Title of Invention

A METHOD FOR THE PREPARATION OF N-METHYL-D-ASPARTATE AND ITS DERIVATIVES

Abstract The present invention relates to a new process for the preparation of compounds of the general formulae wherein R1- R4 are, independently from each other, hydrogen, halogen, hydroxy, amino, nitro. lower-alkyl-sulfonylamido, or acetamido; R5 -R8 are, independently from each other hydrogen, lower-alkyi, halogen, trifluoromethyl or lower-alkoxy; and their pharmaceutically acceptable acid addition salts. . The compounds of formulae 1-a and I-b are NMDA (N-methyl-D-aspartate)-receptor-subtype selective blockers, which have a key function in modulating neuronal activity and plasticity which makes theiTt key players in neediating processes underlying development of CNS including learning and memory formation and function. PRICE: THIRTY RUPEES
Full Text

Case 20631
The present invention relates to a process for the preparation of compounds of the general formulae

wherein
R - R are, independently from each other, hydrogen, halogen, hydroxy,
amino, nitro, lower-alkyl-sulfonylamido, or acetamido;
c o
R -R are, independently from each other hydrogen, lower-alkyl, halogen,
trifluoromethyl or lower-alkoxy;
and their pharmaceutically acceptable acid addition salts.
Particularly, the present invention is concerned with a process for the preparation of compounds of general formula I-a.
The compounds of formulae I-a and I-b are NMDA (N-methyl-D-aspartate)-receptor-subtype selective blockers, which have a key function in modulating neuronal activity and plasticity which makes them key players in mediating processes underlying development of CNS including learning and memory formation and function.
Under pathological conditions of acute and chronic forms of neurodegeneration overactivation of NMDA receptors is a key event for triggering neuronal cell death. NMDA receptors are composed of members from two subunit famihes, namely NR-1 (8 different splice variants) and NR-2 (A to D) originating from different genes. Members from the two subunit families show a distinct distribution in different brain areas. Heteromeric combinations of NR-1 members with different NR-2 subunits result in NMDA receptors, displaying different pharmacological properties. Possible therapeutic indications for
DV/24.01.01

NMDA receptor subtype specific blockers include acute forms of neurodegeneration caused, e.g., by stroke or brain trauma; chronic forms of neurodegeneration such as Alzheimer's disease, Parkinson's disease, Huntington's disease or ALS (amyotrophic lateral sclerosis); neurodegeneration associated with bacterial or viral infections, diseases such as schizophrenia, anxiety and depression and acute/chronic pain.
The compounds of formula I and their pharmaceutically acceptable salts can be prepared by methods known in the art, e.g. in WO 95/25721, for example by processes described below, which comprises

wherein the substituents are as defined above and X signifies a leaving group, and, if desired,
b) converting the compound of formula I obtained into a pharmaceutically
acceptable acid addition salt, and, if desired,
c) converting a racemic mixture at the stage of formula III or at the stage of formula
I into enantiomeric compounds III-a, III-b


or I-a, I-b respectively, thus obtaining optically pure compounds. However, the above processes usually lead to yields of 10% or less of the desired compound due to the poor efficiency of the methods employed, i.e. resolution by crystallization of diasteromeric salts.
It has now been found that the compounds of formula I-a and I-b can be prepared more effectively and with considerably higher yield if manufactured according to the reaction scheme depicted below:


or the corresponding cis configurated enantiomer, 7b, lll-b and l-b, respectively
wherein
are as defined above;
R9 is an amino protecting group, preferably benzyl;
R10 and R10 are independently a carboxylic acid protecting group;
Y and X represent independently a leaving group; and
AZ signifies a mineral acid from the group of HBF4, H2SO4, HPFg,
HBr, HI, HCl, HSbFg or HCIO4, or a strong organic acid from the group of C1-8-alkylSO3H, picric acid, formic acid, a lower alkylcarboxylic acid such as e.g. acetic acid, propionic acid or

trifluoroacetic acid, or a dicarboxylic acid, such as e.g. oxalic acid, succinic acid, maleic acid, tartaric acid or phthalic acid.
Step 5 (asymmetric hydrogenation) and step 6 (deprotection of the ring nitrogen) can be inverted (Step 6* and step 5*).
The new process involves several key steps such as new methods for the preparation of the intermediates of formula II

and of formula III, especially by a new approach for the enantioselective preparation of the intermediate of formula III, i.e. of the intermediates of formulae

a) A new process for the preparation of the compound of formula (4) or of a salt thereof starting with a protected glycin (1) and a butyric acid derivative (2). The new process is considerably shorter and gives much higher yields than the conventionally used processes described in Helv. Chim. Acta, 1954, 20, 178; J. Am. Chem. Soc, 1948, 71, 896 or in I. Chem. Soc. Perkin Trans. 1,1998, 3673.


Y signifies a leaving group;
R10 and R10 signify independently a carboxylic acid protecting group;
R9 signifies an amino protecting group, preferably benzyl; and
AZ is as defined above.
The compound of formula (4) is then transformed according to standard procedures as depicted in reaction scheme 1, by benzylation in position 4 of the piperidine ring to form a compound of formula (5) (step 3). Subsequent decarboxylation and formation of the stable salt yields a compound of formula (6) (step 4).
b) Both the free base of formula (6 and 6bis) and its salts can be submitted to the asymmetric hydrogenation reaction, which proceeds with concomitant dynamic-kinetic resolution. However, due to the limited stability of the free base, according to the invention a salt thereof is preferentially submitted to the asymmetric hydrogenation reaction in the presence of a homogeneous chiral catalyst, a chiral diamine and an organic or an inorganic base (step 5 and step 5*, respectively).


thiophenol (8) and 2-bromoethanol and subsequent transformation with SOCI2 to obtain the highly mutagenic and unstable substituted (2-chloroethylsulfanyl)-benzene. The new and enhanced process for the preparation of intermediates of formula (II) avoids the presence of said highly mutagenic and unstable compound:

wherein X is a leaving group, preferably a halogen, and R1-R4 are as defined above.
The invention is thus concerned with a new process for the preparation of chiral compounds of formula I-a and I-b, respectively, which comprises:
Step 1, reacting a protected amino acid ester (1) with a 4-substituted butyric acid derivative (2) in the presence of a base to form an N-protected alkyloxycarbonylmethyl-amino-butyric acid derivative (3); preferred bases for the present reaction are organic bases such as triethylamine, ethyl-diisopropylamine, or inorganic bases such as K2CO3 or Na2C03. The reaction is carried out in an inert polar solvent preferably in dimethylformamide (DMF), dioxane or acetonitrile. The reaction is carried out at a temperature between 0°C and 120°C, preferably at a temperature between 40°C and 80°C.
Step 2, cyclising the protected alkoxycarbonylmethyl amino butyric acid derivative (3) in a Dieckmann condensation to yield protected alkyl 3-oxo-piperidine carboxylate (4) which is isolated as a salt fi-om a mineral acid or a strong organic acid, preferably as the hydrochloride salt. The reaction is preferably carried out in an apolar aromatic solvent such as toluene at a temperature of about 40°C to about 120°C, preferably at a temperature of about 85°C.

