyrrole, including 3-(SCH3)pyrrole; 2,5-dialkylpyrrole, including 2,5-dimethylpyrrole; 3,4- dihaloalkylpyrrole, including 3,4- bis (trifluoromethyl) pyrrole, 2-alkylpyrrole, including 2-methylpyrrole; 2-alkoxyalkylpyrrole, including 2-methoxymethylpyrrole; 2- alkylthioalkylpyrrole, including 2- methylthiomethylpyrrole; 2- dialkylaminoalkylpyrrole, including 2- dimethylaminomethylpyrrole; alkyl pyrrole 2- acetate, including dimethylaminomethylpyrrole;
alkyl pyrrole 2-acetate, including methyl pyrrole 2-acetate; 2-alkoxyalkoxyalkylpyrrole, including 2- methoxymethoxyethylpyrrole; 3-aryloxyalkylpyrrole, including 3-benzyloxymethylpyrrole; 2- alkoxypyrrole, including 2-methoxypyrrole; 3- alkoxypyrrole, including 3-methoxypyrrole; 3- aryloxypyrrole, including 3-benzyloxypyrrole; 3,4- dialkylpyrrole, and 3-alkylpyrrole, including 3- methylpyrrole and 3,4-dimethylpyrrole; 1,6 and 4,5- alkylidene pyrrole, including 4,5,6,7- tetrahydroindole and 2-methyl-4,5,6,7- tetrahydroindole.
The N-substituent on the pyrrole ring is any moiety that is electron withdrawing and that activates the ring toward cycloaddition with a dienophile. The N-substituent is preferably carbomethoxy, however, other electron withdrawing moieties, including carbobenzyloxy, tert- butoxycarbonyl and optically active alkoxycarbonyl, including (+) and (-)-menthyloxycarbonyl can also be used. ii). Arylsulfonyl (optionally
substituted aryl or
heteroaromatic) acetylene In this process, a compound of the formula aryl -SO2C=C- (optionally substituted aryl or
heteroaromatic) is reacted with the N-(electron withdrawing-substituted) pyrrole or its derivative.
The arylsulfonyl-(optionally substituted aryl or heteroaromatic)-acetylene can be prepared by methods known to those of skill in the art. In one embodiment, described in detail in the Example 26 below, the compound is prepared by reacting the lithium salt of methyl (aryl) sulfone with the desired optionally substituted aryl or
heteroaromatic acid chloride to produce a 1-(aryl or heteroaromatic)-2-arylsulfonylethanone, that is converted to the corresponding acetylene via an enolphosphate intermediate as described in Example 27 below. Any optionally substituted aryl or heteroaromatic acid chloride can be used, including without limitation, the acid chloride of nicotinic acid, isonicotinic acid, 5-chloronicotinic acid, 6- methylnicotinic acid, 6-methoxynicotinic acid, 6- phenylnicotinic acid, 6-methylthionicotinic acid, 2-chloropyridine-4-carboxylic acid, 2,6- dimethylpyridine-4-carboxylic acid, l-methyl-2 (1H) - pyridone-3-carboxylic acid, 6-methylthionicotinic acid, 3-quinolinic acid, 4-quinolinic acid, 7- chloro-3-quinolinic acid, 6-methoxy-3-quinolinic acid, isoquinoline-4-carboxylic acid, 5-chloro- thiophene-2-carboxylic acid, pyrimidine-5- carboxylic acid, 5-methoxyindole-3-carboxylic acid, 1,2,5-thiadiazole-2-carboxylic acid, thiazole-5- carboxylic acid, 2-chloro-thiazole-5-carboxylic acid, and 5-chloropyridazine-2-carboxylic acid.
Substituents that can be positioned on the aromatic or heteroaromatic group include, but are not limited to, alkyl, halo, aryl, alkoxy,
dialkylamino, alkylthio, hydroxy, hydroxyalkyl, and C(O) (alkyl or aryl).
The aryl group attached to the sulfone can be any group that sufficiently activates the
acetylenic group to act as a dienophile toward the activated pyrrole and which does not interfere with the cycloaddition reaction. Nonlimiting examples are phenyl, p-alkylphenyl, including p- methylphenyl; halophenyl, and including p- chlorophenyl, p-fluorophenyl, and p-nitrophenyl. Fluoroalkanesulfonyl, including CF3SO2 and C4F9SO2, can also be used to activate an aryl- or
heteroarylacetylene.
Methods to prepare a wide variety of
arylsulfonyl-(aryl or heteroaromatic)-acetylenes are described in Bhattacharya, S.N., et al,
Organomet . Chem. Synth . 1, 145 (1970), and the reaction of an aryl or heteroaromatic
trimethylsilyl acetylene (Sakamoto, T., et al., Synthesis, 312 (1983)) with tosyl chloride in the presence of a Lewis acid catalyst such as aluminum trichloride.
The process for preparing active compounds through the Diels-Alder reaction of an N- (electron withdrawing-substituted) pyrrole with an
arylsulfonyl (optionally substituted aryl or
heterocyclic) acetylene is set out in detail in the working examples below. These examples are merely illustrative, and not intended to limit the scope of the process or the compounds that can be made according to the process. As discussed above, this is a general method that can be combined with conventional synthetic techniques to provide a wide variety of products, all of which are considered to fall within the scope of the invention. The compounds are numbered as illustrated in Figure 2.
Example 26 Preparation of 1-(2-chloro-5- pyridyl)-2-phenylsulfony1ethanone (9) To a cold solution (-30°C) of 20 g methyl phenyl sulfone in 400 ml dried tetrahydrofuran was added 128 ml 2.5M n-butyllithium (2.4 eq) slowly. The resulting solution was stirred at -30°C for 30 minutes. A solution of 26 g 6-chloronicotinyl chloride in 100 ml tetrahydrofuran was then added during a 20 minute period. After stirring at the same temperature for 30 minutes, the mixture was quenched by addition of sat. ammonium chloride (ca. 100 ml). The organic layer was separated and the aqueous layer extracted with chloroform three times. The combined organic layer was washed with sat. brine and dried over magnesium sulfate. After removal of solvent, the brown solid was triturated with methanol (150 ml) to give 7.06 g of a slightly yellow solid. Another crop of the product (11.75 g) was obtained from the mother liqueur by chromatography on a short silica gel column using 50% ethyl acetate in petroleum ether as the eluent. The total yield is 18.81 g (49.7%). m.p. 152-3°C. MS (CI) m/z 296, 298 (M+1).
In a similar manner, when the acid chlorides of nicotinic acid, isonicotinic acid, 5- chloronicotinic acid, 6-methylnicotinic acid, 6- methoxynicotinic acid, 6-phenylnicotinic acid, 6- methylthionicotinic acid, 2-chloropyridine-4- carboxylic acid, 2,6-dimethylpyridine-4-carboxylic acid, 1-methyl-2(1H)pyridone-3-carboxylic acid, 6- methylthionicotinic acid, 3-quinolinic acid, 4- quinolinic acid, 7-chloro-3-quinolinic acid, 6- methoxy-3-quinolinic acid, isoquinoline-4- carboxylic acid, 5-chloro-thiophene-2-carboxylic acid, pyrimidine-5-carboxylic acid, 5- methoxyindole-3-carboxylic acid, 1,2,4-thiadiazole- 2-carboxylic acid, thiazole-5-carboxylic acid, 2- chloro-thiazole-5-carboxylic acid, 5- chloropyridazine-2-carboxylic acid are used in place of 6-chloronicotinyl chloride in the
condensation reaction, the corresponding
ketosulfones are obtained.
Example 27 Preparation of 2-chloro-5-pyridyl
phenylsulfonyl acetylene (22)
A solution of 3.34 g (11.3 mmol) of 20 in 100 ml dried tetrahydrofuran was added to a suspension of 840 mg 60% sodium hydride (washed with ethyl ether) in 100 ml tetrahydrofuran. After stirring 10 minutes, 1.88 ml (11.3 mmol) diethyl
chlorophosphate was added in one portion. The mixture was stirred at room temperature overnight, then cooled to -78°C, and 1.35 g potassium t- butoxide is added in portions. The brown solution was stirred at -78°C for another 10 minutes and allowed to warm to ca. -30°C. Water was added and the aqueous layer extracted with methylene
chloride. After drying and evaporation in vacuo, the residue was purified on a silica gel column, and eluted with 25% ethyl acetate in petroleum ether. The white solid (1.2 g) obtained after evaporation of solvent has a m.p. 140-141°C.
MS (CI) m/z 278, 280 (M+1), yield 38%.
In a similar manner, when other heterocyclic ketosulfones described in Example 26 are used in place of compound 20, the corresponding acetylenes are obtained.
Example 28 Preparation of N-carbomethoxy pyrrole
(21)
Potassium (5.85 g, 0.15 mol) was added to a solution of 10 ml pyrrole (0.145 mol) in 80 ml hot cyclohexane in several portions. The mixture was refluxed for 1 hour. To this cold solution was added 15 g (0.16 mol) methyl chloroformate slowly. After addition, the mixture was stirred at room temperature for 30 minutes. During this period,
2.5 ml dimethyl sulfoxide was added for catalysis. After quenching with ice-water, the organic layer was separated and the aqueous layer extracted with ether. The combined organic layer was washed with 10% sodium bicarbonate, sat. sodium chloride and dried over magnesium sulfate. Removal of solvent yielded 17.4 g of a liquid. Bulb to bulb
distillation gives 16.5 g N-carbomethoxy pyrrole 21 as a colorless liquid, yield 91%. The product requires storage at -20°C.
In a similar manner, the N-carbomethoxy, N- carbobenzyloxy and N-tert-butoxycarbonyl
derivatives of 2,5-dimethylpyrrole, 3,4- bis (trifluoromethyl) pyrrole, 2-methylpyrrole, 2- methoxymethylpyrrole, 2 -methylthiomethylpyrrole, 2- dimethylaminomethylpyrrole, methyl pyrrole-2- acetate, 2-methoxymethoxyethylpyrrole, 3- benzyloxymethylpyrrole, 2 -methoxypyrrole, 3- methoxypyrrole and 3-benzyloxypyrrole are prepared.
Example 29 Preparation of 7-carbomethoxy-2-(2- chloro-5- pyridyl)-3-phenylsulfonyl-7-aza- bicyclo[2.2.1]-2,5-diene (23)
2-Chloro-5-pyridyl phenylsulfonyl acetylene 22 (1.12 g, 40.3 mmol) was dissolved in 8.0 g N- carbomethoxy pyrrole 21. The mixture was stirred in a covered flask at 80-85°C for 24 hours. After evaporation in vacuo to recover N-carbomethoxy pyrrole, the residue was chromatographed on a silica gel column using 25% to 50% ethyl acetate in petroleum ether as eluent to recover 0.2 g of the acetylene 22 and obtain 1.21 g of a slightly dark product. The crude product was triturated with methanol to yield 0.94 g (58% or 70% according to recovered starting material) of a white solid.
m.p. 101°C. MS(CI) m/z 403, 405 (M+1). When the arylsulfonyl acetylene derivatives described in Example 27 are used in place of compound 22 in this experiment, the corresponding Diels-Alder adducts are obtained. Example 30 Preparation of 7-carbomethoxy-5-(2- chloro-5-aza-bicyclo[2.2.1]hept-2-ene (24)
Compound 23 (0.726 g, 1.9 mmol) was dissolved in 50 ml anhydrous methanol and 7 ml dried
tetrahydrofuran containing 1.0 g (8.0 mmol) of sodium dihydrophosphate. To this mixture was added 3.0 g 6% sodium amalgam in two portions at -20°C under nitrogen. The stirred mixture was allowed to warm spontaneously to room temperature during a 2 hour period and stirred at room temperature for another hour. The upper layer was decanted and the residue washed with methanol. Water and 10% HCl were added to the combined methanolic extracts to bring the pH to 6 and most of the methanol removed in vacuo. The mixture was then extracted with methylene chloride. The combined organic layer was washed with sat. brine and dried over magnesium sulfate. After removal of solvent, the residue was purified on a silica gel column using 33% ethyl acetate in petroleum ether as the eluent to yield 215.3 mg (42.9%) of a colorless oil. 'H-NMR shows that it is a (1:2) mixture of exo and endo isomers. MS (CI) m/z 265, 267 (M+1) . 1HNMR 6.01-6.53 (2H, H5,6), 4.61-4.91 (2H, H1,4). When other Diels-Alder adducts described in Example 29 are treated with sodium amalgam in a similar manner, the
