WO2014124651A1 - Pyrrolidine-2-carboxylic acid derivatives as iglur antagonists - Google Patents

Pyrrolidine-2-carboxylic acid derivatives as iglur antagonists Download PDF

Info

Publication number
WO2014124651A1
WO2014124651A1 PCT/DK2014/050035 DK2014050035W WO2014124651A1 WO 2014124651 A1 WO2014124651 A1 WO 2014124651A1 DK 2014050035 W DK2014050035 W DK 2014050035W WO 2014124651 A1 WO2014124651 A1 WO 2014124651A1
Authority
WO
WIPO (PCT)
Prior art keywords
tert
unsaturated
saturated
pyrrolidine
alkyl
Prior art date
Application number
PCT/DK2014/050035
Other languages
French (fr)
Inventor
Lennart Bunch
Original Assignee
Københavns Universitet
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 Københavns Universitet filed Critical Københavns Universitet
Publication of WO2014124651A1 publication Critical patent/WO2014124651A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings

Definitions

  • the present invention relates to a class of substituted pyrrolidine-2- carboxylic acid derivatives as iGluR antagonist, their salt and solvates, phar- maceutical compositions comprising them, their use as medicament and in therapy, and preparation thereof.
  • the invention relates to a class of substituted pyrrolidine-2-carboxylic acid derivatives as iGluR antagonists, which is useful in the treatment of psychiatric diseases or neurological disorders or a disease or disorder associated with abnormal activities of iGluR receptors.
  • S-glutamate In the mammalian central nervous system (CNS), (S)-glutamate (Glu) functions as the major excitatory neurotransmitter. 1
  • the glutamatergic neurotransmitter system is involved in a vast number of basic neuro- physiological processes such as memory, cognition, as well as neuronal plasticity and development. 2"9
  • psychiatric diseases or neurological disorders such as depression, 10"12 anxiety, 13"15 addiction, 16 migraine, 17 and schizophrenia 18"22 may be directly related to disordered glutamatergic neurotransmis- sion.
  • elevated synaptic Glu levels or excessive Glu signaling is neurotoxic and will ultimately cause neuronal death.
  • neurodegenerative diseases such as Alzheimer's, 27"31 Huntington's, 32 amyotrophic lateral sclerosis (ALS), 33 cerebral stroke, 34 and epilepsy 35 may indeed be the result of a malfunctioning glutamatergic neurotransmitter system which may be reversed by action of small molecule Glu ligands.
  • Glu activates a number of pre- and post-synaptic Glu receptors.
  • the Glu receptors have been grouped in two main classes: the fast acting ionotropic Glu receptors (iGluRs) comprising the three groups AMPA receptors (subunits GluAl-4), kainate (KA) receptors (subunits GluKl-5), and NMDA receptors (subunits GluNl, GluN2A-D and GluN3A-C), 36 and the G-protein coupled metabotropic Glu receptors (mGluRs, subunits mGluRl-8), 37 which produce a slower signal transduction through second messenger systems.
  • iGluRs fast acting ionotropic Glu receptors
  • KA subunit composition GluK3 and GluK5
  • GluK2 and GluK3 Abnormal expression of KA subunit composition (GluK3 and GluK5) in the prefrontal cortex has been observed in schizophrenic sub- jects, 39 but also decreased expression of GluK2 and GluK3 from the medial dorsal thalamus to the dorsolateral prefrontal cortex and other cortical regions may be important to the pathophysiology of schizophrenia.
  • two population studies have suggested altered GluK3 expression (GRIK3 gene) as a risk factor, 41,42 whereas GluK2 (GRIK2 gene) in one Japa- nese study came out short.
  • GluK3 receptor In bipolar disorder the GluK3 receptor is suggested to play a role, 44 but also intervention of GluK2 may constitute a therapeutic target. 45 In an rodent (rat) model of pain, trigeminal caudal nucleus nerve terminals mainly express GluK2/GluK3 subunits, which evidence that differentiated expression of KA receptor subtypes plays a role at the various stages of pain transmission. 46
  • iGluR intradermal receptor
  • GluAl-4, GluKl-5, or GluNl-3 such as GluN2A, GluN2B, GluN2C or GluN2D
  • new antagonists having high affinity and/or high specificity to one and more of the iGluR receptors such as GluAl-4, GluKl-5, or GluNl- 3, such as GluN2A, GluN2B, GluN2C or GluN2D, would therefore be useful in the treatment of disorders and diseases associated with these receptors.
  • Q represents compounds of Formula (la) or (lb);
  • each case may represent if appropriate the presence of at least one double bond between T 2 and (Z 2 or Z 3 ), or between Z 2 and (Zi or Z 3 ), or between Zi and Ti, or between Ti and Z 4; or between Z 4 and T 2 ; or
  • Ti is C, or CH
  • T 2 is C, or CH
  • Zi is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Z 2 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Z 3 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Z 4 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Ri may together with Zi or Z 4 , or
  • Z 2 may together with Zi or Z 3 ,
  • Ri is H, OR 4 , Ci-Ce-alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, COOR 4 , N(OH)H, or NHR 4 ,
  • R 2 is independently selected among R 4 , O, OR 4 , halogen, N(OH)H, N(OH)R 4 , NHR 4 , COR 4 , CONHR 4 , CN, CF 3 , CCI 3 , SH, or S0 2 NHR 4 ,
  • R 3 is independently selected among R 4 , O, OR 4 , or halogen
  • R 4 is independently selected among H, OH, Ci-C 6 -alkyl, C 2 -C 6 - alkenyl, C 2 -C 6 -alkynyl, phenyl, Ci-C 6 -alkylphenyl, or saturated or unsaturated C 5 - or C 6 -cycloalkyl, or saturated or unsaturated Ci-C 6 -alkyl C 5 - or C 6 - heterocyclyl, wherein Ci-C 6 -alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, phenyl, Ci-C 6 - alkylphenyl, or saturated or unsaturated C 5 - or C 6 -cycloalkyl, or saturated or unsaturated Ci-C 6 -alkyl C 5 - or C 6 - heterocyclyl may be substituted with one or more substituents selected from the group comprising d-C 6 -alkyl, Ci-C 6
  • halogen represents CI, Br, or I
  • the compounds of Formula (I) as previously described are stereoisomeric and contain at least two isomeric centers. Thus, depending on the orientation of the stereoisomers, presence of four different diastereomers is possible.
  • compounds of Formula (II) according to Formula (I) wherein
  • Q represents compounds of Formula (Ia l) or (Ib2);
  • Q represents a saturated ring.
  • T 2 is C
  • Zi is CR 2 , N, S, O, or NR 3 ,
  • Z 2 is CR 2 , or N
  • Z 3 is CR 2 , or N
  • Z 4 is CR 2 , or N
  • Ri may together with Zi or Z 4 , or
  • Z 2 may together with Zi or Z 3 ,
  • saturated or unsaturated C 5 - or C 6 -cycloalkyl or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the saturated or unsaturated C 5 - or C 6 -cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR 4 or R 4 ;
  • Ri is H, OR 4 , Ci-Ce-alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, COOR 4 , N(OH)H, or NHR 4 ,
  • R 2 is R 4 , O, OR 4 , halogen, N(OH)H, N(OH)R 4 , NHR 4 , COR 4 , CONHR 4 , or S0 2 NHR 4 ,
  • R 3 is R 4 , O, OR 4 , or halogen
  • R 4 is H, Ci-C 6 -alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, phenyl, Ci-C 6 - alkylphenyl, or saturated or unsaturated C 5 - or C 6 -cycloalkyl, or saturated or unsaturated Ci-C 6 -alkyl C 5 - or C 6 -heterocyclyl, wherein Ci-C 6 -alkyl, C 2 -C 6 - alkenyl, C 2 -C 6 -alkynyl, phenyl, Ci-C 6 -alkylphenyl, or saturated or unsaturated C 5 - or C 6 -cycloalkyl, or saturated or unsaturated Ci-C 6 -alkyl C 5 - or C 6 - heterocyclyl may be substituted with one or more substituents selected from the group comprising Ci-C 6 -alkyl, Ci-C 6 -alkoxy
  • the compound of Formula (I) as previously described can comprise additional ringforming structures wherein
  • Ri may together with Zi or Z 4 , or
  • Z 2 may together with Zi or Z 3 ,
  • Z 5 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Z 5 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Z 6 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • Z 7 is CR 2 , C(R 2 ) 2 , N, S, O, or NR 3 ,
  • R 4 has the same meaning as given above, are also part of the invention.
  • compounds of Formula (I) are selected from the group wherein R 4 is alkyl, benzyl, alkylbiphenyl, preferably propyl, benzyl, or 3-methyl[l,l'-biphenyl].
  • Ci-6 alkyl denotes a straight chain or branched alkyl group with 1, 2, 3, 4, 5 or 6 carbon atoms.
  • Suitable Ci-6 alkyl groups include, for example, methyl, ethyl, propyl (e.g. n-propyl and isopropyl), butyl (e.g n-butyl, iso-butyl, sec-butyl and tert-butyl), pentyl (e.g. n- pentyl), and hexyl (e.g. n-hexyl).
  • Alkenyl may be inter- preted similarly to the term "alkyl”.
  • Alkenyl groups contain at least 1 double bond. Suitable alkenyl groups include ethenyl, propenyl, 1-butenyl, and 2- butenyl.
  • Alkenyl groups contain at least 1 triple bond.
  • saturated or unsaturated C 5 - or C 6 -cycloalkyl denotes cyclic carbon rings comprising 5 or 6 carbon atoms, wherein either a single or double bond between the mutually adjacent carbon atoms exist.
  • Suitable saturated or unsaturated C 5 - or C 6 -cycloalkyl groups include cyclopentane, cyclohexane, cyclopentene, cyclohexene, cyclopenta- di-ene, cyclohhexa-di-ene, and phenyl.
  • saturated or unsaturated heterocyclyl denotes a heterocyclic compound, such as a carbocyclyl group, phenyl group, or aryl residue, having atoms of at least two different elements as members of its ring.
  • Suitable ring atoms in heterocyclic compound may be C, N, S, or O.
  • Heterocyclic compounds according to the present invention may contain 3, 4, 5, 6, 7, 8 or even more rings atoms, preferably 5 or 6 ring atoms.
  • Suitable saturated or unsaturated heterocyclic compounds may include pyrrolidine, pyrrole, tetrahydrofuran, furan, thiolane, thiophene, imida- zolidine, pyrazolidine, imidazole, pyrazole, oxazolidine, isoxazolidine, oxazole, isoxazole, thiazolidine, isothiazolidine, thiazole, isothiazole, dioxolane, dithi- olane, triazoles, furazan, oxadiazole, thiadiazole, dithiazole, tetrazole, piperi- dine, pyridine, oxane, pyran, thiane thiopyran, piperazine, diazines, morpho- line, oxazine, thiomorpholine, thiazine, dioxane, dioxine, dithiane, dithiine, triazin
  • halogen comprises fluorine (F), chlorine (CI), bromine (Br) and iodine (I), more typically F, CI or Br.
  • Tautomers are isomers of organic compounds that readily interconvert by a chemical reaction called tautomerization. This reaction commonly results in the formal migration of a hydrogen atom or proton, accompanied by a switch of a single bond and adjacent double bond.
  • the compounds of the invention have one or more asymmetric centers.
  • Compounds with asymmetric centers give rise to enantiomers (optical isomers), diastereomers (configurational isomers) or both, and it is intended that all of the possible enantiomers and diastereomers in mixtures and as pure or partially purified compounds are included within the scope of this invention.
  • the present invention is meant to encompass all isomeric forms of the compounds of the invention.
  • the present invention includes all stereoisomers of compounds of Formula (I).
  • Compounds of Formula (I) comprises although depending on the choice of T 2 at least one chiral centers, i.e.
  • Diastereomers differ from enantiomers in that these are pairs of stereoisomers that differ in all stereocenters. Diastereomers have different physical properties (unlike enantiomers) and different chemical reactivity. Diastereoselectivity is the preference for the formation of one or more than one diastereomer over the other in an organic reaction.
  • the independent syntheses of the enantiomerically or diastereomeri- cally enriched compounds, or their chromatographic separations, may be achieved as known in the art by appropriate modification of the methodology disclosed herein.
  • Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates that are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers or diastereo- mers are isolated.
  • the separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
  • the coupling reaction is often the formation of salts using an enantiomerically pure acid or base.
  • the diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue.
  • the racemic mixture of the compounds can also be separated directly by chromatographic methods using chiral stationary phases, which methods are well known in the art.
  • any enantiomer or diastereomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
  • Pyrrolidine-2-carboxylic acid derivatives according to the invention can be prepared from the various examples given further below or by con- suiting handbooks within organic chemistry.
  • handbook - although not intending to be limited thereto - are "Organic Chemistry, 2 nd Edition, 2000, by Maitland Jones, Jr., and Organic Chemistry, 6th Edition, Robert T. Morrison, and Robert N. Boyd. These two specifically referred handbooks are hereby incorporated by reference.
  • pharmaceutically acceptable derivative in present context includes pharmaceutically acceptable salts, which indicate a salt which is not harmful to the patient.
  • Such salts include pharmaceutically acceptable basic or acid addition salts as well as pharmaceutically acceptable metal salts, ammonium salts and alkylated ammonium salts.
  • a pharmaceutically acceptable derivative further includes hydrates, polymorphs, esters and prodrugs, or other precursors of a compound which may be biologically metabolized into the active compound, or crystal forms of a compound.
  • Salts and solvates of the compounds of Formula (I) and physiologically functional derivatives thereof which are suitable for use in medicine are those wherein the counter-ion or associated solvent is pharmaceutically acceptable.
  • salts and solvates having non-pharmaceutically acceptable counter-ions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds and their pharmaceutically acceptable salts and solvates.
  • Pharmaceutically acceptable acid addition salts include those formed from hydrochloric, hydrobromic, sulfuric, nitric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, sulfamic, sulfanilic, succinic, oxalic, fumaric, maleic, malic, mandelic, glutamic, aspartic, oxaloacetic, methanesulfonic, ethanesulfonic, arylsulfonic (for example p-toluenesulfonic, benzenesulfonic, naphthalenesulfonic or naphthalenedisulfonic), salicylic, glu- taric, gluconic, tricarballylic, cinnamic, substituted cinnamic (for example, phenyl, methyl, methoxy or halo substituted cinnamic, including
  • Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases such as dicyclohexylamine and [Lambda]/-methyl-D-glucamine.
  • some of the crystalline forms of the compounds may exist as polymorphs and as such are intended to be included in the present invention.
  • some of the compounds may form solvates with water (i.e. hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention.
  • the compounds, including their salts can also be obtained in the form of their hy- drates, or include other solvents used for their crystallization.
  • Organic molecules can form crystals that incorporate water into the crystalline structure without modification of the organic molecule.
  • An organic molecule can exist in different crystalline forms, each different crystalline forms may contain the same number of water molecules pr organic molecule or a different number of water molecules pr organic molecule.
  • administering shall encompass the treatment of the various disorders described with derivatives of the claimed compounds which convert to the active compound in vivo after administration to the subject.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bund- gaard, Elsevier, 1985.
  • antagonist in the present context refers to a substance that does not provoke a biological response itself upon binding to a receptor. Hence, antagonists have affinity to but no efficacy for their cognate recep- tors, and binding will disrupt the interaction and inhibit the function of e.g. an agonist.
  • AMPA receptor denotes a receptor family within the iG- luRs receptors.
  • the AMPA receptor comprises subunits such as, GluAl, GluA2, GluA3, or GluA4.
  • KA denotes a receptor family within iGluRs receptors.
  • KA receptor comprises subunits such as GluKl, GluK2, GluK3, GluK4, or GluK5.
  • NMDA denotes a receptor family within iGluRs receptors.
  • the NMDA receptor comprises subunits such as GluNl, GluN2A, GluN2B, GluN2C, GluN2D, GluN3A, GluN3B, or GluN3C.
  • a receptor antagonist defined by the Formula (I) is thus capable of binding to the GluKl, GluK2, GluK3, GluK4, or GluK5 receptor, respectively.
  • the same considerations apply similarly to receptor antagonists of the AMPA receptor and NMDA receptor subunits.
  • the antagonist may be an antagonist of several different types of receptors, and thus capable of binding to several different types of receptors, such AMPA receptors, KA receptors, and NMDA receptors.
  • the antagonist can also be a selective antagonist, which only binds to and activates one type of receptor.
  • Antagonist may bind reversible or irre- versible depending on the antagonist-receptor complex.
  • the term "IC 50" is commonly used as a measure of antagonist drug potency and reflects the measure of the effectiveness of a compound in inhibiting biological or biochemical function. This quantitative measure indicates how much of a compound of Formula (I) is needed to inhibit 50% of the ac- tivity of a particular receptor. IC 50 can be regarded as the functional strength of the different compounds of Formula (I).
  • IC 50 is not a direct indicator of affinity although the two can be related at least for competitive agonists and antagonists by the Cheng-Prusoff equation.
  • the term 'V refers to the binding affinity, which describe the binding of compounds of Formula (I) to a receptor.
  • composition is intended to encompass a product comprising compounds of Formula (I) in the therapeutically effective amounts, as well as any product which results, directly or indi- rectly, from combinations of the claimed compounds.
  • terapéuticaally effective amount of a compound as used herein refers to an amount sufficient to cure, alleviate, prevent, reduce the risk of, or partially arrest the clinical manifestations of a given disease or disorder and its complications. An amount adequate to accomplish this is de- fined as a “therapeutically effective amount”.
  • Compounds of Formula (I) according to the present invention may be used in pharmaceutical compositions and method for treatment of disorders, diseases in a subject, or conditions associated with the dysfunction of iGluR receptors, e.g. the AMPA receptors, KA receptors and NMDA receptors, and their corresponding subunits, GluAl-4, GluKl-5 and GluNl, GluN2A-D, and GluN3A-C.
  • iGluR or its subtypes such as GluAl-4, GluKl-5, GluNl-3 and its subtypes with antagonists according to the present invention would be helpful in the treatment of disorders and diseases associated with these receptors.
  • treatment refers to the management and care of a patient for the purpose of combating a condition, disease or disorder.
  • the term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound for the purpose of: alleviating or re- lieving symptoms or complications; delaying the progression of the condition, disease or disorder; curing or eliminating the condition, disease or disorder; and/or preventing the condition, disease or disorder, wherein "preventing” or “prevention” is to be understood to refer to the management and care of a patient for the purpose of hindering the development of the condition, dis- ease or disorder, and includes the administration of the active compounds to prevent or reduce the risk of the onset of symptoms or complications.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment. Treatment of animals, such as mice, rats, dogs, cats, cows, sheep and pigs, is, however, also within the scope of the present invention.
  • the present invention relates to compounds of Formula (I) or pharmaceutical compositions thereof, or methods for treatment of diseases or conditions binding one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits to obtain a beneficial therapeutic effect.
  • compounds of Formula (I) or pharmaceutical compositions thereof are used for treatment of diseases or conditions binding one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits to obtain a beneficial therapeu- tic effect.
  • the present invention relates to compounds of Formula (I) or pharmaceutical compositions thereof, or methods for treatment of disorders of the central nervous system, neuro-physiological processes such as memory, cognition; as well as neuronal plasticity and de- velopment, psychiatric diseases or neurological disorders such as depression, anxiety, addiction, pain, migraine, and schizophrenia, and neurodegenerative diseases; such as Alzheimer, Huntington disease, amyotrophic lateral sclerosis (ALS), cerebral stroke, and epilepsy; and diseases including aching, ADHD, Autism, Diabetes, Huntington's disease, ischemia, multiple sclerosis, Parkinson's disease (Parkinsonism), Rasmussen's encephalitis, seizures, AIDS dementia complex, amyotrophic lateral sclerosis, combined systems disease (vitamin B12 deficiency), drug addiction, drug tolerance, drug dependency, glaucoma, hepatic encephalopathy, hydroxybutyric aminoaciduria, hyperho- mocysteinemia and homocysteinuria,
  • Formula (I) is administered to subjects in need of treatment in pharmaceutically effective doses.
  • a therapeutically effective amount of a compound according to the present invention is an amount sufficient to cure, prevent, reduce the risk of, alleviate or partially arrest the clinical manifestations of a given disease or its complications.
  • the amount that is effective for a particular therapeutic purpose will depend on the severity and the sort of the disease as well as on the weight and general state of the subject.
  • the antagonists of the present invention may be administered one or several times per day, such as from 1 to 4 times per day, such as from 1 to 3 times per day, such as from 1 to 2 times per day, wherein administration from 1 to 3 times per day is preferred.
  • the present invention relates to antagonist of compound of Formula (I) which is administered in doses of 0.5-1500 mg/day, preferably 0.5-200 mg/day, more preferably 0.5-60 mg/day, even more preferably 0.5-30 mg/day.
  • the present invention relates to antagonist of compound of Formula (I) suitable for oral, rectal, nasal, pulmonary, buccal, sublingual, transdermal or parenteral administration.
  • any of the processes for preparation of the compounds of the present invention it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Syn- thesis, John Wiley & Sons, 1991, fully incorporated herein by reference.
  • the protecting groups may be removed at a convenient subsequent stage using methods known from the art.
  • the present invention relates to compounds of Formula (I), such as
  • R 3-Ph-Bn Reagents and conditions, a) BH 3 , THF, 0°C to rt, aq work-up, then TBSCI, imidazole, DMF, 0°C to rt (71%); b) For 6b: RBr, K 2 C0 3 , rt in DMF (90%); for 6c,d: RBr, K 2 C0 3 , reflux in acetone (92-95%).
  • the synthesis of analogs 2f-n ( Figure 3) was carried out following the general strategy described for 2a-e (Scheme 1-5). Starting from 5 and appropriately substituted aryl analog 6 analogs 2f-n were prepared in moderate yields.
  • the five new analogs 2a-e were characterized pharmacologically in radio ligand binding assays at native iGluRs (rat synaptosomes) and cloned homomeric subtypes, GluKl-3 (Table 1).
  • 4'-Hydroxy analog 2a displayed a general higher binding affinity for the iGluRs in native tissue, and reduced iGluR class selectivity, as compared to lead structure 1.
  • a 25- and 15-fold higher affinity for AMPA and KA receptors was observed, while a 6- fold higher affinity at NMDA receptors could be demonstrated.
  • LC-MS was performed using an Agilent 1200 series solvent delivery system equipped with an autoinjector coupled to an Agilent 6400 series triple quadrupole mass spectrometer equipped with an electrospray ionization source. Gradients of 10% aqueous acetonitrile + 0.05% formic acid (buffer A) and 90% aqueous acetonitrile + 0.046% formic acid (buffer B) were employed or an Agilent 1200 system using a C18 reverse phase column (Zorbax 300 SB-C18, 21.1 mm - 250 mm) with a linear gradient of the binary solvent system of H 2 0/CH 3 CN/TFA (A: 100/0/0.1 and B: 5/95/0.1) with a flow rate of 20 mL/min.
  • Optical rotation was measured using a Perkin-Elmer 241 spec- trometer, with Na lamp at 589 nm . Melting points were measured using an automated melting point apparatus, MPA100 OptiMelt (SRS) and are uncorrected. Compounds were dry either under high vacuum or freeze dried using a Holm & Halby, Heto LyoPro 6000 freeze drier.
  • the compound 10b (180 mg, 0.46 mmol, 1.00 equiv) was dissolved in AcOH (2.5 mL) and added 2M HCI in Et 2 0 (2.5 mL, 5 mmol, 10.9 equiv). The mixture was left to stir under N 2 at r.t. for 18 h. The mixture was suspended in H 2 0 (10 mL), solidified by cooling on a dry ice/acetone bath and freeze dried to yield 149 mg (99%) of 2b as a slightly off white, crisp solid.
  • the compound 10c (302 mg, 0.68 mmol, 1.00 equiv) was dissolved in AcOH (4 mL) and added 2M HCI in Et 2 0 (4 mL, 8 mmol, 11.8 equiv). The mixture was left to stir under N 2 at RT for 18 h. The mixture was suspended in H 2 0 (20 mL), solidified by cooling on a dry ice/acetone bath and freeze dried to yield 228 mg (88%) of 2c as a slightly off white, crisp solid.
  • Phenol 16 (4.00 g, 12.6 mmol, 1.00 equiv) was dissolved in DMF (25 mL) and added K 2 C0 3 (3.48 g, 25.2 mmol, 2.00 equiv) and propyl bromide (2.30 mL, 3.11 g, 25.3 mmol, 2.01 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was added Et 2 0 (200 mL) and washed with water (3 x 100 mL), brine (75 mL), dried over MgS0 4 , filtered and concentrated in vacuo. The mixture was purified by DCVC (dia.
  • Phenol 16 (3.50 g, 11.0 mmol, 1.00 equiv) was dissolved in acetone (50 mL) and added K 2 C0 3 (3.10 g, 22.4 mmol, 2.04 equiv) and benzyl bro- mide (2.10 mL, 3.02 g, 17.7 mmol, 1.61 equiv). The mixture was left to stir at reflux for 24 h. Piperazine (4.75 g, 55.1 mmol, 5.01 equiv) was added and the mixture refluxed for another 2.5 h. The mixture was cooled to r.t. and concentrated in vacuo.
  • Phenol 16 (3.00 g, 9.46 mmol, 1.00 equiv) was dissolved in acetone (50 mL) and added K 2 C0 3 (2.61 g, 18.9 mmol, 2.00 equiv) and 3- phenylbenzyl bromide (3.52 g, 14.2 mmol, 1.50 equiv). The mixture was left to stir at reflux for 24 h. Piperazine (4.10 g, 47.6 mmol, 5.03 equiv) was added and the mixture refluxed for another 2.5 h. The mixture was cooled to r.t. and concentrated in vacuo.
  • Enone 5 (1.00 g, 3.05 mmol, 1.00 equiv) was dissolved in dry Et 2 0 (3.0 mL) and added dropwise to the cuprate mixture at -78 °C, which resulted in a slight color change to orange. The temperature was raised to -42 °C and the reaction mixture was stirred at this temperature for 1 hour. The dark brown solution with barely any precipitation was quenched by addition of sat. NH 4 CI(aq) (5 mL), allowed to warm up to ambient temperature and then transferred to a separating funnel with brine (30 mL) and EtOAc (30 mL). The organic layer was separated and the blue aqueous layer was extracted with EtOAc (2 x 30 mL).
  • Solution A Thiophene (482 mg, 5.74 mmol, 1.57 equiv) was dissolved in dry Et 2 0 (6 mL) in a dry vial under N 2 . The mixture was cooled to 0 °C and n-BuLi (2.34 mL, 5.85 mmol, 1.60 equiv, 2.50M) added dropwise over the course of 15 min. The mixture was left to stir at 0 °C for 15 min. then at r.t. for 2 h (a white, colloid precipitate was formed).
  • Bromide 6b (1.65 g, 4.59 mmol, 1.25 equiv) was dissolved in dry Et 2 0 (50 mL) in a dry flask under N 2 and cooled to -78 °C.
  • t-BuLi (5.76 mL, 9.22 mmol, 2.52 equiv, 1.60 M) was added dropwise over the course of 20 min.
  • the mixture was left to stir at -78 °C for 45 min. (The solution became slightly colored and unclear) before a suspension of CuCN (410 mg, 4.58 mmol, 1.25 equiv) in dry Et 2 0 (4 mL) was added dropwise over the course of 6 min.
  • the mixture was cooled to 0 °C and dropwise added H 2 0 (5 mL) over the course of 10 min., NaOH (2M, 25 mL) dropwise over the course of 20 min and H 2 0 2 (30%, 5 mL) over the course of 5 min (organic/aqueous ratio important). After 5 min the mixture was remove from the icebath and left to stir at r.t. for 1 h. The mixture was poured into sat. NaHC0 3 (100 mL) and EtOAc (75 mL).
  • the compound 7c (1.66 g, 2.53 mmol, 1.00 equiv) was dissolved in dry THF (20 mL) and added IM BH 3 THF complex (25 mL, 25 mmol, 9.88 equiv) over the course of 5 min.
  • the mixture was refluxed under N 2 for 20 h.
  • the mixture was cooled to 0 °C, added THF (40 mL) and dropwise added H 2 0 (6 mL) over the course of 15 min.
  • NaOH (2M, 30 mL) was dropwise over the course of 15 min and H 2 0 2 (30 %, 10 mL) over the course of 15 min (organic/aqueous ratio important).
  • the compound 7d (1.77 g, 2.42 mmol, 1.00 equiv) was dissolved in dry THF (20 mL) and added 1M BH 3 THF complex (25 mL, 25 mmol, 10.3 equiv) over the course of 5 min.
  • the mixture was refluxed under N 2 for 20 h.
  • the mixture was cooled to 0 °C, added THF (40 mL) and dropwise added H 2 0 (6 mL) over the course of 15 min.
  • NaOH (2M, 30 mL) was dropwise over the course of 15 min and H 2 0 2 (30 %, 10 mL) over the course of 15 min (organic/aqueous ratio important).
  • the compound 8a (448 mg, 0.94 mmol, 1.0 equiv) was dissolved in dry THF (9 mL) under an atmosphere of N 2 and 1M TBAF in THF (2.8 mL, 736 mg, 2.81 mmol, 3.0 equiv) was added at room temperature. The reaction mixture was stirred for two. H 2 0 (20 mL) and sat. aqueous NaHC0 3 (20 mL) were added and then extracted with EtOAc (3 x 30 mL). The combined organic phases were dried over anhydrous Na 2 S0 4 , filtered and evaporated in vacuo to dryness.
  • the compound 8c (967 g, 1.51 mmol, 1.00 equiv) was dissolved in dry THF (15 mL) in a dry vial and added 1M TBAF (6 mL, 6 mmol, 3.97 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was quenched using 50% sat. NaHC0 3 (50 mL) and transferred to a separation funnel containing EtOAc (50 mL).
  • the compound 8d (800 g, 1.11 mmol, 1.00 equiv) was dissolved in dry THF (15 mL) in a dry vial and added 1M TBAF (5 mL, 5 mmol, 4.50 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was quenched using phosphate buffer (50 mL, Ph 7) and transferred to a separation funnel containing EtOAc (50 mL).
  • Suspension A NaI0 4 (1.92 g, 8.98 mmol, 9.98 equiv) was suspended in H 2 0 (6 ml_) and after stirring at r.t. for 5 min. RuCI 3 ⁇ H 2 0 (8 mg, 0.035 mmol, 0.04 equiv) was added. The black suspension was stirred for 1 min at r.t. prior to use.
  • H 2 0 (10 mg, 0.044 mmol, 0.04 equiv) were suspended in H 2 0 (7.5 mL) and stired at r.t. for 1 min. prior to use.
  • Kalivas, P. W. Cocaine and amphetamine-like psychostimulants neurocircuitry and glutamate neuroplasticity. Dialogues in clinical neuros- cience 2007, 9, 389-97.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Psychiatry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pain & Pain Management (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to compounds of Formula (I), combinations and use thereof for disease therapy, or pharmaceutically acceptable salt or solvate thereof, including all tautomers, stereoismers and polymorphs thereof, which are iGlu R inhibitors, and hence are useful in the treatment of psychiatric diseases or neurological disorders or a disease or disorder associated with abnormal activities of iGluR receptors.

