Europäisches Patentamt *EP000966429B1* (19) European Patent Office

Office européen des brevets (11) EP 0 966 429 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Date of publication and mention (51) Int Cl.7: C07C 233/05, A61K 31/16 of the grant of the patent: 04.06.2003 Bulletin 2003/23 (86) International application number: PCT/IL98/00015 (21) Application number: 98900346.2 (87) International publication number: (22) Date of filing: 14.01.1998 WO 98/030536 (16.07.1998 Gazette 1998/28)

(54) STEREOISOMERS, A METHOD FOR THEIR SYNTHESIS AND SEPARATION AND USES THEREOF VALNOCTAMID-STEREOISOMERE, EINE METHODE ZU IHRER HERSTELLUNG UND ABTRENNUNG UND IHRE ANWENDUNGEN STEREO-ISOMERES DU VALNOCTAMIDE, LEUR PROCEDE DE SYNTHESE ET DE SEPARATION, ET LEURS UTILISATIONS

(84) Designated Contracting States: (56) References cited: AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC US-A- 3 056 726 NL PT SE • BAREL, SHIMON ET AL: "Stereoselective (30) Priority: 14.01.1997 IL 12000697 pharmacokinetic analysis of valnoctamide in healthy subjects and in patients with " (43) Date of publication of application: CLIN. PHARMACOL. THER. (ST. LOUIS) (1997), 29.12.1999 Bulletin 1999/52 61(4), 442-449 CODEN: CLPTAT;ISSN: 0009-9236, XP002063383 (73) Proprietor: YISSUM RESEARCH DEVELOPMENT • PISANI, F. ET AL: COMPANY "-valnoctamide interaction in OF THE HEBREW UNIVERSITY OF JERUSALEM epileptic patients: in vitro/in vivo correlation" Jerusalem 91042 (IL) EPILEPSIA (N. Y.) (1993), 34(5), 954-9 CODEN: EPILAK;ISSN: 0013-9580, XP002063379 cited in (72) Inventors: the application • BIALER, Meir • BIALER, M. ET AL: " of a 96222 Jerusalem (IL) isomer, valnoctamide, in healthy • YAGEN, Boris subjects" EUR. J. CLIN. PHARMACOL. (1990), 96757 Jerusalem (IL) 38(3), 289-91 CODEN: EJCPAS;ISSN: 0031-6970, • SPIEGELSTEIN, Ofer XP002063380 cited in the application 90805 Mevasseret Zion (IL) • J. P.CHAMBON ET AL: "Valnoctamide: • ROEDER, Michael Pharmacological data suggesting an D-72810 Gomaringen (DE) antiepileptic activity" NEUROSCI. LETT. • SCHURIG, Volker [SUPPL], vol. 5, 1980, page S327 XP002063381 D-72076 Tubingen (DE) cited in the application

(74) Representative: Remarks: Leson, Thomas Johannes Alois, Dipl.-Ing. et al The file contains technical information submitted Patentanwälte after the application was filed and not included in this Tiedtke-Bühling-Kinne & Partner, specification Bavariaring 4 80336 München (DE)

Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention). EP 0 966 429 B1

Printed by Jouve, 75001 PARIS (FR) EP 0 966 429 B1

Description

Field of the Invention

5 [0001] The present invention generally relates to the individual (2R)(3S) stereoisomer of the drug valnoctamide (a mixture of four stereoisomer kinds, VCD-valmethamide or 2-ethyl-3-methyl pentanamide) obtainable by the process defined in claim 1 useful in treatment of neurological and psychotic disorders such as different kinds of epilepsy and affective disorders, and useful as tranquilizers and to treat pain, and to pharmaceutical compositions containing, as an active ingredient, said stereoisomer. The present invention further relates to a method for stereoselective separation 10 and quantification of the four stereoisomers from a of VCD or plasma of patients treated with the racemic drug. The present invention further relates to a unique method for the synthesis of the individual stereoisomers (2S)(3S) and (2R)(3S).