Step 3, benzylating the protected alkyl 3-oxo-piperidine carboxylate (4) salt. This reaction is well known in the art and can be carried out for example in the presence of a base and an appropriate solvent, such as for example with potassium-tert.-butoxyde in tetrahydrofliran (THF), with NaH in THF, NaOC2H5 in ethanol, K2CO3 in THF or in dimethylformamide (DMF).
Step 4, decarboxylation of the benzylated N-protected alkyl 3-oxo-piperidine carboxylate (5). The decarboxylation reaction is carried out by methods known in the art, for example by heating in the presence of a strong acid such hydrochloric acid, sulfuric acid and the like. The resulting salt is identified by formula (6) or (6bis).
Step 5 and 5*, asymmetric hydrogenation of the salt of formula (6) or (6bis) in presence of a ruthenium complex with a chiral diphosphine ligand, a chiral diamine and an organic or an inorganic base.
Typical of Steps 5 and 5* is the fact that the substrates (6) and (6bis) are racemic compounds which contain weakly acidic protons on the chiral carbon atoms. During the asymmetric hydrogenation the chiral catalyst converts at first only one enantiomer of (6) or (6bis). In the mean time, due to the configurational lability of the chiral center, the other enantiomer is racemized in situ by the base. Since the desired enantiomer of (6) and (6bis) to be hydrogenated is generated continuously from the undesired one, finally 100% yield of the single desired enantiomer of the products (7a) or (Ill-a) can be obtained.
It has been found that the salt of the piperidine derivative of formula (6) is stable and can be hydrogenated in high optical and chemical yields by the process according to the invention. The hydrogenation may also be performed with the salt of the unprotected piperidine derivative of formula (6bis).
The asymmetric hydrogenation is carried out in presence of a ruthenium phosphine complex represented by the formula
Ru(E)2(L)(A) IV
wherein
E is a halogen atom;
L is a chiral diphosphine ligand; and
A is a chiral diamine.
Complexes of type IV can be specifically prepared, isolated and characterized in analogy to the methods described in Angew. Chem. Int. Ed. 1998, 37, 1703-1707 and in the references cited therein, or can be prepared "in situ" from components as described in

above mentioned reference, and be employed without intermediate isolation in the catalytic asymmetric hydrogenation. When the complexes of type IV are prepared "in situ", the amount of chiral diphosphine ligand (L) used in the reaction can vary from 0.5 to 2.5 equivalents relative to ruthenium, preferably from 0.8 to 1.2 equivalents. Analogously the amount of chiral diamine can vary from 0.5 to 2.5 equivalents based on the amount of the ruthenium-complex, preferably 1 to 2 equivalents.
Suitable chiral diphosphine Ugands are known in the art. Such ligands are for example atropisomeric biphenyl-phosphine or binaphthyl-phosphine derivatives. Further ligands which may be useful in the present hydrogenation are l,2-bis(2,5-dimethylphospholano)benzene as described in USP 5,171,892; l-[2-(diphenylphosplvno)-ferrocenyl]ethyl-di-tert.-butyl-phosphine as described in EP 0 564 406; l-[2-(di-(4-trifluoromethyl)phenyl)-phosphino)ferrocenyl]ethyl-di-phenyl-phosphine as in EP 0 646 590; 4,12-bis(diphenylphosphino)-[2.2]paracyclophane (Tetrahedron Letters 1998, 39,4441-4444); 4,4'-Bisdiphenylphosphine-2,2',5,5'-tetramethyl-3,3'-dithiophene (WO 96/01831); 2,2'-Bis-(diphenylphosphinyl)-3,3'-dibenzo[b]thiophene (WO 96/01831); (2R,2'R)-Bis(diphenylphosphino)-(lR,rR)-dicyclopentane and enantiomer (WO 97/47633); l,2-Bis{(lR,2R,4R,5R)-2,5-bis-isopropyl-8-phenylphosphabicyclo[2.2.1]-heptyl}benzene and enantiomer (WO 97/47633); 2,2',3,3'-Tetraphenyl-4,4',5,5'-tetra-methyl-6,6'-bis-phosphanorborna-2,5-dienyl (Chem Eur Journal 1997, 3, 1365-1369); (aR-aR')-2,2'-Bis(a-N,N-dimethylaminopropyl)-(S,S)-l,r-bis(diphenylphosphino)-ferrocene and enantiomer (Tetrahedron: Asymmetry 1999,10, 375-384); and ((5,6),(5',6')-Bis(methylenedioxy)biphenyl-2,2'-diyl)bis(diphenylphosphine) (EP 850945).



Above-mentioned diphosphine ligands are known in the art and can be prepared for example as described in EP 0 398 132 and WO-92/16535 (MeOBIPHEP; 3,5-iPr-MeOBIPHEP), in EP 0 104 375 (BIPHEMP) and in EP 0 580 331 (BINAP).
In order to obtain high yields of the cis-configurated product of formula Ill-a and Ill-b, respectively, in high optical purity it is essential that the reaction is carried out in the presence of a chiral diamine which is in "unlike" configuration to the chiral complex, i.e. it is important that the diphosphine is (S) and the diamine is (R,R) or that the diphosphine is (R) and the diamine is (S,S). The reaction is carried out in presence of chiral diamines as depicted below:


Further suitable chiral diamines are propane- and butanediamines. An especially preferred chiral diamine is DPEN (V), (R,R) or (S,S)-l,2-diphenylethylenediamine. The chiral diamines are commercially available or can be prepared according to known methods.
The hydrogenation is preferably carried out in an organic solvent which is inert under the reaction conditions. As such solvents there can be mentioned, in particular, lower alcohols such as e.g. methanol, ethanol or isopropanol, trifluoroethanol or mixtures of such alcohols with halogenated hydrocarbons such as methylene chloride, chloroform, hexafluorobenzene and the like or with ethers such as diethyl ether, tetrahydrofuran or dioxane. Preferred solvent for the reaction are lower alcohols, especially preferred is isopropanol. The reaction is carried out at a concentration of about 1 to 50%, ideally about 5 to 30%.
The substrate-to-catalyst molar ratio (S/C ratio) is 10-roOO'OOO, preferably 100-SOO'OOO. The hydrogenation is carried out at a pressure of 10^ -10^ Pa, ideally at a pressure of about 105 to 107 Pa and at a temperature of about 0°C to about 50°C, ideally at 20°C to 40°C.
The amount of base present in the reaction is 1.0 - 0.001, preferably 0.05 - 0.2 mol equivalents with respect to the substrate in addition to 1 mol equivalent of base which is necessary to neutralize the acid salt of the substrate of formula (6) or (6bis), respectively.
Preferred bases used in the asymmetric hydrogenation are for example inorganic or organic bases. Preferred inorganic bases are alkali or alkaline earth metal hydroxides.

carbonates, hydrogenocarbonates, alcoholates or silanolates such as for example LiOH, LiOCHj, LiOC2H5, LiOCH(CH3)2, LiOC(CH3)3, NaOH, NaOCHj, NaOCaHj, NaOCH(CH3)2, NaOC(CH3)3, KOH, KOCH3, KOC2H5, KOCH(CH3)2, KOC(CH3)3. KOSi(CH3)3, or CS2CO3 preferred inorganic bases are alcoholates, especially KOC(CH3)3. Preferred organic bases are tertiary amines such as triethylamine, ethyl-diisopropylamine, tripropylamine and the like.
The asymmetric hydrogenation of step 5 can be carried out either batchwise or in a continuous manner.
Step 6, deprotection of the compound of formula (7a) or the isomer (7b) under standard conditions depending on the N protecting group , for example by hydrogenation of the N-benzylated compound in presence of Pd/C to form the unprotected amine Ill-a and Ill-b, respectively.
Step 6*, refers to the deprotection of the compound of formula (6) in analogy to step 6, to compound (6bis) which is then subsequently submitted to the asymmetric hydrogenation (step 5* discussed above).
Step 7, treating the sulfone intermediate of formula II with a base to form the corresponding vinyl sulfone derivative which is subsequently reacted with the piperidine derivative of formula Ill-a or Ill-b to the desired product of formula I-a or I-b, respectively. The reaction is carried out in presence of bases such as triethylamine in solvents such as CH2CI2.
As already mentioned previously sulfones of formula II are usually prepared by halogenation of the hydroxy thioether of formula (9) to form the highly toxic and unstable chlorothioether which is then subsequently oxidized to form the intermediate of formula II. The invention now provides a new process to produce this intermediate of formula II, see Scheme 4 which avoids the formation of highly toxic intermediates. The process according to the invention consists of the following steps:
step a), a substituted thiophenyl derivative (8) is deprotonated in presence of a strong inorganic or organic base and subsequently reacted with 2-haloethanol to form the thioether of formula (9);
step b), oxidation of the thioether (9) in presence of an oxidative agent such as 3-chloroperbenzoic acid (MCPBA), H2O2/ACOH, KMn04, tBuOOH, NMO/OSO4, or oxone® to yield the corresponding sulfone of formula (10); and
Step c), replacement of the hydroxy group of the sulfone derivative by a halogen atom e.g. with SOX'2. wherein X' is halogen, e.g. chlorine, bromine or iodine in the presence of pyridine in dichloromethane.

The term "pharmaceutically acceptable acid addition salts" embraces salts with inorganic and organic acids, such as hydrochloric acid, nitric acid, sulfuric acid, lactic acid, phosphoric acid, citric acid, formic acid, fumaric acid, maleic acid, acetic acid, succinic acid, tartaric acid, methane-sulfonic acid, p-toluenesulfonic acid and the like.
As used herein, the term " alkyl" refers to both straight and branched chain saturated hydrocarbon groups having 1 to 10 carbon atoms; whereas the term "lower alkyl" refers to both straight and branched chain saturated hydrocarbon groups having 1 to 5 carbon atoms, for example, methyl, ethyl, n-propyl, isopropyl, tert.-butyl and the like.
The term "lower-alkyl-sulfonylamido" refers to sulfonamido groups substituted with "lower alkyl" groups as defined above.
The term "cycloalkyl" refers to a cyclic hydrocarbon group having 3 to 7 carbon atoms. Such groups are for example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
The term "optionally substituted phenyl" refers to unsubstituted phenyl or mono, di-or trisubstituted phenyl groups with substituents such as lower alkyl, alkoxy groups or halogenated alkyl such as trifluoromethyl, pentafluoroethyl, or substituted with halogen, hydroxy, amino, dialkylamino or acetamido, or substituted with phenyl or trialkylsilyl and the like.
As used herein "heterocyclic ring" refers to 5 or 6-membered rings containing one or two hetero atoms chosen from O, S and N. Examples of preferred heterocyclic rings are furane, thiophene, pyrrole, pyridine and pyrimidine. The heterocyclic rings may be unsubstituted or substituted with substituents as defined for "substituted phenyl".
As used herein the term "leaving group" refers to conventionally used easily substituted functional groups such as halogen, e.g. chlorine, bromine or iodine, or organic acid residues such as sulfonic acid derivatives, e.g. p-toluene sulfonate, brosylate, methylsulfonate, triflate (trifluoromethylsulfonate) and the like.
Nucleophilic substitution of the leaving groups (steps 1 and 7) are carried out by methods known in the art, e.g. in inert organic solvents under basic conditions. "Inert organic solvents" refers to polar solvents such as dimethylformamide (DMF), dimethyl-acetamide (DMA), dimethylsulfoxide (DMSO), or to alcohols such as methanol, ethanol or isopropanol, or to cyclic ethers such as dioxane or tetrahydrofurane (THF), or to halogenated hydrocarbons such as dichloromethane, or to aromatic hydrocarbons such as toluene, or to nitriles, such as acetonitrile, or to mixtures of the named solvents. As bases are used inorganic or organic bases. Preferred inorganic bases are alkali or alkaline earth