Description

Pyrrolidine-2-carboxylic acid derivatives as iGluR antagonists Field of the invention
The present invention relates to a class of substituted pyrrolidine-2- carboxylic acid derivatives as iGluR antagonist, their salt and solvates, phar- maceutical compositions comprising them, their use as medicament and in therapy, and preparation thereof. In particular, the invention relates to a class of substituted pyrrolidine-2-carboxylic acid derivatives as iGluR antagonists, which is useful in the treatment of psychiatric diseases or neurological disorders or a disease or disorder associated with abnormal activities of iGluR receptors.
Background
In the mammalian central nervous system (CNS), (S)-glutamate (Glu) functions as the major excitatory neurotransmitter.1 The glutamatergic neurotransmitter system is involved in a vast number of basic neuro- physiological processes such as memory, cognition, as well as neuronal plasticity and development.2"9 Thus, psychiatric diseases or neurological disorders such as depression,10"12 anxiety,13"15 addiction,16 migraine,17 and schizophrenia18"22 may be directly related to disordered glutamatergic neurotransmis- sion. Moreover, elevated synaptic Glu levels or excessive Glu signaling is neurotoxic and will ultimately cause neuronal death.23"26 Thus, it is believed that neurodegenerative diseases such as Alzheimer's,27"31 Huntington's,32 amyotrophic lateral sclerosis (ALS),33 cerebral stroke,34 and epilepsy35 may indeed be the result of a malfunctioning glutamatergic neurotransmitter system which may be reversed by action of small molecule Glu ligands.1
Once released from the pre-synaptic neuron into the synapse, Glu activates a number of pre- and post-synaptic Glu receptors. On the basis of the pharmacological profile and ligand selectivity, the Glu receptors have been grouped in two main classes: the fast acting ionotropic Glu receptors (iGluRs) comprising the three groups AMPA receptors (subunits GluAl-4), kainate (KA) receptors (subunits GluKl-5), and NMDA receptors (subunits GluNl, GluN2A-D and GluN3A-C),36 and the G-protein coupled metabotropic Glu receptors (mGluRs, subunits mGluRl-8),37 which produce a slower signal transduction through second messenger systems. For example, disturbances of expression of KA receptors and function of KA receptors has been suggested to be linked to severe neurological- and psychiatric diseases.38 Abnormal expression of KA subunit composition (GluK3 and GluK5) in the prefrontal cortex has been observed in schizophrenic sub- jects,39 but also decreased expression of GluK2 and GluK3 from the medial dorsal thalamus to the dorsolateral prefrontal cortex and other cortical regions may be important to the pathophysiology of schizophrenia.40 Furthermore, two population studies have suggested altered GluK3 expression (GRIK3 gene) as a risk factor,41,42 whereas GluK2 (GRIK2 gene) in one Japa- nese study came out short.43 In bipolar disorder the GluK3 receptor is suggested to play a role,44 but also intervention of GluK2 may constitute a therapeutic target.45 In an rodent (rat) model of pain, trigeminal caudal nucleus nerve terminals mainly express GluK2/GluK3 subunits, which evidence that differentiated expression of KA receptor subtypes plays a role at the various stages of pain transmission.46
To this date only selective antagonists for the GluKl subtype has been reported.47
Figure imgf000003_0001
(S)-Glutamate 1 LY466195 UBP310
(Glu) (CNG-10100)
Chemical structures of (S)-Glu, rationally designed iGluR anta- gonist 1 (CNG-10100) and examples of selective high-affinity GluKl antagonists LY46619548 and UBP310.
Hence, there is a strong need present for novel selective antagonists for iGluR or its subtypes such as GluAl-4, GluKl-5, or GluNl-3, such as GluN2A, GluN2B, GluN2C or GluN2D, which can be used to elucidate the role and function of iGluR receptors under both physiological and pathological conditions. Thus, new antagonists having high affinity and/or high specificity to one and more of the iGluR receptors such as GluAl-4, GluKl-5, or GluNl- 3, such as GluN2A, GluN2B, GluN2C or GluN2D, would therefore be useful in the treatment of disorders and diseases associated with these receptors.
Summary of the invention With this background, it is an object of the present invention in a first embodiment to provide a compound of Formula (I)
Figure imgf000004_0001
and pharmaceutically acceptable derivatives, as well as all tautomers and stereoisomers of compound of Formula (I), wherein
Q represents compounds of Formula (la) or (lb);
Figure imgf000004_0002
— in each case may represent if appropriate the presence of at least one double bond between T2 and (Z2 or Z3), or between Z2 and (Zi or Z3), or between Zi and Ti, or between Ti and Z4; or between Z4 and T2; or
Ti is C, or CH,
T2 is C, or CH,
Zi is CR2, C(R2)2, N, S, O, or NR3,
Z2 is CR2, C(R2)2, N, S, O, or NR3,
Z3 is CR2, C(R2)2, N, S, O, or NR3,
Z4 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, and Z3 cannot represent adjacent O or S;
Ri may together with Zi or Z4, or
Z2 may together with Zi or Z3,
form a saturated or unsaturated C5- or C6-cycloalkyl, or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the satu- rated or unsaturated C5- or C6-cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR4 or R4,
Ri is H, OR4, Ci-Ce-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, COOR4, N(OH)H, or NHR4,
R2 is independently selected among R4, O, OR4, halogen, N(OH)H, N(OH)R4, NHR4, COR4, CONHR4, CN, CF3, CCI3, SH, or S02NHR4,
R3 is independently selected among R4, O, OR4, or halogen,
R4 is independently selected among H, OH, Ci-C6-alkyl, C2-C6- alkenyl, C2-C6-alkynyl, phenyl, Ci-C6-alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- heterocyclyl, wherein Ci-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, phenyl, Ci-C6- alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- heterocyclyl may be substituted with one or more substituents selected from the group comprising d-C6-alkyl, Ci-C6- alkoxy, aryl, halogen, and amine,
halogen represents CI, Br, or I, and
with the proviso that Zi, Z2, Z3 and Z4 are not all CH, and Ti and T2 are not both C, when Ri is OH.
In one particular embodiment of the present invention, the compounds of Formula (I) as previously described are stereoisomeric and contain at least two isomeric centers. Thus, depending on the orientation of the stereoisomers, presence of four different diastereomers is possible. Thus, compounds of Formula (II) according to Formula (I), wherein
Figure imgf000005_0001
(II),
wherein
Q represents compounds of Formula (Ia l) or (Ib2);
Figure imgf000006_0001
, are also part of the invention.
In one particular embodiment of the present invention, Q represents a saturated ring.
Thus, compounds according to Formula (I), wherein
Q represents
.T2
z4- ^z3
/
Figure imgf000006_0002
wherein
Ti is C,
T2 is C,
Zi is CR2, N, S, O, or NR3,
Z2 is CR2, or N, Z3 is CR2, or N,
Z4 is CR2, or N,
wherein the residues Zi, Z2, and Z3 cannot represent adjacent O or
S;
Ri may together with Zi or Z4, or
Z2 may together with Zi or Z3,
form a saturated or unsaturated C5- or C6-cycloalkyl, or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the saturated or unsaturated C5- or C6-cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR4 or R4;
Ri is H, OR4, Ci-Ce-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, COOR4, N(OH)H, or NHR4,
R2 is R4, O, OR4, halogen, N(OH)H, N(OH)R4, NHR4, COR4, CONHR4, or S02NHR4,
R3 is R4, O, OR4, or halogen,
R4 is H, Ci-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, phenyl, Ci-C6- alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6-heterocyclyl, wherein Ci-C6-alkyl, C2-C6- alkenyl, C2-C6-alkynyl, phenyl, Ci-C6-alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- heterocyclyl may be substituted with one or more substituents selected from the group comprising Ci-C6-alkyl, Ci-C6-alkoxy, aryl, halogen, and amine, halogen represents CI, Br, or I, and
with the proviso that Zi, Z2, Z3 and Z4 are not all CH, and Ti and T2 are not both C when Ri is OH,
are also part of the invention.
In a second embodiment of the present invention, the compound of Formula (I) as previously described can comprise additional ringforming structures wherein
Ri may together with Zi or Z4, or
Z2 may together with Zi or Z3,
form a saturated or unsaturated C5- or C6-cycloalkyl, or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the satu- rated or unsaturated C5- or C6-cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR4 or R4.
In particular, in yet a further embodiment, compounds of Formula (I) as previously described, which are selected from the group consisting of
Figure imgf000008_0001
(IV),
wherein
Z5 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, Z3 and Z5 cannot represent adjacent O
Figure imgf000008_0002
Figure imgf000009_0001
wherein
Z5 is CR2, C(R2)2, N, S, O, or NR3,
Z6 is CR2, C(R2)2, N, S, O, or NR3,
Z7 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, Z3, Z5, Z6, Z7 cannot represent adjacent
O or S,
— , Ti, T2, Zi, Z2, Z3, Z4, R2, R2, R3, R4 have the same meaning as given above,
are also part of the invention.
In another embodiment of the present invention, the compounds Formula (I) as previously described, which is selected from the group consisting of
Figure imgf000009_0002
wherein
R4 has the same meaning as given above, are also part of the invention.
In one particular embodiment of the invention, compounds of Formula (I) are selected from the group wherein R4 is alkyl, benzyl, alkylbiphenyl, preferably propyl, benzyl, or 3-methyl[l,l'-biphenyl].
Each of the described embodiments of the present invention is to be construed as disclosing the present invention either individually or in combination with the other embodiments.
Detailed description of the invention
In the following the present invention is described in more detail. All individual features and details can be individually applied to each embodiment and aspect of the compounds of Formula (I), its preparations, its formulations, its methods and its use.
The term "Ci-6 alkyl", unless specifically limited, denotes a straight chain or branched alkyl group with 1, 2, 3, 4, 5 or 6 carbon atoms. Suitable Ci-6 alkyl groups include, for example, methyl, ethyl, propyl (e.g. n-propyl and isopropyl), butyl (e.g n-butyl, iso-butyl, sec-butyl and tert-butyl), pentyl (e.g. n- pentyl), and hexyl (e.g. n-hexyl).
The term "Ci-6 alkenyl", unless specifically limited, may be inter- preted similarly to the term "alkyl". Alkenyl groups contain at least 1 double bond. Suitable alkenyl groups include ethenyl, propenyl, 1-butenyl, and 2- butenyl.
The term "Ci-6 alkynyl", unless specifically limited, may be interpreted similarly to the term "alkyl". Alkenyl groups contain at least 1 triple bond.
The term "saturated or unsaturated C5- or C6-cycloalkyl", unless specifically limited, denotes cyclic carbon rings comprising 5 or 6 carbon atoms, wherein either a single or double bond between the mutually adjacent carbon atoms exist. Suitable saturated or unsaturated C5- or C6-cycloalkyl groups include cyclopentane, cyclohexane, cyclopentene, cyclohexene, cyclopenta- di-ene, cyclohhexa-di-ene, and phenyl.
The term "saturated or unsaturated heterocyclyl", unless specifically limited, denotes a heterocyclic compound, such as a carbocyclyl group, phenyl group, or aryl residue, having atoms of at least two different elements as members of its ring. Suitable ring atoms in heterocyclic compound may be C, N, S, or O. Heterocyclic compounds according to the present invention may contain 3, 4, 5, 6, 7, 8 or even more rings atoms, preferably 5 or 6 ring atoms. Suitable saturated or unsaturated heterocyclic compounds may include pyrrolidine, pyrrole, tetrahydrofuran, furan, thiolane, thiophene, imida- zolidine, pyrazolidine, imidazole, pyrazole, oxazolidine, isoxazolidine, oxazole, isoxazole, thiazolidine, isothiazolidine, thiazole, isothiazole, dioxolane, dithi- olane, triazoles, furazan, oxadiazole, thiadiazole, dithiazole, tetrazole, piperi- dine, pyridine, oxane, pyran, thiane thiopyran, piperazine, diazines, morpho- line, oxazine, thiomorpholine, thiazine, dioxane, dioxine, dithiane, dithiine, triazine, trioxane, or tetrazine.
The term "halogen" comprises fluorine (F), chlorine (CI), bromine (Br) and iodine (I), more typically F, CI or Br.
All possible tautomers of the claimed compounds are included in the present invention. Tautomers are isomers of organic compounds that readily interconvert by a chemical reaction called tautomerization. This reaction commonly results in the formal migration of a hydrogen atom or proton, accompanied by a switch of a single bond and adjacent double bond.
As compounds of Formula (I) contains at least 2 two asymmetric carbons, there are up to 4 possible configurations, which cannot all be non- superimposable mirror images of each other.
The compounds of the invention have one or more asymmetric centers. Compounds with asymmetric centers give rise to enantiomers (optical isomers), diastereomers (configurational isomers) or both, and it is intended that all of the possible enantiomers and diastereomers in mixtures and as pure or partially purified compounds are included within the scope of this invention. The present invention is meant to encompass all isomeric forms of the compounds of the invention. The present invention includes all stereoisomers of compounds of Formula (I). Compounds of Formula (I) comprises although depending on the choice of T2 at least one chiral centers, i.e. at the second position of pyrrolidine, a COOH group, and at the third position of pyrrolidine, a 5- or 6-membered ring, as indicated by the Q-group. Diastereomers differ from enantiomers in that these are pairs of stereoisomers that differ in all stereocenters. Diastereomers have different physical properties (unlike enantiomers) and different chemical reactivity. Diastereoselectivity is the preference for the formation of one or more than one diastereomer over the other in an organic reaction.
The independent syntheses of the enantiomerically or diastereomeri- cally enriched compounds, or their chromatographic separations, may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates that are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers or diastereo- mers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The coupling reaction is often the formation of salts using an enantiomerically pure acid or base. The diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods using chiral stationary phases, which methods are well known in the art.
Alternatively, any enantiomer or diastereomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
Pyrrolidine-2-carboxylic acid derivatives according to the invention can be prepared from the various examples given further below or by con- suiting handbooks within organic chemistry. Examples of such handbook - although not intending to be limited thereto - are "Organic Chemistry, 2nd Edition, 2000, by Maitland Jones, Jr., and Organic Chemistry, 6th Edition, Robert T. Morrison, and Robert N. Boyd. These two specifically referred handbooks are hereby incorporated by reference.
The term "pharmaceutically acceptable derivative" in present context includes pharmaceutically acceptable salts, which indicate a salt which is not harmful to the patient. Such salts include pharmaceutically acceptable basic or acid addition salts as well as pharmaceutically acceptable metal salts, ammonium salts and alkylated ammonium salts. A pharmaceutically acceptable derivative further includes hydrates, polymorphs, esters and prodrugs, or other precursors of a compound which may be biologically metabolized into the active compound, or crystal forms of a compound. Salts and solvates of the compounds of Formula (I) and physiologically functional derivatives thereof which are suitable for use in medicine are those wherein the counter-ion or associated solvent is pharmaceutically acceptable. However, salts and solvates having non-pharmaceutically acceptable counter-ions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds and their pharmaceutically acceptable salts and solvates.
Pharmaceutically acceptable acid addition salts include those formed from hydrochloric, hydrobromic, sulfuric, nitric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, sulfamic, sulfanilic, succinic, oxalic, fumaric, maleic, malic, mandelic, glutamic, aspartic, oxaloacetic, methanesulfonic, ethanesulfonic, arylsulfonic (for example p-toluenesulfonic, benzenesulfonic, naphthalenesulfonic or naphthalenedisulfonic), salicylic, glu- taric, gluconic, tricarballylic, cinnamic, substituted cinnamic (for example, phenyl, methyl, methoxy or halo substituted cinnamic, including 4-methyl and 4-methoxycinnamic acid), ascorbic, oleic, naphthoic, hydroxynaphthoic (for example 1- or 3-hydroxy-2-naphthoic), naphthaleneacrylic (for example naphthalene-2-acrylic), benzoic, 4-methoxybenzoic, 2- or 4- hydroxybenzoic, 4-chlorobenzoic, 4-phenylbenzoic, benzeneacrylic (for example 1,4- benzene- diacrylic), isethionic acids, perchloric, propionic, glycolic, hydroxyethanesul- fonic, pamoic, cyclohexanesulfamic, salicylic, saccharinic and trifluoroacetic acid.
Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases such as dicyclohexylamine and [Lambda]/-methyl-D-glucamine.
Furthermore, some of the crystalline forms of the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e. hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention. The compounds, including their salts, can also be obtained in the form of their hy- drates, or include other solvents used for their crystallization. Organic molecules can form crystals that incorporate water into the crystalline structure without modification of the organic molecule. An organic molecule can exist in different crystalline forms, each different crystalline forms may contain the same number of water molecules pr organic molecule or a different number of water molecules pr organic molecule.
The term "administering" shall encompass the treatment of the various disorders described with derivatives of the claimed compounds which convert to the active compound in vivo after administration to the subject. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bund- gaard, Elsevier, 1985.
The term "antagonist" in the present context refers to a substance that does not provoke a biological response itself upon binding to a receptor. Hence, antagonists have affinity to but no efficacy for their cognate recep- tors, and binding will disrupt the interaction and inhibit the function of e.g. an agonist.
The term "AMPA receptor" denotes a receptor family within the iG- luRs receptors. The AMPA receptor comprises subunits such as, GluAl, GluA2, GluA3, or GluA4.
The term "KA" denotes a receptor family within iGluRs receptors. The
KA receptor comprises subunits such as GluKl, GluK2, GluK3, GluK4, or GluK5.
The term "NMDA" denotes a receptor family within iGluRs receptors. The NMDA receptor comprises subunits such as GluNl, GluN2A, GluN2B, GluN2C, GluN2D, GluN3A, GluN3B, or GluN3C.
A receptor antagonist defined by the Formula (I) , is thus capable of binding to the GluKl, GluK2, GluK3, GluK4, or GluK5 receptor, respectively. The same considerations apply similarly to receptor antagonists of the AMPA receptor and NMDA receptor subunits.
The antagonist may be an antagonist of several different types of receptors, and thus capable of binding to several different types of receptors, such AMPA receptors, KA receptors, and NMDA receptors.
The antagonist can also be a selective antagonist, which only binds to and activates one type of receptor. Antagonist may bind reversible or irre- versible depending on the antagonist-receptor complex. The term "IC50" is commonly used as a measure of antagonist drug potency and reflects the measure of the effectiveness of a compound in inhibiting biological or biochemical function. This quantitative measure indicates how much of a compound of Formula (I) is needed to inhibit 50% of the ac- tivity of a particular receptor. IC50 can be regarded as the functional strength of the different compounds of Formula (I).
IC50 is not a direct indicator of affinity although the two can be related at least for competitive agonists and antagonists by the Cheng-Prusoff equation.
The term 'V refers to the binding affinity, which describe the binding of compounds of Formula (I) to a receptor.
As used herein, the term "pharmaceutical composition" is intended to encompass a product comprising compounds of Formula (I) in the therapeutically effective amounts, as well as any product which results, directly or indi- rectly, from combinations of the claimed compounds.