15 Background of the Invention

[0002] Epilepsy is an ancient disease which affects about 1% of the global population. Despite the progress in an- tiepileptic therapy, about 25% of the epileptic patients continue to suffer from uncontrolled seizures and toxicity. At present, there are four major epileptic drugs in use: , , carbamazepine and valproic 20 . Valproic acid (VPA) is one of the major antiepileptic drugs. It has two major side effects, teratogenicity and hepatotoxicity, which have been associated with therapy. Valnoctamide (VCD-valmethamide or 2-ethyl-3 methyl pentanamide) is an isomer of a valpromide (VPD) and is sold as an over- the-counter drug in several European countries. It has been available clinically for many decades and is 25 still used as a mild tranquilizer and occasionally as an antiepileptic drug (Chambon JP, Perio A, Neurosci. Lett [Suppl] 5: S327-S327, 1980). Valpromide (VPD) is used as an and agent, and in humans it is a of valproic acid (VPA). VPD may also be useful in treatment of neurological disorders and psychotic or affective disorders such as convulsions and epilepsy, and useful as tranquilizers and to treat pain. Recently, interest in VCD was revived by the observation that this compound possesses marked anticonvulsant activity in animal models. 30 Both VCD and VPD are three times more potent as than VPA. In addition, VPD, unlike VPA, has not been found to be teratogenic in animals possibly because it contains a carboxamide moiety instead of a carboxylic acid. However, in humans VPD acts as a prodrug to VPA and therefore its superiority over VPA in animal models does not have clinical implications. Unlike VPD, VCD acts as a drug on its own in both animals and humans and undergoes only slow and very little 35 transformation to its corresponding (less active) valnoctic acid (VCA). Based on its anticonvulsant potency, metabolic stability and lack of teratogenicity, VCD was viewed as having potential to become a new antiepileptic drug. It was recently found, however, that VCD is an inhibitor of the epoxide hydrolase. This inhibition was regarded as a drawback to the development of racemic VCD as a new antiepileptic drug. In the present invention, for the first time, characterization of the pharmacokinetics (PK) of the four stereoisomers of 40 VCD in humans was performed. This characterization clearly demonstrates that VCD pharmacokinetics is stereose- lective, with one isomer exhibiting a much higher clearance and a shorter half-life compared with the other stereoi- somers. Stereoselective pharmacokinetics has been demonstrated previously with different drugs, such as and , but this is the first time that PK stereoselectivity has been shown for an amide of an aliphatic short- chain . 45 [0003] The present invention relates to the (2R)(3S) stereoisomer of valnoctamide obtainable by the process defined in claim 1 useful in treatment of neurological and psychotic disorders such as different kinds of epilepsy and affective disorders, and useful as tranquilizers and to treat pain. The present invention further relates to a method for stereose- lective separation and quantification of the four stereoisomers from a racemic mixture of VCD and to a method for the synthesis of the 2S, 3S and 2R,3S VCD stereoisomers. 50 [0004] The stereoisomers have all of the benefits, and more, of the racemic drug of valnoctamide, but do not have any of its drawbacks, making them preferable for use in the treatment of convulsions and epilepsy and as an active ingredient in pharmaceutical compositions for the treatment of the latter. The present invention further relates to a method for separating the four stereoisomers of racemic valnoctamide. This method is a new stereoselective gas chroinatography-mass spectrometry (GC-MS) assay which allows specific and 55 sensitive quantitation of valnoctamide stereoisomers in human plasma. This method is the only one available for in- vestigating the pharmacokinetics and pharmacodynamics of valnoctamide stereoisomers in plasma samples, investi- gations which may have important practical implications for the development of a new anticonvulsant drug for humans. The present invention further relates to a unique method for the synthesis of the individual stereoisomers (2S) (3S)

2 EP 0 966 429 B1

and (2R) (3S).

Summary of the Invention

5 [0005] The present invention relates to the (2R)(3S) stereoisomer of valnoctamide (one of four isomers of valnocta- mide) obtainable by the process defined in claim 1 useful in treatment of neurological and psychotic disorders such as different kinds of epilepsy and affective disorders, and useful as tranquilizers and to treat pain, and to pharmaceutical compositions, containing as an active ingredient at least the (2R)(3S) stereoisomer of valnoctamide obtainable by the process defined in claim 1, useful as anticonvulsant drugs or tranquilizers. 10 The present invention further relates to a method for separating valnoctamide isomers comprising subjecting a mixture of racemic valnoctamide to gas chromatography preferably with oven temperature programming conditions; 50°C for 1 minute, increasing at and of 80°C / minute until 100°C, holding for a minute then reaching final temperature of 250°C at a rate of 40°C / minute, the injector temperature is 240°C and the GC-MS transfer line temperature is 250°C; inlet pressure is 5 psi with a linear flow rate 20 cm / second and the GC carrier gas is helium. Following, the isomers are 15 separated on a chiral stationary phase column, preferably consisting of octakis (3-O-butanoyl-2,6-di-o-pentyl)-γ-cyclo- dextrin chemically linked via a 6-mono-octamethylene spacer to a dimethylpolysiloxane and being coated on a fused silica capillary column of 25 m x 250µm coated by 0.25 µm octakis (3-O-butanoyl-2,6-di-o-pentyl)-γ-cyclodextrin. The present invention farther relates to a method for the stereoselective synthesis of the valnoctamide stereoisomers (2S,3S) and (2R,3S) comprising; 20 (a) synthesizing (3S)-methyl valeric acid from L-isoleucine; (b) subsequently synthesizing of (3S)-methyl valeroyl chlorid; and (c) subsequently synthesizing (2S,3S) valnoctic acid or subsequently synthesizing (2R,3S) valnoctic acid; (d) subsiquently synthesizing (2S,3S) valnoctamide or (2R,3S) valnoctamid. 25 Detailed Description of the Invention