corresponding substituted
7-aza-bicyclo[2.2.1]hept-2-enes are obtained.
Example 31 Preparation of
7-carbomethoxy-2-(2-chloro-5- pyridyl)-7-aza-bicyclo[2.2.1]heptane
(25)
Compound 24 (178.4 mg, 0.674 mmol) (mixture of isomers) was dissolved in 10 ml methanol containing 5 mg 10% Pd-C. The mixture was hydrogenated under 1 atm. of hydrogen. After 18 ml of hydrogen was absorbed (5 minutes), the catalyst was removed by filtration and methanol removed in vacuo to give 165 mg (92%) of colorless oil. 1H-NMR indicates that it is a (1:2) mixture of exo and endo isomers. MS(CI) m/z 267, 269 (M+1). 1H-NMR 4.21-4.44 (2H, H1,4). In a similar manner, other substituted
7-aza-bicyclo [2.2.1] hept-2-enes described in
Example 30 are hydrogenated to the corresponding substituted 7-aza-bicyclo[2.2.1]heptane analogs. Example 32 Preparation of racemic epibatidine (19) and endo-epibatidine (19')
Compound 25 (90 mg, 0.338 mmol) was dissolved in 2.5 ml hydrobromic acid (33% in acetic acid). The mixture was stirred at room temperature for 20 hours. After evaporation of the mixture in vacuo the residue was dissolved in water and extracted with ethyl ether to recover the starting material (26 mg). The aqueous layer was neutralized with potassium hydroxide to pH 11 and extracted with methylene chloride. The combined organic layer was washed with saturated brine and dried over
magnesium sulfate. After removal of the solvent, the 56 mg residue was chromatographed on silica gel column using ethyl acetate, methylene chloride and sat. ammonia methanol (2:1:0.03) to give 18 mg (25%) of epibatidine (19) m.p. 50-51° and 20 mg (28.4%) of endo-epibatidine (19'). The spectral data for these compounds is provided in Table 3.
Figure imgf000058_0001
The N-acetyl derivatives of epibatidine can be prepared from epibatidine and acetic anhydride in the presence of triethylamine. Likewise, other N-substituted 7-azabicyclo[2.2.1]heptanes described in Example 31 are deprotected to the corresponding free amine. The amines are readily acylated to the amide, alkylated to the tertiary amine and
quaternary ammonium derivatives by using
conventional methods. The amines also form stable and water-soluble salts with organic and inorganic acids as preferred in the pharmaceutical
formulation.
Example 33 Preparation of 7-carbomethoxy-2-(2- methoxypyridyl)-7-aza-bicyclo[2.2.1] heptane (29)
7-Carbomethoxy-2-(2-chloro-5-pyridyl)- 7-aza-bicyclo[2.2.1]heptane 25 (20 mg, 0.076 mmol) was dissolved in 1.0 ml methanol containing 12.8 mg (0.2 mmol) potassium hydroxide. The mixture was refluxed for one hour, then concentrated and partitioned between ethyl ether and water. The aqueous layer was extracted with ether again and the combined organic layer was washed with sat. sodium bicarbonate, and dried over magnesium sulfate. Removal of solvent yielded a 10 mg residue. H1-NMR shows it is a 1:2 mixture of exo and endo isomers of the title compound. H1-NMR 3.92, 3.90 (2s, Py-OCH3), 3.71, 3.66 (2s, NCOOCH3).
Example 34 Preparation of deschloro analogues of epibatidine (30)
N-carbomethoxy-5-(2-chloro-5-pyridyl)- 7-aza-bicyclo[2.2.1]hept-2-ene 25 (16 mg) was dissolved in 3 ml methanol containing 7 mg 10% palladium on carbon. The mixture was hydrogenated under a slightly elevated pressure of hydrogen for one hour. After removal of catalyst and solvent, the residue was partitioned between ether and aqueous sodium bicarbonate. The aqueous layer was extracted with ether and the combined organic layer was dried over magnesium sulfate. Removal of solvent gave 10 mg of 7 -carbomethoxy-2-(3-pyridyl)- 7-azanorbornane (12) . MS (Cl) m/z 233 (M+1), H1-NMR 3.72, 3.66 (2s, N-COOCH3).
Example 35 Preparation of 5,6-dehydro analogs of epibatidine
When the N-acylated 7-aza-bicyclo[2.2.1] hept-5-ene derivatives prepared in Example 30 are acid hydrolyzed under conditions similar to that described in Example 32, the corresponding
5,6-dehydro analogs of epibatidine (19) and its endo-isomer (19') are obtained.
Example 36 Preparation of 1,4-dimethyl-2- (6-chloro-3-pyridyl)-3-phenylsulfonyl
-7-carbomethoxy-7-azabicyclo[2.2.1]hept-2,5-diene
A mixture of 0.14 g (0.5 mmol)
2-chloro-5-pyridyl phenylsulfonyl acetylene (22) and 0.7 g 2,5-dimethyl-N-carbomethoxypyrrole (31) was heated and maintained at 85°C for 48 hour. The excess pyrrole (31) was removed in vacuo and the dark residue chromatographed on silica gel using 25%-33% ethyl acetate in petroleum ether as eluent, yielding 76 mg (35%) of the title compound. MS (CI) m/z 431, 433 (M+1). H1-NMR 6.79, 6.55 (AB J=5.4Hz, H5,6), 3.52 (s, 3H, N-COOCH3), 1.96, 1.68 (2s, 6H, 2CH3).
Example 37 Preparation of benzoyl phenylsulfonyl methane (32)
A procedure similar to the preparation of compound 20 was used. The product was obtained in 60% yield as a white crystal (crystallized from carbon tetrachloride). m.p. 91-93°C (lit, m.p.
93-94°C).
When the acid chloride of 4-chlorobenzoic acid, 3-methoxybenzoic acid,
3,4-methylenedioxybenzoic acid, 3,4,5- trimethoxybenzoic acid, 3-trifluoromethylbenzoic acid, 3-dimethylaminobenzoic acid,
4-methylthiobenzoic acid, 4- methylsulfinylbenzoic acid, 4-methylsulfonylbenzoic acid, 3, 5-difluorobenzoic acid, 2-naphthoic acid,
4-dimethylamino-2-naphthoic acid,
6-methoxy-2-naphthoic acid, 2-phenylpropionic acid and 2-(3,4-methylenedioxyphenyl) propionic acid are used in place of benzoyl chloride above, the corresponding substituted ketosulfones are
prepared.
Example 38 Preparation of phenyl phenylsulfonyl acetylene (34)
A procedure similar to the preparation of compound 22 was used. Chromatography of the crude product on silica gel using 5% ethyl acetate in petroleum ether as the eluent yielded 20% of the acetylene 34 as a solid.
Using a similar procedure, the other
ketosulfones described in Example 37 are converted to the corresponding substituted aryl and aralkyl acetylenic derivatives.
Example 39 Preparation of 7-carbomethoxy-2- phenyl-3-phenylsulfonyl-7-azanorborna -2,5-diene (35)
Phenyl phenylsulfonyl acetylene 34 (84.3 mg, 0.35 mmol) was mixed with 0.42 g of N-carbomethoxy pyrrole (21). The mixture was heated to and maintained at 85°C for 48 hours. After removal of the excess pyrrole, the residue was chromatographed on silica gel column and eluted with 25-33% ethyl acetate in petroleum ether to give 30 mg (23%) of the adduct as a colorless oil. MS(CI) m/z
368 (M+1). H1-NMR 7.05(s, 2H, H5,6), 5.51, 5.48(2s, 2H, H1,4), 3.5(br.s. 3H, N-COOCH3).
Using a similar procedure, cycloadditions of substituted pyrroles described in Example 28 and substituted acetylenic derivatives prepared in Example 38 give the corresponding
7-aza-bicyclo[2.2.1]hepta-2,5-diene adducts.
Example 40 Preparation of 2-phenyl-7-aza-bicyclo
[2 .2 .1] heptane (36)
The bicyclic adduct 35 was reductively
desulfonated, hydrogenated and acid hydrolyzed as described in Examples 30, 31 and 32 to yield 36.
Similarly, the other bicyclic adducts in Example 39 are converted to the corresponding 2-substituted aryl-7-aza-bicyclo[2.2.1]heptanes.
Example 41 Preparation of
2-phenyl-7-aza-bicyclo[2.2.1]hept-
5-ene (37)
The bicyclic adduct 35 is reductively
desulfonated and acid hydrolyzed as described in Examples 30 and 32 to yield 37. Similarly, the other bicyclic adducts in Example 39 are converted to the corresponding 2-substituted aryl-7-aza- bicyclo[2.2.1]hept-5-enes.
Example 42 Preparation of 5 and/or 6 substituted
2-aryl (or heteroaryl)-7-aza- norbornanes from the corresponding
7-N-acyl or 7-aza-2-aryl (or
heteroaryl)-norborn-5-enes
The 5 and/or 6-substituents are introduced by functioning the 5,6-double bond through
conventional reactions, e.g., additions, hydroboration, epoxidation followed by ring opening with nucleophiles (alkoxide, amine, azide,
alkylsulfide, halide, hydroxide, etc.).
Example 43 Preparation of 3-methyl-7-aza-2-exo- (2-chloro-5-pyridyl)bicyclo[2.2.1] heptane (38)
7-Carbomethoxy-2-(2-chloro-5-pyridyl)- 3-phenylsulfonyl-7-azabicyclo[2.2.1]hept-2,5-diene (23) is hydrogenated in methanol containing 10% Pd-C until both double bonds are saturated. The product, 7-carbomethoxy-2-(2-chloro-5-pyridyl)-3- phenylsulfonyl-7-aza-bicyclo[2.2.1]heptane 39, is dissolved in dry tetrahydrofuran and treated with n-butyl lithium (1.1 eq) at -30 to 0°C, followed by methyl iodide (1-1 eq) in tetrahydrofuran. The reaction mixture is then stirred at room
temperature and poured into iced water. The product is extracted with ether and washed with water. After drying and evaporation of the ether solution, the crude product is chromatographed on a silica gel column, using a mixture of petroleum ether and ethyl acetate (3:1 by volume) to yield stereoisomers of
7-carbomethoxy-2-(2-chloro-5-pyridyl)-3-methyl- 3-phenylsulfonyl-7-aza-bicyclo[2.2.1]heptane (40). The alkylation products are each treated with sodium amalgam as in Example 30 to remove the phenylsulfonyl group, followed by acid cleavage of the 7-carbomethoxy group as in Example 32 to yield isomeric 3-methyl analogs of compound 8 and 8'.
Similarly, when methyl iodide is replaced by ethyl bromide, allyl bromide, benzyl chloride, methoxymethyl chloride and methoxyethyl
methanesulfonate, and corresponding 3-ethyl, 3- allyl, 3-benzyl, 3-methoxymethyl and 3-methoxyethyl derivatives are obtained. Other 2 -aryl or 2 -heteroaryl derivatives of 7-N-acyl-7-aza-3-phenylsulfonyl-bicyclo[2.2.1] hepta-2, 5-diene described in Example 29 are likewise hydrogenated, converted to the sulfonyl carbanion, alkylated, desulfonated and deacylated to give the corresponding 3 -alkyl or aralkyl analogs.
Example 44 Preparation of 7-methyl-7-aza- 2 -exo-(2-chloro-5-pyridyl)
bicyclo[2.2.1]heptane (41)
Epibatidine 19 prepared in Example 32 is alkylated with methyl iodide (1.1 eq) in dry tetrahydrofuran at room temperature, followed by the usual isolation procedure, to give the
7-N-methyl derivative.
Similarly, alkylation with ethyl iodide, isopropyl bromide, allyl bromide, cyclopropylmethyl bromide, benzyl chloride, 4-methoxybenzyl chloride, 3,4-dimethoxybenzyl chloride, phenethyl bromide, propargyl bromide, hydroxyethyl chloride and methoxyethyl iodide yield the corresponding
7-N-alkylated derivatives.
Other substituted 7-aza-bicyclo[2.2.1]heptane analogs described in the examples above are
alkylated to their 7-N-alkyl is derivatives in the same manner.
The N-acetyl derivative of epibatidine in Example 7 is reduced to the N-ethyl derivative by the treatment of lithium aluminum hydride in dry tetrahydrofuran at room temperature. Similarly, the 7-N-propionyl, N-benzoyl, N-phenylacetyl and N-2-furoyl derivative of epibatidine are reduced to the corresponding 7-propyl, 7-benzyl, 7-phenethyl and 7-(2-furfuryl) derivatives. Example 45 Resolution of racemic compounds