The term "therapeutically effective amount" of a compound as used herein refers to an amount sufficient to cure, alleviate, prevent, reduce the risk of, or partially arrest the clinical manifestations of a given disease or disorder and its complications. An amount adequate to accomplish this is de- fined as a "therapeutically effective amount".
Compounds of Formula (I) according to the present invention may be used in pharmaceutical compositions and method for treatment of disorders, diseases in a subject, or conditions associated with the dysfunction of iGluR receptors, e.g. the AMPA receptors, KA receptors and NMDA receptors, and their corresponding subunits, GluAl-4, GluKl-5 and GluNl, GluN2A-D, and GluN3A-C. Thus, targeting iGluR or its subtypes such as GluAl-4, GluKl-5, GluNl-3 and its subtypes with antagonists according to the present invention would be helpful in the treatment of disorders and diseases associated with these receptors.
The terms "treatment" and "treating" as used herein refer to the management and care of a patient for the purpose of combating a condition, disease or disorder. The term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound for the purpose of: alleviating or re- lieving symptoms or complications; delaying the progression of the condition, disease or disorder; curing or eliminating the condition, disease or disorder; and/or preventing the condition, disease or disorder, wherein "preventing" or "prevention" is to be understood to refer to the management and care of a patient for the purpose of hindering the development of the condition, dis- ease or disorder, and includes the administration of the active compounds to prevent or reduce the risk of the onset of symptoms or complications.
The term "subject" refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment. Treatment of animals, such as mice, rats, dogs, cats, cows, sheep and pigs, is, however, also within the scope of the present invention.
In a certain embodiment, the present invention relates to compounds of Formula (I) or pharmaceutical compositions thereof, or methods for treatment of diseases or conditions binding one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits to obtain a beneficial therapeutic effect.. Optionally, compounds of Formula (I) or pharmaceutical compositions thereof are used for treatment of diseases or conditions binding one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits to obtain a beneficial therapeu- tic effect..
In yet a certain embodiment, the present invention relates to compounds of Formula (I) or pharmaceutical compositions thereof, or methods for treatment of disorders of the central nervous system, neuro-physiological processes such as memory, cognition; as well as neuronal plasticity and de- velopment, psychiatric diseases or neurological disorders such as depression, anxiety, addiction, pain, migraine, and schizophrenia, and neurodegenerative diseases; such as Alzheimer, Huntington disease, amyotrophic lateral sclerosis (ALS), cerebral stroke, and epilepsy; and diseases including aching, ADHD, Autism, Diabetes, Huntington's disease, ischemia, multiple sclerosis, Parkinson's disease (Parkinsonism), Rasmussen's encephalitis, seizures, AIDS dementia complex, amyotrophic lateral sclerosis, combined systems disease (vitamin B12 deficiency), drug addiction, drug tolerance, drug dependency, glaucoma, hepatic encephalopathy, hydroxybutyric aminoaciduria, hyperho- mocysteinemia and homocysteinuria, hyperprolinemia, lead encephalopathy, leber's disease, MELAS syndrome, MERRF, mitochondrial abnormalities (and other inherited or acquired biochemical disorders), neuropathic pain syndromes (e.g. causalgia or painful peripheral neuropathies), nonketotic hyperglycinemia, olivopontocerebellar atrophy, essential tremor, Rett syndrome, sulfite oxidase deficiency, Wernicke's encephalopathy or cancer.
According to the present invention, the antagonist of compound of
Formula (I) is administered to subjects in need of treatment in pharmaceutically effective doses. A therapeutically effective amount of a compound according to the present invention is an amount sufficient to cure, prevent, reduce the risk of, alleviate or partially arrest the clinical manifestations of a given disease or its complications. The amount that is effective for a particular therapeutic purpose will depend on the severity and the sort of the disease as well as on the weight and general state of the subject. The antagonists of the present invention may be administered one or several times per day, such as from 1 to 4 times per day, such as from 1 to 3 times per day, such as from 1 to 2 times per day, wherein administration from 1 to 3 times per day is preferred.
In a preferred embodiment, the present invention relates to antagonist of compound of Formula (I) which is administered in doses of 0.5-1500 mg/day, preferably 0.5-200 mg/day, more preferably 0.5-60 mg/day, even more preferably 0.5-30 mg/day.
In a preferred embodiment, the present invention relates to antagonist of compound of Formula (I) suitable for oral, rectal, nasal, pulmonary, buccal, sublingual, transdermal or parenteral administration.
During any of the processes for preparation of the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Syn- thesis, John Wiley & Sons, 1991, fully incorporated herein by reference. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.
In a preferred embodiment, the present invention relates to compounds of Formula (I), such as
(1) (2S,3R)-2-Carboxy-3-(3-carboxy-4-hydroxyphenyl)pyrrolidin-l-ium- 2,2,2-trifluoroacetate,
(2) (2S,3R)-3-(3-Carboxy-5-propoxyphenyl)pyrrolidine-2-carboxylic acid hydrochloride,
(3) (2S,3R)-3-(3-(Benzyloxy)-5-carboxyphenyl)pyrrolidine-2-carboxylic acid hydrochloride,
(4) (2S,3R)-3-(3-([l,l'-Biphenyl]-3-ylmethoxy)-5- carboxyphenyl)pyrrolidine-2-carboxylic acid, (2S,3R)-2-Carboxy-3-(3- carboxy-5-hydroxyphenyl)pyrrolidin-l-ium 2,2,2-trifluoroacetate,
(5) (2S,3R)-tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(2,2- dimethyl-4H-benzo[d][l,3]dioxin-6-yl)-5-oxopyrrolidine-l-carboxylate,
(6) (2S,3R)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(3- (((tert-butyldimethylsilyl)oxy)-methyl)-5-propoxyphenyl)-5-oxopyrrolidine-l- carboxylate,
(7) (2S,3R)-tert-Butyl 3-(3-(benzyloxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((tert-butyldime- thylsilyl)oxy)methyl)-5-oxopyrrolidine-l-carboxylate,
(8) (2S,3R)-tert-Butyl 3-(3-([l,l'-biphenyl]-3-ylmethoxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((tert- butyldimethylsilyl)oxy)methyl)-5-oxopyrrolidine-l-carboxylate,
(9) (2S,3R)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(2,2- dimethyl-4H-benzo[d][l,3]-dioxin-6-yl)pyrrolidine-l-carboxylate,
(10) (2S,3R)-tert-Butyl- 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(3- (((tert-butyldimethylsilyl)oxy)-methyl)-5-propoxyphenyl)pyrrolidine-l- carboxylate,
(11) (2S,3R)-tert-Butyl-3-(3-(benzyloxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((tert- butyldimethylsilyl)oxy)methyl)pyrrolidine-l-carboxylate
(12) (2S,3R)-tert-Butyl- 3-(3-([l,l'-biphenyl]-3-ylmethoxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)-phenyl)-2-(((tert- butyldimethylsilyl)oxy)methyl)pyrrolidine-l-carboxylate,
(13) (2S,3R)-tert-Butyl 3-(2,2-dimethyl-4H-benzo[d][l,3]dioxin-6-yl)-2- (hydroxymethyl)-pyrrolidine-l-carboxylate,
(14) (2S,3R)-tert-Butyl 2-(hydroxymethyl)-3-(3-(hydroxymethyl)-5- propoxyphenyl)pyrrolidine-l-carboxylate,
(15) (2S,3R)-tert-Butyl 3-(3-(benzyloxy)-5-(hydroxymethyl)phenyl)-2- (hydroxymethyl)pyrrolidine- l-carboxylate,
(16) (2S,3R)-tert-Butyl 3-(3-([l,l'-biphenyl]-3-ylmethoxy)-5- (hydroxymethyl)phenyl)-2-(hydroxymethyl)-pyrrolidine-l-carboxylate,
(17) (2S,3R)-l-(tert-Butoxycarbonyl)-3-(2,2-dimethyl-4-oxo-4H- benzo[d][l,3]dioxin-6-yl)pyrrolidine-2-carboxylic acid,
(18) (2S,3R)-l-(tert-butoxycarbonyl)-3-(2,2-dimethyl-4H- benzo[d][l,3]dioxin-6-yl)pyrrolidine-2-carboxylic acid,
(19) (2S,3R)-l-(tert-Butoxycarbonyl)-3-(3-carboxy-5- propoxyphenyl)pyrrolidine-2-carboxylic acid,
(20) (2S,3R)-3-(3-(Benzyloxy)-5-carboxyphenyl)-l-(tert- butoxycarbonyl)pyrrolidine-2-carboxylic acid, and
(21) (2S,3R)-3-(3-([l,l'-Biphenyl]-3-ylmethoxy)-5-carboxyphenyl)- l-(tert-butoxycarbonyl)-pyrrolidine-2-carboxylic acid Examples
Results and discussion
Based on ligand conformational analysis and the vast structural information on the GluA2 & GluKl ligand binding domains, (2S,3£)-3-(3- carboxyphenyl)pyrrolidine-2-carboxylic acid (1, CNG-10100) was design as a broad-range iGluR antagonist.49 Succeeding its synthesis, pharmacological characterization of 1 showed low-to-medium range micromolar binding affinity to AMPA, KA and NMDA receptors, and antagonist function in a non- desensitizing functional GluKl assay.49 Thus 1 defined a new lead structure for the discovery of subtype selective iGluR antagonists.
From our modeling studies introduction of substituents on the phenyl ring was attractive. Introduction of a hydroxyl group in the 4'-position (compound 2a, Figure 2) would immediately enhance the ligand's ability to engage in favorable hydrogen bonding interactions with amino acid residues AAA, AAA (GluKl numbering) as well as key water molecules trapped in the receptor. However, this process could also be net-disfavored, which would then induce subtype or group selectivity rather than enhanced overall binding affinity.
Figure 2. Chemical structures of rationally design analogs 2a- e
Figure imgf000020_0001
2a (2b) 2c (2d) (2e)
(CNG-10104) (CNG-10107)
Chemistry
The synthesis commenced with double protection of commercially available optically pure (S)-glutaminol (3), to give 4 in high yield (Scheme 1). Subsequently, introduction of the double bond was carried out under standard selenylation conditions, to give enone 5. For target compound 2a, the synthesis of suitably protected phenylbromide 6a was carried out starting from commercially available phenolic alcohol 12 (Scheme 2). With enone 5 and phenyl bromide 6a in hand, the copper catalyzed conjugate addition reaction was carried out as previously described to give the desired product 7a as a single diastereomer (70% yield, Scheme 1). Reduction of the endo- cyclic carbonyl group by borane gave pyrrolidinone 8a in low-to-acceptable yield (32-46%), with the byproduct being reductive opening of isopropylidene group (determined by LC-MS). At this stage a shortcut was attempted : Full deprotection of 8a in p-TsOH/MeOH at rt gave the expected phenolic diol 13 in high yield (Scheme 3). However, oxidation to the corresponding diacid 14 was unsuccessful although several conditions were tried : RuCl3-NaI04, TEM- PO-NaOCI-NaCI02, Pd-C/O^NaBH^ PDC in DMF, and KMn04 in acetone (Scheme 3). Returning to the planned pathway (Scheme 1), ruthenium catalyzed oxidation of 8a gave the desired acid-lactone 10a together with acid 11, in a 1 : 2 ratio. The two products were easily separated using flash chromatography after which acid-lactone 10a was fully deprotected in TFA/H20 to give target compound 2a.
Scheme 1. Synthesis of 2a (CNG-10104)
Figure imgf000021_0001
9a 10a 11 2a
(CNG-10104)
Reagents and conditions, a) TBSCI, imidazole, DCM, rt, 24h, aq workup, then BOC20, DMAP, ACN, rt, 16h (two steps, 91%); b) LHMDS, PhSeCI, THF, -78 °C, then 30% H202, EtOAc, 0 °C to rt (two steps, 64%); c) 6a, tert- BuLi, CuCN, Et20, -78 to -42 °C (70%); d) BH3, THF, reflux, 31/2h, then H20, NaOH, H202, rt, 31/2h (46%); e) TBAF, THF, rt, 2h (87%); f) NaI04, RuCI3, 0 °C, l1/2h (10a: 22%, 11 : 40%); g) 10a only, TFA: H20 (1 : 1), rt, 16h (59%).
cheme thesis of bromide 6a
Figure imgf000021_0002
12 6a
Reagents and conditions, a) 2,2-dimethoxypropane, ZnCI2, acetone, 40
Figure imgf000021_0003
Scheme 3. Attempted oxidation of phenolic diol 12
Figure imgf000021_0004
13
Reagents and conditions, a) p-TsOH, MeOH, rt, 5h (71%).
The synthesis of target structures 2b-e (Scheme 4) followed the strategy described for 2a with only few changes Firstly, suitably protected phenyl bromides 6b-d were prepared starting from commercially available acid 15 in two (three) steps (Scheme 5). Due to the bulkiness of the phenyl bromides 6b-d, conjugate addition to enone 5 was carried out by use of a mixed-cyano Gilman cuprate, as previously described by us to be advantageous for this specific enone.50 Reduction of the endo-carbonyl group gave pyrrolidinones 8a-d in acceptable yields (51-60%), and removal of the TBS groups provided the free diols 9b-d in high yield. Subsequent ruthenium catalyzed oxidation to give diacids lOb-d was carried out at 0 °C to prevent over oxidation of the electron rich phenyl ring giving L-trans-2,3-dicarboxy-pyrrolidinone 17 (Scheme 4), which was otherwise identified as the major byproduct. Finally removal of the BOC-group under acidic water-free conditions gave target compounds 2b-d, as the HCI salts. Analog 2e which comprised a free 5'- phenol was obtained by hydrogenation over Pd/C of 2c to give the desired product in 63% after purification on HPLC.
Figure imgf000022_0001
Figure imgf000022_0002
Reagents and conditions, a) 6b-d, tert-BuLi, CuCN, ThLi, Et20, -78 to -
42 °C (42-62%); b) BH3, THF, reflux, 20h, then H20, NaOH, H202, rt, lh (51- 60%); c) TBAF, THF, rt, (92-94%); d) NaI04, RuCI3, 0 °C, 2h (66-83%); e) 2 M HCI in Et20, AcOH, rt, 18h (87-99%). f) 2c only, H2 (g), Pd/C (10%), AcOH, rt, 6d (44%).
Scheme 5. Synthesis of substituted phenylbromides 6b-d
Figure imgf000022_0003
15 16 6b: R = n-Pr
6c: R = Bn
6d: R = 3-Ph-Bn Reagents and conditions, a) BH3, THF, 0°C to rt, aq work-up, then TBSCI, imidazole, DMF, 0°C to rt (71%); b) For 6b: RBr, K2C03, rt in DMF (90%); for 6c,d: RBr, K2C03, reflux in acetone (92-95%). The synthesis of analogs 2f-n (Figure 3) was carried out following the general strategy described for 2a-e (Scheme 1-5). Starting from 5 and appropriately substituted aryl analog 6 analogs 2f-n were prepared in moderate yields.
Figure imgf000023_0001
Pharmacological characterization
The five new analogs 2a-e were characterized pharmacologically in radio ligand binding assays at native iGluRs (rat synaptosomes) and cloned homomeric subtypes, GluKl-3 (Table 1). 4'-Hydroxy analog 2a displayed a general higher binding affinity for the iGluRs in native tissue, and reduced iGluR class selectivity, as compared to lead structure 1. In details, a 25- and 15-fold higher affinity for AMPA and KA receptors was observed, while a 6- fold higher affinity at NMDA receptors could be demonstrated. But most interestingly, 2a displayed a 10-fold increase in binding affinity for homomeric GluK3 (0.87 μΜ), while affinity for homomeric GluKl and -GluK2 was essentially unchanged. This 5-fold preference for GluK3 over GluKl is indeed a unique binding affinity profile for a KA antagonist and encourages future SAR investigations of the 4'-position for the discovery of a fully selective GluK3 antagonist. Furthermore, as the binding affinity profile of the 5'-hydroxy ana- log 2e is essentially unchanged compared to 1, strengthen the interest in the 4'-position as a point of modification to obtain selectivity for the GuK3 subtype.
The pharmacological profiles of 5'-ethers 2b-d at native iGluRs show that affinity for AMPA receptors is increased (2 to 10 fold) with increasing steric bulk of the 5'-substituent, whereas for KA receptors only a 2-3 fold increase in observed. At the NMDA receptors the affinity was unaffected. At cloned homomeric GluKl-3 the three analogs 2b-d displayed only modest changes in affinity as compared to 1 (Table 1) : A general 4-fold increase was observed for 2b, whereas 2c only displayed a 4-fold higher affinity for GluK2,3. Analog 2d displayed slightly lower affinity for GluKl, mid micromo- lar affinity for GluK2, and similar affinity for GluK3. Together these data confirms that the 5'-sidechain is directed into the spacious compartment in the receptors located between the Dl and D2 domain. However, by targeting differences in amino acid residues in this area of the receptors, it may be possi- ble to achieve class or subtype selectivity.
The pharmacological characterization of 2f and 2g which comprise a halogen in the 4-position, showed a surprising shift in profile to selectivity for the NMDA receptors over both AMPA and KA receptors (Table 1). The pharmacological profile of analog 2j proved that a heterocycle can also be ac- commodated by the iGlu receptors (Table 1).
Table 1. Chemical structures and pharmacological characterization of 1 and 2a-e at native iGluR as well as cloned homomeric GluKl- 3 subtypes. All values in μΜ AM PA KA NMDA GluKl GluK2 GluK3 GluKl:K3
IC50 IC50 K| K| K| ratio la
51 22 6.0 4.3 > 100 8.1 0.5
2.0 1.4 1.0 4.8 10- 0.87
2a 5.5
[5.71±0.03] [5.87±0.07] [6.02±0.11] ± 1.5 100 ±0.09
25 7.2 5.0 1.7 56 2.7
2b 0.6
[20;30] [6.4;8.0] [3.9;6.4] ±0.05 ± 1.4 ± 0.3
2c 6.3 6.7 4.1 3.7 32 2.1
1.8
[5.20±0.04] [5.18±0.06] [5.41±0.10] ± 0.8 ± 2.5 ± 0.4
4.9 12 4.0 8.7 68 8.6
2d 1.0
[5.34±0.09] [4.92±0.03] [5.41±0.08] ± 1.2 ± 7.6 ± 0.6
54 34 5.4 8.3 8.5
2e > 100 1.0
[4.28±0.05] [4.52±0.11] [5.27±0.04] ± 0.2 + 1.2
59 4.6
2f > 100
[4.23 ± 0.05] [5.34 ± 0.04]
0.63 154 131
2g >100 >100 > 100
[6.22 ±0.10] ± 13 ± 13
40 20 3.8
2j
[4.40±0.05] [4.71±0.05] [5.42±0.01]
17 -10 1-10 1.2
b
a Values taken from reference 49. Agonist activity was shown in functional assays This section provides specific examples of selected targets molecules. However, it is to be understood that these examples outline the synthetic routes for other target molecules not specifically disclosed.
All reagents were obtained from commercial suppliers and used without further purification. Dry solvents were obtained differently. THF was distilled over sodium/benzophenone. Et20 was dried over neatly cut sodium . All solvents were tested for water content using a Carl Fisher apparatus. Water - or air sensitive reactions were conducted in flame dried glassware under n i- trogen with syringe-septum cap technique. Purification by DCVC (dry column vacuum chromatography) was performed with silica gel size 25-40μηι (Merck, Silica gel 60). For TLC was used Merck TLC Silica gel F254 with appropriate spray reagents: KMn04 or Molybdenum blue. 1H NMR and 13C NMR spectra were obtained on a Varian Mercury Plus (300 MHz) and a Varian Gemini 2000 instrument (75 MHz), respectively. HPLC was done using Agilent Prep HPLC systems with Agilent 1100 series pump, Agilent 1200 series diode array, mu ltiple wavelength detector (G1365B), and Agilent PrepHT High Performance Preparative Cartridge Column (Zorbax, 300 SB-C18 Prep HT, 21.2 x 250 mm, 7 μηι). Preparative HPLC was performed using Spectraseries UV100 with a JASCO 880-PU HPLC pump and an XTerra®Prep MS C18 ,( ΙΟμηι, 10X300 mm) column. LC-MS was performed using an Agilent 1200 series solvent delivery system equipped with an autoinjector coupled to an Agilent 6400 series triple quadrupole mass spectrometer equipped with an electrospray ionization source. Gradients of 10% aqueous acetonitrile + 0.05% formic acid (buffer A) and 90% aqueous acetonitrile + 0.046% formic acid (buffer B) were employed or an Agilent 1200 system using a C18 reverse phase column (Zorbax 300 SB-C18, 21.1 mm - 250 mm) with a linear gradient of the binary solvent system of H20/CH3CN/TFA (A: 100/0/0.1 and B: 5/95/0.1) with a flow rate of 20 mL/min. Optical rotation was measured using a Perkin-Elmer 241 spec- trometer, with Na lamp at 589 nm . Melting points were measured using an automated melting point apparatus, MPA100 OptiMelt (SRS) and are uncorrected. Compounds were dry either under high vacuum or freeze dried using a Holm & Halby, Heto LyoPro 6000 freeze drier.
(2S,3 ?)-2-Carboxy-3-(3-carboxy-4-hydroxyphenyl)pyrrolidin- l-ium-2,2,2-trifluoroacetate (2a). The carboxylic acid compound (44 mg, 0.112 mmol, 1.00 equiv) was dissolved in TFA: H20 (1 : 1) (4 mL) and stirred overnight at room temperature. The clear and colorless reaction mixture was evaporated in vacuo and co-evaporated with MeCN (5 x 10 mL) and then lyophilized overnight to re- move the water. The brownish viscous oil (34 mg, 83%) was trituated with a few mL of ice-cold Et20 by which the product precipitated. The trituation was repeated once. The product was isolated upon drying in high vacuum to an off-white solid (24 mg, 59%). MS (m/z) calcd. for Ci2Hi4N05 [M+H]+ 252.1, found 252.1. ^ NMR (400 MHz, DMSO-d6) δ 7.77 (s, 1H), 7.36 (d, J = 8.1 Hz, 1H), 6.81 (d, J = 8.6 Hz, 1H), 4.10-4.04 (m, 2H), 3.42 (t, J = 8.4 Hz, 2H), 3.26-3.18 (m, 1H), 3.15-3.06 (m, 2H), 2.36-2.27 (m, 1H), 2.13-2.05 (m, 1H), 2.00- 1.91 (m, 1H); 13C NMR (100 MHz, DMSO-d6) δ 171.6, 171.5, 170.0, 169.7, 160.9, 133.3, 129.4, 129.0, 116.7, 64.6, 60.0, 46.8, 45.5, 45.3, 43.1, 33.5, 26.8. [a]25 D -65.7° (c = 0.067, 10% DMSO in H20). Mp 172- 178 °C (decomp.).
(2S,3/?)-3-(3-Carboxy-5-propoxyphenyl)pyrrolidine-2- carboxylic acid hydrochloride (2b).