metal hydroxides, carbonates, hydrogenocarbonates, alcoholates or silanolates. Preferred organic bases are tertiary amines such as triethylamine, ethyl-diisopropylamine, tripropylamine and the Uke.
The term "amino protecting group" refers in the scope of the present invention to groups such as those employed in peptide chemistry for example to benzyl, tert.-butoxy-carbonyl, allyloxy carbonyl and the like; to a substituted alkoxycarbonyl group such as trichloroethoxycarbonyl etc.; to an optionally substituted aralkyloxycarbonyl group, for example, p-nitrobenzyloxycarbonyl or benzyloxycarbonyl; to an aralkyl group such as trityl or benzhydryl; to an alkanoyl group such as formyl or acetyl; to a halogen-alkanoyl group such as chloroacetyl, bromoacetyl, iodoacetyl or trifluoroacetyl; or to a silyl protective group such as the trimethylsilyl group. Especially preferred amino protecting are benzyl, tert.-butoxycarbonyl (BOC) and benzyloxycarbonyl (Z). For the hydrogenation step (step 5) to be highly enantioselective it is essential that the protected nitrogen is basic, therefore benzyl is an especially preferred protecting group.
The term "carboxylic acid protecting group" refers in the scope of the present invention to protecting groups conventionally used to replace the acidic proton of a carboxylic acid. Examples of such groups are described in Greene, T., Protective Groups in Organic Synthesis, Chapter 5, pp. 152-192 (John Wiley and Sons, Inc. 1981). Preferably these examples include methoxymethyl, methylthiomethyl, 2,2,2-trichloroethyI, 2-haloethyl, 2-(trimethylsilyl)ethyl, methyl, ethyl, isopropyl, tert.-butyl, allyl, benzyl, triphenylmethyl (trityl), benzhydryl, p-nitrobenzyl, p-methoxybenzyl, trimethylsilyl, triethylsilyl, tert.-butyldimethylsilyl, i-propyl-dimethylsilyl. Preferred are benzhydryl, tert.-butyl, p-nitrobenzyl, p-methoxybenzyl and allyl. Especially preferred carboxylic protecting groups are methyl, ethyl, tert. butyl or benzyl.
Suitable protecting groups and methods for their cleavage will be familiar to any person skilled in the art, although of course there can be used only those protecting groups which can be cleaved off by methods under the conditions of which other structural elements in the compounds are not affected.
The term "oxygen acid or complex acid" signifies in the scope of the present invention acids from the group H2SO4, HCIO4, HBr04, HIO4, HNO3, H3PO4, CF3SO3H or C6H5SO3H as well as halogen complexes with the elements boron, phosphorus, arsenic, antimony or bismuth. HCIO4, CF3SO3H, HPFe, HBF4, HB(Ph)4, HB(3,5-(CF3)2-C6H3)4, HSbFe and HAsFe are preferred representatives with HSbF6 and HBF4 being most preferred.
The following abbreviations are used in the description of the examples:





R ' and R ' are independently a carboxylic acid protecting group; and
Y represents a leaving group;
to a compound of formula (3) in the presence of a base, which is triethylamine, ethyl-diisopropyl-amine, K3CO3 or NaiCO3, and in a solvent which is DMF, dioxane or acetonitrile; and
b) cyclising the obtained protected alkoxycarbonylmethyl amino butyric acid derivative

wherein the symbols are as defined above; to a compound of formula (4) in an apolar aromatic solvent, which is toluene, and
c) ben/.ylating the protected alkyl 3-oxo-piperidine carhoxylate (4) salt
wherein
AZ signifies a mineral acid or a strong organic acid
to a compound of formula (5) in the presence of a base and a solvent, which are potassium-tert-butox)'de in THF, NaH in THF, NaOC:H5 in ethanol or K2CO3 in THF,
and
d) decarbox\'lating the benzylated protected alkyl 3-oxo-piperidine carboxylate (5)

wherein the symbols are as defined above; to a compound of formula (6) or (6bis) in presence of a strong acid which is hydrochloric acid or sulfuric acid; and



wherein
R - R' are as defined above; and
X is a leaving group;
which is previously treated with a base, which is triethylamine in CH2CI2, to form the corresponding reactive vinyl suifone intermediate

in the presece of a base to obtain the desired compound of formula I.
The invention will now be described more in detail with reference to embodiments given by way of example in which:

Example 1 (step 1) Preparation of 4-(benzyl-ethoxycarbonylmethyl-amino)-butyTic acid ethyl ester

Example 1.1
A solution of 4.9 ml (25.8 mmol) N-benzylglycine ethyl ester and 7.7 ml (51.7 mmol) ethyl-4-bromobutyrate in 40 ml dimethylformamide (DMF) was treated at room temperature with 9.0 ml (64.3 mmol) triethylamine. The reaction mixture was heated to 65°C and stirred for 23 h, subsequently cooled to room temperature and DMF was evaporated. The residue was treated with 100 ml water and 100 ml ethyl acetate. The phases were separated after extraction, the organic phase washed twice with total 100 ml water and the combined organic phase dried over Na2SO4. The solvent was removed under reduced pressure and the residue purified by chromatography over silica gel (hexane: ethyl acetate = 9:1) to yield 7.1 g (88.8%) product as yellow oil. MS(ISP):308(100, [M + H]+)
Example 1.2
To a solution of 2.571 (17.25 mol) ethyl-4-bromobutyrate in 101 dioxane was added at 100°C 1.72 kg (8.54 mol) N-benzylglycine ethyl ester. The reaction mixture was treated under reflux dropwise over a period of 6 h with 3.101 (22.24 mol) triethylamine and subsequently stirred under reflux for 16 h. The suspension was cooled to 50°C and treated with 101 toluene, stirred at 0°C for 1 h and afterwards filtered. The filtrate was concentrated to yield 3.08 kg crude product.
Example 2 (Step 2):
Preparation of rac-ethyl N-benzyl-3-oxo-4-piperidinecarboxylate hydrochloride