The substituted 7-aza-bicyclo[2.2.1]heptane derivatives are resolved to their optical isomers by conventional methods including chromatography on a chiral column, fractional crystallization of diastereomeric salts of chiral acids and separation of the chiral ester or amide derivatives followed by regeneration of the optically pure enantiomers. (See Optical Resolution Procedures for Chemical Compounds, Vol. 1, Amines, by P. Newman, 1980
Optical Resolution Information Center, N.Y. 10471.)
Example 46 Resolution of racemic epibatidine
(19).
To a solution of racemic epibatidine 19 and triethylamine ( 1.1 eq) in methylene chloride is added (-)-menthyl chloroformate (1.1 eq). The reaction mixture is stirred at room temperature for 6 hours, washed with iced water and dried over magnesium sulfate. After evaporation of solvent, the residue is chromatographed on a silica gel column, using a mixture of petroleum ether and ethyl acetate (5:1 by volume) to yield a mixture of two diastereoisomers of 7-N-(-)-menthyloxycarbonyl derivatives of d- and 1-epibatidine. Separation of the diastereoisomers by HPLC on a chiral column and treatment of each isomer with HBr/AcOH as in
Example 32 yields the corresponding d and
1-epibatidine.
Example 47 Preparation of optical isomers of
substituted 7-aza-bicyclo[2.2.1] heptane derivatives from chiral intermediates
N-carbo-(-)-menthyloxy pyrrole is prepared from pyrrole and (-)-menthyl chloroformate by the method described above. The chiral pyrrole is treated with the sulfonyl acetylene 22 or 34 as in Example 29 to give a diastereoisomeric mixture of the chiral cycloadduct 7-aza-bicyclo[2.2.1]hepta -2,5-diene derivative. After treatment with sodium amalgam as in Example 30, the diastereoisomeric mixture of 2-exo-aryl-7-aza-bicyclo[2.2.1]
hepta-5-ene derivatives is obtained. These diastereomers are separated by chromatography to give the d and 1 enantiomers . The optically active intermediates are each reduced and treated with HBr/AcOH to yield optically active epibatidine enantiomers. Similarly, other substituted
7-aza-bicyclo[2,2,1] heptane analogs are prepared from the corresponding chiral pyrroles and chiral cycloadducts.
Example 48 Preparation of benzo[5a,6a]
epibatidine (39)
Scheme 4 illustrates the preparation of compound 39.
Figure imgf000066_0001
a) Preparation of N-methanesulfonyl isoindole (40)
Sodium hydride (0.88g) was suspended in 3 ml dimethyl formamide. To this stirred solution was added methanesulfonamide (0.95 g, 10 mmol) in 5 ml dimethyl formamide dropwise under nitrogen. After stirring at 60°C for 0.5 hours, a solution of 2.64g (10 mmol) α , α'-dibromo-o-xylene in 7 ml DMF was added at a rate appropriate to maintain the
temperature at 60-70 °C. The mixture was stirred at room temperature for another hour, then quenched by pouring into water. The resulting precipitate was collected and washed with water, petroleum ether and ether successively. Weight 1.57g (80%). 1H-NMR δ2.37 (s, 3H, -CH3), 4.709 (s, 4H, 2CH2).
7.25 - 7.35 (m. 4H, ArH). b) Preparation of 2-(6-chloro-3-pyridyl)-3- phenylsulfonyl-1,4-dihydronaphthalene- 1,4-imine (41)
Potassium t-butoxide (560 mg, 5.0 mmol) was dissolved in 3 ml DMSO under nitrogen. To this stirred solution was added 197 mg (1.0 mmol) N- methanesulfonyl isoindole in portions. After addition, the mixture was stirred at room
temperature for 1.5 hours and quenched by addition of 3 ml water. After extraction with 45 ml ether, the combined organic layer was washed with
saturated brine and dried over magnesium sulfate for 10 minutes. After filtration, the filtrate was combined with 83 mg (0.3 mmol) 1-(6-chloro-3- pyridyl)-2-phenylsulfonyl acetylene 22. The reaction mixture was stirred at room temperature overnight to evaporate in vacuo and chromatographed on silica gel column. Eluting with a mixed solvent (ethyl acetate, methylene chloride and ammonia in methanol) gave 108 mg blue residue. The color material was removed by washing the acidified material. After basification and extraction with ether, 62 mg of pure compound 41 was obtained as a foam. Yield 52%. MS (CI), 395, 397 (M+1). 1H-NMR (CDCl3) : δ5.242(d, J=1.5Hz, 1H), 5.362 (d, J=0.9Hz, 1H). (H1 or H4). c) Preparation of exo and endo-benzo
[5a, 6a] epibatidine (39)
Compound 41 (54 mg, 0.137 mmol) was dissolved in a mixture of 3 ml methanol and 1 ml
tetrahydrofuran. The solution was cooled to -20°C and 66 mg 6% sodium amalgam was added. The mixture was stirred for 2 hours. The excess reagent was decomposed by water and the liquid layer was decanted out. After concentration of the liquid in vacuo, the residue was extracted with methylene chloride (3x5 ml). The combined organic layer was washed with saturated brine and dried over
magnesium sulfate. After removal of solvent, the residue was separated on preparative thin layer chromatography with 33% methylene chloride in ethyl acetate to give 5.5 mg exo-benzo [5a, 6a]
epibatidine and 8.5 mg endo-benzo [5a, 6a]
epibatidine. Both isomers are an oil. Yields are 15% and 25% respectively. MS (CI), 257, 259 (M+1). 1H-NMR (CDCI3), (for exoisomer). 2.753 (dd, J=4.8, 8.4 Hz, 1H, H2), 4.371 (s, 1H, H1), 4.656 (d, J=4 Hz, 1H, H4).
Example 49 Preparation of N-methyl-benzo [5a, 6a] epibatidine (42)
Scheme 5 illustrates a method for the
production of N-methyl-benzo [5a, 6a] epibatidine
Figure imgf000069_0001
a) Preparation of N-methyl isoindole (43)
N-methyl isoindole was prepared according to the method set forth in B. Zeeh and K. H. Konig, Synthesis 1972, 45. b) Preparation of 2-(6-chloro-3-pyridyl)-3- phenylsulfonyl-1,4-dihydronaphthalene- 1,4-imine (44)
N-methyl isoindole (91 mg, 0.7 mmol) was mixed with 1-(6-chloro-3-pyridyl)-2-phenylsulfonyl acetylene 22 (139 mg, 0.5 mmol) in ethyl ether.
After stirring at room temperature for 1 hour, the mixture was concentrated and chromatographed on silica gel column, eluting with ethyl acetate.
This gave 204 mg of compound 44 as a clear oil. Yield 100%. MS (CI), 409, 411 (M+1). H1-NMR (CDCl3). δ 2.36 (br, 3H, -NCH3), 4.805 (s, 1H), 4.93 (br.s., 1H), (H1, or H4). c) Preparation of N-methyl-benzo [5a, 6a] epibatidine (42)
Compound 44 (125 mg, 0.306 mmol) was dissolved in 10 ml methanol together with 4 ml
tetrahydrofuran. The solution was cooled to -20°C and 216 mg sodium dihydrophosphate was added to the solution followed by 1.0g 6% sodium amalgam. The mixture was then stirred at room temperature for 3 hours and quenched with water. The organic layer was decanted out and concentrated in vacuo . The residue was extracted with methylene chloride (2x10 ml). The combined organic layer was washed with saturated brine and dried over magnesium sulfate. After removal of solvent, the residue was
chromatographed on silica gel column eluting with
50% ethyl acetate in petroleum ether. This gave 19 mg (19%) exo-N-methyl-benzo [5a, 6a] epibatidine.
Further elution with a mixed solvent (ethyl
acetate, methylene chloride and ammonia in
methanol) yielded 55 mg (66%) of the endo-isomer. Total yield 85%. MS (CI), 271, 273 (M+1). H1-NMR (CDCl3), (for exoisomer): 2.679 (dd, J=4.5, 8.7Hz, 1H, H2), 3.935 (s, 1H, H1), 4.203 (d, J=4.0Hz, 1H, H4), 2.072 (s, 3H, NCH3). Example 50 Preparation of N-formamidinyl
epibatidine dihydrochloride (45)
Scheme 6 shows the preparation of compound 45
Figure imgf000070_0001
Racemic-epibatidine 19 (42 mg, 0.2 mmol) was mixed with 77 mg (0.7 mmol) freshly prepared ethyl formamidinate hydrochloride and 129 mg (1.0 mmol) diisopropyl ethylamine in 1 ml acetonitrile. After stirring at room temperature for 48 hours, the mixture was acidified with 1.0 M hydrogen chloride in ether. After evaporation in vacuo, the residue was separated on silica gel preparative thin layer chromatography, using a solvent system of 25% methanol in chloroform, to give 25 mg of the compound 45 as a hygroscopic solid. Yield 36%. MS (CI), 236, 238 (free base M+1). H1-NMR (CD3OD). δ 3.40 (M, 1H, H2). Example 51
The process of Example 50 was repeated with the replacement of ethyl formamidinate by S-methyl pseudothiourea, S-methyl-N-methyl pseudothiourea, S-methyl-N-nitro pseudothiourea, or methyl
acetamidinate to form the N-guanidyl, N-methyl- guanidyl, N-nitroguanidyl and N-acetamidinyl epibatidine.
Example 52 Preparation of N-formamidinyl
deschloroepibatidine dihydrochloride (46)
N-Formamidinyl epibatidine (12 mg, 0.04 mmol) 45 was dissolved in 2 ml methanol containing 5 mg 10% palladium on carbon. After hydrogenation under 1 atm hydrogen for 3 hours, the catalyst was removed by filtration. The filtrate was
concentrated in vacuo to give 10 mg compound 46 as a hygroscopic solid. Yield 100%. MS(CI), 202 (M+1 -2HCl). H1-NMR (CD3OD), δ 3.5 (M, 1H, H2). Example 53 Preparation of 1-methyl epibatidine (47), and 4-methyl epibatidine (48)
Figure imgf000072_0001
a) Preparation of 2-methylpyrrole (49)
2-Methylpyrrole was prepared according to the method set forth in J. Org. Chem. .28., 3052. b) Preparation of N-t-butoxycarbonyl-2- methylpyrrole (50)
2-Methyl pyrrole (2.5g) was dissolved in 6 ml tetrahydrofuran, and was slowly added to a
suspension of 2.4g 60% sodium hydride (washed with ether) in 30 ml tetrahydrofuran. A solution of 7.6g di-t-butyl-dicarbonate in 20 ml of the same solvent was added to this cooled mixture. After shaking occasionally for 3 hours, it was decomposed carefully with water, and extracted with ether. The combined organic layer was washed with
saturated brine and dried over magnesium sulfate. Removal of the solvent gave 6g residue. Bulb-to- bulb distillation gave 4.5g slightly yellow oil (ca. 80°C/5mmHg). Yield 80%. MS (CI), 183 (M+2). H1-NMR (CDCl3) δ 1.584 (s, 9H, 3CH3), 2.421 (s, 3H, CH3). c) Preparation of 1- (and 4) -methyl-2-(6- chloro-3-pyridyl)-3-phenylsulfonyl-7-t- butoxycarbonyl-7-azanorborna-2,5-diene
(51)
Compound 50 (10 mmol, 1.8g) was mixed with 1- (6-chloro-3-pyridyl)-2-phenylsulfonyl acetylene (22) 555 mg (2.0 mmol). The mixture was heated at 78°C in a tightly covered flask under nitrogen for 24 hours. The mixture was separated on silica gel column eluting with 25% of ethyl acetate in petroleum ether. After recovery of 1.5g of compound 50 and 120 mg compound 22, 636 mg of compound 51 was obtained as a yellow oil. Yield 69.3%. 1H-NMR showed that the oil is a 2:1 mixture of 1-methyl isomer and 4-methyl isomer. MS (CI), 459, 461. (M+1). H1-NMR (CDCl3), (for major isomer): 1.37 (s, 9H, 3CH3), 1.748 (s, 3H, CH3), 5.45 (d, J=3Hz, 1H, H4). (For the minor isomer), 1.346 (s, 9H, 3CH3), 1.958 (s, 3H, CH3), 5.26 (d, 1H, J=3Hz, H1). d) Preparation of N-t-Boc-1 (and 4) -methyl epibatidine (52)
Compound 51 (1.0 mmol, 459 mg) was dissolved in a mixture of 20 ml methanol and 10 ml