The compound 10b (180 mg, 0.46 mmol, 1.00 equiv) was dissolved in AcOH (2.5 mL) and added 2M HCI in Et20 (2.5 mL, 5 mmol, 10.9 equiv). The mixture was left to stir under N2 at r.t. for 18 h. The mixture was suspended in H20 (10 mL), solidified by cooling on a dry ice/acetone bath and freeze dried to yield 149 mg (99%) of 2b as a slightly off white, crisp solid. H NMR (300 MHz, DMSO) : δ 0.98 (3H, t, J = 7 Hz), 1.73 (2H, m), 2.01 (1H, m), 2.36 (1H, m), 3.23 (1H, m), 3.41 (1H, m), 3.54 (1H, m), 3.96 (2H, m), 4.17 (1H, m), 7.28 (1H, m), 7.30 (1H, m), 7.51 (1H, m); 13C NMR (75 MHz, DMSO) : δ 10.5, 22.1, 33.6, 45.2, 47.3, 64.3, 69.2, 112.9, 118.6, 120.8, 132.2, 142.5, 158.7, 166.8, 169.6; LCMS : m/z [M + H] + : calc: 293.1, found : 294.2; HPLC: purity254 = 99.4 % OR: [a]35 D: +42.09° (c = 0.43, abs EtOH : H20 (3 : 1)).
(2S,3/?)-3-(3-(Benzyloxy)-5-carboxyphenyl)pyrrolidine-2- carboxylic acid hydrochloride (2c).
The compound 10c (302 mg, 0.68 mmol, 1.00 equiv) was dissolved in AcOH (4 mL) and added 2M HCI in Et20 (4 mL, 8 mmol, 11.8 equiv). The mixture was left to stir under N2 at RT for 18 h. The mixture was suspended in H20 (20 mL), solidified by cooling on a dry ice/acetone bath and freeze dried to yield 228 mg (88%) of 2c as a slightly off white, crisp solid. H NMR (300 MHz, DMSO) : δ 2.01 (1H, m), 2.37 (1H, m), 3.05 - 3.48 (2H, m), 3.56 (1H, q, J = 9 Hz), 4.04 (0.1H, d, J = 11 Hz), 4.19 (0.9H, d, J = 8 Hz), 5.15 (2H, s), 7.28-7.49 (7H, m), 7.56 (1H, m), 9.53 (1H, vbs); 13C NMR (75 MHz, DMSO) : δ 33.6, 45.3, 47.2, 64.2, 69.5, 113.3, 119.1, 121.2, 127.7, 127.8, 128.3, 132.2, 136.6, 142.5, 158.4, 166.8, 169.5; LCMS: m/z [M + H] + : calc: 341.1, found : 342.2; HPLC: purity254 = 96.6 %; OR: [a]35 D: -11.46° (c = 0.41, abs EtOH : DMSO (3: 1)).
(2S,3 ?)-3-(3-([l,l'-Biphenyl]-3-ylmethoxy)-5- carboxyphenyl)pyrrolidine-2-carboxylic acid hydrochloride (2d).
The compound lOd (247 mg, 0.48 mmol, 1.0 equiv) was dissolved in AcOH (4 mL) and added 2M HCI in Et20 (4 mL, 8 mmol, 16.7 equiv). The mixture was left to stir under N2 at r.t. for 18 h. The mixture was suspended in H20 (20 mL), solidified by cooling on a dry ice/acetone bath and freeze dried to yield 188 mg (87%) of 2d as a slightly off white, crisp solid. H NMR (300 MHz, DMSO): δ 1.98 (1H, m), 2.35 (1H, m), 3.22 (1H, m), 3.38 (1H, m), 3.54 (1H, q, J = 8 Hz), 4.15 (1H, d, J = 9 Hz), 5.20 (2H, s), 7.30 - 7.70 (11H, m), 7.74 (1H, s); 13C NMR (75 MHz, DMSO) : δ 33.5, 45.2, 47.1, 64.7, 69.4, 113.0, 119.3, 121.2, 126.1, 126.2, 126.6, 126.8, 127.5, 128.9, 129.0, 132.3, 137.3, 139.7, 140.2, 142.8, 158.3, 166.8, 169.5, 201.9; LCMS: m/z [M + H]+ : calc: 417.2, found : 418.2; HPLC: purity254 = 99.9 %; OR: [a]35 D: + 37.50 (c = 0.40°, abs EtOH : DMSO (3 : 1)).
(2S,3/?)-2-Carboxy-3-(3-carboxy-5-hydroxyphenyl)pyrrolidin- 1-ium 2,2,2-trifluoroacetate (2e).
The O-benzyl analog 2c (91 mg, 0.24 mmol, 1.00 equiv) was dissolved in AcOH (7 mL) and an atmosphere of N2 was applied. Pd/C (10 mg) was added followed by purging with H2(g). The reaction was stirred for 6 days after which HPLC did not show further change in reaction outcome. The mixture was diluted with MeCN (50 mL), filtered through a syringe filter and eva- porated in vacuo. The residue was co-evaporated with MeCN (3 x 50 mL) and the crude product was dried in high vacuum overnight affording a pale brown glass (57 mg, 83%). The product was further purified by preparative HPLC to yield a clear, colorless oil (30 mg, 44%). ^ NMR (300 MHz, NaOD) : δ 2.10 - 2.24 (1H, m), 2.41 - 2.53 (1H, m), 3.40 - 3.51 (1H, m), 3.53 - 3.64 (2H, m), 4.25 (1H, d, J = 9 Hz), 7.04 (1H, t, J = 2 Hz), 7.29 (1H, dd, J = 1, 2 Hz), 7.47 (1H, t, J = 1 Hz); 13C NMR (75 MHz, NaOD) : δ 33.6, 46.4, 48.5, 66.0, 116.1, 120.4, 121.0, 132.2, 141.5, 156.6, 170.2, 172.1; LCMS : m/z [M + H]+ : calc: 252.1, found : 252.1; HPLC: purity254 = 100 %; OR: [a]35 D: + 52.64° (c = 0.35, 2M NaOH).
(2S,3R)-2-carboxy-3-(3-carboxy-4-fluorophenyl)pyrrolidin-l- ium chloride (2f).
Compound (2g) was prepared following general procedure described in Scheme 1-4. The oily residue was dissolved in HCI 1M (10 mL) and the solvent was evaporated (3 times) to afford the corresponding HCI salt (42 mg). mp 196.8→dec. ^ NMR (D20 + dioxane, 400 MHz) δ 7.97 (dd, J = 6.9, 2.6 Hz, 1H), 7.69 (ddd, J = 8.6, 4.6, 2.5 Hz, 1H), 7.31 (dd, J = 11.0, 8.6 Hz, 1H), 4.36 (d, J = 9.7 Hz, 1H), 3.77 - 3.66 (m, 2H), 3.57 (ddd, J = 11.8, 10.2, 6.9 Hz, 1H), 2.59 (dtd, J = 13.9, 7.0, 3.3 Hz, 1H), 2.29 (dtd, J = 13.4, 10.4, 8.2 Hz, 1H). 13C NMR (D20+dioxane) δ 172.0, 168.4, 162.5, 160.8, 135.34, 135.31, 135.2, 135.1, 131.5, 119.2, 119.1, 118.2, 118.1, 67.2, 65.9, 47.8, 46.3, 33.5. MS (m/z) calcd. for Ci2Hi3FN04 [M + H]+ 254.1, found 254.1.
(2S,3R)-3-(3-carboxy-4-chlorophenyl)pyrrolidine-2-carboxylic acid (2g).
Compound (2g) was prepared following general procedure described in Scheme 1-4. The final crude product was recrystallized from H20/acetone to afford IPS-068 (89 mg, 0.291 mmol, 56% yield) as an off-white solid, mp 239.4-241.3 °C. ^ NMR (D20 + dioxane) δ 7.85 (d, J = 1.8 Hz, 1H), 7.68 - 7.49 (m, 2H), 4.37 (d, J = 9.7 Hz, 1H), 3.71 (ddd, J = 11.8, 9.2, 5.1 Hz, 2H), 3.62 - 3.52 (m, 1H), 2.60 (dtd, J = 13.9, 7.1, 3.4 Hz, 1H), 2.35 - 2.23 (m, 1H). 13C NMR (D20+dioxane) δ 171.9, 170.7, 138.3, 132.6, 131.9, 131.8, 131.7, 130.3, 65.7, 47.8, 46.2, 33.4. MS (m/z) calcd. for Ci2Hi3CIN04 [M + H]+ 270.0, found 270.0.
(2S,3R)-3-(3-carboxy-4-methylphenyl)pyrrolidine-2-carboxylic acid hydrochloride (2h)
Compound (2h) was prepared following general procedure described in Scheme 1-4. The crude reaction mixture was purified by preparative HPLC (0- 20 % B in A) to afford the title compound (21 mg, 22 % over two steps) after 3 evaporations from 1M HCI to isolate as a white HCI salt. H NMR (400 MHz, D20) δ: 7.78 (d, J = 2.0 Hz, 1H), 7.45 (dd, J = 7.9, 2.1 Hz, 1H), 7.29 (d, J = 7.9 Hz, 1H), 4.39 (d, J = 9.8 Hz, 1H), 3.70 - 3.61 (m, 2H), 3.51 (ddd, J = 11.7, 10.2, 6.9 Hz, 1H), 2.50 (dtd, J = 14.0, 7.1, 3.2 Hz, 1H), 2.44 (s, 3H), 2.26 - 2.17 (m, 1H) 13C NMR (100 MHz, D20) δ: 171.54, 170.63, 139.15, 135.72, 132.31, 131.28, 130.16, 129.04, 64.56, 47.25, 45.81, 32.82, 20.04. LC-MS (m/z) calcd for Ci3Hi6N04 [M + H+], 250.3; found, 250.1.
5-((2S,3R)-2-carboxypyrrolidin-3-yl)-2-oxo-l,2- dihydropyridine-3-carboxylic acid hydrochloride (2i)
Compound (2i) was prepared following general procedure described in Scheme 1-4. The crude reaction mixture was purified by preparative HPLC (0^20 % B in A) to afford the title compound (7.10 mg, 28 % over two steps) after 3 evaporations from 1M HCI to isolate as a white HCI salt. H NMR (400 MHz, D20) δ: 8.63 (d, J = 2.6 Hz, 1H), 8.00 (d, J = 2.6 Hz, 1H), 4.38 (d, J = 10.0 Hz, 1H), 3.76 - 3.65 (m, 2H), 3.55 (td, J = 11.1, 6.8 Hz, 1H), 2.59 (dtd, J = 13.8, 7.1, 3.0 Hz, 1H), 2.28 (dtd, J = 13.7, 10.6, 8.2 Hz, 1H). 13C NMR (100 MHz, D20) δ: 170.44, 167.93, 163.99, 146.40, 140.48, 119.94, 116.61, 63.97, 45.48, 43.83, 31.76. LC-MS (m/z) calcd for CiiHi3N205 [M + H+], 253.2; found, 253.1.
(2S,3R)-3-(3-carboxy-5-(pyridin-4-yl)phenyl)pyrrolidine-2- carboxylic acid hydrochloride (2j)
Compound (2j) was prepared following general procedure described in Scheme 1-4. The crude reaction mixture was purified by preparative HPLC (0^20 % B in A) to afford the title compound (18.0 mg, 9 % over two steps) after 3 evaporations from 1M HCI to isolate as a white HCI salt. H NMR (400 MHz, D20) δ: 8.84 (d, J = 7.0 Hz, 2H), 8.36 (t, J = 1.7 Hz, 1H), 8.34 (d, J = 7.0 Hz, 2H), 8.24 (t, J = 1.4 Hz, 1H), 8.18 (t, J = 1.9 Hz, 1H), 4.58 (d, J = 10.1 Hz, 1H), 3.95 - 3.85 (m, 1H), 3.77 (ddd, J = 11.6, 8.2, 3.1 Hz, 1H), 3.61 (td, J = 11.8, 7.0 Hz, 1H), 2.67 (dtd, J = 13.9, 7.0, 3.2 Hz, 1H), 2.45 - 2.29 (m, 1H). 13C NMR (100 MHz, D20) δ: 170.65, 168.79, 156.56, 141.32, 140.63, 135.63, 132.22, 131.88, 131.46, 131.43, 128.54, 124.79, 64.71, 47.53, 45.77, 32.95. ; LC-MS (m/z) calcd for Ci7Hi7N204 [M + H+], 313.3; found, 313.2.
(2S,3R)-3-(3-carboxy-5-(pyridin-3-yl)phenyl)pyrrolidine-2- carboxylic acid hydrochloride (2k) Compound (2k) was prepared following general procedure described in Scheme 1-4. The crude reaction mixture was purified by preparative HPLC (0^20 % B in A) to afford the title compound (18.0 mg, 9 % over two steps) after 3 evaporations from 1M HCI to isolate as a white HCI salt. H NMR (400 MHz, D20) δ: 9.11 (d, J = 2.1 Hz, 1H), 8.89 (dt, J = 8.3, 1.7 Hz, 1H), 8.83 (d, J = 5.8 Hz, 1H), 8.28 (t, J = 1.6 Hz, 1H), 8.21 - 8.17 (m, 2H), 8.04 (t, J = 1.6 Hz, 1H), 4.56 (d, J = 10.0 Hz, 1H), 3.88 (td, J = 10.3, 7.4 Hz, 1H), 3.76 (ddd, J = 11.6, 8.3, 3.2 Hz, 1H), 3.60 (ddd, J = 11.9, 10.2, 6.9 Hz, 1H), 2.66 (dtd, J = 14.0, 7.1, 3.2 Hz, 1H), 2.42 - 2.32 (m, 1H). 13C NMR (100 MHz, D20) δ: 170.68, 169.06, 145.03, 140.52, 140.02, 139.38, 139.05,
134.73, 131.81, 131.74, 129.91, 128.00, 127.60, 64.76, 47.56, 45.78, 32.97. LC-MS (m/z) calcd for Ci7Hi7N204 [M + H+], 313.3; found, 313.2.
(2S,3R)-3-(3-carboxy-5-(pyridin-2-yl)phenyl)pyrrolidine-2- carboxylic acid hydrochloride (21)
Compound (21) was prepared following general procedure described in
Scheme 1-4, (30.6 mg, 48 % over two steps) as a white HCI salt. H NMR (400 MHz, D20) δ: 8.84 (ddd, J = 6.0, 1.6, 0.7 Hz, 1H), 8.70 (td, J = 8.0, 1.6 Hz, 1H), 8.41 (t, J = 1.6 Hz, 1H), 8.35 (dd, J = 8.1, 1.0 Hz, 1H), 8.32 (t, J =
I.6 Hz, 1H), 8.17 (t, J = 1.8 Hz, 1H), 8.09 (ddd, J = 7.4, 5.9, 1.2 Hz, 1H), 4.55 (d, J = 10.0 Hz, 1H), 3.91 (td, J = 10.3, 7.4 Hz, 1H), 3.77 (ddd, J =
I I.6, 8.2, 3.2 Hz, 1H), 3.61 (ddd, J = 11.8, 10.2, 6.9 Hz, 1H), 2.68 (dtd, J = 13.9, 7.0, 3.2 Hz, 1H), 2.44 - 2.33 (m, 1H). 13C NMR (100 MHz, D20) δ: 170.68, 169.06, 145.03, 140.52, 140.02, 139.38, 139.05, 134.73, 131.81,
131.74, 129.91, 128.00, 127.60, 64.76, 47.56, 45.78, 32.97. LC-MS (m/z) calcd for Ci7Hi7N204 [M + H+], 313.3; found, 313.2.
(2S,3R)-3-(3-carboxy-5-(pyrimidin-5-yl)phenyl)pyrrolidine-2- carboxylic acid hydrochloride (2m)
Compound (2m) was prepared following general procedure described in Scheme 1-4. The crude reaction mixture was purified by preparative HPLC (0^20 % B in A) to afford the title compound (16.2 mg, 6 % over two steps) after 3 evaporations from 1M HCI to isolate as a white HCI salt. H NMR (400 MHz, D20) <5: 9.32 (s, 1H), 9.22 (d, J = 1.4 Hz, 2H), 8.00 - 7.98 (m, 2H), 7.89 (t, J = 1.7 Hz, 1H), 4.47 (d, J = 9.9 Hz, 1H), 3.76 (td, J = 10.3, 7.5 Hz, 1H), 3.66 (ddd, J = 11.7, 8.2, 3.2 Hz, 1H), 3.50 (ddd, J = 12.0, 10.3, 6.8 Hz, 1H), 2.54 (dtd, J = 13.9, 7.1, 3.1 Hz, 1H), 2.23 (dtd, J = 13.5, 10.4, 8.2 Hz, 1H). 13C NMR (100 MHz, D20) δ: 170.18, 168.52, 154.94, 152.31, 140.47, 134.04, 132.28, 131.68, 131.28, 130.10, 127.58, 64.35, 47.23, 45.83, 32.86. LC-MS (m/z) calcd for Ci6Hi6N304 [M + H+], 314.3; found, 314.1.
(2S,3R)-3-(3-carboxy-5-(2-oxo-l,2-dihydropyrimidin-5- yl)phenyl)pyrrolidine-2-carboxylic acid hydrochloride (2n)
Compound (2n) was prepared following general procedure described in Scheme 1-4. The crude reaction mixture was purified by preparative HPLC (0- 20 % B in A) followed by recrystallization from H20/acetone to afford the title compound (18 mg, 10 % over two steps) after 3 evaporations from 1M HCI to isolate as an off-white HCI salt. ^ NMR (400 MHz, D20) δ: 8.91 (s, 2H), 8.18 (t, J = 1.7 Hz, 1H), 8.14 (t, J = 1.5 Hz, 1H), 7.87 (t, J = 1.6 Hz, 1H), 4.41 (d, J = 9.7 Hz, 1H), 3.82 - 3.77 (m, 1H), 3.74 (ddd, J = 11.6, 8.1, 3.2 Hz, 1H), 3.59 (ddd, J = 12.3, 10.3, 6.8 Hz, 1H), 2.64 (dtd, J = 13.8, 7.0, 3.1 Hz, 1H), 2.36 (dtd, J = 13.5, 10.3, 8.1 Hz, 1H).13C NMR (100 MHz, D20) δ: 171.59, 169.46, 164.45, 156.45, 153.91, 140.59, 132.85, 131.65, 130.54, 128.42, 126.65, 65.51, 47.92, 45.67, 32.98. LC-MS (m/z) calcd for Ci6Hi6N305 [M + H+], 330.3; found, 330.2.
(S)-tert-Butyl-2-(((tert-butyldimethylsilyl)oxy)methyl)-5- oxopyrrolidine-l-carboxylate (4).
(a) To a solution of commercially available (S)-pyroglutaminol (3)
(1.00 g, 8.69 mmol, 1.0 equiv) in DCM (10 mL) was added imidazole (1.48 g, 21.7 mmol, 2.5 equiv) and TBDMS-CI (1.56 g, 10.4 mmol, 1.2 equiv). The mixture was stirred at r.t. for 24 hours. The solution was diluted with Et20 (100 mL) and the organic phase was washed with H20 (2 x 100 mL) and brine (2 x 100 mL) respectively. The organic phase was dried over MgS04, filtered and concentrated in vacuo to yield a yellow oil (1.90 g). (b) The oil (1.90 g, 8.2 mmol, 10.0 equiv) was dissolved in MeCN (70 mL), cooled to 0° C and added DMAP (0.11 g, 0.83 mmol, 1.0 equiv) and Boc20 (3.60 g, 16.5 mmol, 2.0 equiv). The reaction mixture was left to stir at r.t. overnight. The organic phase was washed with brine (3 x 100 mL) and dried over MgS04. The solution was filtered and concentrated in vacuo to yield a dark, red oil, which was purified using flash chromatography (EtOAc : heptane (1 : 9)) affording 4 as a thick, yellow oil (2.61 g, 91%). ^ NMR (300 mHz, CDCI3) : δ 0.04 (3H, s), 0.05 (3H, s), 0.88 (9H, s), 1.54 (9H, s), 1.95 - 2.20 (2H, m), 2.37 (1H, ddd, J = 18, 9, 2 Hz), 2.71 (1H, dt, J = 18, 11 Hz), 3.68 (1H, dd, J = 10, 2 Hz), 3.91 (1H, dd, J = 11, 4 Hz), 4.16 (1H, m). 13C NMR (75 MHz, CDCI3) : δ -5.4, -5.3, 18.3, 21.3, 26.0, 28.2, 32.5, 59.0, 64.4, 82.7,
150.0, 174.9 OR: [a]22 D: -71.57° (c = 0.55, EtOAc); TLC: Rf: 0.13 (EtOAc: heptane (1 :9)).
(S)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-5-oxo-
2,5-dihydro-lH-pyrrole-l-carboxylate (5).
(a) Pyrrolidone 4 (7.00 g, 21.2 mmol, 1.00 equiv) was dissolved in dry THF (12.5 mL), cooled to -78 °C and dropwise added 1M LiHMDS (48.0 mL, 48.0 mmol, 2.26 equiv) over the course of one hour. The mixture was left to stir at -78 °C for lh after which a solution of PhSeCI (5.50 g, 28.7 mmol, 1.35 equiv) in dry THF (8 mL) was added dropwise over the course of 30 min. The mixture was left to stir at -78 °C for 45 min, before quenched with sat. NH4CI (10 mL) and stirred for 10 min. The mixture was transferred to a separation funnel containing sat. NaHC03 (100 mL) and the flask flushed with Et20 (2 x 50 mL), which was also transferred to the separation funnel. The organic phase was isolated and filtered through a plug of celite to remove undissolvable material. The aqueous phase was reextracted with 2 x 75 mL Et20 and the combined organic phases was washed with brine (75 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (7 cm, 75 mL fractions, 0-20% EtOAc in heptanes) to yield an orange solid (7.52 g). (b) The selenyl diastereomers were dissolved in EtOAc (60 mL), cooled to 0 °C and added 30 % H202 (15 mL) over the course of 10 min. The mixture was left at 0 °C for 15 min before stirred at r.t. for one hour. The mixture was poured in to a separation funnel containing sat. NaHC03 (100 mL). The phases were separated and the aqueous phase extracted with EtOAc (2 x 75 mL). The combined organic phases was washed with brine (75 mL), dried over MgS04, concentrated in vacuo and purified using DCVC (diameter 7 cm, 75 mL fractions, 0-30% EtOAc in heptanes) to yield enone 5 as a white solid (4.42 g, 64%). ^ NMR (300 mHz, CDCI3) : δ 0.01 (3H, s), 0.05 (1.5H, s), 0.06 (1.5H, s), 0.88 (9H,s), 1.57 (9H, s), 3.72 (1H, dd, J = 7, 10 Hz), 4.16 (1H, dd, J = 4, 10 Hz), 4.60 (1H, m), 6.13 (1H, dd, J = 1.5, 6 Hz), 7.26 (1H, dd, J = 2, 6 Hz); 13C NMR (75 MHz, CDCI3) : δ -5.5, -5.4, 18.1, 25.7, 28.2, 62.4, 63.6, 82.9, 127.0, 149.4, 149.7, 169.4; LCMS: m/z [M + H] + : calc:
228.1, found : 228.0; TLC: Rf : 0.25 (EtOAc: heptanes (1 :4)); OR: [a]31 D: - 173.35° (c = 0.77, abs EtOH). 6-Bromo-2,2-dimethyl-4H-benzo[d][l,3]dioxane (6a).
A round-bottomed flask was charged with 4-bromo-2- (hydroxymethyl)phenol (5.46 g, 26.88 mmol, 1.00 equiv) and anhydrous ZnCI2 (9.89 g, 72.58 mmol, 2.70 equiv) and an atmosphere of nitrogen was applied. Dry acetone (40 mL) was added via a syringe followed by 2,2- dimethoxypropane (16.49 mL, 13.99 g, 134.39 mmol, 5.00 equiv). The reaction mixture was heated to 40 °C and stirred for l1/_ hours and then cooled to ambient temperature. Sat. aqueous Na2C03 (50 mL) was added to quench the reaction and filtered by suction to remove ZnC03. The solid was washed with EtOAc (40 mL). The biphasic filtrate was transferred to a separatory funnel and the yellow organic layer was separated. The colorless aqueous layer was extracted with EtOAc (2 x 20 mL). The combined organic layers were quickly dried over anhydrous MgS04, filtered and evaporated in vacuo to dryness. The crude product was purified by dry column vacuum chromato- graphy (10% EtOAc in heptane) affording the desired product as a yellow liquid. To remove excess solvents the product was co-evaporated with DCM (3 x 25 mL) followed by drying in high vacuum overnight (5.73 g, 88%). The product 6a crystallized upon storage in the refrigerator. Rf 0.57 (15% EtOAc in heptane, KMn04 staining). ^ NMR (CDCI3) : δ 7.23 (dd, J = 7.4, 1.5 Hz, 1H), 7.08 (d, J = 1.5 Hz, 1H), 6.68 (d, J = 8.5 Hz, 1H), 4.79 (s, 2H), 1.52 (s, 6H).
((3-Bromo-5-propoxybenzyl)oxy)(fert-butyl)dimethylsilane
(6b).
Phenol 16 (4.00 g, 12.6 mmol, 1.00 equiv) was dissolved in DMF (25 mL) and added K2C03 (3.48 g, 25.2 mmol, 2.00 equiv) and propyl bromide (2.30 mL, 3.11 g, 25.3 mmol, 2.01 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was added Et20 (200 mL) and washed with water (3 x 100 mL), brine (75 mL), dried over MgS04, filtered and concentrated in vacuo. The mixture was purified by DCVC (dia. = 6 cm, 75 mL fractions, 0 - 10% EtOAc in heptanes) to yield 6b as a clear, colorless oil (4.09 g, 90%). ^ NMR (300 MHz, CDCI3) : δ 0.12 (6H, s), 0.96 (9H, s), 1.04 (3H, t, J = 8 Hz), 1.81 (2H, sextet, 3 = 1 Hz), 3.90 (2H, t, J = 7Hz), 4.67 (2H, s), 6.81 (1H, s), 6.92 (1H, s), 7.02 (1H, s); 13C NMR: (75 MHz, CDCI3) : δ -5.0, 10.7, 18.7, 22.7, 26.2, 64.4, 69.9, 111.1, 116.2, 121.1, 122.6, 144.7, 159.9; TLC: Rf : 0.77 (EtOAc: heptanes (1 : 10)). ((3-(Benzyloxy)-5-bromobenzyl)oxy)(£er£-butyl)dimethylsilane
(6c).
Phenol 16 (3.50 g, 11.0 mmol, 1.00 equiv) was dissolved in acetone (50 mL) and added K2C03 (3.10 g, 22.4 mmol, 2.04 equiv) and benzyl bro- mide (2.10 mL, 3.02 g, 17.7 mmol, 1.61 equiv). The mixture was left to stir at reflux for 24 h. Piperazine (4.75 g, 55.1 mmol, 5.01 equiv) was added and the mixture refluxed for another 2.5 h. The mixture was cooled to r.t. and concentrated in vacuo. The mixture was added water (50 mL) and EtOAc (50 mL) and transferred to a separation funnel containing EtOAc (100 mL) and sat. NH4CI (100 mL). After separation the aqueous phase was reextracted with EtOAc (50 mL). The combined organic phases was washed with brine (80 mL), dried over MgS04, filtered and concentrated in vacuo. The mixture was purified by DCVC (dia. = 7 cm, 75 mL fractions, 0 - 8 % EtOAc in heptanes) to yield 7 as a clear, colorless oil (4.25 g, 10.4 mmol, 95%). H NMR (300 MHz, CDCI3) : δ 0.12 (6H, s), 0.96 (9H, s), 4.68 (2H, s), 5.05 (2H, s), 6.90 (1H, s), 7.02 (1H, s), 7.06 (1H, s), 7.30-7.45 (5H, m); 13C NMR (75 MHz, CDCI3) : δ -5.01, 18.6, 26.2, 64.3, 70.3, 111.3, 116.6, 121.5, 122.6, 127.5, 128.1, 128.6, 136.5, 144.8, 159.5; TLC: Rf : 0.65 (EtOAc: heptanes (1 : 10)).
((3-([l,l'-Biphenyl]-3-ylmethoxy)-5-bromobenzyl)oxy)(£er£- butyl)dimethylsilane (6d).
Phenol 16 (3.00 g, 9.46 mmol, 1.00 equiv) was dissolved in acetone (50 mL) and added K2C03 (2.61 g, 18.9 mmol, 2.00 equiv) and 3- phenylbenzyl bromide (3.52 g, 14.2 mmol, 1.50 equiv). The mixture was left to stir at reflux for 24 h. Piperazine (4.10 g, 47.6 mmol, 5.03 equiv) was added and the mixture refluxed for another 2.5 h. The mixture was cooled to r.t. and concentrated in vacuo. The mixture was added water (50 mL) and EtOAc (50 mL) and transferred to a separation funnel containing EtOAc (100 mL) and sat. NH4CI (100 mL). After separation the aqueous phase was reex- tracted with EtOAc (50 mL). The combined organic phases was washed with brine (80 mL), dried over MgS04, filtered and concentrated in vacuo. The mixture was purified using DCVC (dia. = 7 cm, 75 mL fractions, 0 - 8% EtOAc in heptanes) to yield 6d as a clear, colorless oil (4.21 g, 92%). H NMR (300 MHz, CDCI3) : δ 0.11 (6H, s), 0.96 (9H, s), 4.68 (2H, s), 5.11 (2H, s), 6.92 (1H, s), 7.02-7.09 (2H, m), 7.32-7.51 (5H, m), 7.53-7.67 (4H, m); 13C NMR (75 MHz, CDCI3) : δ -4.99, 18.6, 26.2, 64.3, 70.3, 111.3, 116.6, 121.6, 122.6, 126.3, 126.4, 127.0, 127.2, 127.5, 128.8, 129.1, 137.0; TLC: Rf : 0.58 (EtOAc: heptanes (1 : 10)).