Example 2.1
A solution of 2.8 g (9.1 mmol) 4-(benzyl-ethoxycarbonylmethyl-amino)-butyric acid ethyl ester in 18 ml toluene was treated at room temperature with 980.0 mg (13.7 mmol) sodium ethoxide. The reaction mixture was heated to 85°C and stirred for 3.5 h. After cooling to room temperature the reaction mixture was poured onto 50 ml ice-water, treated with 50 ml toluene and subsequently extracted. The aqueous phase was extracted with total 100 ml ethyl acetate, the organic phase washed twice with total 100 ml water and the combined organic phase dried over Na2SO4. The solvent was removed under reduced pressure to yield 2.2 g crude product. This crude product was treated with 3.7 ml of a solution of HCl in methyl alcohol 20%-weight and subsequently the solvent was rempved under reduced pressure to yield 2.4 g crude product as white crystals. The crystals were dissolved under reflux in 30 ml isopropanol and the solution cooled to room temperature and stirred at this temperature for 2 h. The formed crystals were separated on a filter funnel and washed with 3 ml isopropanol (4°C) and dried to yield 1.8 g (67.5%) product as white crystals. MS(ISP):262(100, [M + H]+).
Example 2.2
A solution of 2.02 kg (5.60 mol) crude 4-(benzyl-ethoxycarbonylmethyl-amino)-butyric acid ethyl ester from Example 1.2. in 10 1 toluene was treated at room temperature with 0.79 kg (11.03 mol) sodium ethoxide (exothermic). The reaction mixture was heated to 85°C and stirred for 3.5 h. The so formed suspension was cooled to room temperature and treated with 5 1 toluene and 0.5 kg dicalite speedex. After neutralization by slowly addition of 0.7 1 acetic acid the suspension was filtered. The filtrate was concentrated to a volume of 9 1 and treated with 1.41 (6.86 mol) HCl in ethanol (4.9 M). After formation of crystals the ethanol was exchanged under reduced pressure by addition of 8 1 toluene. The so formed suspension was treated with 5 1 toluene, stirred at 0°C for 16 h and subsequently filtered. The crystals were dried to yield 1.62 kg (94%) product.
Example 3 (Step 3):
Preparation of rac-l,4-dibenzyl-3-oxo-piperidine-4-carboxylic acid ethyl ester


A mixture of 38.3 g (341.0 mmol) potassium tert.-butoxide and 625 ml absolute tetrahydrofiiran was stirred at room temperature for 0.5 h. The resulting milky solution was cooled to 0°C and then 50.0 g (168.0 mmol) ethyl N-benzyl-3-oxo-4-piperidine carboxylate hydrochloride were added via a powder dropping funnel. The temperature was kept below 5°C. The mixture was warmed up to room temperature and further stirred for 1 h resulting in a yellow solution. After cooling to 0°C, a solution of 30.2 g (176.0 mmol) benzyl bromide in 20.0 ml of absolute tetrahydrofiiran was dropwise added in 0.5 h. A maximum temperature of 2°C was observed. The reaction mixture was warmed to room temperature and stirred for 4 h. The reaction solution was cooled to 0°C and 200 ml saturated ammonium chloride solution was slowly added. After extraction and phase^ separation, the aqueous phase was extracted twice with 100 ml of ethyl acetate. The combined organic phase were washed twice with 100 ml of saturated sodium chloride solution, dried over Na2SO4, the solvent evaporated under reduced pressure and the residue dried to yield 58.3 g (99.3%) crude product. MS (ISP): 352 (100, [M + H]""), 174 (15).
Example 4 (Step 4):
Preparation of rac-l,4-dibenzyl-3-oxo-piperidine hydrochloride

A solution of 118.0 g (336 mmol) crude rac-l,4-dibenzyl-3-oxo-piperidine-4-carboxylic acid ethyl ester in 118.0 ml of absolute ethanol was cooled to 0°C and subsequent 405 ml (4.9 mol) 37% hydrochloric acid was cautiously added. The reaction temperature was kept below 7°C. Finally the mixture was heated under reflux for 19 h. To the dark brown solution were added some crystals of rac-l,4-dibenzyl-3-oxo-piperidine hydrochloride, then the mixture was allowed to cooled to room temperature and further stirred for 2 h. The resulting crystals, after recuperation on a Buchner funnel and washing twice with 60 ml of deionized water, were dried to yield 102.2 g crude product. Then 400 ml of ethyl acetate was added to the crude product and the mixture refluxed for 2 h and cooled afterwards to room temperature. The resulting beige suspension was filtered, the crystals were washed twice with 50 ml of ethyl acetate and dried to yield 82.2 g (78 % over two steps) product.
MS (ISP): 280 (100, [M + H]^), 262 (9). M.p. 202 - 203°C.

Example 5 (Step 5): Preparation of cis-l,4-dibenzyl-3-hydroxy-piperidine

Example 5.1
In the glove box (O2 content a) A 25-mg sample was dissolved in 0.8 ml of pyridine and silylated with 0.2 ml of commercial N,0-bis-(trimethylsilyl)-acetamide (BSA)+5% trimethylchlorosilane (TMS)-solution. Gas chromatographic analysis on a Permaphase PVMS/54 column showed complete conversion and a cis/trans ratio of 99:1. Retention times: 10.65 min (trans-l,4-dibenzyl-piperidin-3-ol), 10.80 min (cis-l,4-dibenzyl-piperidin-3-ol), 11.15 min (rac-l,4-dibenzyl-piperidin-3-one).
b) ca. 0.6 ml sample of reaction mixture was taken up in water/ethyl acetate mixture and treated with a 5% ammonium chloride solution. The organic phase was dried (Na2SO4). HPLC analysis of an aliquot containing ca. 5-10 mg of product confirmed that the cis/trans-ratio was 99:1 and showed the enantiomeric purity (ee) of (S,S)- cis-l,4-dibenzyl-piperidin-3-ol to be 91%. Column: 2 x Chiralpak AS, (250 x 4.6 mm), Daicel Chemicals Industries, Cat. No. 20025; Mobile phase: 1% (v/v) ethanol in n-

hexane; flow 1.2 ml/min; pressure 6x10^ Pa. The following retention times were observed: 11.2 min cis-(R,R)-product; 12.5 min cis-(S,S)-product; 17.5 and 19.5 min starting material (two enantiomers), 17.5 and 19.5 min trans-product (two enantiomers).
Example 5.2 (scaled-up)
In the glove box (O2 content Examples 5.3-5.12
The experiments in Table 1 have been carried out in analogy to Example 5.1 using various
chiral diphosphines with S-configuration.^