tetrahydrofuran. The solution was stirred and cooled to -20°C. To this solution was added 720 mg sodium dihydrophosphate followed by 1.5g (6.0 mmol) 6% sodium amalgam. After stirring at room
temperature for 2 hours, another 0.8g of 6% sodium amalgam was added and stirring was continued for another 2 hours. The excess reagent was decomposed by water, and the solution was decanted out. After concentration of the solution at ambient temp in vacuo, the residue was extracted with methylene chloride (4x15 ml). The combined organic layer was washed with saturated brine and dried over
magnesium sulfate. After removal of solvent, the residue (372 mg) was hydrogenated under latm hydrogen in the presence of 8.4 mg platinum oxide for 2 hours. The catalyst was removed by
filtration and the filtrate was concentrated in vacuo to a residue (360 mg). Separation took place on a silica gel column eluting with 17% ethyl acetate in petroleum ether. 95 mg of the endo- isomers and 65 mg of the exo-isomers were obtained. Total yield 50%. MS (Cl), 323, 325 (M+1). H1-NMR (CDCl3) (for exo isomer major), 2.78 (dd, 1H,
J=5.4Hz, 7.8Hz, H2), 4.45 (t, 1H, J=4.5Hz, H4). e) Preparation of 1-methyl epibatidine (47) and 4-methyl epibatidine (48)
The exo-isomer of compound 52 (65 mg) was dissolved in 5 ml methylene chloride. To this - cooled solution (0°C) was added 2.5 ml
trifluoroacetic acid. The resulting pink solution was then stirred at room temperature for 1.5 hours. After neutralization with 4.5g potassium carbonate in 10 ml water, the organic layer was separated and the aqueous layer was extracted with methylene chloride. The combined organic layer was washed with saturated brine and dried over magnesium sulfate. Removal of solvent and separation on silica gel preparative thin layer chromatography developing with a mixed solvent (ethyl acetate, methylene chloride and ammonia in methanol) gave 6 mg of 4-methyl epibatidine 48 and 12 mg 1-methyl epibatidine 47. Total yield 40.2% MS (CI), 223, 225 (M+1). H1-NMR (CDCl3), (for 1-methyl
epibatidine, major, exo-isomer). δ 2.657 (dd, J=4.8, 8.7Hz, 1H, H2), 3.694 (t, J=4.7Hz, 1H, H4). (For 4-methyl epibatidine, minor exo-isomer):
2.887 (dd, J=4.7Hz, 1H, H2), 3.486 (d, J=4.5Hz, 1H, H1)) Example 54 Preparation of 2-(2-fluoro-5- pyridyl)-7-azanorbornane (53)
Figure imgf000075_0001
a) Preparation of 1-(2-fluoro-5-pyridyl)-2- phenylsulfonyl ethanone (54) The method set forth in Example 26 was used, replacing 6-chloronicotinyl chloride with 6- fluoronicotinyl chloride (see Anderson et al; J.
Med. Chem, 1990, 33(6) 1667), providing compound 54 as a white crystal, mp. 127-128°C. Yield 72%.
MS (CI), 280 (M+1). H1-NMR (CDCl3). δ 2.70 (s, 2H,
CH2) . b) Preparation of 1-(2-Fluoro-5-pyridyl)-2- phenylsulfonyl acetylene (55)
Use of the method set forth in Example 27 gave compound 55 in 62% yield from compound 54 as a white solid. mp. 97-98.5°C. MS (CI) 262 (M+1). c) Preparation of 7-carbomethoxy-2-(2- fluoro-5-pyridyl)-3-Phenylsulfonyl-7- azabicyclo[2.2.1]-hepta-2,5-diene (56) Use of the method set forth in Example 29 gave compound 56 in 66% yield plus 22% of recovered acetylene 55. Compound 56 is a white cubic
crystal, mp. 85-87°C. MS(CI) 387 (M+1). H1-NMR (CDCl3), 3.446 (br.s., 3H, CH3), 5.459 (d, J=7.2Hz, 2H, H1,4). d) Preparation of 7-carbomethoxy-5-(2- fluoro-5-pyridyl)-7- azabicyclo[2.2.1]hept-2-ene (57)
Use of the method set forth in Example 30 gave compound 57 as a 1:2.5 mixture of exo and endo isomers in a total yield of 64% from compound 56. MS (CI) 249 (M+1). H1-NMR (CDCl3), (for endo-isomer). 3.682 (s, 3H, OCH3), (for exo-isomer), 3.655 (s, 3H, OCH3). e) Preparation of 7-carbomethoxy-2-(2- fluoro-5-pyridyl)-7- azabicyclo[2.2.1]heptane (58)
Use of the method set forth in Example 31 gave compound 58 as a colorless oil in a yield of 93.3% from compound 57. MS (CI) 251 (M+1). H1-NMR (CDCl3), (for endo-isomer), δ 3.722 (s, OCH3) , (for exo- isomer) δ 3.671 (s, 3H, OCH3). f. Preparation of 2-(2-fluoro-5-pyridyl)-7- azanorbornane (53) The method set forth in Example 32 was used to produce 23 mg (16.2%) of the exo-isomer of compound 53 and 54.8 mg (38%) of the endo isomer of compound 53, as an oil from 185 mg of Compound 58 (0.74 mmol). MS(CI) 193 (M+1). 1H-NMR (CDCl3). δ 2.763 (dd, J=.8, 9.0Hz, 1H, H2), 3.532 (s, 1H, H1), 3.769 (t, J=3.6Hz, 1H, H4). (For endo-isomer). δ 3.324 (dt, J=12Hz, 5.7Hz, 1H, H2), 3.779 (q, J=5.1Hz, 2H, H1,4). Example 55 Preparation of 2-(2-chloro-3- pyridyl)-7-azanorbornane (59)
Figure imgf000077_0001
a) Preparation of 1-(2-chloro-3-pyridyl)-2- phenylsulfonyl ethanone (60) Use of the method set forth in Example 26 gave compound 60 in 74% yield from 2-chloronicotinyl chloride as white solid, mp. 103-104°C. MS (CI) 296, 297 (M+1). H1-NMR (CDCl3) δ 4.871 (s, 2H, -CH2-) b) Preparation of 1-(2-chloro-3-pyridyl)-2- phenylsulfonyl acetylene (61)
Use of the method set forth in Example 27 gave compound 61 in 27% yield from compound 60 as a white solid, mp. 90-94°C. MS (CI) 278, 280 (M+1). c) Preparation of 7-carbomethoxy-2-(2- chloro-3-pyridyl)-3-phenylsulfonyl-7- azabicyclo[2.2.1]hepta-2,5-diene (62)
Use of the method set forth in Example 29 gave compound 62 in 62.4% from 61 as an oil. MS(CI) 403, 405 (M+1). H1-NMR (CDCl3), δ 3.612 (s, 3H, OCH3). 5.429 (t, J=2.1Hz, 1H), 5.497 (t, J=2.1Hz, 1H). d) Preparation of 7-carbomethoxy-5-(2- chloro-3-pyridyl)-7- azabicyclo[2.2.1]hept-2-ene (63)
Use of the method set forth in Example 30, gave compound 63 as the exo-isomer, 12%, and the endo-isomer, 35%. MS (Cl) 265, 267 (M+1). H1-NMR (CDCl3) (for exo-isomer). δ 3.66 (s, 3H, OCH3), 6.502 (br.s. 2H, H5,6). H1-NMR (CDCl3) (for endo- isomer). δ 3.686 (s, 3H, OCH3), 4.882, 5.029
(2br.s. 2H, H1,4). 5.88, 6.544 s., 2H, H5,6). e) Preparation of 7-carbomethoxy-2-(2- chloro-3-pyridyl)-7- azabicyclo[2.2.1]heptane (64)
Using the method set forth in Example 31, the exo-compound 63 was hydrogenated to give compound 64 in quantitative yield. MS(CI) 267, 269 (M+1). H1-NMR (CDCl3) δ 3.277 (dd, J=4.5, 8.4Hz, 1H, H2). 3.654 (s, 3H, OCH3) . f) Preparation of 2-(2-chloro-3-pyridyl)-7- azanorbornane (59)
Use of the method set forth in Example 32, gave compound 59 from exo-compound 64, in 41% yield as an oil. MS (Cl) 209, 211 (M+1). H1-NMR (CDC13) δ 3.162 (dd, J=4.8, 8.7Hz, 1H, H2), 3.681 (s, 1H), 3.795 (t, J=3.6Hz, 1H) (H1, H4) .
Example 56 Preparation of 2-(2-chloro-4- pyridyl)-7-azabicyclo[2.2.1] heptane (65)
Figure imgf000079_0001
a) Preparation of 1-(2-chloro-4-pyridyl)-2- phenylsulfonylethanone (66)
Using the method set forth in Example 26, where 2-chloroisonicotinyl chloride (see Anderson et al., J. Med. Chem. 1990, 33(b), 1667) was used instead of 6-chloronicotinyl chloride, compound 66 was obtained in 51% yield as a white crystal, mp. 124-125.5°C (methanol). MS (CI) 296, 298 (M+1). b) Preparation of 1-(2-chloro-4-pyridyl)-2- phenylsulfonyl acetylene (67)
Using the method set forth in Example 27, compound 67 was obtained in 54% yield from compound
66 as a white crystal, mp. 78-79°C. MS (CI) 278,
280 (M+1). c) Preparation of 7-carbomethoxy-2-(2- chloro-4-pyridyl)-3-phenylsulfonyl-7- azabicyclo[2.2.1]hepta-2,5-diene (68)
Using the method set forth in Example 29, compound 68 was obtained from compound 67 in 68% yield as a slightly brown oil. MS (CI) 403,
405 (M+1). H1-NMR (CDCl3) δ 3.502 (br.s. 3H, OCH3), 5.420, 5.483 (25, 2H, H1,4), 7.065 (s, 2H, H5,6). d) Preparation of 7-carbomethoxy-5-(2- chloro-4-pyridyl)-7- azabicyclo[2.2.1]hept-2-ene (69)
Using the method set forth in Example 30, compound 69 was obtained from the desulfonation of compound 68 in 13.6% yield as a 1:2 mixture of exo- and endo-isomers. MS(CI) 265, 267 (M+1). 1H-NMR (CDCl3), (for endo-isomer) δ 3.682 (s, 3H, OCH3), (for exo-isomer). δ 3.665 (s, 3H, OCH3). e) Preparation of 7-carbomethoxy-2-(2- chloro-4-pyridyl)-7- azabicyclo[2.2.1]heptane (70)
Using the method set forth in Example 31, compound 70 was obtained from the hydrogenation of compound 69 in 95% yield. MS (CI) 267, 269 (M+1). 1H-NMR (CDCI3) (for endo-isomer), δ 3.694 (s, 3H, OCH3), (for exo-isomer). δ 3.655 (s, 3H, OCH3). f) Preparation of 2-(2-chloro-4-pyridyl)-7- azabicyclo[2.2.1]heptane (65) Using the method set forth in Example 32, compound 65 was obtained from the deprotection of compound 70 in 23.6% (exo-isomer). MS(CI) 209, 211 (M+1). 1H-NMR (CDCI3), δ 2.738 (dd, J=9.0,
5.1Hz, 1H, H2), 3.629 (d, J=2.4Hz, 1H), 3.791
(br.s., 1H). Some endo-isomer can be isolated.
Example 57 Preparation of disodium 7- epibatidinylphosphate (71)
Figure imgf000081_0001
Epibatidine (40.0 mg) was dissolved in 3 ml phosphorous oxychloride and the mixture was
refluxed for 3 hours in the absence of moisture.
The excess reagent was removed in vacuo to give 100 mg 7-epibatidinyl phosphoryl dichloride as a brown oily residue. To 28 mg of this residue in 2 ml tetrahydrofuran was added 2 ml 1M sodium hydroxide in ice bath. The mixture was stirred at room temperature for another 4 hours. After evaporation of the organic solvent, the aqueous solution was washed with ethyl ether (2x5 ml). The aqueous layer was then evaporated in vacuo to ca. 0.5 ml and left to stand at room temperature for several hours to give compound 71 as a white crystal.
Yield 14 mg (80%). 1H-NMR(D2O) δ2.745 (p, J=4.5Hz, 1H, H2), 3.723 (br.s., 1H), 3.920 (br.s., 1H).
7.357 (d, J=8.4Hz, 1H). 8.073 (dd, J=2.4, 8.4Hz, 1H), 8.263 (d, J=2.4Hz, 1H). 31P-NMR (D2O). 5.332. Chlorosulfonic acid or other N-sulfate reagents can be used in place of phosphorus oxychloride, under these reaction conditions to prepare the N-sulfate derivative of epibatidine and analogs thereto. Example 58 Preparation of 2,3-dehydroepibatidine (72)
Scheme 7 shows the production of compound 72.
Figure imgf000082_0001
a) Preparation of 7-carbo-t-butoxy-2-(2-chloro-5- pyridyl)-3-phenylsulfonyl-7- azabicyclo[2.2.1]hepta-2,5-diene (73)
Using the method set forth in Example 29, compound 73 was obtained from the Diels-Alder reaction of 1-(2-chloro-5-pyridyl)-2- phenylsulfonylacetylene 22 with N-carbo-t-butoxy pyrrole (N-t-Boc-pyrrole) in 64% yield as a white solid, mp. 133-134°C. MS (CI) 445, 447 (M+1). b) Preparation of 7-t-boc-2-(2-chloro-5- pyridyl)-3-phenylsulfonyl-7- azabicyclo[2.2.1]hept-2-ene (73)
Adduct 73 (445 mg) was dissolved in a mixture of 20 ml methanol and 10 ml tetrahydrofuran
containing 8 mg platinum oxide. After
hydrogenation under latm hydrogen for 3 hours, thee catalyst was removed by filtration. The filtrate was concentrated in vacuo to give 440 mg residue. It was solidified after trituration in methanol. Yield 98%. MS (CI) 447, 449 (M+1). H1-NMR (CDCl3) δ 1.266 (s, 9H, C(CH3)3), 4.905, 4.945 (2br.s., 2H, H2 , 4) . c) Preparation of 2-(2-chloro-5-pyridyl)-3- phenylsulfonyl-7-azabicyclo[2.2.1]hept- 2-ene (75) Using the method set forth in Example 53e, the t-Boc of compound 74 was easily deprotected by trifluoro acetic acid at 0°C to give compound 75 in 95.4% yield as a white solid. MS (CI) 347,