(2S,3/?)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3- (salicyl alcohol isopropylidene acetal-6-yl)-5-oxopyrrolidine-l- carboxylate (7a). A dry round-bottomed flask was charged with the bromide 6a (1.86 g, 7.63 mmol, 2.50 equiv) and a nitrogen atmosphere was applied thoroughly. Dry Et20 (25 mL) was added via a syringe and the clear, pale yellow solution was cooled to -78 °C. tert-BuLi in pentane (10.16 mL, 978 mg, 15.27 mmol, 5.00 equiv, 1.50M) was added dropwise over the course of 25 minutes. The reaction mixture became milky due to precipitation. After 10 minutes of stirring at -78 °C, a suspension of CuCN (342 mg, 3.82 mmol, 1.25 equiv) in dry Et20 (2.4 mL) was added portion wise. The resulting suspension was stirred at -78 °C for 5 minutes and then at -42 °C for 10 min, after which it was re-cooled to -78 °C. Enone 5 (1.00 g, 3.05 mmol, 1.00 equiv) was dissolved in dry Et20 (3.0 mL) and added dropwise to the cuprate mixture at -78 °C, which resulted in a slight color change to orange. The temperature was raised to -42 °C and the reaction mixture was stirred at this temperature for 1 hour. The dark brown solution with barely any precipitation was quenched by addition of sat. NH4CI(aq) (5 mL), allowed to warm up to ambient temperature and then transferred to a separating funnel with brine (30 mL) and EtOAc (30 mL). The organic layer was separated and the blue aqueous layer was extracted with EtOAc (2 x 30 mL). The combined organic layers were dried over anhydrous MgS04, filtered and eva- porated in vacuo to dryness. Purification by dry column vacuum chromatography (0→20% EtOAc in heptane) afforded the desired product 7a as a pale yellow oil (1.05 g, 70%). Rf 0.40 (25% EtOAc in heptane, PMA staining). ^ NMR (CDCI3) : δ 6.98 (dd, J = 8.5, 2.2 Hz, 1H), 6.78 (dd, J = 5.1, 3.2 Hz, 2H), 4.82 (s, 2H), 4.05-3.97 (m, 2H), 3.80 (dd, J = 9.9, 1.7 Hz, 1H), 3.38 (dt, J = 9.4, 2.5, 1H), 3.15 (dd, J = 17.7, 9.5 Hz, 1H), 2.50 (dd, J = 17.6, 2.5 Hz, 1H), 1.56 (s, 6H), 1.55 (s, 9H), 0.93 (s, 9H), 0.10 (s, 3H), 0.09 (s, 3H). 13C NMR (CDCI3) : δ 174.1, 150.1, 149.8, 136.1, 126.2, 122.2, 119.7, 117.5, 99.9, 99.5, 83.1, 67.0, 63.6, 60.9, 40.1, 38.2, 28.2, 25.9, 24.9, 24.8, 18.3, -5.4. MS (m/z) calcd. for C2iH34N04Si [M-Boc + H]+ 392.2, found 392.2. [a]25D -20.7° (c = 0.28, EtOH). (2S,3/?)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3- (3-(((£er£-butyldimethylsilyl)oxy)-methyl)-5-propoxyphenyl)-5- oxopyrrolidine-l-carboxylate (7b).
Solution A: Thiophene (482 mg, 5.74 mmol, 1.57 equiv) was dissolved in dry Et20 (6 mL) in a dry vial under N2. The mixture was cooled to 0 °C and n-BuLi (2.34 mL, 5.85 mmol, 1.60 equiv, 2.50M) added dropwise over the course of 15 min. The mixture was left to stir at 0 °C for 15 min. then at r.t. for 2 h (a white, colloid precipitate was formed).
Bromide 6b (1.65 g, 4.59 mmol, 1.25 equiv) was dissolved in dry Et20 (50 mL) in a dry flask under N2 and cooled to -78 °C. t-BuLi (5.76 mL, 9.22 mmol, 2.52 equiv, 1.60 M) was added dropwise over the course of 20 min. The mixture was left to stir at -78 °C for 45 min. (The solution became slightly colored and unclear) before a suspension of CuCN (410 mg, 4.58 mmol, 1.25 equiv) in dry Et20 (4 mL) was added dropwise over the course of 6 min. The mixture was left to stir at -78 °C for 10 min, then 10 min at -42 °C before recooled to -78 °C. To the mixture was added dropwise Solution A (7.1 mL, 4.6 mmol, 1.25 equiv) over the course of 20 min. and left to stir at - 78 °C for 10 min, then at -42 °C for 10 min before recooled to -78 °C. A solution of enone 5 (1.20 g, 3.66 mmol, 1.00 equiv) dissolved in Et20 (10 mL) was dropwise added over the course of 25 min. (The mixture instantly turned highly yellow). The mixture was left to stir at -78 °C for 10 min., then at -42 °C for 45 min before quenched using sat. NH4CI (8 mL). The mixture was transferred to a separation funnel containing sat. NaHCC>3 (50 mL) and EtOAc (50 mL). The phases were separated and the aqueous phase extracted with EtOAc (2 x 50 mL). The combined organic phases were washed with brine (50 mL), dried over MgS04, filtered and concentrated in vacuo. Purification by DCVC (0 - 12 % EtOAc in heptanes and 0 - 5 % EtOAc in PhMe) to yield 1.38 g (62%) of 7b as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 0.08 (3H, s), 0.10 (3H, s), 0.12 (6H, s), 0.92 (9H, s), 0.96 (9H, s), 1.04 (3H, t, J = 8 Hz), 1.54 (9H, s), 1.81 (2H, h, J = 7 Hz), 2.54 (1H, dd, J = 18, 3 Hz), 3.12 (1H, dd, J = 18, 10 Hz), 3.40 (1H, dt, J = 10, 2 Hz), 3.79 (1H, dd, J = 11, 2 Hz), 3.91 (2H, t, J = 7 Hz), 4.00 (1H, dd, J = 10, 4 Hz), 4.07 (1H, m), 4.68 (2H, s), 6.60 (1H, m), 6.68 (1H, s), 6.80 (1H, m); 13C NMR (75 MHz, CDCI3) : δ -5.2, -5.0, 10.7, 18.4, 18.6, 22.8, 26.1, 26.2, 28.3, 38.9, 40.0, 63.8, 64.9, 66.8, 69.6, 83.0, 110.5, 111.6, 116.0, 143.9, 145.6, 149.8, 159.7, 174.1; LCMS: m/z [M + H]+ : calc: 607.4, found : 508.1; TLC: Rf = 0.49 (EtOAc : toluene (1 : 10)); OR: [a]23 D: -22.33° (c = 0.30, abs EtOH).
(2S,3/?)-tert-Butyl 3-(3-(benzyloxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((£er£-butyldime- thylsilyl)oxy)methyl)-5-oxopyrrolidine-l-carboxylate (7c).
Solution A: Thiophene (518 mg, 6.16 mmol, 1.56 equiv) was dissolved in dry Et20 (8 mL) in a dry vial under N2. The mixture was cooled to 0 °C and dropwise added 2.29M n-BuLi (2.74 mL, 6.27 mmol, 1.59 equiv) over the course of 15 min. The mixture was left to stir at 0 °C for 15 min. before re- moved from the ice bath and stirred at r.t. for 2 h (a white, coloide precipitate was formed).
Bromide 6c (2.01 g, 4.93 mmol, 1.25 equiv) was dissolved in dry Et20 (60 mL) in a dry flask under N2 and cooled to -78 °C. t-BuLi (6.18 mL, 9.89 mmol, 2.51 equiv, 1.60M) was added dropwise over the course of 15 min and the mixture was left to stir at -78 °C for 30 min. (The solution became slightly yellow colored and unclear) before a suspension of CuCN (440 mg, 4.91 mmol, 1.25 equiv) in dry Et20 (4 mL) was added dropwise over the course of 5 min. The mixture was left to stir at -78 °C for 10 min, then 10 min at -42 °C before recooled to -78 °C. To the mixture was added dropwise Solution A (9.0 mL, 4.9 mmol, 1.34 equiv) over the course of 15 min. and left to stir at -78 °C for 10 min, then at -42 °C for 10 min before recooled to -78 °C. A solution of enone 5 (1.29 g, 3.94 mmol, 1.00 equiv) dissolved in Et20 (4 mL) was dropwise added over the course of 15 min. (The mixture instantly turned highly yellow). The mixture was left to stir at -78 °C for 15 min., then at -42 °C for 40 min before quenched using sat. NH4CI (10 mL). The mixture was transferred to a separation funnel containing sat. NaHCC>3 (75 mL) and EtOAc (75 mL). The phases were separated and the aqueous phase was extracted with EtOAc (2 x 50 mL). The combined organic phases were washed with brine (75 mL), dried over MgS04, filtered and concentrated in vacuo. Purification by DCVC (dia. = 7, 75 mL fractions, 0 - 10 % EtOAc in heptanes) to yield 1.11 g (43%) of 7c as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 0.09 (3H, s), 0.10 (3H, s), 0.11 (6H, s), 0.93 (9H, s), 0.95 (9H, s), 1.54 (9H, s), 2.53 (1H, dd, J = 18, 2 Hz), 3.13 (1H, dd, J = 18, 10 Hz), 3.31 (1H, dm, J = 9 Hz), 3.79 (1H, dd, J = 11, 2 Hz), 4.00 (1H, dd, J = 11, 4 Hz), 4.07 (1H, m), 4.69 (2H, s), 5.05 (2H, s), 6.67 - 6.73 (2H, m), 6.89 (1H, s), 7.28 - 7.45 (5H, m). 13C NMR (75 MHz, CDCI3) : δ -5.0, -4.8, 18.6, 18.8, 26.3, 26.4, 28.5, 39.1, 40.2, 64.0, 65.1, 67.0, 70.2, 83.2, 111.0, 112.2, 116.6, 127.7, 128.2, 128.8, 137.0, 144.2, 145.8, 150.0, 159.5, 174.1; LCMS: m/z [M + H] + : calc: 655.4, found : 556.3; TLC: Rf = 0.44 (EtOAc : heptanes (1 : 4)); OR: [a]35 D: -22.22° (c = 0.89, abs. EtOH).
(2S,3/?)-tert-Butyl 3-(3-([ l,l'-biphenyl]-3-ylmethoxy)-5- (((£er£-butyldimethylsilyl)oxy)-methyl)phenyl)-2-(((£er£- butyldimethylsilyl)oxy)methyl)-5-oxopyrrolidine-l-carboxylate (7d).
Solution A: Thiophene (700 mg, 8.32 mmol, 1.58 equiv) was dissolved in dry Et20 (8 mL) in a dry vial under N2.The mixture was cooled to 0 °C and dropwise added n-BuLi (3.70 mL, 8.47 mmol, 1.60 equiv, 2.29 M) over the course of 20 min. The mixture was left to stir at 0 °C for 15 min. then at r.t. for 2 h (a white, coloide precipitate was formed).
Bromide 6d (3.17 g, 6.56 mmol, 1.24 equiv) was dissolved in dry Et20 (80 mL) in a dry flask under N2 and cooled to -78 °C. t-BuLi (8.28 mL, 13.25 mmol, 2.51 equiv, 1.60M) was then added dropwise over the course of 30 min. The mixture was left to stir at -78 °C for 45 min. (The solution became yellow colored) before a suspension of CuCN (590 mg, 6.59 mmol, 1.25 equiv) in dry Et20 (4 mL) was added dropwise over the course of 15 min. The mixture was left to stir at -78 °C for 10 min (mixture turned wine red), then 15 min at -42 °C before recooled to -78 °C over the course of 10 min. The mixture was dropwise added Solution A (9.9 mL, 6.6 mmol, 1.25 equiv) over the course of 30 min. and left to stir at -78 °C for 10 min, then at -42 °C for 10 min before recooled to -78 °C. A solution of enone 5 (1.73 g, 5.28 mmol, 1.00 equiv) dissolved in Et20 (4 mL) was dropwise added over the course of 15 min. (The mixture turned slightly yellow). The mixture was left to stir at - 78 °C for 15 min., then at -42 °C for 90 min before quenched using sat. NH4CI (12 mL). The mixture was transferred to a separation funnel containing sat. NaHC03 (75 mL) and EtOAc (75 mL). The phases were separated and the aqueous phase extracted with EtOAc (2 x 50 mL). The combined organic phases were washed with brine (75 mL), dried over MgS04, filtered and concentrated in vacuo. Purification by DCVC (0 - 10% EtOAc in heptanes and 0 - 4% EtOAc in PhMe) to yield 1.62 g (42%) of 7d as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 0.10 (3H, s), 0.11 (3H, s), 0.12 (6H, s), 0.94 (9H, s), 0.96 (9H, s), 1.55 (9H, s), 2.57 (1H, dd, J = 18, 3 Hz), 3.16 (1H, dd, J = 18, 9 Hz), 3.44 (1H, dm, J = 10 Hz), 3.81 (1H, dd, J = 10, 2 Hz), 4.02 (1H, dd, J = 10, 4 Hz), 4.10 (1H, m), 4.71 (2H, s), 5. 13 (2H, s), 6.74 (2H, m), 6.94 (1H, m), 7.33-7.52 (5H, m), 7.53-7.70 (4H, m). 13C NMR (75 MHz, CDCI3) : δ -5.2, -5.0, 18.4, 18.6, 26.1, 26.2, 28.3, 38.9, 40.1, 63.8, 64.9, 66.8, 70.1, 83.1, 110.6, 112.1, 116.4, 126.3, 126.4, 126.9, 127.2, 127.4, 128.8, 129.1, 137.3, 140.8, 141.6, 144.0, 145.7, 149.8, 159.3; LCMS : m/z [M + H]+ : calc: 731.4, found : 632.4; TLC : Rf = 0.38 (EtOAc : Heptanes (1 : 4)); OR: [a]31 D: -22.68 (c = 0.71 g/100 mL; Abs. EtOH).
(2S,3/?)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3- (2,2-dimethyl-4H-benzo[d] [ l,3]-dioxin-6-yl)pyrrolidine-l- carboxylate (8a).
A round-bottomed flask charged with the conjugated product 7a (558 mg, 1.14 mmol, 1.00 equiv) and equipped with a condenser and an atmosphere of nitrogen was applied. Through the septum, dry THF (15 mL) and 1M BH3-THF (7.5 mL, 648 mg, 7.54 mmol, 6.00 equiv) were added via syringes and the colorless reaction mixture was heated to reflux (80 °C) for 31/2 hours. Then cooled to 0 °C and THF (10 mL), H20 (1 mL), 2M aqueous NaOH (13 mL) and 30% H202 (4 mL) were added sequentially. Stirred for 31/2 hours at room temperature and then quenched by addition of sat. aqueous NaHC03 (30 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over anhydrous MgS04, filtered and evaporated in vacuo to dryness. Purification by dry column vacuum chromatography (0→15% EtOAc in heptane) afforded the desired product 8a as a colorless, viscous oil (248 mg, 46%). Rf 0.47 (25% EtOAc in heptane, PMA staining). ^ NMR (CDCI3) : δ 7.05-6.96 (m, 1H), 6.81-6.75 (m, 2H), 4.83 (s, 2H), 4.01 (dd, J = 10.3, 3.7 Hz, 1H), 3.79-3.60 (m, 4H), 3.45 (td, J = 7.2, 5.4 Hz, 1H), 3.37-3.26 (m, 1H), 2.30-2.16 (m, 1H), 1.94- 1.79 (m, 1H), 1.56 (s, 6H), 1.50 (s, 9H), 0.91 (s, 9H), 0.06 (s, 6H); 13C NMR (CDCI3) : δ (rotamers present) 154.2, 149.6, 135.7, 135.1, 127.0, 126.8, 123.2, 123.1, 119.2, 117.0, 99.4, 79.4, 65.7, 65.5, 62.8, 61.4, 61.0, 47.0, 46.4, 46.0, 44.9, 33.0, 31.8, 28.7, 26.0, 24.9, 18.3, -5.2; MS (m/z) calcd. for C2iH36N03Si [M-Boc + H]+ 378.3, found 378.2; [a]25D +9.7° (c = 0.68, EtOH).
(2S,3/?)-tert-Butyl- 2-(((tert-butyldimethylsilyl)oxy)methyl)-3- (3-(((£er£-butyldimethylsilyl)oxy)-methyl)-5- propoxyphenyl)pyrrolidine-l-carboxylate (8b). 7c The compound 7b (1.43 g, 2.35 mmol, 1.0 equiv) was dissolved in dry THF (25 mL) and added IM BH3 THF complex (25 mL, 25 mmol, 10.6 equiv) over the course of 5 min. The mixture was refluxed under N2 for 20 h. The mixture was cooled to 0 °C and dropwise added H20 (5 mL) over the course of 10 min., NaOH (2M, 25 mL) dropwise over the course of 20 min and H202 (30%, 5 mL) over the course of 5 min (organic/aqueous ratio important). After 5 min the mixture was remove from the icebath and left to stir at r.t. for 1 h. The mixture was poured into sat. NaHC03 (100 mL) and EtOAc (75 mL). After separation the aqueous phase was extracted with EtOAc (2 x 75 mL) and the combined organic phases was washed with brine, dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 4 cm, 30 mL fractions, 0 - 5% EtOAc in PhMe) to yield 820 mg (59%) of 8b as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 0.06 (6H, s), 0.12 (6H, s), 0.91 (9H, s), 0.96 (9H, s), 1.04 (3H, t, J = 7 Hz), 1.49 (9H, s), 1.81 (2H, h, J = 7 Hz), 1.82 - 1.95 (1H, m), 2.17 - 2.32 (1H, m), 3.25 - 3.42 (1H, m), 3.49 (1H, m), 3.55-3.80 (3H, m), 3.80 - 3.87 (0.5H, m), 3.91 (2H, t, J = 6 Hz), 3.98-4.07 (0.5H, m), 6.66 (1H, m), 6.73 (1H, s), 6.75 (1H, m). 13C NMR (75 MHz, CDCI3) : δ -5.1, -4.9, 10.8, 18.4, 18.7, 22.8, 26.1, 26.2, 28.8, 31.8, 32.8, 45.7, 46.7, 47.2, 61.5, 62.9, 65.1, 65.5, 65.7, 69.5, 79.1, 79.5, 109.9, 110.2, 112.3, 117.1, 117.4, 143.1, 143.2, 144.9, 145.4, 154.2, 154.3, 159.4. LCMS: m/z [M + H] + : calc: 593.4, found : 494.4 TLC: Rf = 0.44 (EtOActoluene (1 : 20)); OR: [a]23 D: +11.48° (c = 0.30, abs EtOH).
(2S,3 ?)-tert-Butyl-3-(3-(benzyloxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((£er£-butyldime- thylsilyl)oxy)methyl)pyrrolidine-l-carboxylate (8c). 7c
The compound 7c (1.66 g, 2.53 mmol, 1.00 equiv) was dissolved in dry THF (20 mL) and added IM BH3 THF complex (25 mL, 25 mmol, 9.88 equiv) over the course of 5 min. The mixture was refluxed under N2 for 20 h. The mixture was cooled to 0 °C, added THF (40 mL) and dropwise added H20 (6 mL) over the course of 15 min. NaOH (2M, 30 mL) was dropwise over the course of 15 min and H202 (30 %, 10 mL) over the course of 15 min (organic/aqueous ratio important). After 5 min the mixture was remove from the icebath and left to stir at r.t. for 1 h. The mixture was poured into sat. NaH- C03 (100 mL) and EtOAc (75 mL). After separation the aqueous phase was extracted with EtOAc (2 x 75 mL) and the combined organic phases was washed with brine (100 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 4 cm, 30 mL fractions, 0 - 2% EtOAc in PhMe) to yield 976 mg (60%) of 8c as a clear, colorless oil. H NMR (300 MHz, CDCI3) : δ 0.08 (6H, s), 0.12 (6H, s), 0.93 (9H, s), 0.97 (9H, s), 1.51 (9H, s), 1.85-2.00 (1H, m), 2.18 - 2.35 (1H, m), 3.27 - 3.43 (1H, m), 3.46 - 3.55 (1H, m), 3.58 - 3.80 (3H, m), 3.86 (0.5H, m), 4.05 (0.5H, dd, J = 10, 4 Hz), 4.71 (2H, s), 5.06 (2H, s), 6.71-6.80 (2H, m), 6.86 (1H, m), 7.30- 7.47 (5H, m). 13C NMR (75 MHz, CDCI3) : δ -5.1, -4.9, 18.4, 18.7, 26.1, 26.2, 28.8, 31.8, 32.8, 45.7, 46.7, 47.2, 61.5, 62.8, 65.0, 65.5, 65.6, 70.0, 76.7, 77.2, 77.6, 79.2, 79.5, 110.2, 110.4, 112.7, 117.5, 117.7, 127.5, 127.9, 128.6, 136.99, 137.03, 143.2, 143.3, 145.0, 145.4, 154.1, 154.2, 159.1; LCMS: m/z [M + H]+ : calc: 641.4, found : 542.4; TLC: Rf = 0.20 (EtOAc : toluene (1 : 40)); OR: [a]35 D: +3.20° (c = 0.50, abs EtOH).
(2S,3/?)-tert-Butyl- 3-(3-([ l,l'-biphenyl]-3-ylmethoxy)-5- (((£er£-butyldimethylsilyl)oxy)methyl)-phenyl)-2-(((£er£- butyldimethylsilyl)oxy)methyl)pyrrolidine-l-carboxylate (8d).
The compound 7d (1.77 g, 2.42 mmol, 1.00 equiv) was dissolved in dry THF (20 mL) and added 1M BH3 THF complex (25 mL, 25 mmol, 10.3 equiv) over the course of 5 min. The mixture was refluxed under N2 for 20 h. The mixture was cooled to 0 °C, added THF (40 mL) and dropwise added H20 (6 mL) over the course of 15 min. NaOH (2M, 30 mL) was dropwise over the course of 15 min and H202 (30 %, 10 mL) over the course of 15 min (organic/aqueous ratio important). After 5 min the mixture was remove from the icebath and left to stir at r.t. for 1 h. The mixture was poured into sat. NaH- C03 (100 mL) and EtOAc (75 mL). After separation the aqueous phase was extracted with EtOAc (2 x 75 mL) and the combined organic phases was washed with brine (100 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 4 cm, 30 mL fractions, 0 - 2% EtOAc in PhMe) to yield 887 mg (51%) of 8d as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 0.10 (6H, s), 0.14 (6H, s), 0.95 (9H, s), 0.99 (9H, s), 1.53 (9H, s), 1.94 (1H, m), 2.28 (1H, m), 3.38 (1H, m), 3.55 (1H, m), 3.62 - 3.86 (3H, m), 3.90 (0.5H, m), 4.08 (0.5H, dd, J = 10, 4 Hz), 4.74 (2H, s), 5.14 (2H, s), 6.78 - 6.84 (2H, m), 6.91 (1H, m), 7.33 - 7.52 (5H, m), 7.55 - 7.72 (4H, m). 13C NMR (75 MHz, CDCI3): δ -5.1, -4.9, 18.4, 18.6, 26.1, 26.2, 28.8, 31.8, 32.8, 45.7, 46.7, 47.2, 61.5, 62.8, 65.0, 65.5, 65.6, 70.0, 79.2, 79.5, 110.1, 110.3, 112.8, 117.6, 117.8, 126.35, 126.44, 126.8, 127.2, 127.4, 128.8, 129.0, 137.49, 137.54, 140.9, 141.6, 143.26, 143.34, 145.0, 145.4, 154.15, 154.23, 159.1; LCMS: m/z [M + H] + : calc: 717.4, found : 618.4; TLC: Rf = 0.21 (EtOActoluene (1 :40)); OR: [a]35 D: +1.94° (c = 0.67, abs EtOH).
(2S,3/?)-tert-Butyl 3-(2,2-dimethyl-4H-benzo[d] [l,3]dioxin-6- yl)-2-(hydroxymethyl)-pyrrolidine-l-carboxylate (9a).
The compound 8a (448 mg, 0.94 mmol, 1.0 equiv) was dissolved in dry THF (9 mL) under an atmosphere of N2 and 1M TBAF in THF (2.8 mL, 736 mg, 2.81 mmol, 3.0 equiv) was added at room temperature. The reaction mixture was stirred for two. H20 (20 mL) and sat. aqueous NaHC03 (20 mL) were added and then extracted with EtOAc (3 x 30 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and evaporated in vacuo to dryness. Purified by dry column vacuum chromatography (15→35% EtOAc in heptane), followed by drying in high vacuum overnight afforded the desired alcohol as a colorless, viscous oil, which crystallized to a white solid upon drying in high vacuum (295 mg, 87%). ^ NMR (CDCI3) : δ 7.02 (dd, J = 8.4, 2.3 Hz, 1H), 6.84 (broad s, 1H), 6.76 (d, J = 8.3 Hz, 1H), 5.13 (broad d, J = 8.0 Hz, 1H), 4.82 (s, 2H), 3.86 (t, J = 7.8 Hz, 1H), 3.73 (t, J = 9.4 Hz, 2H), 3.64-3.57 (m, 1H), 3.33 (dt, J = 10.6, 6.3 Hz, 1H), 2.84-2.76 (m, 1H), 2.15-2.08 (m, 1H), 1.99-1.85 (m, 1H), 1.55 (s, 6H), 1.51 (s, 9H). 13C NMR (CDCI3) : δ 156.7, 150.1, 132.2, 127.2, 123.5, 119.4, 117.2, 99.5, 80.5, 67.2, 66.0, 60.9, 47.3, 47.1, 33.1, 28.5, 24.8, 24.7. MS (m/z) calcd. for C16H22NO5 [M-fBu + H]+ 308.2, found 308.1. [a]25 D +14.2° (c = 0.86, EtOH). Mp 110-112 °C.
(2S,3R)-tert-Buty\ 2-(hydroxymethyl)-3-(3-(hydroxymethyl)-5- propoxyphenyl)pyrrolidine-l-carboxy-late (9b).
Compound 8b (1.30 g, 2.19 mmol, 1.00 equiv) was dissolved in dry THF (12 mL) in a dry vial and added 1M TBAF (8.80 mL, 8.80 mmol, 4.02 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was quenched using 50% sat. NaHC03 (50 mL) and transferred to a separation funnel containing EtOAc (25 mL). The aqueous phase was extracted with EtOAc (2 x 50 mL) and the combined organic phases was washed with brine (50 mL), dried over MgS04, filtered concentrated in vacuo and purified using DCVC (dia = 3 cm, 25 mL fractions, 0 - 90% EtOAc in heptanes) to yield 736 mg (92%) of diol 9b as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ
I.05 (3H, t, J = 7 Hz), 1.51 (9H, s), 1.81 (2H, h, J = 7 Hz), 1.90 - 2.07 (2H, m), 2.15 (1H, m), 2.86 (1H, m), 3.35 (1H, m), 3.62 (1H, dd, J = 11, 7 Hz), 3.68 - 3.90 (3H, m), 3.92 (2H, t, J = 7 Hz), 4.65 (2H, s), 5.07 (1H, m), 6.70 (1H, m), 6.78 - 6.84 (2H, m). 13C NMR (75 MHz, CDCI3) : δ 10.8, 22.8, 28.7, 33.1, 47.4, 48.1, 65.4, 66.2, 67.2, 69.7, 80.7, 111.3, 113.6, 118.2, 142.5, 142.9, 156.8, 159.7; LCMS: m/z [M + H] + : calc: 365.2, found : 310.2; TLC: Rf = 0.37 (EtOAc : heptanes (3 : 1)); OR: [a]23 D: +23.85° (c = 0.31, abs EtOH).
(2S,3/?)-tert-Butyl 3-(3-(benzyloxy)-5-
(hydroxymethyl)phenyl)-2-(hydroxymethyl)pyrrolidine-l- carboxylate (9c).
The compound 8c (967 g, 1.51 mmol, 1.00 equiv) was dissolved in dry THF (15 mL) in a dry vial and added 1M TBAF (6 mL, 6 mmol, 3.97 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was quenched using 50% sat. NaHC03 (50 mL) and transferred to a separation funnel containing EtOAc (50 mL). The aqueous phase was reextracted with EtOAc (2 x 50 mL) and the combined organic phases was washed with brine (75 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 4 cm, 30 mL fractions, 0 - 100% EtOAc in heptanes) to yield 572 mg (92%) of diol 9c as a clear, colorless oil. 1H NMR (300 MHz, CDCI3) : δ 1.51 (9H, s), 1.89 -
2.08 (2H, m), 2.14 (1H, m), 2.87 (1H, m), 3.34 (1H, m), 3.61 (1H, dd, J =
II, 7 Hz), 3.67 - 3.90 (3H, m), 3.95 (1H, m), 4.65 (2H, s), 5.06 (2H, s),
5.09 (1H, m), 6.79 (1H, m), 6.85 (1H, m), 6.88 (1H, m), 7.28 - 7.45 (5H, m). 13C NMR (75 MHz, DMSO) : δ 28.2, 30.5, 31.2, 44.5, 45.3, 45.8, 46.0,
60.7, 60.8, 62.8, 65.5, 69.0, 78.4, 78.5, 110.4, 111.8, 117.4, 127.5, 127.7,
128.3, 137.0, 144.2, 144.8, 145.0, 153.2, 153.6, 158.3; LCMS: m/z [M +
H]+ : calc: 413.2, found : 358.2; TLC: Rf = 0.42 (EtOAc : heptanes (3 : 1));