In addition to 1 molar equivalent for the neutralization of the hydrochloride. Ruthenium complex prepared in situ from [RuCl2(diphosphine)(DMF)n] + DPEN. of the cis-product with (S,S)-configuration.
Ruthenium complex prepared in situ from [RuCl((S)-BIPHEMP)(benzene)]Cl and (R,R)-DPEN
Examples 5.13-5.15
The experiments in Table 2 have been carried out under following conditions: 185-ml autoclave, 1.3 g scale, c = 5% in isopropanol, 20°C, 4x10^ Pa hydrogen. The ruthenium catalyst has been prepared in the glove box by stirring the complex [RuCl2((S)-(3,5-Xyl)-MeOBIPHEP)(DMF)n] with 1 molar equiv. of chiral diamine at room temperature for 1 h.


ee of the cis-product with (S,S)-configuration; ' S/base = 10. Example 5.23
In a manner analogous to Example 5.1,41.06 g of rac-l,4-dibenzyl-piperidin-3-one hydrochloride was asymmetrically hydrogenated in the presence of 1.4 mg of [RuCl2((S)-3,5-iPr-MeOBIPHEP)((R,R)-DPEN)], S/C= lOO'OOO for 16 hours to afford after work-up (S,S)- cis-l,4-dibenzyl-piperidin-3-ol of 97% ee.
Example 5.24
In a manner analogous to Example 5.23, but at a temperature of 30°C, 41.06 g of rac-1,4-dibenzyl-piperidin-3-one hydrochloride was asymmetrically hydrogenated in the presence of 3.0 mg of [Rua2((S)-3,5-iPr-MeOBIPHEP)((R,R)-DPEN)], S/C= SO'OOO, to afford after work-up (S,S)- cis-l,4-dibenzyl-piperidin-3-ol of 96% ee.
Examples 5.25-5.28
The experiments in Table 4 have been carried out under the following conditions: 30 ml autoclave, 0.45 g of rac-l,4-dibenzyI-piperidin-3-one, c = 10% in isopropanol, 20°C, 4x10* Pa hydrogen. Catalyst: [RuCl2((S)-3,5-iPr-MeOBIPHEP)((R,R)-DPEN)], S/C= 300, S/base = 5 (in addition to 1 molar equivalent for the neutralization of the hydrochloride).



Preparation of (3S, 4S)-4-benzyl-piperidine-3-ol (work-up after hydrogenation and deprotection of the amino group):
Example 6.1
The reaction mixture from Example 5.1 was treated with 300 ml ethyl acetate, 170 ml water, 50 ml 5% aqueous NH4CI solution and solid NaCl to saturate the aqueous phase. After phase separation, the aqueous phase was extracted twice with 200 ml ethyl acetate and the organic phases were washed twice with 150 ml brine. The combined organic phase was dried over Na2SO4, the solvent evaporated under reduced pressure to yield 35.6 g crude material as light-yellow crystals. This material was dissolved in 400 ml ethanol, treated at room temperature with 6.7 g (6.3 mmol) Pd/C 10% and stirred under H2 at 55°C for 2 h. Subsequent filtration and evaporation of the solvent yielded 25.3 g crude product as amorphous material. This crude product was dissolved at 100°C in 100 ml toluene, cooled to 65°C and treated with 125 ml hexane. The so formed suspension was cooled to 35°C and treated again with 125 ml hexane. The suspension was stirred for 48 h at 0°C and filtered afterwards to yield 10.0 g (ee = 99.8%) product as white crystals. The mother liquor (13.9 g) was dissolved at 90°C in 45 ml toluene treated at 55°C with 55 ml hexane, cooled to 45°C and added again 55 ml hexane. The suspension was stirred 16 h at room temperature and 3 h at 0°C, filtered to yield 9.7 g (ee = 99.9%) product as white crystals. (Overall yield 81.5%)
MS (EI): 191 (100, [M]), 118 (76), 91 (44), 30 (100). M.p. 91.5-92.5°C.
Example 6.2 (work-up after hydrogenation, deprotection of the amino group and crystallization in presence of a resolving agent)
The reaction mixture from Example 5.1 was treated with 10 ml 5% aqueous NH4CI solution and concentrated under reduced pressure to a total volume of 50 ml. This residue

was treated with 50 ml water, 16 g NaCl, 50 ml 5% aqueous NH4CI solution and extracted twice with 100 ml ethyl acetate. The organic phases were washed twice with 100 ml brine, the combined organic phase was dried over Na2SO4 and the solvent evaporated under reduced pressure. The residue was dissolved in 105 ml ethanol, treated at room temperature with 1.8 g (1.7 mmol) Pd/C 10% and stirred under H2 at 55°C for 2.5 h. Subsequent filtration and evaporation of the solvent yielded 6.2 g crude product as yellow oil. The crude product was dissolved in 180 ml methanol and treated at 65°C with a solution of 6.2 g (15.73 mmol) of (+)-di-0,0'-p-tolyl-D-tartaric acid in 25 ml methanol. The suspension was cooled to room temperature, stirred 48 h at this temperature, cooled to 0°C and stirred for 2 h. The suspension was filtered to yield 9.2 g (ee = 98.9%) salt as white crystals. 9^09 g of this product was dissolved in 460 ml methanol, stirred 1 h under reflux, cooled slowly to room temperature and stirred for 16 h at room temperature The suspension was cooled to 0°C, treated with 460 ml diethyl ether and stirred for 3 h. The suspension was filtered to yield 8.65 g (72.5 %) salt. 4.02 g of this salt was dissolved in 40 ml CH2CI2 and treated with 20 ml aqueous NaOH IN. After extraction and phase separation, the aqueous phase was extracted twice with 20 ml CH2CI2 and the combined organic phase dried over Na2SO4. The solvent was removed under reduced pressure to yield 2.0 g (100%, ee = 99.6%) product. Overall yield was 72.5%.
Example 7 (Step 7):
Preparation of (3S, 4S)-4-benzyl-l-[2-(4-hydroxy-benzenesulfonyl)-ethyl]-piperidin-3-ol

Example 7.1
A solution of 5.5 g (24.4 mmol) 4-(2-chloro-ethane-sulfonyl)-phenol in 50 ml CH2CI2 was treated at 37°C with 3.7 ml (26.4 mmol) triethylamine and stirred for 3.25 h. Afterwards a solution of 4.5 g (22.0 mmol) (3S, 4S)-4-benzyl-piperidin-3-ol in 40 ml CH2CI2 was added over a time period of 15 min. The reaction mixture was stirred at 37°C for 3 h, cooled to room temperature treated with 80 ml water and solid NaCl. After extraction and phase separation the water phases were extracted 3 times with 70 ml CH2CI2. The combined organic phase was dried over Na2SO4, concentrated to a volume of 70 ml, treated with 75 ml toluene and concentrated to a volume of ca. 100 ml. After 3 d at 0°C the suspension was concentrated to 50 ml and filtered to yield 9.2 g crude product as white crystals. Chromatographic purification on Si02 (CH2Cl2/tert.-butylmethyl ether (tBME)= 19/1) yielded 7.2 g ( 87.1%) product as white powder.