349 (M+1). H1-NMR (CDCl3) δ4.423 (d, J=4.2Hz, 1H), 4.500 (d, J=3.6Hz, 1H) (H1,4). d) Preparation of 2,3-dehydroepibatidine
(72)
Compound 75 (365 mg) was desulfonated using the method set forth in example 30 to give 23 mg of compound 72 as a colorless oil. Yield 19%. MS(CI) 207, 209 (M+1). H1-NMR (CDCl3) δ 4.323 (s, 1H, H1), 4.574 (d, J=3.0Hz, 1H, H4), 6.560 (d, J=2.4Hz, 1H, H3).
Example 59 Preparation of Chloroethylepibatidine (76)
Figure imgf000084_0001
Using the method set forth in Example 44, epibatidine 19 was alkylated with 1-chloro-2- bromoethane to give compound 76 in a 35% yield as a clear oil. MS (Cl) 271,273, 275 (M+1). H1-NMR
(CDCl3). δ 3.225, 3.476 (25, 2H, H1,4), 3.568 (t, J=6.6Hz, 2H).
Example 60 Preparation of 2-(2-hydroxy-5- pyridyl)-7-azanorbornane (77)
Compound 53 (8.5 mg, 0.044 mmol) was dissolved in 1 ml tert-butanol. To this solution was added 1 ml 2M potassium hydroxide. After reflux for 20 hours and evaporation of butanol, the mixture was adjusted with 1M hydrochloric acid to pH 6-7.
Evaporation of solvent in vacuo and purification of product with silica gel preparative thin layer chromatography developing with 20% 7N ammonia methanol in chloroform gave 4.2 mg compound 77 as an oil. Yield 50%. MS (CI) 191 (M+1). 1H-NMR
(CDCI3) δ 2.554 (br.s., 1H, H2), 3.503; 3.743
(2br.s., 2H, H14). Example 61 Preparation of 2-(2-methylthio-5- pyridyl)-7-azanorbornane (78)
Figure imgf000085_0002
Using the method set forth in Example 33, compound 78 was obtained in 28% yield from sodium methylmercaptanide in ethanol as a colorless oil. MS (CI) 221, 223 (M+1). 1H-NMR (CDCl3) δ 2.542 (s, 3H, SCH3), 2.757 (dd, J=5.1, 8.7Hz, 1H, H2), 3.546, 3.781 (2br.s., 2H, H1,4).
Example 62 Preparation of 5,6- bis (trifluoromethyl)
deschloroepibatidine (79)
Scheme 8 shows the preparation of compound 79.
Figure imgf000085_0001
a) Preparation of 7-t-Boc-1,2- bis (trifluoromethyl)-7- azabicyclo[2.2.1]hepta-2,5-diene (80)
Compound 80 was prepared according to the procedure set forth in J. Leroy et al, Synthesis, 1982 313.
(b) Preparation of 7-t-Boc-2,3- bis(trifluoromethyl)-5-(pyridyl)-7- azabicyclo[2.2.1]hept-2-ene (81) Compound 80 (165 mg, 0.5 mmol) and 105 mg 3- iodopyridine (0.5 mmol) were dissolved in 1 ml dimethyl formamide containing 9 mg palladium acetate, 21 mg triphenyl phosphine, 120 mg
piperidine and 60 mg 88% formic acid. The mixture was stirred at 60-70°C under nitrogen for 1.5 hours and at room temperature overnight. The solvent was removed in vacuo and the residue was partitioned between methylene chloride and water. The organic layer was separated and the aqueous layer was extracted with methylene chloride. The combined organic layer was washed with saturated brine and dried over magnesium sulfate. After removal of solvent in vacuo, the residue (218 mg) was
separated in silica gel column eluting with 20% ethyl acetate in petroleum, to give 48 mg unstable compound 81 as a red oil. MS(CI) 409 (M+1). Yield 23%. 1H-NMR(CDCl3) δ 1.427 (s, 9H, OC(CH3)3), 2.974 (dd, J=4.2, 8.4Hz, 1H, H2), 4.906, 5.147 (2br.s., 2H, H1,4).
The 5-(2-chloro-5-pyridyl) analog was obtained by replacing the iodopyridine in the above reaction with 2-chloro-5-iodopyridine. c) Preparation of 2,3-bis(trifluoromethyl)- 5-pyridyl-7-azabicyclo[2.2.1]hepta-2-ene (82)
Using the method set forth in Example 53e, compound 81 was easily deprotected with
trifluoroacetic acid to give compound 82 in 90% yield. 1H-NMR (CDCl3). δ 2.02 (dd, J=8.4, 2.1Hz, 2H, H3), 2.88 (dd, J=4.8, 8.4Hz, 1H, H2), 4.36, 4.63 (2br.s., 2H, H1.4).
The 5-(2-chloro-5-pyridyl) analog was obtained in the manner set forth above. d) Preparation of 5,6-bis(trifluoromethyl) deschloroepibatidine (79)
Compound 82 was hydrogenated under high pressure of hydrogen, providing compound 79.
5,6-bis (trifluoromethyl) epibatidine was obtained in the manner set forth above.
IV. Pharmaceutical Compositions
Humans, equine, canine, bovine and other animals, and in particular, mammals, suffering from pain can be treated by administering to the patient an effective amount of one or more of the
above-identified compounds or a pharmaceutically acceptable derivative or salt thereof in a
pharmaceutically acceptable carrier or diluent. The active materials can be administered by any appropriate route, for example, orally,
parenterally, intravenously, intradermally,
subcutaneously, or topically, in liquid, cream, gel or solid form.
As used herein, the term pharmaceutically acceptable salts or complexes refers to salts or complexes that retain the desired biological activity of the above-identified compounds and exhibit minimal undesired toxicological effects. Nonlimiting examples of such salts are (a) acid addition salts formed with inorganic acids (for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, naphthalenedisulfonic acid, and polygalacturonic acid; (b) base addition salts formed with metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, sodium, potassium, and the like, or with a cation formed from ammonia,
N,N-dibenzylethylene-diamine, D-glucosamine, tetraethylammonium, or ethylenediamine; or (c) combinations of (a) and (b); e.g., a zinc tannate salt or the like.
The active compound is included in the
pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount without causing serious toxic effects in the patient treated. A preferred dose of the active compound for all of the above-mentioned conditions is in the range from about 0.0001 to 20 mg/kg, preferably 0.001 to 2 mg/kg per day, more generally 0.05 to about 0.5 mg per kilogram body weight of the recipient per day. A typical topical dosage will range from 0.001% to 0.5% wt/wt in a suitable carrier. The effective dosage range of the pharmaceutically acceptable derivatives can be calculated based on the weight of the parent compound to be delivered. If the derivative exhibits activity in itself, the
effective dosage can be estimated as above using the weight of the derivative, or by other means known to those skilled in the art. The compound is conveniently administered in any suitable unit dosage form, including but not limited to one containing 0.001 to 1000 mg, preferably 0.01 to 500 mg of active ingredient per unit dosage form. A oral dosage of 0.1 to 200 mg is usually convenient.
The active ingredient can be administered by the intravenous injection of a solution or
formulation of the active ingredient, optionally in saline, or an aqueous medium or administered as a bolus of the active ingredient.
The concentration of active compound in the drug composition will depend on absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the
compositions, and that the concentration ranges set forth herein are exemplary only and are not
intended to limit the scope or practice of the claimed composition. The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be
incorporated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant
materials can be included as part of the
composition.
The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid,
Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange
flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or other enteric agents.
The active compound or pharmaceutically acceptable salt or derivative thereof can be administered as a component of an elixir,
suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
The active compound or pharmaceutically acceptable derivatives or salts thereof can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antibiotics, antifungals, antiinflammatories, or antiviral compounds. Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline
solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS).
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art . The materials can also be obtained commercially from Alza Corporation.
V. Analgesic Activity of 7-Azabicyclo[2.2.1]
-heptanes and -heptenes
A wide variety of biological assays have been used to evaluate the ability of a compound to act as an analgesic. Any of these known assays can be used to evaluate the analgesic ability of the compounds disclosed herein. The Straub-tail reaction, which is characteristic of opiate alkaloids, has been used as an assay for opiate agonists and antagonists. The assay is described in detail in Br. J. of Pharmacol . 1969, 36, 225. Another accepted assay for analgesic activity is the hot plate analgesia assay, described in J. of Pharmacol . Exp. Therap. 1953, 107, 385. An assay for the evaluation of the ability of a compound to bind to an opiate receptor is described in Mol . Pharmacol . 1974, 10 , 868.
In addition to their potent central analgesic effects, some of the substituted
7-aza-bicyclo[2.2.1]-heptanes and -heptenes
described herein also possess varying degrees of peripheral anti-inflammatory and analgesic effects which are useful for therapeutic applications. The following assays for the evaluation peripheral anti-inflammatory activities are described in
Barber, A. and Gottschlich, R., Opioid Agonists nd Antagonists: An Evaluation of Their Peripheral Actions in Inflammation, Medicinal Research Review, Vol. 12, No.5, 525-562 (September, 1992) : paw hyperalgesia in rat that has been induced by prostaglandin E2 or carrageenan; inflamed knee joint in cat that has been induced by carrageenan, bradykinin or PGE2; formalin test in mouse or rat that has been induced by formalin; neurogenic inflammation in rat, cat or guinea pig that has been induced by antidromic stimulation of sensory nerves; and the writhing test in mouse that is induced by acetic acid, phenylbenzoquinone, prostaglandin or bradykinin; and adjuvant arthritis in rat that is induced by Freund's adjuvant. Example 63 Evaluation of Analgesic Activity
Table 4 provides the analgesic activity measured as ED50 (μg/Kg) for selected compounds disclosed herein, as determined using the Straub- Tail assay, as describe by J. Daly et al. J. Am.
Chem. Soc , 1980, 102, 830; T. F. Spande, et al. J. Am. Chem. Soc. 1992, 114, 3475; T. Li, et al.
Bioorganic and Medicinal Chemistry Letters 1993, 3, 2759.
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Example 64 Evaluation of Nicotinic Receptor Binding Activity
7-aza-bicyclo[2.2.1] -heptanes and -heptenes were evaluated for their ability to bind to the acetylcholine nicotinic receptor using a standard binding assay, e.g. X. Zhang and A. Nordberg, Arch . Pharmacol . , 348, 28 (1993); R. E. Middleton and J. B. Cohen, Biochemistry, 30, 6987 (1991), with nicotine sulfate as the reference compound, rat cortex as the tissue substrate, and a [3H]-NMCI radioligand. The results are provided in table 5.
Figure imgf000098_0001
Modifications and variations of the present invention will be obvious to those skilled in the art from the foregoing detailed description of the invention. Such modifications and variations are intended to come within the scope of the appended claims.