[0006] The present invention relates to the (2R)(3S) stereoisomer of the drug valnoctamide (VCD-valmethamide or 2-ethyl-3-methyl pentanamide) obtainable by the process defined in claim 1, useful in treatment of neurological and 30 psychotic disorders such as different kinds of epilepsy and affective disorders), and useful as tranquilizers and to treat pain, and to pharmaceutical compositions containing, as an active ingredient, said stereoisomer. VCD, as compared with the well-known antiepileptic drug VPA, is retained in the body for a relatively long period and the plasma levels of its corresponding acid (VCA) are much lower than those of the parent drug. In contrast to VPD, which has a high (metabolic) extraction ratio by the liver, where it undergoes extensive first-pass to its 35 corresponding acid VPA, VCD does not have a high affinity for hepatic metabolism. This is reflected by its low oral clearance and its half-life, which is ten times higher than that of VPD (Bialer et al., Eur. J. Clin. Pharmacol. 38: 289-291, 1990). Based on its anticonvulsant potency, metabolic stability and lack of teratogenicity, VCD was viewed as having potential to become a new antiepileptic drug. However, it was recently found (Pisani et aL, Epilepsia 34:954-959, 1993) that 40 VCD is an inhibitor of the enzyme epoxide hydrolase. The inhibition of this enzyme, resulting in the decreased clearance of CBZ-E in patients who were treated with the antiepileptic drug carbamazepine (CBZ) together with VCD, caused in these patients signs suggestive of CBZ intoxication. This inhibition was regarded as a drawback to the development of racemic VCD as a new antiepileptic drug, but since VCD has two chiral centers (as depicted in Figure 1), and there are two pairs of enantiomers in the VCD molecule, 45 there is, a possibility that its pharmacokinetics (PK) and pharmacodynamics (PD), including toxicological potential and epoxide hydrolase inhibition, are stereoselective. by itself, PK stereoselectivity could lead to PD stereoselectivity, and have considerable implications for the further development of an antiepileptic drug. The method for the stereoselective separation and quantification of the four stereoisomers from a racemic mixture of VCD or plasma of patients treated with the racemic drug, of the present invention was developed based on this back- 50 ground. This method is a new stereoselective gas chromatography-mass spectrometry (GC-MS) assay which allows the specific and sensitive quantitation of VCD stereoisomers. The present invention further relates to a method for the stereoselective synthesis of the valnoctamide stereoisomers (2S,3S) and (2R,3S). The said method of synthesis comprises the following steps;

55 (a) synthesizing (3S)-methyl valeric acid (see figure 6 (I)) from L-isoleucine; (b) synthesizing of (3S)-methyl valeroyl chloride (see figure 7 (II)); (c) synthesizing (2S,3S) valnoctic acid (see figure 8 (III')) or subsequently synthesizing (2R,3S) valnoctic acid (see figure 8 (III));

3 EP 0 966 429 B1

(d) synthesizing (2S,3S) valnoctamide (see figure 8 (V')) or (2R,3S) valnoctamide (see figure 8 (V)).

[0007] The said invention will be further illustrated by the following experiments. These experiments do not intend to limit the scope of the invention, but to demonstrate and clarify it only. 5 The method of separation of the present invention was applied to investigate the pharmacokinetics of VCD stereoi- somers in plasma samples from seven healthy subjects and six epileptic patients.

Healthy subjects:

10 [0008] Seven healthy male volunteers, aged 20-31 years, weighing 61 to 85 kg, received 2 x 200 mg valnoctamide tablets (Nirvanil@) at 08:00 hours after an overnight fast Venous blood samples were taken through an indwelling catheter 0, 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12 and 24 hours after dosing. Food was witheld for 4 hours after dosing.