OR: [a]35 D: +12.27° (c = 0.44, abs EtOH).
(2S,3/?)-tert-Butyl 3-(3-([ l,l'-biphenyl]-3-ylmethoxy)-5-
(hydroxymethyl)phenyl)-2-(hydroxymethyl)-pyrrolidine-l- carboxylate (9d).
The compound 8d (800 g, 1.11 mmol, 1.00 equiv) was dissolved in dry THF (15 mL) in a dry vial and added 1M TBAF (5 mL, 5 mmol, 4.50 equiv). The mixture was left to stir at r.t. for 18 h. The mixture was quenched using phosphate buffer (50 mL, Ph 7) and transferred to a separation funnel containing EtOAc (50 mL). The aqueous phase was reextracted with EtOAc (2 x 50 mL) and the combined organic phases was washed with brine (50 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 4 cm, 30 mL fractions, 0 - 90 % EtOAc in heptanes) to yield 512 mg (94%) of diol 9d as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 1.51 (9H, s), 1.97 (IH, m), 2.14 (IH, m), 2.60-3.00 (2H, m), 3.34 (IH, m), 3.63 (IH, dd, J = 11, 7 Hz), 3.65 (2H, m), 3.96 (IH, m), 4.63 (2H, s), 5.10 (2H, s), 5.24 (IH, m), 6.81 (IH, m), 6.86 (IH, s), 6.91 (IH, m), 7.32-7.50 (5H, m), 7.53-7.60 (4H, m). 13C NMR (75 MHz, CDCI3) : δ 28.6, 32.9, 47.2, 47.9, 64.9, 65.9, 67.0, 70.1, 80.7, 111.3, 113.7, 118.5, 126.4, 126.5, 126.8, 127.2, 127.4, 128.8, 129.0, 137.3, 140.8, 141.5, 142.5, 143.2, 156.7, 159.2; LCMS: m/z [M + H] + : calc: 489.3, found : 434.2; TLC: Rf = 0.39 (EtOAc : heptanes (3 : 1)); OR: [a]35 D: + 10.19 0 (c = 0.53, abs EtOH).
(2S,3 ?)-l-(tert-Butoxycarbonyl)-3-(2,2-dimethyl-4-oxo-4H- benzo[d] [l,3]dioxin-6-yl)pyrrolidine-2-carboxylic acid (10a) and (2S,3 ?)-l-(tert-butoxycarbonyl)-3-(2,2-dimethyl-4H- benzo[d] [l,3]dioxin-6-yl)pyrrolidine-2-carboxylic acid (11).
To an ice-cooled mixture of the protected alcohol (150 mg, 0.413 mmol, 1.0 equiv) in MeCN (2.5 mL) and EtOAc (2.5 mL) was added dropwise NaI04 (883 mg, 4.13 mmol, 10.0 equiv) and RuCI3 H20 (4 mg, 0.017 mmol, 0.04 equiv) dissolved in H20 (4 mL). The reaction was stirred at 0 °C for V/2 hours. The brownish reaction mixture was filtered through a plug of Celite® and washed with EtOAc (15 mL). The filtrates were transferred to a separating funnel and the organic layer was isolated. The aqueous layer was extracted with EtOAc (2 x 15 mL) and the combined organic phases were washed with brine (25 mL) and dried over anhydrous Na2S04. The crude product was isolated as a dark brown oil by evaporation in vacuo (179 mg). Purification by dry column vacuum chromatography (5→ 25% EtOAc in heptane containing 1% AcOH) afforded compound 10a and 11, respectively. For 10a: White foam (36 mg, 22%). Rf 0.25 (50% EtOAc in heptane + 1% AcOH). ^ (CDCI3) : δ 7.88-7.84 (m, IH), 7.49 (t, J = 7.7 Hz, IH), 6.98-6.94 (m, IH), 4.36 (d, J = 5.9 Hz, 0.5H), 4.25 (d, J = 7.0 Hz, 0.5H), 3.86-3,73 (m, 2H), 3.64-3.45 (m, IH), 2.40-2.30 (m, IH), 2.13-2.02 (m, IH), 1.76 (s, 6H), 1.54 (s, 5H), 1.45 (s, 4H). 13C NMR (CDCI3) : δ 196.3, 183.3, 174.9, 174.4, 173.7, 172.8, 161.1, 155.6, 154.9, 153.6, 135.5, 135.2, 134.9, 127.8, 127.5, 117.6, 113.3, 106.5, 81.1, 80.8, 65.6, 65.3, 61.4, 61.2, 49.8, 49.0, 48.1, 47.3, 46.3, 45.9, 45.4, 32.6, 32.4, 29.7, 28.4, 28.2, 25.8 (sever- al rotamers present! ). MS (m/z) calcd. for Ci5Hi8N05 [M-Boc + H]+ 292.1, found 292.1. [a]25 D +21.8° (c 0.21 EtOH). Mp 200 °C (decomp.). For 11 : White solid (62 mg, 40%). Rf 0.35 (50% EtOAc in heptane + 1% AcOH, KMn04 staining). ^ NMR (CDCI3) : δ 10.77 (broad s, 1H), 7.04 (d, J = 8.0 Hz, 1H), 6.85 (s, 1H), 6.78 (d, J = 8.3 Hz, 1H), 4.84 (s, 2H), 4.36 (broad s, 0.5H), 4.21 (d, J = 6.1 Hz, 0.5H), 3.81-3.41 (m, 3H), 2.38-2.22 (m, 1H), 2.04- 1.94 (m, 1H), 1.55 (s, 6H), 1.51 (s, 4H), 1.44 (s, 5H). 13C NMR (CDCI3) : δ 178.1, 155.3, 153.6, 150.2, 132.5, 132.0, 126.6, 126.5, 123.1, 123.0, 119.4, 117.2, 99.5, 81.1, 80.7, 65.8, 60.8, 49.3, 47.3, 46.3, 46.0, 33.0, 32.6, 28.5, 28.3, 24.8 (several rotamers present! ). MS (m/z) calcd. for Ci5H20NO4 [M-Boc + 2H]+ 278.1, found 278.0. [a]25 D +49.4° (c = 0.23, abs EtOH). Mp 128- 130 °C.
(2S,3/?)-l-(£er£-Butoxycarbonyl)-3-(3-carboxy-5- propoxyphenyl)pyrrolidine-2-carboxylic acid (10b).
Suspension A : NaI04 (1.92 g, 8.98 mmol, 9.98 equiv) was suspended in H20 (6 ml_) and after stirring at r.t. for 5 min. RuCI3 H20 (8 mg, 0.035 mmol, 0.04 equiv) was added. The black suspension was stirred for 1 min at r.t. prior to use.
Diol 9b (276 mg, 0.90 mmol, 1.00 equiv) was dissolved in CH3CN (5 ml_) and EtOAc (5 ml_), cooled to 0 °C and dropwise added suspension A. The flask containing suspension A was washed with H20 (2 ml_), which was added the mixture. The mixture was left to stir at 0 ° C for 1.5 h. The mixture was filtered through a plug of celite and the plug was afterwards washed with EtOAc (2 x 5 ml_). The organic phases were pooled in a separation funnel and added H20 (20 ml_). After separation of the two phases the aqueous phase was extracted with EtOAc (2 x 20 ml_). The pooled organic phases was washed with brine (20 ml_), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 3 cm, 30 ml_ fractions, 0 - 50% EtOAc in heptanes containing 2 % AcOH) to yield 196 mg (66%) of diacid 10b as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 1.06 (3H, t, J = 7 Hz), 1.44 (5H, s), 1.52 (4H, s), 1.83 (2H, h, J = 7 Hz), 2.09 (1H, m), 2.35 (1H, m), 3.50 - 3.86 (3H, m), 3.97 (2H, t, J = 7 Hz), 4.30 (0.6H, d, J = 7 Hz), 4.46 (0.4H, d, J = 6 Hz), 7.06 (1H, m), 7.49 (1H, m), 7.59 (1H, m), 11.0 (2H, bs). 13C NMR (75 MHz, CDCI3) : δ 28.4, 28.6, 32.7, 33.1, 46.2, 46.4, 48.1, 49.8, 65.2, 65.7, 69.9, 81.2, 81.3, 113.7, 113.8, 119.7, 119.9, 121.0, 121.1, 130.9, 131.0, 142.1, 142,6, 153.8, 155.0, 159.4, 171.4, 171.5, 176.7, 178.3; LCMS: m/z [M + H] + : calc: 393.2, found : 294.1; TLC: Rf = 0.36 (EtOAc: heptanes: AcOH (40: 20: 1)).
(2S,3/?)-3-(3-(Benzyloxy)-5-carboxyphenyl)-l-(£er£- butoxycarbonyl)pyrrolidine-2-carboxylic acid (10c).
Suspension A: NaI04 (2.40 g, 11.22 mmol, 10.0 equiv) and RuCI3
H20 (10 mg, 0.044 mmol, 0.04 equiv) were suspended in H20 (7.5 mL) and stired at r.t. for 1 min. prior to use.
Diol 9c (462 mg, 1.12 mmol, 1.00 equiv) was dissolved in CH3CN (10 mL) and EtOAc (10 mL), cooled to 0 °C and dropwise added suspension A over the course of 15 min. The flask containing suspension A was washed with H20 (7.5 mL), which was added the mixture over the course of 5 min. The mixture was left to stir at 0 °C for 2 h. The mixture was filtered through a plug of celite and the plug was afterwards washed with EtOAc (2 x 20 mL). The organic phases were pooled in a separation funnel and added H20 (40 mL). After separation of the two phases the aqueous phase was extracted with EtOAc (2 x 50 mL). The pooled organic phases was washed with brine (50 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 3 cm, 30 mL fractions, 0 - 50% EtOAc in heptanes containing 2% AcOH) to yield 410 mg (83%) of diacid 10c as a clear, colorless oil. ^ NMR (300 MHz, CDCI3) : δ 1.44 (4.5H, s), 1.52 (4.5H, s), 2.06 (1H, m), 2.34 (1H, m), 3.45 - 3.85 (3H, m), 4.28 (0.5H, d, J = 7 Hz), 4.44 (0.5H, d, J = 6 Hz), 5.10 (2H, s), 7.14 (1H, t, J = 7 Hz), 7.30-7.47 (5H, m), 7.57-7.64 (2H, m), 7.75-9.40 (2H, vbs). 13C NMR (75 MHz, DMSO) : δ 28.0, 28.2, 32.1, 32.6, 46.0, 48.0, 49.1, 65.1, 65.4, 69.4, 79.1, 113.3, 118.5, 118.7, 120.6, 120.7, 127.7, 127.8, 128.3, 132.2, 136.6, 142.7, 143.1, 152.7, 153.2, 158.4, 166.8, 172.8, 173.3; LCMS : m/z [M + H] + : calc: 441.2, found : 342.2; TLC: Rf = 0.36 (EtOAc: heptanes: AcOH (10: 5: 0.1)); OR: [a]35 D: +49.74° (c = 0.39, abs EtOH).
(2S,3 ?)-3-(3-([l,l'-Biphenyl]-3-ylmethoxy)-5-carboxyphenyl)- l-(£er£-butoxycarbonyl)-pyrrolidine-2-carboxylic acid (lOd). Suspension A : NaI04 (1.71 g, 11.46 mmol, 16.4 equiv) and R11CI3 ' H20 (8 mg, 0.035 mmol, 0.05 equiv) were suspended in H20 (6 ml_) and stired at r.t. for 1 min. prior to use.
Diol 9d (342 mg, 0.699 mmol, 1.00 equiv) was dissolved in CH3CN (8 mL) and EtOAc (8 ml_), cooled to 0 ° C and dropwise added suspension A over the course of 15 min. The flask containing suspension A was washed with H20 (6 mL), which was added the mixture over the course of 5 min. The mixture was left to stir at 0 °C for 2 h. The mixture was filtered through a plug of ce- lite and the plug was afterwards washed with EtOAc (2 x 15 mL). The organic phases were pooled in a separation funnel and added H20 (40 mL). After separation of the two phases the aqueous phase was extracted with EtOAc (2 x 50 mL). The pooled organic phases was washed with brine (50 mL), dried over MgS04, filtered, concentrated in vacuo and purified using DCVC (dia = 3 cm, 30 mL fractions, 0 - 45% EtOAc in heptanes containing 2% AcOH) to yield 250 mg (69%) of diacid lOd as a clear, colorless oil. H NMR (300 MHz, CDCI3) : δ 1.44 (4.5H, s), 1.52 (4.5H, s), 2.07 (1H, m), 2.35 (1H, m), 3.45 - 3.86 (3H, m), 4.29 (0.5H, d, J = 8 Hz), 4.45 (0.5H, d, J = 6 Hz), 5.16 (2H, s), 7.17 (1H, t, J = 2 Hz), 7.31-7.50 (5H, m), 7.54-7.69 (6H, m). 13C NMR (75 MHz, CDCI3) : δ 28.5, 28.7, 32.8, 33.1, 46.3, 46.6, 47.8, 50.0, 65.4, 65.7, 70.5, 81.3, 81.8, 114.1, 120.3, 121.7, 122.0, 126.6, 126.7, 127.2, 127.3, 127.6, 128.9, 129.2, 131.1, 136.8, 140.8, 141.8, 142.3, 142.9, 153.7, 155.6, 159.2, 171.3, 175.5, 177.4, 178.3; LCMS : m/z [M + H] + : calc: 517.2, found : 418.2; TLC: Rf = 0.40 (EtOAc: heptanes: AcOH (10 : 5 : 0.1)); OR: [a]35 D: +42.73° (c = 0.33, abs EtOH).
3-Bromo-5-(((£er£-butyldimethylsilyl)oxy)methyl)phenol (16) .
(a)
Commercially available acid 15 (10.0 g, 46.8 mmol, 1.00 equiv) was dissolved in dry THF (60 mL), under N2, cooled to 0 ° C. To this solution was added THF · BH3 complex (70 mL, 70 mmol, 1.50 equiv) over the course of 30 min (gas formation). After stirring at 0 °C for 15 min the reaction mixture was left to stir at r.t. over night. The mixture was transferred to a separation funnel containing phosphate buffer (100 mL, pH 7) and added Et20 (100 mL). The phases were separated and the aqueous phase extracted with EtOAc (2 x 75 mL). The combined organic phases was washed with brine (80 mL), dried over MgS04, filtered and concentrated in vacuo to yield a white solid, (b) The solid was dissolved in DMF (20 mL) and cooled 0 °C under N2. Imidazole (4.13 g, 60.7 mmol, 1.30 equiv) was added followed by portion wise addition of TBDMS-CI (7.95 g, 52.7 mmol, 1.13 equiv) over 1 min. The mixture was left to stir at 0 °C for 15 min and then 10 min at r.t. before DMF (10 mL) was added due to extensive formation of solid material. The mixture was left to stir at r.t. over night. The mixture was transferred to a separation funnel containing Et20 (100 mL), EtOAc (100 mL) and sat. NaHC03 (100 mL). The organic phase was washed with water (3 x 100 mL), brine (100 mL), dried over MgS04, filtered and concentrated in vacuo. The mixture was purified by DCVC (dia. = 6 cm, 50 mL fractions, 0-30% EtOAc in heptanes) three times to yield 16 as a clear, colorless oil (10.4 g, 71 %). ^ NMR (300 MHz, CDCI3) : δ 0.13 (6H, s), 0.96 (9H, s), 6.66 (2H, s), 5,64 (1H, s), 6.74 (1H, s), 6.86 (1H, s), 7.01 (1H, s). 13C NMR (75 MHz, CDCI3) : δ -5.0, 18.7, 26.2, 64.3, 112.0, 117.4, 121.5, 122.6, 144.8, 156.3; TLC: Rf : 0.20 (EtOAc: heptanes (1 : 10)).
References
(1) Meldrum, B. S. Glutamate as a Neurotransmitter in the Brain: Review of Physiology and Pathology. J. Nutr. 2000, 130, 1007-1007.
(2) Bleakman, D. ; Lodge, D. Neuropharmacology of AM PA and kai- nate receptors. Neuropharmacology 1998, 37, 1187-1204.
(3) Chittajallu, R. ; Braithwaite, S. P. ; Clarke, V. R. J. ; Henley, J. M. Kainate receptors: su bun its, synaptic localization and function. Trends in Pharmacological Sciences 1999, 20, 26-35.
(4) Lerma, J. ; Paternain, A. V. ; Rodriguez-Moreno, A. ; Lopez- Garcia, J. C. Molecular physiology of kainate receptors. Physiological Reviews
2001, 81, 971-998.
(5) Rozas, J. L ; Paternain, A. V. ; Lerma, J. Noncanonical signaling by ionotropic kainate receptors. Neuron 2003, 39, 543-53.
(6) Pinheiro, P. ; Mulle, C. Kainate receptors. Cell and Tissue Re- search 2006, 326, 457-482.
(7) Lovinger, D. M. Neurotransmitter roles in synaptic modulation, plasticity and learning in the dorsal striatum. Neuropharmacology 2010, 58, 951-961.
(8) Anggono, V. ; Huganir, R. L. Regulation of AMPA receptor traf- ficking and synaptic plasticity. Current Opinion in Neurobiology 2012, 22,
461-469.
(9) Kalivas, P. W. Cocaine and amphetamine-like psychostimulants: neurocircuitry and glutamate neuroplasticity. Dialogues in clinical neuros- cience 2007, 9, 389-97.
(10) McCarthy, D. 1 ; Alexander, R.; Smith, M. A. ; Pathak, S. ; Kanes,
S. ; Lee, C.-M.; Sanacora, G. Glutamate-based depression GBD. Medical Hypotheses 2012, 78, 675-681.
(11) Sanacora, G. ; Treccani, G. ; Popoli, M. Towards a glutamate hypothesis of depression : An emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2012, 62, 63-77.
(12) Chaki, S. ; Okubo, T.; Sekiguchi, Y. Non-monoamine-based approach for the treatment of depression and anxiety disorders. Recent Patents CNS Drug Discov. 2006, 1, 1-27. (13) Harvey, B. H. ; Shahid, M. Metabotropic and ionotropic gluta- mate receptors as neurobiological targets in anxiety and stress- related disorders: focus on pharmacology and preclinical translational models. Pharmacol Biochem Behav 2012, 100, 775-800.
(14) Riaza Bermudo-Soriano, C ; Perez-Rodriguez, M. M. ; Vaquero-
Lorenzo, C ; Baca-Garcia, E. New perspectives in glutamate and anxiety. Pharmacology Biochemistry and Behavior 2012, 100, 752-774.
(15) Michelson, D. ; Levine, L. R. ; Dellva, M. A. ; Mesters, P. ; Schoepp, D. D. ; Dunayevich, E. ; Tollefson, G. D. Clinical studies with mGluR2/3 agonists: LY354740 compared with placebo in patients with generalized anxiety disorder. Neuropharmacology 2005, 49, 257-257.
(16) Lea, P. M. th; Faden, A. I. Metabotropic glutamate receptor su btype 5 antagonists MPEP and MTEP. CNS drug reviews 2006, 12, 149-66.
(17) Vikelis, M. ; Mitsikostas, D. D. The role of glutamate and its re- ceptors in migraine. CNS Neurol. Disord. Drug Targets 2007, 6, 251-7.
(18) Javitt, D. C. Glutamatergic theories of schizophrenia. Isr J Psychiatry Relat Sci 2010, 47, 4-16.
(19) Javitt, D. C. Glutamate and schizophrenia : phencyclidine, N- methyl-D-aspartate receptors, and dopamine-glutamate interactions. Int. Rev. Neurobiol.2007, 78, 69-108.
(20) Fell, M. J. ; McKinzie, D. L ; Monn, J. A. ; Svensson, K. A. Group II metabotropic glutamate receptor agonists and positive allosteric modulators as novel treatments for schizophrenia. Neuropharmacology 2012, 62, 1473-1483.
(21) Rotaru, D. C ; Lewis, D. A. ; Gonzalez-Burgos, G. The role of glutamatergic inputs onto parvalbumin-positive interneurons: relevance for schizophrenia. Rev Neurosci 2012, 23, 97-109.
(22) Coyle, J. T. ; Basu, A. ; Benneyworth, M. ; Balu, D. ; Konopaske, G. Glutamatergic synaptic dysregulation in schizophrenia : therapeutic impl i- cations. Handb Exp Pharmacol 2012, 267-295.
(23) Almeida, A. ; Heales, S. J. ; Bolanos, J. P. ; Medina, J. M. Glutamate neurotoxicity is associated with nitric oxide-mediated mitochondrial dysfunction and glutathione depletion. Brain research 1998, 790, 209-16.
(24) Gu, B. ; Nakamichi, N. ; Zhang, W. -S. ; Nakamura, Y. ; Kambe, Y. ; Fukumori, R. ; Takuma, K. ; Yamada, K. ; Takarada, T. ; Taniura, H. ; Yone- da, Y. Possible protection by notoginsenoside Rl against glutamate neurotoxicity mediated by N-methyl-D-aspartate receptors composed of an NR1/NR2B subunit assembly. J. Neurosci. Res. 2009, 87, 2145-2156.
(25) Kumar, A. ; Singh, R. L ; Babu, G. N. Cell death mechanisms in the early stages of acute glutamate neurotoxicity. Neurosci. Res. 2010, 66,
271-278.
(26) Miao, Y. ; Dong, L.-D. ; Chen, J.; Hu, X.-C ; Yang, X.-L; Wang, Z. Involvement of calpain/p35-p25/Cdk5/NMDAR signaling pathway in gluta- mate-induced neurotoxicity in cultured rat retinal neurons. PLoS ONE 2012, 7, e42318.
(27) Danysz, W.; Parsons, C. G. Alzheimer's disease, β-amyloid, glutamate, NMDA receptors and memantine--searching for the connections. Br. J. Pharmacol. 2012, 167, 324-352.
(28) McKeage, K. Memantine: a review of its use in moderate to se- vere Alzheimer's disease. CNS Drugs 2009, 23, 881-97.
(29) Kavirajan, H. Memantine: a comprehensive review of safety and efficaOcy. Expert opinion on drug safety 2009, 8, 89-109.
(30) Francis, P. T. Glutamatergic approaches to the treatment of cognitive and behavioural symptoms of Alzheimer's disease. Neurodegener Dis. 2008, 5, 241-3.
(31) Hu, N.-W. ; Ondrejcak, T.; Rowan, M. J. Glutamate receptors in preclinical research on Alzheimer's disease: update on recent advances. Pharmacol. Biochem. Behav. 2012, 100, 855-862.
(32) Fan, M. M. ; Raymond, L. A. N-methyl-D-aspartate (NMDA) re- ceptor function and excitotoxicity in Huntington's disease. Progress in neurobiology 2007, 81, 272-93.
(33) Corona, J. C ; Tovar-y-Romo, L. B. ; Tapia, R. Glutamate excitotoxicity and therapeutic targets for amyotrophic lateral sclerosis. Expert opinion on therapeutic targets 2007, 11, 1415-28.
(34) Muir, K. W. Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Current Opinion In Pharmacology 2006, 6, 53- 60.
(35) Alexander, G. M. ; Godwin, D. W. Metabotropic glutamate receptors as a strategic target for the treatment of epilepsy. Epilepsy Research 2006, 71, 1-22. (36) Traynelis, S. F.; Wollmuth, L. P.; McBain, C. J.; Menniti, F. S.; Vance, K. M.; Ogden, K. K.; Hansen, K. B.; Yuan, H.; Myers, S. J.; Dingle- dine, R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010, 62, 405-496.
(37) Vaidyaa, A.; Jainb, S.; Jaina, A. K.; Agrawala, A.; Kashawa, S.
K.; Jaina, S. K.; Agrawala, R. K. Metabotropic Glutamate Receptors: A Review on Prospectives and Therapeutic Aspects. Mini Rev Med Chem 2012.
(38) Jane, D. E.; Lodge, D.; Collingridge, G. L. Kainate receptors: Pharmacology, function and therapeutic potential. Neuropharmacology 2009, 56,90-113.
(39) Meador-Woodruff, J. H.; Davis, K. L; Haroutunian, V. Abnormal kainate receptor expression in prefrontal cortex in schizophrenia. Neuropsy- chopharmacology 2001, 24, 545-552.
(40) Sodhi, M. S.; Simmons, M.; McCullumsmith, R.; Haroutunian, V.; Meador-Woodruff, J. H. Glutamatergic gene expression is specifically reduced in thalamocortical projecting relay neurons in schizophrenia. Biol Psychiatry 2011, 70, 646-54.
(41) Ahmad, Y.; Bhatia, M. S.; Mediratta, P. K.; Sharma, K. K.; Negi, H.; Chosdol, K.; Sinha, S. Association between the ionotropic glutamate re- ceptor kainate3 (GRIK3) Ser310Ala polymorphism and schizophrenia in the Indian population. World J Biol Psychiatry 2009, 10, 330-3.
(42) Djurovic, S.; Kahler, A. K.; Kulle, B.; Jonsson, E. G.; Agartz, I.; Le Hellard, S.; Hall, H.; Jakobsen, K. D.; Hansen, T.; Melle, I.; Werge, T.; Steen, V. M.; Andreassen, O. A. A possible association between schizophrenia and GRIK3 polymorphisms in a multicenter sample of Scandinavian origin (SCOPE). SchizophrRes 2009, 107, 242-8.
(43) Shibata, H.; Shibata, A.; Ninomiya, H.; Tashiro, N.; Fukumaki, Y. Association study of polymorphisms in the GluR6 kainate receptor gene (GRIK2) with schizophrenia. Psychiatry Res 2002, 113, 59-67.
(44) Schiffer, H. H.; Heinemann, S. F. Association of the human kainate receptor GluR7 gene (GRIK3) with recurrent major depressive disorder. American Journal of Medical Genetics Part B-Neuropsychiatric Genetics 2007, 144B, 20-26.
(45) Peng, L; Li, B.; Du, T.; Wang, F.; Hertz, L. Does conventional anti-bipolar and antidepressant drug therapy reduce NMDA-mediated neuron- al excitation by down regulating astrocytic GluK2 function? Pharmacol Bio- chem Behav 2012, 100, 712-25.
(46) Samengo, I.; Curro, D.; Navarra, P.; Barrese, V.; Taglialatela, M.; Martire, M. Molecular and pharmacological evidence for a facilitatory func- tional role of pre-synaptic GluK2/3 kainate receptors on GABA release in rat trigeminal caudal nucleus. Eur J Pain 2012, 16, 1148-57.
(47) Larsen, A. M.; Bunch, L. Medicinal Chemistry of Competitive Kainate Receptor Antagonists. ACS Chemical Neuroscience 2010, 2, 60-74.
(48) Weiss, B.; Alt, A.; Ogden, A. M.; Gates, M.; Dieckman, D. K.; Clemens-Smith, A.; Ho, K. H.; Jarvie, K.; Rizkalla, G.; Wright, R. A.; Calliga- ro, D. O.; Schoepp, D.; Mattiuz, E. L; Stratford, R. E.; Johnson, B.; Salhoff, C; Katofiasc, M.; Phebus, L. A.; Schenck, K.; Cohen, M.; Filla, S. A.; Ornstein, P. L; Johnson, K. W.; Bleakman, D. Pharmacological characterization of the competitive GLU(K5) receptor antagonist decahydroisoquinoline LY466195 in vitro and in vivo. Journal of Pharmacology and Experimental Therapeutics 2006, 318, 772-781.
(49) Larsen, A. M.; Venskutonyte , E. A.; Nielsen, B.; Pickering, D. S.; Bunch, L. Discovery of a New Class of Ionotropic Glutamate Receptor Antagonists by the Rational Design of (2S ,3R)-3-(3- Carboxyphenyl)-pyrrolidine-2-carboxylic Acid. ACS Chemical Neuroscience 2011, 2, 107-114.
(50) Bunch, L; Krogsgaard-Larsen, P.; Madsen, U. Mixed cyano- gilman cuprates - Advances in conjugate addition to alpha, beta -unsaturated pyroglutaminol. Synthesis 2002, 31-33.