Claims

We claim :
1. An -azabicyclo[2.2.1]-heptane or -heptene compound of the formula:
Figure imgf000100_0001
wherein:
R1 and R4 are independently hydrogen, alkyl, including CH3; alkylhydroxy, including CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; carboalkoxy, including carbomethoxy; allyl, aryl and thioalkyl, including -SCH3;
R3, R5 and R6 are independently hydrogen, alkyl, including -CH3; alkylhydroxy, including -CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; thioalkyl, including -SCH3; halo, including Cl, F; haloalkyl, including CF3; NH2, alkylamino or dialkylamino, including -N(CH3)2 and -NHCH3; cyclic dialkylamino, including
Figure imgf000101_0001
amidine, cyclic amidine including
Figure imgf000101_0002
and their N-alkyl derivatives;
Figure imgf000101_0003
-CO2H; CO2alkyl, including -CO2CH3; -C(O) alkyl, including -C(O)CH3; -CN, -C(O)NH2, -C (O)NH (alkyl), -C(O)N(alkyl)2, including -C (O)N(CH3)2; allyl, -SO2 (alkyl), -SO2aryl, including -SO2(C6H5),
-S(O) alkyl, -S(O)aryl, aryl, heteroaryl;
Figure imgf000101_0004
R5 and R6 together can be alkylidene or haloalkylidene, including -CH2- and -CF2-; epoxide (-O-); episulfide (-S-); imino (-N(alkyl)- or -N(H)-) or a fused aryl or heteroaryl ring
including a fused phenyl ring;
R2 is independently hydrogen, alkyl, including CH3; alkenyl including -CH2-HC=CH2; alkylhydroxy, including -CH2-OH; alkyloxyalkyl including -CH2-O- (alkyl), alkylamine, including -CH2NH2; carboxylate, C(O)Oalkyl, including CO2Me; C(O)Oaryl,
C(O)Oheteroaryl, COOaralkyl, -CN, Q, C(O)Q, - alkyl (Q), -alkenyl (Q), -alkynyl (Q), -O- (Q) -S-Q, - NH-Q or -N(alkyl)-Q;
R2 and R3 together can be -C (O) -N(R8) -C (O) or CH(OH) -N(R8)-C(O)- wherein R8 can be alkyl, aryl including phenyl, or heteroaryl;
R7 is hydrogen, alkyl, including CH3, or CH2CH3; alkyl substituted with one or more halogens, including CH2CH2Cl; -CH2- (cycloalkyl), including -CH2-(cyclopropyl); -CH2CH=CH2, -CH2CH2(C6H5),
alkylhydroxy, including CH2CH2OH, alkylamino (alkyl)2, including CH2CH2N(CH3)2; alkyloxyalkyl,
alkylthioalkyl, aryl, dialkyl to form a quarternary ammonium including
Figure imgf000102_0001
or
Figure imgf000103_0001
wherein R9 is hydrogen or alkyl;
wherein Y' is CN, NO2, alkyl, OH, -O-alkyl; wherein Z is O or S;
wherein R10 and R11 are each independently -O-, -OH, -O-alkyl, -O-aryl, -NH2, -NH (alkyl),
-N (alkyl)2, -NH(aryl) and -N(aryl)2
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
and wherein the Q moiety can be optionally substituted with 1 to 3 W substituents; and
W is alkyl, including CH3; halo, including Cl and F; aryl, heteroaryl, OH, oxyalkyl, including -OCH3; SH, thioalkyl, including -SCH3; -SO (alkyl) including -SOCH3; -SO2alkyl, including -SO2CH3;
-OCH2CH=CH2, -OCH2(C6H5), CF3, CN, alkylenedioxy, including -methylenedioxy-; -CO2H, -CO2alkyl, including -CO2CH3; -OCH2CH2OH, -NO2, -NH2, -NH (alkyl), including -NHCH3; -N (alkyl)2, including -N(CH3)2;
-NHC(O) alkyl, including -NHC(O)CH3; -SO2CF3, or
-NHCH2aryl, including -NHCH2 (C6H5); and wherein
the - - - indicates an optional double bond.
2. The compound of claim 1, wherein R' is selected from the group consisting of methyl, allyl, methylcyclopropyl, methylcyclobutyl,
phenethyl, hydroxyethyl, methoxyethyl,
methylthioethyl, dimethylaminopropyl and
(4-methoxybenzyl).
3. The compound of claim 1 selected from the group consisting of 2-exo-(3-pyridyl); 2- endo-(3-pyridyl); 7-methyl-2-exo-(3-pyridyl); 7- cyclopropylmethyl-2-exo- (3-pyridyl); 2-exo-(6- chloro-3-pyridyl); 2-exo-(6-fluoro-3-pyridyl) and 7-phenethyl-2-exo-(3-pyridyl).
4. The compound of claim 1 selected from the group consisting of 2-exo-(4-pyridyl);
7-methyl-2-exo-(4-pyridyl); 7- allyl-2-exo-(4-pyridyl); and
7-cyclopropylmethyl-2-exo-(4-pyridyl).
5. The compound of claim 1 selected from the group consisting of
2-exo-(3-chloro-4-pyridyl); 7-cyclopropylmethyl-2-
2-exo-(3-chloro-4-pyridyl); and
7-phenethyl-2-exo-(3-chloro-4-pyridyl).
6. The compound of claim 1 selected from the group consisting of 2-exo-(2-fluoro-5-pyridyl); 2-exo-(2-methoxy-5- pyridyl); 2 - exo-(2-methylthio-5-pyridyl);
2-exo-(2-methyl-5-pyridyl);
2-exo-(2-dimethylamino-5-pyridyl), and the 7- cyclopropylmethyl derivatives of these compounds.
7. The compound of claim 1, wherein:
the 2-substituent is selected from the group consisting of the exo and endo isomers of phenyl; (3-chlorophenyl); (3-dimethylaminophenyl); (3-trifluoromethylphenyl); (3,4,- methylenedioxyphenyl); (3,4-dimethoxyphenyl);
(4-fluorophenyl); (4-hydroxyphenyl);
(4-methylthiophenyl); (4-methylsulfonylphenyl), (3,5-difluorophenyl); (chlorophenyl); (2-naphthyl); (7-methoxy-2-naphthyl); (5-chloro-2-thiophenyl); (chloro-5-thiazolyl); (4-pyrimidyl);
(2-chloro-5-pyrimidyl); (5-chloro-2-pyridazinyl); (1,2,4-thiadiazolyl); (5-dimethylamino-2-furyl); 2-(5-indolyl); 2-(5-fluoro-3-indolyl);
2-(5-methoxy-3-indolyl); 2-(4-chlorobenzyl);
2-(5-chloro-3-pyridylmethyl); 2-(4-pyridylmethyl); 2-nicotinyl; 2-(6-chloronicotinyl); 2-isσnicotinyl; 2-(3-chloro-isonicotinyl); 2-(4-chlorobenzoyl); 2- (4-dimethylaminobenzoyl); 2-(3,4-dimethoxybenzoyl); and
the 7 substituent is selected from group consisting of hydrogen, methyl, cyclopropylmethyl, allyl and phenethyl.
8. The compound of claim 1, wherein R' and R4, are independently selected from the group consisting of methyl, hydroxymethyl, methoxymethyl, carbomethoxy, allyl, benzyl, (4-fluorobenzyl), and (4 -methoxybenzyl).
9. The compound of claim 1, wherein R3 is selected from the group consisting of methyl, hydroxymethyl, methoxymethyl, carbomethoxy,
carboxy, carbamyl, cyano, acetyl, aminomethyl, dimethylaminomethyl, methylthiomethyl, phenylsulfonyl, methanesulfonyl, benzyl, and allyl
10. The compound of claim 1, wherein R5 and R6 are selected from the group consisting of trifluoromethyl, methoxy, methyl; carbomethoxy, hydroxymethyl, methoxymethyl, chloro, hydroxy.
11. The compound of claim 1 that has a double bond between C2 and C3.
12. The compound of claim 1 that has a double bond between C5 and C6.
13. The compound of claim 1, wherein: the 2-substituent is selected from the group consisting of 2-exo-(3-pyridyl), 2-exo-(6-chloro-3- pyridyl), and 2-exo-(6-fluoro-3-pyridyl); and
the 7 substituent is selected from the group consisting of
s
N-phosphate, (N)-SO3H, , and their salts;
Figure imgf000109_0003
Figure imgf000109_0001
14. A method for imparting analgesia to a mammal, comprising administering an effective amount of an azabicyclo[2.2.1]-heptane or -heptene compound of the formula:
wherein:
Figure imgf000109_0002
R1 and R4 are independently hydrogen, alkyl, including CH3; alkylhydroxy, including CH2OH;
alkyloxyalkyl, including
-CH2OCH3; alkylthioalkyl, including -CH2SCH3;
alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and
CH2N(CH3)2; oxyalkyl, including -OCH3; carboalkoxy, including carbomethoxy; allyl, aryl and thioalkyl, including -SCH3;
R3, R5 and R6 are independently hydrogen, alkyl, including -CH3; alkylhydroxy, including -CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; thioalkyl, including -SCH3; halo, including Cl, F; haloalkyl, including CF3; NH2, alkylamino or
dialkylamino, including -N(CH3)2 and -NHCH3; cyclic dialkylamino, including
Figure imgf000110_0001
amidine, cyclic amidine including
Figure imgf000111_0001
and their N-alkyl derivatives;
Figure imgf000111_0002
-CO2H; CO2alkyl, including -CO2CH3; -C(O) alkyl, including -C(O)CH3; -CN, -C(O)NH2, -C(O)NH (alkyl), -C(O)N(alkyl)2, including -C(O)N(CH3)2; allyl,
-SO2 (alkyl), -SO2aryl, including -SO2(C6H5),
-S(O) alkyl, -S(O)aryl, aryl, heteroaryl;
Figure imgf000111_0003
R5 and R6 together can be alkylidene or
haloalkylidene, including -CH2- and -CF2-; epoxide (-O-); episulfide (-S-); imino (-N(alkyl)- or
-N(H)-) or a fused aryl or heteroaryl ring
including a fused phenyl ring;
R2 is independently hydrogen, alkyl, including CH3; alkenyl including -CH2-HC=CH2; alkylhydroxy, including -CH2-OH; alkyloxyalkyl including -CH2-O- (alkyl), alkylamine, including -CH2NH2; carboxylate, C (O) Oalkyl, including CO2Me ; C (O)Oaryl,
C(O)Oheteroaryl, COOaralkyl, -CN, Q, C(O)Q, - alkyl (Q), -alkenyl (Q), -alkynyl (Q), -O- (Q) -S-Q, - NH-Q or -N(alkyl)-Q; R2 and R3 together can be -C (O) -N (R8) -C (O) or CH(OH) -N(R8) -C(O) - wherein R8 can be alkyl, aryl including phenyl, or heteroaryl;
R7 is hydrogen, alkyl, including CH3, or CH2CH3; alkyl substituted with one or more halogens, including CH2CH2Cl; -CH2- (cycloalkyl), including -CH2- (cyclopropyl); -CH2CH=CH2, -CH2CH2 (C6H5),
alkylhydroxy, including CH2CH2OH, alkylamino (alkyl)2, including CH2CH2N (CH3)2; alkyloxyalkyl,
alkylthioalkyl, aryl, dialkyl to form a quarternary ammonium including
c
Figure imgf000112_0001
or y
Figure imgf000113_0001
wherein R9 is hydrogen or alkyl;
wherein Y' is CN, NO2, alkyl, OH, -O-alkyl; wherein Z is O or S;
wherein R10 and R11 are each independently -O-, -OH, -O-alkyl, -O-aryl, -NH2, -NH (alkyl),
-N (alkyl)2, -NH(aryl) and -N(aryl)2;
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
and wherein the Q moiety can be optionally substituted with 1 to 3 W substituents; and
W is alkyl, including CH3; halo, including Cl and F; aryl, heteroaryl, OH, oxyalkyl, including -OCH3; SH, thioalkyl, including -SCH3; -SO (alkyl) including -SOCH3; -SO2alkyl, including -SO2CH3;
-OCH2CH=CH2, -OCH2(C6H5), CF3, CN, alkylenedioxy, including -methylenedioxy-; -CO2H, -CO2alkyl, including -CO2CH3; -OCH2CH2OH, -NO2, -NH2, -NH (alkyl), including -NHCH3; -N (alkyl) 2, including -N(CH3)2; -NHC(O) alkyl, including -NHC(O)CH3; -SO2CF3, or -NHCH2aryl, including -NHCH2 (C6H5) ; and wherein
the - - - indicates an optional double bond.