MS (ISP): 398 (8, [M + Na]^). 376 (100, [M + H]+), 358 (12). M.p. 155.5-156.2°C.
Example 7.2
As in example 7.1. but instead of chromatographic purification, the crude product is
purified by crystallization (dissolving in methanol, afterwards exchange methanol by
toluene).
Example 8 (step a of Scheme 4) Preparation of 4-(2-hydroxy-ethylsulfanyl)-phenol

A solution of 5.0 g (35.7 mmol) 4-mercaptophenol in 50 ml methyl alcohol was treated at -5°C dropwise over a period of 30 min with 39.2 ml (39.2 mmol) aqueous NaOH IN and stirred Ih at -5°C. Afterwards a solution of 5.2 ml (39.2 mmol) 2-bromo ethanol in 16.5 ml methyl alcohol was added at -5°C dropwise over a period of 15 min. The reaction mixture was stirred for 21 h at room temperature, concentrated and the residue treated with 10 ml water and 30 ml tBME. After extraction and phase separation, the organic phase was washed with 20 ml saturated NaHC03 and 20 ml brine. The combined organic phases were dried over Na2SO4 and the solvent was removed under reduced pressure to yield 6.03 g crude product. The crude product was dissolved in 18 ml tBME at 40°C and subsequently treated dropwise with 25 ml hexane. The so formed suspension was stirred 16 h at room temperature and 1 h at 4°C. The crystals were separated on a filter funnel and washed with 5 ml hexane (4°C) to yield 4.8 g (77.7 %) product as white crystals. MS (ISN): 229 (100, [M + OAc]"), 169 (29, [M - H]"). M.p.: 71.5-72.0°C
Example 9 (step b of Scheme 4) Preparation of 4-(2-hydroxy-ethansulfonyl)-phenol

A solution of 5.0 g (28.5 mmol) 4-(2-hydroxy-ethylsulfanyl)-phenol in 25 ml methyl alcohol was treated at 10°C in parts over 20 min with 26.3 g (42.8 mmol) oxone ®. The suspension was stirred at room temperature (exothermic reaction) for 2 h, filtrated and the filtrate treated with 1 ml aqueous sodium hydrogen sulfite solution (38-40%). The pH of the reaction mixture was adjusted to 7 with 2 ml aqueous NaOH (28%), the suspension filtrated and the filtrate evaporated. The residue was treated with 20 ml toluene and

subsequent the solvent evaporated. This procedure was repeated two times to yield 6.81 g
crude product as white crystals.
MS (EI): 202 (9, [M]), 174 (13), 157 (30), 109 (32), 94 (100)
M.p.: 125.9-127.6°C.
Example 10 (step c of Scheme 4) Preparation of 4-(2-chloro-ethansulfonyl)-phenol

A solution of 6.81 g crude 4-(2-hydroxy-ethansulfonyl)-phenol in 35 ml CH2CI2 was » treated at room temperature with 5.3 ml (65.9 mmol) pyridine. To the reaction mixture was added at 0°C dropwise over 15 min a solution of 4.2 ml (57.1 mmol) thionyl chloride in 10 ml CH2CI2. After 65 h at room temperature the reaction mixture was treated with 35 ml brine, extracted and the organic phases were washed twice with total 100 ml aqueous half saturated NaCl solution. The combined organic phases were dried over Na2SO4 and the solvent was removed under reduced pressure to yield 6.77 g crude product. The crude product was dissolved in 2.5 ml CH2CI2 and 25 ml toluene, stirred 24 h at 50°C, 24 h at room temperature and 48 h at 0°C. The so formed suspension was filtered to yield 5.44 g (86.5% over 2 steps) product as white crystals. MS (El): 220 (17, [M]), 157 (100), 109 (18), 94 (17), 93 (60), 65 (41). M.p.: 72.5-73.5°C.
Example 11 (step 6* of Scheme 1) Preparation of rac-4-benzyl-pyperidine-3-one hydrochloride

A suspension consisting of 50.0 g of rac-l,4-dibenzyl-3-oxo-piperidine hydrochloride (0.158 mol), 0.50 1 of water and 10.0 g of 5% palladium on charcoal (Pd/C) was stirred under a hydrogen atmosphere until 3.51 of gas had been consumed (1 h). Then the Pd/C was filtered off and rinsed with water, the filtrate was evaporated and the residue taken up in isopropanol. The crystallization started spontaneously and was completed at 5°C over night. The precipitate was filtered off, the filter cake was rinsed with cold isopropanol and dried to constant weight (0.2 mbar, room temperature, 2 h) to afford 30 g of rac-4-benzyl-pyperidine-3-one hydrochloride as light yellow crystals.
Elemental analysis:


Example 12.1
In the glove box (O2 content
The samples were analyzed as follows:
A 20 mg sample was transformed into its trifluoroacetate by treatment with trifluoroacetic anhydride in pyridine and analyzed by gas chromatography on a chiral column (BGB-174, commercially available from BGB Analytic AG, Laufrainweg 139, CH-4469 Anwil, 15 m x 0.25 mm, 100° - 220°C, programmed with 2°C/min). Retention times: 35.19 min (R,R)-cis-4-benzyl-3-hydroxy-piperidine, 35.78 min (S,S)-cis-4-benzyl-3-hydroxy-piperidine, 37.87 and 39.20 min trans-4-benzyl-3-hydroxy-piperidine.
Example 12.2
The examples of Table 6 have been carried out in analogy to Example 12.1 in a 35 ml autoclave on a 0.34 g scale at a substrate-to-catalyst ratio of 300 (Exp. No. 12.2.1- 12.2.4) or in a 185 ml autoclave on a 9 g scale at a substrate-to-catalyst ratio of 50'000 (Exper. No. 12.2.5 - 12.2.7) using various chiral diphosphines with S-configuration.