Claims

P A T E N T C L A I M S
1. A compound of Formula (I)
Figure imgf000055_0001
and pharmaceutically acceptable derivatives, as well as all tautomers and stereoisomers of compound of Formula (I), wherein
Q represents compounds of Formula (la) or (lb);
Figure imgf000055_0002
— in each case may represent if appropriate the presence of at least one double bond between T2 and (Z2 or Z3), or between Z2 and (Zi or Z3), or between Zi and Ti, or between Ti and Z4; or between Z4 and T2; or
Ti is C, or CH,
T2 is C, or CH,
Zi is CR2, C(R2)2, N, S, O, or NR3,
Z2 is CR2, C(R2)2, N, S, O, or NR3,
Z3 is CR2, C(R2)2, N, S, O, or NR3,
Z4 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, and Z3 cannot represent adjacent O or S;
Ri may together with Zi or Z4, or
Z2 may together with Zi or Z3,
form a saturated or unsaturated C5- or C6-cycloalkyl, or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the satu- rated or unsaturated C5- or C6-cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR4 or R4,
Ri is H, OR4, Ci-Ce-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, COOR4, N(OH)H, or NHR4,
R2 is independently selected among R4, O, OR4, halogen, N(OH)H, N(OH)R4, NHR4, COR4, CONHR4, CN, CF3, CCI3, SH, or S02NHR4,
R3 is independently selected among R4, O, OR4, or halogen,
R4 is independently selected among H, OH, Ci-C6-alkyl, C2-C6- alkenyl, C2-C6-alkynyl, phenyl, Ci-C6-alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- heterocyclyl, wherein Ci-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, phenyl, Ci-C6- alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- heterocyclyl may be substituted with one or more substituents selected from the group comprising d-C6-alkyl, Ci-C6- alkoxy, aryl, halogen, and amine,
halogen represents CI, Br, or I, and
with the proviso that Zi, Z2, Z3 and Z4 are not all CH, and Ti and T2 are not both C, when Ri is OH.
2. A compound according to claim 1, comprising Formula (II)
Figure imgf000056_0001
(II),
wherein
Q represents compounds of Formula (Ia l) or (Ib2);
Figure imgf000057_0001
— in each case may represent if appropriate the presence of at least one double bond between T2 and (Z2 or Z3), or between Z2 and (Zi or Z3), or between Zi and Ti, or between Ti and Z4; or between Z4 and T2; or
Ti is C, or CH,
T2 is C, or CH,
Zi is CR2, C(R2)2, N, S, O, or NR3,
Z2 is CR2, C(R2)2, N, S, O, or NR3,
Z3 is CR2, C(R2)2, N, S, O, or NR3,
Z4 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, and Z3 cannot represent adjacent O or
S;
Ri may together with Zi or Z4, or
Z2 may together with Zi or Z3,
form a saturated or unsaturated C5- or C6-cycloalkyl, or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the saturated or unsaturated C5- or C6-cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR4 or R4;
Ri is H, OR4, Ci-Ce-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, COOR4, N(OH)H, or NHR4,
R2 is independently selected among R4, O, OR4, halogen, N(OH)H, N(OH)R4, NHR4, COR4, CONHR4, or S02NHR4,
R3 is independently selected among R4, O, OR4, or halogen,
R4 is independently selected among H, d-C6-alkyl, C2-C6-alkenyl, C2- C6-alkynyl, phenyl, Ci-C6-alkylphenyl, or saturated or unsaturated C5- or C6- cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6-heterocyclyl, wherein Ci-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, phenyl, Ci-C6-alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- eterocyclyl may be substituted with one or more subs- tituents selected from the group comprising d-C6-alkyl, Ci-C6-alkoxy, aryl, halogen, and amine,
halogen represents CI, Br, or I, and
with the proviso that Zi, Z2, Z3 and Z4 are not all CH, and Ti and T2 are not both C, when Ri is OH.
3. A compound according to claims 1 or 2, wherein
Q represents
Figure imgf000058_0001
Figure imgf000058_0002
wherein
Ti is C,
T2 is C,
Zi is CR2, N, S, O, or NR3,
Z2 is CR2, or N,
Z3 is CR2, or N,
Z4 is CR2, or N,
wherein the residues Zi, Z2, and Z3 cannot represent adjacent O or
S; Ri may together with Zi or Z4, or
Z2 may together with Zi or Z3,
form a saturated or unsaturated C5- or C6-cycloalkyl, or a saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, wherein the satu- rated or unsaturated C5- or C6-cycloalkyl, or the saturated or unsaturated heterocyclyl containing 5 or 6 ring atoms, may be substituted with one or more substituents selected from the group comprising OR4 or R4;
Ri is H, OR4, Ci-Ce-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, COOR4, N(OH)H, or NHR4,
R2 is R4, O, OR4, halogen, N(OH)H, N(OH)R4, NHR4, COR4, CONHR4, or S02NHR4,
R3 is R4, O, OR4, or halogen,
R4 is H, Ci-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, phenyl, Ci-C6- alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6-heterocyclyl, wherein Ci-C6-alkyl, C2-C6- alkenyl, C2-C6-alkynyl, phenyl, Ci-C6-alkylphenyl, or saturated or unsaturated C5- or C6-cycloalkyl, or saturated or unsaturated Ci-C6-alkyl C5- or C6- heterocyclyl may be substituted with one or more substituents selected from the group comprising Ci-C6-alkyl, Ci-C6-alkoxy, aryl, halogen, and amine, halogen represents CI, Br, or I, and
with the proviso that Zi, Z2, Z3 and Z4 are not all CH, and Ti and T2 are not both C when Ri is OH .
4. A compound according to any of the claims 1 to 3, wherein Ri may together with Zi or Z4, or Z2 may together with Zi or Z3, form a satu- rated or unsaturated C5- or C6-cycloalkyl or a 5- or 6- membered heterocyclyl ring, the heterocyclyl ring containing one, two, three or four heteroatoms selected from the group comprising sulfur, oxygen and nitrogen .
5. A compound according to claim 4, wherein the 5- or 6-membered ring is selected from the group consisting of pyrrolidine, pyrrole, tetrahydro- furan, furan, thiolane, thiophene, imidazolidine, pyrazolidine, imidazole, pyra- zole, oxazolidine, isoxazolidine, oxazole, isoxazole, thiazolidine, isothiazoli- dine, thiazole, isothiazole, dioxolane, dithiolane, triazoles, furazan, oxadia- zole, thiadiazole, dithiazole, tetrazole, piperidine, pyridine, oxane, pyran, thiane thiopyran, piperazine, diazines, morpholine, oxazine, thiomorpholine, thiazine, dioxane, dioxine, dithiane, dithiine, triazine, trioxane, or tetrazine, and wherein the 5- or 6-membered heterocyclic ring may be substituted with 1 to 3 substituents.
6. A compound according to claims 4 or 5
Figure imgf000060_0001
(IV),
wherein
Z5 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, Z3 and Z5 cannot represent adjacent O or S,
— , Ti, T2, Zi, Z2, Z3, Z4, R2, R3, R4 have the same meaning as giv- en under claim 1.
7. A compound according to claims 4 or 5
Figure imgf000060_0002
(V),or
Figure imgf000061_0001
wherein
Z5 is CR2, C(R2)2, N, S, O, or NR3,
Z6 is CR2, C(R2)2, N, S, O, or NR3,
Z7 is CR2, C(R2)2, N, S, O, or NR3,
wherein the residues Zi, Z2, Z3, Z5, Z6, Z7 cannot represent adjacent
O or S,
— , Ti, T2, Zi, Z2, Z3, Z4, R2, R2, R3, R4 have the same meaning as given under claim 1.
8. A compound according to any of the claims 1 to 7, wherein R2 is
OR4, halogen, N(OH)H, N(OH)R4, NHR4, COR4, CONHR4, or S02NHR4.
9. A compound according to any of the claims 1 to 8, wherein R4 is H, Ci-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, or Ci-C6-alkylphenyl, wherein Ci- C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, or Ci-C6-alkylphenyl may be substi- tuted with one to four substituents selected from the group comprising Ci-C6- alkyl, Ci-C6-alkoxy, aryl, halogen, and amine.
10. A compound according to any of the claims 1 to 5, and claims 8 or 9,
Figure imgf000062_0001
wherein
R4 has the same meaning as given under claim 1.
11. A compound according to claim 10, wherein
R4 is alkyl, benzyl, alkylbiphenyl, preferably propyl, benzyl, or 3- methyl[l,l'-biphenyl].
12. A compound according to any of the claims 1 to 11 selected from the group consisting of
(1) (2S,3R)-2-Carboxy-3-(3-carboxy-4-hydroxyphenyl)pyrrolidin-l-ium- 2,2,2-trifluoroacetate,
(2) (2S,3R)-3-(3-Carboxy-5-propoxyphenyl)pyrrolidine-2-carboxylic acid hydrochloride,
(3) (2S,3R)-3-(3-(Benzyloxy)-5-carboxyphenyl)pyrrolidine-2-carboxylic acid hydrochloride,
(4) (2S,3R)-3-(3-([l,l'-Biphenyl]-3-ylmethoxy)-5- carboxyphenyl)pyrrolidine-2-carboxylic acid, (2S,3R)-2-Carboxy-3-(3- carboxy-5-hydroxyphenyl)pyrrolidin-l-ium 2,2,2-trifluoroacetate,
(5) (2S,3R)-tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(2,2- dimethyl-4H-benzo[d][l,3]dioxin-6-yl)-5-oxopyrrolidine-l-carboxylate, (6) (2S,3R)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(3- (((tert-butyldimethylsilyl)oxy)-methyl)-5-propoxyphenyl)-5-oxopyrrolidine-l- carboxylate,
(7) (2S,3R)-tert-Butyl 3-(3-(benzyloxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((tert-butyldime- thylsilyl)oxy)methyl)-5-oxopyrrolidine-l-carboxylate, (8) (2S,3R)-tert-Butyl 3-(3-([l,l'-biphenyl]-3-ylmethoxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((tert- butyldimethylsilyl)oxy)methyl)-5-oxopyrrolidine-l-carboxylate,
(9) (2S,3R)-tert-Butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(2,2- dimethyl-4H-benzo[d][l,3]-dioxin-6-yl)pyrrolidine-l-carboxylate,
(10) (2S,3R)-tert-Butyl- 2-(((tert-butyldimethylsilyl)oxy)methyl)-3-(3- (((tert-butyldimethylsilyl)oxy)-methyl)-5-propoxyphenyl)pyrrolidine-l- carboxylate,
(11) (2S,3R)-tert-Butyl-3-(3-(benzyloxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)-2-(((tert- butyldimethylsilyl)oxy)methyl)pyrrolidine-l-carboxylate
(12) (2S,3R)-tert-Butyl- 3-(3-([l,l'-biphenyl]-3-ylmethoxy)-5-(((tert- butyldimethylsilyl)oxy)methyl)-phenyl)-2-(((tert- butyldimethylsilyl)oxy)methyl)pyrrolidine-l-carboxylate,
(13) (2S,3R)-tert-Butyl 3-(2,2-dimethyl-4H-benzo[d][l,3]dioxin-6-yl)-2- (hydroxymethyl)-pyrrolidine-l-carboxylate,
(14) (2S,3R)-tert-Butyl 2-(hydroxymethyl)-3-(3-(hydroxymethyl)-5- propoxyphenyl)pyrrolidine-l-carboxylate,
(15) (2S,3R)-tert-Butyl 3-(3-(benzyloxy)-5-(hydroxymethyl)phenyl)-2- (hydroxymethyl)pyrrolidine- l-carboxylate,
(16) (2S,3R)-tert-Butyl 3-(3-([l,l'-biphenyl]-3-ylmethoxy)-5- (hydroxymethyl)phenyl)-2-(hydroxymethyl)-pyrrolidine-l-carboxylate,
(17) (2S,3R)-l-(tert-Butoxycarbonyl)-3-(2,2-dimethyl-4-oxo-4H- benzo[d][l,3]dioxin-6-yl)pyrrolidine-2-carboxylic acid,
(18) (2S,3R)-l-(tert-butoxycarbonyl)-3-(2,2-dimethyl-4H- benzo[d][l,3]dioxin-6-yl)pyrrolidine-2-carboxylic acid,
(19) (2S,3R)-l-(tert-Butoxycarbonyl)-3-(3-carboxy-5- propoxyphenyl)pyrrolidine-2-carboxylic acid,
(20) (2S,3R)-3-(3-(Benzyloxy)-5-carboxyphenyl)-l-(tert- butoxycarbonyl)pyrrolidine-2-carboxylic acid, and
(21) (2S,3R)-3-(3-([l,l'-Biphenyl]-3-ylmethoxy)-5-carboxyphenyl)-l-(tert- butoxycarbonyl)-pyrrolidine-2-carboxylic acid.
13. A compound according to any one of the preceding claims for use as a medicament.
14. A pharmaceutical composition comprising a compound according to any one of claims 1 to 12 in combination with one or more therapeutically acceptable diluents or carriers.
15. A compound according to any one of claims 1 to 12 or a pharmaceutical composition according to claim 14 or a method for treatment of dis- eases or conditions binding one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits to obtain a beneficial therapeutic effect.
16. A method for treating a disease or disorder mediated by one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits, wherein the disease or disorder is selected from the group consisting of psychiatric diseases or neurological disorders or a disease or disorder associated with abnormal activities of one or more of the GluAl, GluA2, GluA3, GluA4, GluKl, GluK2, GluK3, GluK4, GluK5, GluN2A, GluN2B, GluN2C or GluN2D receptor subunits in a pa- tient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound according to any of claims 1 to 12, or a pharmaceutically acceptable salt or solvate thereof, optionally together with a pharmaceutically acceptable carrier.
17. A compound according to any one of claims 1 to 12 or a pharma- ceutical composition according to claims 14 to 15 or a method for treatment of disorders of the central nervous system, neuro-physiological processes such as memory, cognition; as well as neuronal plasticity and development, psychiatric diseases or neurological disorders such as depression, anxiety, addiction, pain, migraine, and schizophrenia, and neurodegenerative diseas- es; such as Alzheimer, Huntington disease, amyotrophic lateral sclerosis (ALS), cerebral stroke, and epilepsy; and diseases including aching, ADHD, Autism, Diabetes, Huntington's disease, ischemia, multiple sclerosis, Parkinson's disease (Parkinsonism), Rasmussen's encephalitis, seizures, AIDS dementia complex, amyotrophic lateral sclerosis, combined systems disease (vitamin B12 deficiency), drug addiction, drug tolerance, drug dependency, glaucoma, hepatic encephalopathy, hydroxybutyric aminoaciduria, hyperho- mocysteinemia and homocysteinuria, hyperprolinemia, lead encephalopathy, leber's disease, MELAS syndrome, MERRF, mitochondrial abnormalities (and other inherited or acquired biochemical disorders), neuropathic pain syn- dromes (e.g. causalgia or painful peripheral neuropathies), nonketotic hyper- glycinemia, olivopontocerebellar atrophy, essential tremor, Rett syndrome, sulfite oxidase deficiency, Wernicke's encephalopathy or cancer.
18. A pharmaceutical composition according to any of the claims 14 to 17 wherein the pharmaceutical composition is administered in doses of 0.5-1500 mg/day, preferably 0.5-200 mg/day, more preferably 0.5-60 mg/day, even more preferably 0.5-30 mg/day.
19. A pharmaceutical composition according to any of the claims 14 to 18 for oral, rectal, nasal, pulmonary, buccal, sublingual, transdermal or parenteral administration.
PCT/DK2014/050035 2013-02-15 2014-02-13 Pyrrolidine-2-carboxylic acid derivatives as iglur antagonists WO2014124651A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361765343P 2013-02-15 2013-02-15
US61/765,343 2013-02-15

Publications (1)