15. The method of claim 14, wherein the compound is administered in an amount ranging between 0.002 and 10 mg/kg per day.
16. The method of claim 14, wherein the compound is administered in an amount ranging between 0.02 and 0.2 mg/kg per day.
17. The method of claim 14, wherein the compound is applied topically in a dosage ranging between 0.001% and 0.5% wt/wt in a carrier suitable for topical administration.
18. The method of claim 14, wherein the compound is administered by intravenous injection.
19. The method of claim 14, wherein the compound is administered orally.
20. The method of claim 14, wherein the compound is administered topically.
21. A method for the treatment of inflammatory conditions in a mammal, comprising administering to a mammal an effective amount of an azabicyclo[2.2.1] -heptane or -heptene compound of the formula:
wherein :
Figure imgf000118_0001
R1 and R4 are independently hydrogen, alkyl, including CH3; alkylhydroxy, including CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; carboalkoxy, including carbomethoxy; allyl, aryl and thioalkyl, including -SCH3;
R3, R5 and R6 are independently hydrogen, alkyl, including -CH3; alkylhydroxy, including -CH2OH;
alkyloxyalkyl, including -CH2OCH3; alkylthioalkyl, including -CH2SCH3; alkylamino, including -CH2NH2; alkylaminoalkyl or alkylaminodialkyl, including CH2NH(CH3) and CH2N(CH3)2; oxyalkyl, including -OCH3; thioalkyl, including -SCH3; halo, including Cl, F; haloalkyl, including CF3; NH2, alkylamino or
dialkylamino, including -N(CH3)2 and -NHCH3; cyclic dialkylamino, including
Figure imgf000118_0002
amidine, cyclic amidine including
Figure imgf000119_0001
and their N-alkyl derivatives;
Figure imgf000119_0002
-CO2H; CO2alkyl, including -CO2CH3; -C(O) alkyl, including -C(O)CH3; -CN, -C(O)NH2, -C (O)NH (alkyl), -C(O)N (alkyl)2, including -C (O)N(CH3)2; allyl,
-SO2 (alkyl), -SO2aryl, including -SO2 (C6H5) ,
-S(O) alkyl, -S(O)aryl, aryl, heteroaryl;
or
Figure imgf000119_0003
R5 and R6 together can be alkylidene or
haloalkylidene, including -CH2- and -CF2-; epoxide (-O-); episulfide (-S-); imino (-N(alkyl)- or
-N(H)-) or a fused aryl or heteroaryl ring
including a fused phenyl ring;
R2 is independently hydrogen, alkyl, including CH3; alkenyl including -CH2-HC=CH2; alkylhydroxy, including -CH2-OH; alkyloxyalkyl including -CH2-O- (alkyl), alkylamine, including -CH2NH2; carboxylate, C(O)Oalkyl, including CO2Me; C(O)Oaryl,
C(O)Oheteroaryl, COOaralkyl, -CN, Q, C(O)Q, - alkyl (Q), -alkenyl (Q), -alkynyl (Q), -O- (Q) -S-Q, - NH-Q or -N (alkyl) -Q;
R2 and R3 together can be -C (O) -N (R8) -C (O) or CH(OH)-N(R8)-C(O)- wherein R8 can be alkyl, aryl including phenyl, or heteroaryl;
R7 is hydrogen, alkyl, including CH3, or CH2CH3; alkyl substituted with one or more halogens, including CH2CH2Cl; -CH2-(cycloalkyl), including -CH2-(cyclopropyl); -CH2CH=CH2, -CH2CH2(C6H5),
alkylhydroxy, including CH2CH2OH, alkylamino (alkyl)2, including CH2CH2N(CH3)2; alkyloxyalkyl,
alkylthioalkyl, aryl, dialkyl to form a quarternary ammonium including
Figure imgf000120_0001
or
k
Figure imgf000121_0001
wherein R9 is hydrogen or alkyl;
wherein Y' is CN, NO2, alkyl, OH, -O-alkyl; wherein Z is O or S;
wherein R10 and R11 are each independently -O-, -OH, -O-alkyl, -O-aryl, -NH2, -NH(alkyl),
-N(alkyl)2, -NH(aryl) and -N(aryl)2;
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
and wherein the Q moiety can be optionally substituted with 1 to 3 W substituents; and
W is alkyl, including CH3; halo, including Cl and F; aryl, heteroaryl, OH, oxyalkyl, including -OCH3; SH, thioalkyl, including -SCH3; -SO (alkyl) including -SOCH3; -SO2alkyl, including -SO2CH3;
-OCH2CH=CH2, -OCH2(C6H5), CF3, CN, alkylenedioxy, including -methylenedioxy-; -CO2H, -CO2alkyl, including -CO2CH3; -OCH2CH2OH, -NO2, -NH2, -NH (alkyl), including -NHCH3; -N (alkyl) , including -N(CH3)2; -NHC(O) alkyl, including - C(O)CH3; -SO2CF3, or -NHCH2aryl, including -NHCH2 (C6H5) ; and wherein
the - - - indicates an optional double bond.
22. The method of claim 21, wherein the compound is administered in an amount ranging between 0.002 and 10 mg/kg per day.
23. The method of claim 21, wherein the compound is administered in an amount ranging between 0.02 and 0.2 mg/kg per day.
24. The method of claim 21, wherein the compound is applied topically in a dosage ranging between 0.001% and 0.5% wt/wt in a carrier suitable for topical administration.
25. The method of claim 21, wherein the compound is administered by intravenous injection.
26. The method of claim 21, wherein the compound is administered orally.
27. The method of claim 21, wherein the compound is administered topically.
28. A process for the preparation of 7- azabicyclo[2.2.1]-heptanes and -heptenes,
comprising the steps of
(i) combining a pentaammineosmium (II) complex of an optionally substituted pyrrole and a dipolarophile to form an optionally substituted 7-azabicyclo[2.2.1]heptene osmium complex; and then (ii) removing the pentaammineosmium from the 7azabicyclo[2.2.1]heptene.
29. The process of claim 28, wherein the pyrrole is selected from the group consisting of
2,5-dialkylpyrrole, 2-alkylpyrrole, 3-alkylpyrrole, 1-alkylpyrrole, 3,4-dialkylpyrrole, pyrrole,
1-silylated pyrrole, (1, 2, or 3)-alkoxy or amino pyrrole, 2,3-dialkoxypyrrole, 2,5-dialkoxypyrrole, and 3,4-dialkoxypyrrole.
30. The process of claim 28, wherein the dipolarophile is Z1-C=C-Z2, wherein Z1 and Z2 are electron-withdrawing groups.
31. The process of claim 30, wherein Z' and Z2 are independently selected from the group consisting of CO(alkyl, aryl or heteroaryl), C(O)H, CO2(alkyl, aryl, or heteroaryl), or S02(alkyl, aryl, or heteroaryl), or wherein Z' and Z2 are together
(CO)2O or (CO) 2NR8, wherein R12 is alkyl including CH3 or C2H5; aryl including phenyl, or heteroaryl.
32. The process of claim 28, wherein the dipolarophile is selected from the group consisting of 3-vinyl-pyridine, N-methylated and
6-carboxylated pyridyl acrylates, alkyl acrylate, alkyl methacrylate, pyridyl substituted vinyl sulfones, acrylonitriles, anhydrides, maleimides, alpha-methylene-δ-butyrolactone, maleate, and fumarate.
33. The process of claim 28, wherein pentaammineosmium (II) is generated in situ by the reduction of pentaammineosmium (III) with a one electron reducing agent that has a reducing
potential of less than -0.75 volts versus hydrogen.
34. The process of claim 28, wherein the counteranion of the pentaammineosmium is selected from the group consisting of CF3SO3-, PF6-, and (alkyl or aryl) SO3-.
35. The process of claim 32, wherein the reducing agent is selected from the group
consisting of magnesium, zinc, aluminum, sodium, lithium, and cobaltacene.
36. The process of claim 33, wherein the optionally substituted pyrrole, pentaammineosmium
(III) , and reducing agent are stirred at a
temperature ranging between 0 and 50 degrees C, until the desired organometallic complex is formed.
37. The process of claim 28, further comprising derivatizing functional groups on the 7-azabicyclo[2.2.1]heptene while pentaammineosmium is complexed to the pi-orbital of the heptene moiety.
38. The process of claim 28, further
comprising reducing the optionally substituted 7-azabicyclo[2.2.1]heptene to an optionally
substituted 7-azabicyclo[2.2.1]heptane.
39. A process for the preparation of
optionally substituted 7-azabicyclo[2-2.1]-heptanes and -heptenes, comprising the steps of combining a N- (electron withdrawing substituted)-optionally substituted pyrrole with an arylsulfonyl (optionally substituted aryl or heterocyclic) acetylene.
40. The process of claim 39, wherein the pyrrole is selected from the group consisting of 3,4-di (CF3) pyrrole, 3-(thioalkyl)pyrrole,
2,5-dialkylpyrrole, 3,4-bis(trifluoromethyl)- pyrrole, 2-alkylpyrrole, 2-alkoxyalkylpyrrole, 2-alkylthioalkylpyrrole, 2- dialkylaminoalkylpyrrole, alkylpyrrole 2-acetate, 2-alkoxyalkoxyalkylpyrrole, 3 -aryloxyalkylpyrrole, 2-alkoxypyrrole, 3-alkoxypyrrole, 3-aryloxypyrrole, 3, 4-dialkylpyrrole, and 3 -alkylpyrrole.
41. The process of claim 39, wherein the N-electron-withdrawing group is selected from the group consisting of carbomethoxy, carbobenzyloxy, and tert-butoxycarbonyl.
42. A pharmaceutical composition comprising an effective amount to impart analgesia to a mammal of the compound of claim 1 or its pharmaceutically acceptable salt, in a
pharmaceutically acceptable carrier or diluent.
43. A pharmaceutical composition comprising an effective amount to treat
inflammation in a mammal of the compound of claim 1 or its pharmaceutically acceptable salt, in a pharmaceutically acceptable carrier or diluent.
44. The method of claim 14, wherein the mammal is a human.
45. The method of claim 21, wherein the mammal is a human.
46. The pharmaceutical composition of claim 43, wherein the mammal is a human.
PCT/US1994/003573 1993-04-01 1994-04-01 7-azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents WO1994022868A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP94912381A EP0691971A1 (en) 1993-04-01 1994-04-01 7-azabicyclo- 2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents
US08/532,584 US6117889A (en) 1994-04-01 1994-04-01 7-Azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents
JP6522396A JPH08511768A (en) 1993-04-01 1994-04-01 7-azabicyclo- [2.2.1] -heptane and -heptene derivatives as analgesics and anti-inflammatory agents
AU64971/94A AU695682B2 (en) 1993-04-01 1994-04-01 7-azabicyclo-(2.2.1)-heptane and -heptene derivatives as analgesics and anti-inflammatory agents
US09/023,844 US6255490B1 (en) 1993-04-01 1998-02-13 7-azabicyclo[2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4144593A 1993-04-01 1993-04-01
US08/041,445 1993-04-01