Example 12.3
The examples of Table 7 have been carried out in analogy to Example 12.1 in a 185 ml autoclave on a 9 g scale at 20°C, under 40 bar of hydrogen pressure, at a 20% concentration, for 20 h, with S/base = 5 (base was KOtBu) and a S/C= 50*000 using [RuCl2((S)-(3,5-iPr)-MeOBIPHEP)((R,R)-DPEN)] as the catalyst. Various parameters have been changed.





WE CLAIM
1. ^ Method for the preparation of compounds of formula

wherein
R1 - R4 are, independently from each other, hydrogen, halogen, hydroxy,
amino, nitro, lower-alkyl-sulfonylamido, or acetamido;
R5-R8 are, independently from each, other hydrogen, lower-alkyl,
halogen, trifluoromethyl or lower-alkoxy';
and their pharmaceutically acceptable acid addition salts of hydrochloric acid, nitric acid, sulfuric acit, lactic acid, phosphoric acid, citric acid, formic acid, fumaric acid, maleic acid, acetic acid, succinic acid, tartaric acid, methane-sulphonic acid or p-toluenesulfonic acid, which process comprises
a) reacting a protected amino acid ester (1)

with a 4-substituted but)'ric acid derivative (2)

wherein
R'9 is an amino protecting group, preferably benzy;

are independently a carboxylic acid protecting group; and
represents a leaving group; to a compound of formula (3) in the presence of a base, which is triethylamine, ethyl-diisopropyl-amine, K2CO3 or Na2CO3, and in a solvent which is DMF, dioxane or acetonitrile; and
b) cyclising the obtained protected alkoxycarbonylmethyl amino butyric acid derivative

wherein the symbols are as defmed above; to a compound of formula (4) in an apolar aromatic solvent, which is toluene, and
c) benzylating the protected alkyl 3-oxo-piperidine carbox}1ate (4) salt

wherein
AZ signifies a mineral acid or a strong organic acid
to a compound of formula (5) in the presence of a base and a solvent, which are potassium-tert-butoxyde in THF, NaH in THF, NaOC2H5 in ethanol or K2CO3 in THE, and
d) decarboxylating the benzylated protected alkyl 3-oxo-piperidine carboxylate (5)

wherein the symbols are as defined above; to a compound of formula (6) or (6bis) in presence of a strong acid which is hydrochloric acid or sulfuric acid; and

e) asymmetrically hydrogenating the salt of formula (6) or (6bis)

wherein R"^ is an amino protecting group; to a compound of formula (7a) or (7b) in presence of a ruthenium complex with a chiral diphosphine ligand, a chiral diamine and a organic or an inorganic base; and
f) deprotecting the compound of formula (7a) or (7b)

wherein the symbols are as defined above; to a compound of formula III-a or III-b by hydrogenation of the N-benzylated compound in the presence of Pd/C, or in the alternative
g) deprotecting the compound of formula (6) to a compound of formula (6bis); and
subsequently
h) reacting the piperidine derivative of formula III-a or III-b

wherein the symbols are as defined above; with the sulfone derivative of formula II


wherein
R1 - R4 are as defined above; and
X is a leaving group;
which is previously treated with a base, which is triethylamine in CH2CI2, to form the corresponding reactive vinyl sulfone intermediate

in the presence of a base to obtain the desired compound of formula I.
2. The method according to claim 1, wherein a compound of formula I is prepared, which
is

3. The method according to claim le), wherein the asymmetric h)'drogenation is
characterized in that the ruthenium complex is a complex of formula
Ru(E)2(L)(A) IV
wherein
E is a halogen atom;
L is a chiral diphosphine ligand; and



The method according to claim 3, characterized in that the chiral diamine is a compound of formula V.
6. The method according to claim 3, wherein the configuration of the chiral diamine is "unlike" to the chiral complex, i.e. the chiral diphosphine ligand is (S) and the diamine is (R,R) or that the diphosphine is (R) and the diamine is (S,S).
7. The method according to claim 3, wherein the amount of chiral diamine used in the reaction is 0.5 to 2.5 equivalents based on the Ru-complex.
8. The method according to claim 3, wherein the base is present in the amount of 1.0 - 0.001 mol equivalents, preferably 0.05 - 0.2 mol equivalents with respect to the substrate in addition to 1 mol equivalent for the neutralization of the acid salt of the substrate (6 or 6bis, respectively).
9. The method according to claim 8, wherein the base is potassium tert.-butylate.
10. A method for the preparation of compounds of formula substantially herein above described and exemplified


Documents:

0253-mas-2001 abstract.jpg

0253-mas-2001 abstract.pdf

0253-mas-2001 others.pdf

0253-mas-2001 claims.pdf

0253-mas-2001 correspondence others.pdf

0253-mas-2001 correspondence po.pdf

0253-mas-2001 description (complete).pdf

0253-mas-2001 form-1.pdf

0253-mas-2001 form-26.pdf

0253-mas-2001 form-3.pdf

0253-mas-2001 form-4.pdf

0253-mas-2001 form-5.pdf

0253-mas-2001 petition.pdf

253.jpg


Patent Number 193610
Indian Patent Application Number 253/MAS/2001
PG Journal Number 08/2007
Publication Date 23-Feb-2007
Grant Date 07-Dec-2005
Date of Filing 20-Mar-2001
Name of Patentee F. HOFFMANN-LA ROCHE AG
Applicant Address 124 GRENZACHERSTRASSE, CH 4070 BASLE.
Inventors:
# Inventor's Name Inventor's Address
1 PIUS WALDMEIER 10 BACHSTRASSE, CH-4317 WEGENSTETTEN.
2 YVO CRAMERI 103 HOHESTRASSE, CH-4104 OBERWILL.
3 MICHELANGLELO SCALONE 14 BASLERSTRASSE, CH-4127 BIRSFELDEN.
4 ULRICH WIDMER 13 ALLEEWEG CH-4310 RHEINFELDEN.
PCT International Classification Number C07D211/00
PCT International Application Number N/A
PCT International Filing date
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 00106210.8 2000-03-22 EUROPEAN UNION