Publication Number Publication Date
WO2014124651A1 true WO2014124651A1 (en) 2014-08-21

Family

ID=50156523

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2014/050035 WO2014124651A1 (en) 2013-02-15 2014-02-13 Pyrrolidine-2-carboxylic acid derivatives as iglur antagonists

Country Status (1)

Country Link
WO (1) WO2014124651A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017007938A1 (en) * 2015-07-09 2017-01-12 Janssen Pharmaceutica Nv Substituted 4-azaindoles and their use as glun2b receptor modulators
US9745253B2 (en) 2015-03-13 2017-08-29 Forma Therapeutics, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
CN107892694A (en) * 2017-12-05 2018-04-10 河南师范大学 A kind of preparation method of the Activity of Pyrazolidinones containing caged scaffold
US9981950B2 (en) 2014-08-15 2018-05-29 Janssen Pharmaceuticals, Inc. Triazoles as NR2B receptor inhibitors
US10071988B2 (en) 2016-02-10 2018-09-11 Janssen Pharmaceutica Nv Substituted 1,2,3-triazoles as NR2B-selective NMDA modulators
US10155727B2 (en) 2014-08-15 2018-12-18 Janssen Pharmaceuticals, Inc. Pyrazoles
CN109574982A (en) * 2019-01-22 2019-04-05 安徽德信佳生物医药有限公司 A kind of method of solid acid catalysis synthesis Vilantro intermediate
US10617676B2 (en) 2016-10-06 2020-04-14 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GluN2B receptor modulators
US11008302B2 (en) 2018-04-04 2021-05-18 Janssen Pharmaceutica Nv Substituted pyridine and pyrimidines and their use as GluN2B receptor modulators
US11161846B2 (en) 2019-06-14 2021-11-02 Janssen Pharmaceutica Nv Substituted pyrazolo[4,3-b]pyridines and their use as GluN2B receptor modulators
US11214563B2 (en) 2019-06-14 2022-01-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyrazines and their use as GluN2B receptor modulators
US11447503B2 (en) 2019-06-14 2022-09-20 Janssen Pharmaceutica Nv Pyridine carbamates and their use as GLUN2B receptor modulators
US11459336B2 (en) 2019-06-14 2022-10-04 Janssen Pharmaceutica Nv Pyrazine carbamates and their use as GluN2B receptor modulators
US11530210B2 (en) 2019-06-14 2022-12-20 Janssen Pharmaceutica Nv Substituted heteroaromatic pyrazolo-pyridines and their use as GLUN2B receptor modulators
US11618750B2 (en) 2019-06-14 2023-04-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyridine amides and their use as GluN2B receptor modulators

Non-Patent Citations (57)

* Cited by examiner, † Cited by third party
Title
"Design of Prodrugs", 1985, ELSEVIER
"Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
AHMAD, Y.; BHATIA, M. S.; MEDIRATTA, P. K.; SHARMA, K. K.; NEGI, H.; CHOSDOL, K.; SINHA, S.: "Association between the ionotropic glutamate receptor kainate3 (GRIK3) Ser3l0Ala polymorphism and schizophrenia in the Indian population", WORLD J BIOL PSYCHIATRY, vol. 10, 2009, pages 330 - 3
ALEXANDER, G. M.; GODWIN, D. W.: "Metabotropic glutamate receptors as a strategic target for the treatment of epilepsy", EPILEPSY RESEARCH, vol. 71, 2006, pages 1 - 22, XP025114927, DOI: doi:10.1016/j.eplepsyres.2006.05.012
ALMEIDA, A.; HEALES, S. J.; BOLANOS, J. P.; MEDINA, J. M.: "Glutamate neurotoxicity is associated with nitric oxide-mediated mitochondrial dysfunction and glutathione depletion", BRAIN RESEARCH, vol. 790, 1998, pages 209 - 16
ANGGONO, V.; HUGANIR, R. L: "Regulation of AMPA receptor trafficking and synaptic plasticity", CURRENT OPINION IN NEUROBIOLOGY, vol. 22, 2012, pages 461 - 469
ANN M. LARSEN ET AL: "Medicinal Chemistry of Competitive Kainate Receptor Antagonists", ACS CHEMICAL NEUROSCIENCE, vol. 2, no. 2, 16 February 2011 (2011-02-16), pages 60 - 74, XP055114657, ISSN: 1948-7193, DOI: 10.1021/cn1001039 *
BLEAKMAN, D.; LODGE, D: "Neuropharmacology of AMPA and kainate receptors", NEUROPHARMACOLOGY, vol. 37, 1998, pages 1187 - 1204, XP027254554
BUNCH, L.; KROGSGAARD-LARSEN, P.; MADSEN, U: "Mixed cyano- gilman cuprates - Advances in conjugate addition to alpha,beta-unsaturated pyroglutaminol", SYNTHESIS, 2002, pages 31 - 33
CHAKI, S.; OKUBO, T.; SEKIGUCHI, Y.: "Non-monoamine-based approach for the treatment of depression and anxiety disorders", RECENT PATENTS CNS DRUG DISCOV., vol. 1, 2006, pages 1 - 27
CHITTAJALLU, R.; BRAITHWAITE, S. P.; CLARKE, V. R. J.; HENLEY, J. M.: "Kainate receptors: subunits, synaptic localization and function", TRENDS IN PHARMACOLOGICAL SCIENCES, vol. 20, 1999, pages 26 - 35, XP004156962, DOI: doi:10.1016/S0165-6147(98)01286-3
CORONA, J. C.; TOVAR-Y-ROMO, L. B.; TAPIA, R.: "Glutamate excitotoxicity and therapeutic targets for amyotrophic lateral sclerosis.", EXPERT OPINION ON THERAPEUTIC TARGETS, vol. 11, 2007, pages 1415 - 28
COYLE, J. T.; BASU, A.; BENNEYWORTH, M.; BALU, D.; KONOPASKE, G.: "Handb Exp Pharmacol", 2012, article "Glutamatergic synaptic dysregulation in schizophrenia: therapeutic implications", pages: 267 - 295
DANYSZ, W.; PARSONS, C. G.: "Alzheimer's disease, [3-amyloid, glutamate, NMDA receptors and memantne--searchng for the connections", BR. J. PHARMACOL., vol. 167, 2012, pages 324 - 352
DJUROVIC, S.; KAHLER, A. K.; KULLE, B.; JONSSON, E. G.; AGARTZ, I.; LE HELLARD, S.; HALL, H.;; JAKOBSEN, K. D.; HANSEN, T.; MELLE,: "A possible association between schizophrenia and GRIK3 polymorphisms in a multicenter sample of Scandinavian origin (SCOPE", SCHIZOPHR RES, vol. 107, 2009, pages 242 - 8, XP026002036, DOI: doi:10.1016/j.schres.2008.10.010
FAN, M. M.; RAYMOND, L. A.: "N-methyl-D-aspartate (NMDA) receptor function and excitotoxicity in Huntington's disease", PROGRESS IN NEUROBIOLOGY, vol. 81, 2007, pages 272 - 93, XP022023409, DOI: doi:10.1016/j.pneurobio.2006.11.003
FELL, M. J.; MCKINZIE, D. L.; MONN, J. A.; SVENSSON, K. A.: "Group II metabotropic glutamate receptor agonists and positive allosteric modulators as novel treatments for schizophrenia", NEUROPHARMACOLOGY, vol. 62, 2012, pages 1473 - 1483, XP028447903, DOI: doi:10.1016/j.neuropharm.2011.06.007
FRANCIS, P. T.: "Glutamatergic approaches to the treatment of cognitive and behavioural symptoms of Alzheimer's disease", NEURODEGENER DIS., vol. 5, 2008, pages 241 - 3
GU, B.; NAKAMICHI, N.; ZHANG, W.-S.; NAKAMURA, Y.; KAMBE, Y.; FUKUMORI, R.; TAKUMA, K.; YAMADA, K.; TAKARADA, T.; TANIURA, H.: "Possible protection by notoginsenoside Rl against glutamate neurotoxicity mediated by N-methyl-D-aspartate receptors composed of an NR1/NR2B subunit assembly", J. NEUROSCI. RES., vol. 87, 2009, pages 2145 - 2156
HARVEY, B. H.; SHAHID, M.: "Metabotropic and ionotropic glutamate receptors as neurobiological targets in anxiety and stress-related disorders: focus on pharmacology and preclinical translational models", PHARMACOL BIOCHEM BEHAV, vol. 100, 2012, pages 775 - 800, XP028433939, DOI: doi:10.1016/j.pbb.2011.06.014
HU, N.-W.; ONDREJCAK, T.; ROWAN, M. J.: "Glutamate receptors in preclinical research on Alzheimer's disease: update on recent advances", PHARMACOL. BIOCHEM. BEHAV., vol. 100, 2012, pages 855 - 862, XP028433946, DOI: doi:10.1016/j.pbb.2011.04.013
JANE D E ET AL: "Kainate receptors: Pharmacology, function and therapeutic potential", NEUROPHARMACOLOGY, PERGAMON PRESS, OXFORD, GB, vol. 56, no. 1, 1 January 2009 (2009-01-01), pages 90 - 113, XP025846207, ISSN: 0028-3908, [retrieved on 20080828], DOI: 10.1016/J.NEUROPHARM.2008.08.023 *
JANE, D. E.; LODGE, D.; COLLINGRIDGE, G. L: "Kainate receptors: Pharmacology, function and therapeutic potential", NEUROPHARMACOLOGY, vol. 56, 2009, pages 90 - 113, XP025846207, DOI: doi:10.1016/j.neuropharm.2008.08.023
JAVITT, D. C.: "Glutamate and schizophrenia: phencyclidine, N-methyl-D-aspartate receptors, and dopamine-glutamate interactions", INT. REV. NEUROBIOL., vol. 78, 2007, pages 69 - 108, XP009113288, DOI: doi:10.1016/S0074-7742(06)78003-5
JAVITT, D. C: "Glutamatergic theories of schizophrenia", ISR J PSYCHIATRY RELAT SCI, vol. 47, 2010, pages 4 - 16
KALIVAS, P. W.: "Cocaine and amphetamine-like psychostimulants: neurocircuitry and glutamate neuroplasticity", DIALOGUES IN CLINICAL NEUROSCIENCE, vol. 9, 2007, pages 389 - 97
KAVIRAJAN, H.: "Memantine: a comprehensive review of safety and effica0cy", EXPERT OPINION ON DRUG SAFETY, vol. 8, 2009, pages 89 - 109, XP008111981, DOI: doi:10.1517/1474033080258420
KUMAR, A.; SINGH, R. L.; BABU, G. N.: "Cell death mechanisms in the early stages of acute glutamate neurotoxicity", NEUROSCI. RES., vol. 66, 2010, pages 271 - 278, XP026903445, DOI: doi:10.1016/j.neures.2009.11.009
LARSEN, A. M.; BUNCH, L.: "Medicinal Chemistry of Competitive Kainate Receptor Antagonists", ACS CHEMICAL NEUROSCIENCE, vol. 2, 2010, pages 60 - 74, XP055114657, DOI: doi:10.1021/cn1001039
LARSEN, A. M.; VENSKUTONYTE , E. A.; NIELSEN, B.; PICKERING, D. S.; BUNCH, L.: "Discovery of a New Class of Ionotropic Glutamate Receptor Antagonists by the Rational Design of (2S ,3R)-3-(3-Carboxyphenyl)-pyrrolidine-2-carboxylic Acid", ACS CHEMICAL NEUROSCIENCE, vol. 2, 2011, pages 107 - 114, XP055114702, DOI: doi:10.1021/cn100093f
LEA, P. M. TH; FADEN, A. I: "Metabotropic glutamate receptor subtype 5 antagonists MPEP and MTEP", CNS DRUG REVIEWS, vol. 12, 2006, pages 149 - 66
LERMA, J.; PATERNAIN, A. V.; RODRIGUEZ-MORENO, A.; LOPEZ-GARCIA, J. C.: "Molecular physiology of kainate receptors", PHYSIOLOGICAL REVIEWS, vol. 81, 2001, pages 971 - 998
LOVINGER, D. M.: "Neurotransmitter roles in synaptic modulation, plasticity and learning in the dorsal striatum", NEUROPHARMACOLOGY, vol. 58, 2010, pages 951 - 961
MAITLAND JONES, JR.: "Organic Chemistry", 2000
MCCARTHY, D. J.; ALEXANDER, R.; SMITH, M. A.; PATHAK, S.; KANES, S.; LEE, C.-M.; SANACORA, G.: "Glutamate-based depression GBD", MEDICAL HYPOTHESES, vol. 78, 2012, pages 675 - 681
MCKEAGE, K.: "Memantine: a review of its use in moderate to severe Alzheimer's disease", CNS DRUGS, vol. 23, 2009, pages 881 - 97, XP008123715
MEADOR-WOODRUFF, J. H.; DAVIS, K. L.; HAROUTUNIAN, V.: "Abnormal kainate receptor expression in prefrontal cortex in schizophrenia", NEUROPSYCHOPHARMACOLOGY, vol. 24, 2001, pages 545 - 552
MELDRUM, B. S: "Glutamate as a Neurotransmitter in the Brain: Review of Physiology and Pathology", J. NUTR., vol. 130, 2000, pages 1007 - 1007
MIAO, Y.; DONG, L.-D.; CHEN, J.; HU, X.-C.; YANG, X.-L.;; WANG, Z.: "Involvement of calpain/p35-p25/CdkS/NMDAR signaling pathway in glutamate-induced neurotoxicity in cultured rat retinal neurons", PLOS ONE, vol. 7, 2012, pages E42318
MICHELSON, D.; LEVINE, L. R.; DELLVA, M. A.; MESTERS, P.; SCHOEPP, D. D.; DUNAYEVICH, E.; TOLLEFSON, G. D.: "Clinical studies with mGluR2/3 agonists: LY354740 compared with placebo in patients with generalized anxiety disorder", NEUROPHARMACOLOGY, vol. 49, 2005, pages 257 - 257
MUIR, K. W.: "Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists", CURRENT OPINION IN PHARMACOLOGY, vol. 6, 2006, pages 53 - 60, XP028058422, DOI: doi:10.1016/j.coph.2005.12.002
PENG, L.; LI, B.; DU, T.; WANG, F.; HERTZ, L: "Does conventional anti-bipolar and antidepressant drug therapy reduce NMDA-mediated neuron al excitation by downregulating astrocytic GluK2 function?", PHARMACOL BIOCHEM BEHAV, vol. 100, 2012, pages 712 - 25
PINHEIRO, P.; MULLE, C.: "Kainate receptors", CELL AND TISSUE RESEARCH, vol. 326, 2006, pages 457 - 482
RIAZA BERMUDO-SORIANO, C.; PEREZ-RODRIGUEZ, M. M.; VAQUERO-LORENZO, C.; BACA-GARCIA, E.: "New perspectives in glutamate and anxiety", PHARMACOLOGY BIOCHEMISTRY AND BEHAVIOR, vol. 100, 2012, pages 752 - 774, XP028433938, DOI: doi:10.1016/j.pbb.2011.04.010
ROBERT T. MORRISON; ROBERT N. BOYD: "Organic Chemistry"
ROTARU, D. C.; LEWIS, D. A.; GONZALEZ-BURGOS, G.: "The role of glutamatergic inputs onto parvalbumin-positive interneurons: relevance for schizophrenia", REV NEUROSCI, vol. 23, 2012, pages 97 - 109
ROZAS, J. L.; PATERNAIN, A. V.; LERMA, J.: "Noncanonical signaling by ionotropic kainate receptors", NEURON, vol. 39, 2003, pages 543 - 53
SAMENGO, I.; CURRO, D.; NAVARRA, P.; BARRESE, V.; TAGLIALATELA, M.; MARTIRE, M.: "Molecular and pharmacological evidence for a facilitatory functional role of pre-synaptic GluK2/3 kainate receptors on GABA release in rat trigeminal caudal nucleus", EUR J PAIN, vol. 16, 2012, pages 1148 - 57
SANACORA, G.; TRECCANI, G.; POPOLI, M.: "Towards a glutamate hypothesis of depression: An emerging frontier of neuropsychopharmacology for mood disorders", NEUROPHARMACOLOGY, vol. 62, 2012, pages 63 - 77
SCHIFFER, H. H.; HEINEMANN, S. F: "Association of the human kainate receptor GluR7 gene (GRIK3) with recurrent major depressive disorder", AMERICAN JOURNAL OF MEDICAL GENETICS PART B-NEUROPSYCHIATRIC GENETICS, 2007, pages 20 - 26
SHIBATA, H.; SHIBATA, A.; NINOMIYA, H.; TASHIRO, N.; FUKUMAKI, Y: "Association study of polymorphisms in the GluR6 kainate receptor gene (GRIK2) with schizophrenia", PSYCHIATRY RES, vol. 113, 2002, pages 59 - 67
SODHI, M. S.; SIMMONS, M.; MCCULLUMSMITH, R.; HAROUTUNIAN, V.; MEADOR-WOODRUFF, J. H.: "Glutamatergic gene expression is specifically reduced in thalamocortical projecting relay neurons in schizophrenia", BIOL PSYCHIATRY, vol. 70, 2011, pages 646 - 54, XP028292313, DOI: doi:10.1016/j.biopsych.2011.02.022
T. W. GREENE; P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
TRAYNELIS, S. F.; WOLLMUTH, L. P.; MCBAIN, C. J.; MENNITI, F. S.; VANCE, K. M.; OGDEN, K. K.; HANSEN, K. B.; YUAN, H.; MYERS, S. J: "Glutamate receptor ion channels: structure, regulation, and function", PHARMACOL REV, vol. 62, 2010, pages 405 - 496
VAIDYAA, A.; JAINB, S.; JAINA, A. K.; AGRAWALA, A.; KASHAWA, S. K.; JAINA, S. K.; AGRAWALA, R. K.: "Metabotropic Glutamate Receptors: A Review on Prospectives and Therapeutic Aspects", MINI REV MED CHEM, 2012
VIKELIS, M.; MITSIKOSTAS, D. D: "The role of glutamate and its receptors in migraine", CNS NEUROL. DISORD. DRUG TARGETS, vol. 6, 2007, pages 251 - 7, XP009113486, DOI: doi:10.2174/187152707781387279
WEISS, B.; ALT, A.; OGDEN, A. M.; GATES, M.; DIECKMAN, D. K.; CLEMENS-SMITH, A.; HO, K. H.; JARVIE, K.; RIZKALLA, G.; WRIGHT, R. A: "Pharmacological characterization of the competitive GLU(K5) receptor antagonist decahydroisoquinoline LY466195 in vitro and in vivo", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 318, 2006, pages 772 - 781

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE49517E1 (en) 2014-08-15 2023-05-02 Janssen Pharmaceuticals, Inc. Pyrazoles
US10323021B2 (en) 2014-08-15 2019-06-18 Janssen Pharmaceuticals, Inc. Triazoles as NR2B receptor inhibitors
US9981950B2 (en) 2014-08-15 2018-05-29 Janssen Pharmaceuticals, Inc. Triazoles as NR2B receptor inhibitors
US10155727B2 (en) 2014-08-15 2018-12-18 Janssen Pharmaceuticals, Inc. Pyrazoles
US9745253B2 (en) 2015-03-13 2017-08-29 Forma Therapeutics, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
US11919839B2 (en) 2015-03-13 2024-03-05 Valo Health, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
US10266487B2 (en) 2015-03-13 2019-04-23 Forma Therapeutics, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
US10508077B2 (en) 2015-03-13 2019-12-17 Forma Therapeutics, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
US10988441B2 (en) 2015-03-13 2021-04-27 Valo Early Discovery, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
US9963447B2 (en) 2015-07-09 2018-05-08 Janssen Pharmaceutica Nv Substituted 4-azaindoles and their use as GluN2B receptor modulators
US10377753B2 (en) 2015-07-09 2019-08-13 Janssen Pharmaceutica Nv Substituted 4-azaindoles and their use as GluN2B receptor modulators
EA033197B1 (en) * 2015-07-09 2019-09-30 Янссен Фармацевтика Нв SUBSTITUTED 4-AZAINDOLES AND THEIR USE AS GluN2B RECEPTOR MODULATORS
WO2017007938A1 (en) * 2015-07-09 2017-01-12 Janssen Pharmaceutica Nv Substituted 4-azaindoles and their use as glun2b receptor modulators
US10766880B2 (en) 2016-02-10 2020-09-08 Janssen Pharmaceutica Nv Substituted 1,2,3-triazoles as NR2B-selective NMDA modulators
US10071988B2 (en) 2016-02-10 2018-09-11 Janssen Pharmaceutica Nv Substituted 1,2,3-triazoles as NR2B-selective NMDA modulators
US10233173B2 (en) 2016-02-10 2019-03-19 Janssen Pharmaceutica Nv Substituted 1,2,3-triazoles as NR2B-selective NMDA modulators
US11207298B2 (en) 2016-10-06 2021-12-28 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GLUN2B receptor modulators
US10617676B2 (en) 2016-10-06 2020-04-14 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GluN2B receptor modulators
US11759455B2 (en) 2016-10-06 2023-09-19 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GLUN2B receptor modulators
CN107892694A (en) * 2017-12-05 2018-04-10 河南师范大学 A kind of preparation method of the Activity of Pyrazolidinones containing caged scaffold
CN107892694B (en) * 2017-12-05 2020-03-20 河南师范大学 Preparation method of pyrazolidinone compound containing bridged ring structure
US11008302B2 (en) 2018-04-04 2021-05-18 Janssen Pharmaceutica Nv Substituted pyridine and pyrimidines and their use as GluN2B receptor modulators
CN109574982A (en) * 2019-01-22 2019-04-05 安徽德信佳生物医药有限公司 A kind of method of solid acid catalysis synthesis Vilantro intermediate
CN109574982B (en) * 2019-01-22 2020-11-27 安徽德信佳生物医药有限公司 Method for synthesizing vilanterol intermediate under catalysis of solid acid
US11447503B2 (en) 2019-06-14 2022-09-20 Janssen Pharmaceutica Nv Pyridine carbamates and their use as GLUN2B receptor modulators
US11530210B2 (en) 2019-06-14 2022-12-20 Janssen Pharmaceutica Nv Substituted heteroaromatic pyrazolo-pyridines and their use as GLUN2B receptor modulators
US11618750B2 (en) 2019-06-14 2023-04-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyridine amides and their use as GluN2B receptor modulators
US11459336B2 (en) 2019-06-14 2022-10-04 Janssen Pharmaceutica Nv Pyrazine carbamates and their use as GluN2B receptor modulators
US11214563B2 (en) 2019-06-14 2022-01-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyrazines and their use as GluN2B receptor modulators
US11161846B2 (en) 2019-06-14 2021-11-02 Janssen Pharmaceutica Nv Substituted pyrazolo[4,3-b]pyridines and their use as GluN2B receptor modulators
US11993587B2 (en) 2019-06-14 2024-05-28 Janssen Pharmaceutica Nv Substituted pyrazolo-pyrazines and their use as GluN2B receptor modulators

Similar Documents

Publication Publication Date Title
WO2014124651A1 (en) Pyrrolidine-2-carboxylic acid derivatives as iglur antagonists
JP6531042B2 (en) Spirolactam NMDA receptor modulators and uses thereof
AU2009205072C1 (en) Condensed aminodihydrothiazine derivative
JP5139307B2 (en) Prolinamide derivatives as sodium channel modulators
KR102462288B1 (en) Spiro-lactam and bis-spiro-lactam NMDA receptor modulators and uses thereof
KR101282464B1 (en) Prolinamide derivatives as sodium channel modulators
EP3416960A1 (en) Novel compounds
JP5139306B2 (en) Quaternary .ALPHA.-aminocarboxamide derivatives as regulators of voltage-gated sodium channels
CA2948161C (en) Pyrrolidine gpr40 modulators for the treatment of diseases such as diabetes
KR20150110784A (en) Spiro-lactam nmda receptor modulators and uses thereof
KR20150110586A (en) Spiro-lactam nmda receptor modulators and uses thereof
CN109111385B (en) 5-HT 2A Receptor inhibitor and preparation method and application thereof
JP2024019396A (en) Spiro-lactam nmda receptor modulators and uses thereof
CA2778194C (en) 2-oxo-1-pyrrolidinyl imidazothiadiazole derivatives
JP6908623B2 (en) Tetrahydroisoquinoline derivative
KR20230127254A (en) Substituted macrocyclic compounds and related therapeutic methods
JP2023513373A (en) P2X3 modulators
KR20220127839A (en) Compounds with KHK inhibitory effect
RU2573399C2 (en) New octahydrothienoquinoline derivative, pharmaceutical composition containing derivative, and using them
JP6483105B2 (en) Piperazine derivatives and their use as medicaments
WO2014049133A1 (en) New positive allosteric modulators of nicotinic acetylcholine receptor
US11534434B2 (en) Xanomeline derivatives and methods for treating neurological disorders
KR20120101551A (en) Bicyclic thiazoles as allosteric modulators of mglur5 receptors
WO2015003723A1 (en) Substituted 4-proline derivatives as iglur antagonists
IL303036A (en) Malt-1 modulators

Legal Events

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

Ref document number: 14705966

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 14705966

Country of ref document: EP

Kind code of ref document: A1