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US4144593A Continuation-In-Part 1993-04-01 1993-04-01

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US08/296,463 Continuation-In-Part US5817679A (en) 1993-04-01 1994-08-25 7-Azabicyclo 2.2.1!-heptane and -heptene derivatives as cholinergic receptor ligands

Publications (1)

Publication Number Publication Date
WO1994022868A1 true WO1994022868A1 (en) 1994-10-13

Family

ID=21916549

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/003573 WO1994022868A1 (en) 1993-04-01 1994-04-01 7-azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents

Country Status (7)

Country Link
EP (1) EP0691971A1 (en)
JP (1) JPH08511768A (en)
CN (1) CN1052726C (en)
AU (1) AU695682B2 (en)
CA (1) CA2159723A1 (en)
HU (1) HUT74380A (en)
WO (1) WO1994022868A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2713641A1 (en) * 1993-12-09 1995-06-16 Egyt Gyogyszervegyeszeti Gyar Derivatives of epi-epibatidine.
EP0778835A1 (en) * 1994-08-25 1997-06-18 University Of Virginia 7-azabicyclo 2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
US5726189A (en) * 1996-05-03 1998-03-10 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Method for imaging nicotinic acetylcholinergic receptors in the brain using radiolabeled pyridyl-7-azabicyclo 2.2.1!heptanes
EP0955301A2 (en) * 1998-04-27 1999-11-10 Pfizer Products Inc. 7-aza-bicyclo[2.2.1]-heptane derivatives, their preparation and use according to their affinity for neuronal nicotinic acetylchloline receptors
US6060473A (en) * 1993-04-01 2000-05-09 Ucb S.A. - Dtb 7-azabicyclo[2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
US6077846A (en) * 1993-09-10 2000-06-20 Ucb, S.A. Epibatidine and derivatives thereof as cholinergic receptor agonists and antagonists
CN1060781C (en) * 1995-03-13 2001-01-17 赫彻斯特股份公司 Phosphonomonoester nucleic acids, process for their preparation, and their use
WO2001049690A1 (en) * 2000-01-07 2001-07-12 University Of Bristol 9-azabicyclo[4.2.1]non-2-ene derivatives as ligands for nicotinic acetylcholine receptors

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2014364693B2 (en) * 2013-12-18 2018-12-06 Novassay Sa Gamma-aminobutyric acid (GABA) analogues for the treatment of pain and other disorders

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993018037A1 (en) * 1992-03-03 1993-09-16 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Epibatidine and derivatives, compositions and methods of treating pain

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993018037A1 (en) * 1992-03-03 1993-09-16 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Epibatidine and derivatives, compositions and methods of treating pain

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 116, no. 7, 1992, Columbus, Ohio, US; abstract no. 55131n, M.J. KUHAR ET AL.: "3--beta--(4-iodophenyl-tropan-2-beta-carboxylic acid methyl ester tartrate and related compounds as cocaine receptor -binding ligands" page 447; *
E.J. COREY ET AL.: "Stereocontrolled total synthesis of (+)- and (-)-epibatidine", JOURNAL OF ORGANIC CHEMISTRY, vol. 58, no. 21, 8 October 1993 (1993-10-08), EASTON US, pages 5600 - 5602 *
M.E. JUNG ET AL.: "Intramolecular Diels-Alder chemistry of pyrroles", JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL COMMUNICATIONS, 1984, LETCHWORTH GB, pages 630 - 632 *
T.D. SPANDE ET AL.: "Epibatidine: A novel (Chloropyridyl)azabicycloheptane with potent analgesic activity from Ecuadorian poison frog", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 114, no. 9, 1992, WASHINGTON, DC US, pages 3475 - 3478 *
U.S. PAT. APPL. US 564755 (KUHAR ET AL.) (01-08-91) *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6060473A (en) * 1993-04-01 2000-05-09 Ucb S.A. - Dtb 7-azabicyclo[2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
US6077846A (en) * 1993-09-10 2000-06-20 Ucb, S.A. Epibatidine and derivatives thereof as cholinergic receptor agonists and antagonists
US6177451B1 (en) 1993-09-10 2001-01-23 Ucb, S.A. Epibatidine and derivatives thereof as nicotine cholinergic receptor agonists
FR2713641A1 (en) * 1993-12-09 1995-06-16 Egyt Gyogyszervegyeszeti Gyar Derivatives of epi-epibatidine.
EP0778835A1 (en) * 1994-08-25 1997-06-18 University Of Virginia 7-azabicyclo 2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
EP0778835A4 (en) * 1994-08-25 1999-02-03 Univ Virginia 7-azabicyclo 2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
CN1060781C (en) * 1995-03-13 2001-01-17 赫彻斯特股份公司 Phosphonomonoester nucleic acids, process for their preparation, and their use
US5726189A (en) * 1996-05-03 1998-03-10 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Method for imaging nicotinic acetylcholinergic receptors in the brain using radiolabeled pyridyl-7-azabicyclo 2.2.1!heptanes
US5969144A (en) * 1996-05-03 1999-10-19 The United States Of America As Represented By The Department Of Health And Human Services Radiolabeled pyridyl-7-azabicyclo[2,2,1]heptanes
EP0955301A2 (en) * 1998-04-27 1999-11-10 Pfizer Products Inc. 7-aza-bicyclo[2.2.1]-heptane derivatives, their preparation and use according to their affinity for neuronal nicotinic acetylchloline receptors
EP0955301A3 (en) * 1998-04-27 2001-04-18 Pfizer Products Inc. 7-aza-bicyclo[2.2.1]-heptane derivatives, their preparation and use according to their affinity for neuronal nicotinic acetylcholine receptors
WO2001049690A1 (en) * 2000-01-07 2001-07-12 University Of Bristol 9-azabicyclo[4.2.1]non-2-ene derivatives as ligands for nicotinic acetylcholine receptors

Also Published As

Publication number Publication date
JPH08511768A (en) 1996-12-10
AU695682B2 (en) 1998-08-20
CA2159723A1 (en) 1994-10-13
HU9502863D0 (en) 1995-11-28
EP0691971A1 (en) 1996-01-17
AU6497194A (en) 1994-10-24
CN1052726C (en) 2000-05-24
CN1133045A (en) 1996-10-09
HUT74380A (en) 1996-12-30

Similar Documents

Publication Publication Date Title
US6255490B1 (en) 7-azabicyclo[2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
US6117889A (en) 7-Azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents
AU775426B2 (en) New compounds
CN116003321A (en) Novel heterocyclic derivatives useful as SHP2 inhibitors
EP2991977B1 (en) C-linked heterocycloalkyl substituted pyrimidines and their uses
US6060473A (en) 7-azabicyclo[2.2.1]-heptane and -heptene derivatives as cholinergic receptor ligands
JP4284181B2 (en) Adenosine A2a receptor antagonist
US20230357277A1 (en) Kras g12d inhibitors
CN109996792B (en) Heterocyclic compounds having activity as modulators of muscarinic M1 and/or M4 receptors in the treatment of CNS diseases and pain
EP3209651B1 (en) Carbazole derivatives
KR101070075B1 (en) SUBSTITUTED 8-PERFLUOROALKYL-6,7,8,9-TETRAHYDROPYRIMIDO[1,2-a]PYRIMIDIN-4-ONE DERIVATIVES
CA2811714A1 (en) Imidazotriazinone compounds
WO2005110410A2 (en) Kinase inhibitors as therapeutic agents
KR20120060867A (en) Heterocyclic compounds as janus kinase inhibitors
JP2006507241A (en) Biaryl diazabicycloalkanamides as nicotinic acetylcholine agonists
CA2140009A1 (en) Benzodiazepine derivatives
RU2190612C2 (en) Novel derivatives of indole-2,3-dione-3-oxime
CN110913855A (en) Imidazole-containing ALK2 kinase inhibitors
KR20070043008A (en) The use of n-aryl diazaspiracyclic compounds in the treatment of addiction
KR20070090922A (en) Nicotinic acetylcholine receptor ligands
JPH0267282A (en) Tetracyclic ketaone
WO1994022868A1 (en) 7-azabicyclo-[2.2.1]-heptane and -heptene derivatives as analgesics and anti-inflammatory agents
WO2011080444A1 (en) Novel (heterocycle/condensed piperidine)-(piperazinyl)-1-alcanone or (heterocycle/condensed pyrrolidine)-(piperazinyl)-1-alcanone derivatives, and use thereof as p75 inhibitors
JP2007515479A (en) Nicotinic acetylcholine receptor ligand
AU708112B2 (en) 7-azabicyclo{2.2.1}-heptane and -heptene derivatives as cholinergic receptor ligands

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 94192237.5

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA CN HU JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2159723

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1994912381

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 08532584

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1994912381

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1994912381

Country of ref document: EP