Patients: 15 [0009] Six epileptic patients (four men and two women aged 23-35 years, with a body weight of 56-85kg) were studied. All had been receiving a constant carbamazepine dose (800-1200 mg/day) for at least three months. One patient was also receiving phenytoin (250 mg/day) and a second patient phenobarbital (150 mg/day). Each patient received add-on VCD for eight consecutive days at a dosage of 400 mg on day 1 (single dose day), followed by 200 20 mg three times daily (tid) for seven days. On the last day, only the morning and the midday doses were administered. Blood samples for plasma VCD levels were collected at 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12 and 24 hours after the first and the last morning dose. Additional samples were taken at times 0, 0.5, 1, and 2 hours after the morning dose on the last treatment day.

25 Plasma handling and analytical assays:

[0010] Plasma samples were separated immediately by centrifugation at 2,000g for 10 minutes and stored at -20°C until assaying (VCD is stable at -20°C). The VCD plasma samples were analyzed by a gas chromatograph (HP 5890 series II)-mass spectrometer (HP 5989A)-GC-MS data system equipped with chiral capillary column. The mass spec- 30 trometer was operated in El mode at 70eV, the source temperature of the MS was maintained at 200°C and the quad- ruple at 100°C. Mass units and their relative abundance were calibrated with perfluorotributylamine (PFTBA). Spectra were collected in the range of 35 to 350 MHz, and the chiral analysis started 18 minutes after injection. Gas chromatograph (GC) oven temperature programming conditions were as follows: 50°C for 1 minute, then increas- ing at a rate of 8°C/minute until 100°C, holding for a minute then 4°C/minute until reaching a final temperature of 250°C. 35 The injector temperature is 240°C and the GC-MS transfer line temperature is 250°C. Inlet pressure is 5 psi with a linear flow rate 20 cm / second. Helium was used as the carrier gas. Chromatographic resolution of the VCD stereoisomers was accomplished by a direct separation on a chiral stationary phase (CSP) column. The CSP consisted of a Chirasil-γ-Dex (octakis (3-O-butanoyl-2,6-di-o-pentyl)-γ-cyclodextrin chemically linked via a 6-mono-octamethylene spacer to a dimethylpolysiloxane. The 0.25 µm Chirasil-γ-Dex was coat- 40 edona25mx250µm (id) fused silica capillary column. This CSP of a cyclodextrin derivative allows complete baseline resolution of the four stereoisomers ( α = 1.04, Rs = 2.0-2.4) in 24 minutes as depicted in Figure 2. Intra and inter-day variability in the retention times of the four VCD stereoisomers was less than 10%. A calibration curve was established by spiking drug-free plasma samples with known amounts of racemic VCD at a concentration range of 0.2 mg/L to 8 mg/L (or 0.05 mg/L to 2 mg/L of each of the four VCD stereoisomers). 45 Reagents and materials:

[0011] VCD (racemic mixture) was obtained from Clin Midy (Italy). Propylisopropylacetamide (PID), previously syn- thesized by Haj-Yehia and Bialer (Pharm Res 6:683 -9,1989), was used as internal standard (IS). Stock solution of 50 VCD (racemic mixture) was prepared by dissolving the drug in chloroform at a concentration of 5 mg/mL The internal standard (PID) was dissolved in chloroform at a concentration of 10 mg/mL. The stock solutions were stored at -20°C.

Pharmacokinetic and statistical analysis:

55 [0012] Pharmacokinetic parameters were calculated by noncompartmental analysis according to statistical moment theory. Peak plasma concentrations (Cmax) and peak times (tmax) were derived directly from the measured values. The linear terminal slopes (λ) of the log-concentration of each VCD stereoisomer versus time curves were calculated by the peeling procedure. Half life (t1/2) values were calculated as 0.693/ λ. Areas under the plasma concentration

4 EP 0 966 429 B1

versus time curve (AUC), after single dosing, were calculated by the trapezoidal rule with extrapolation to infinity by dividing the last measured concentration by the terminal slope. During repeated administration AUC was calculated over the last two dosing intervals by using the trapezoidal rule. Mean residence time (MRT) was calculated after single dosing as AUMC/AUC, in which AUMC is the area under the concentration x time product versus time curve from zero 5 to infinity. Following single dosing, oral clearance (CL/F, in which F is the oral availability) and apparent volume of distribution at steady state (Vss/F) were calculated as Dose/AUC and as MRT x CL/F, respectively. Following repeated dosing, CL/F was calculated from the quotient of the dose (200 mg) and the AUC obtained during one dosing interval (defined as one-half of the AUC calculated over two consecutive dosing intervals). Comparison of the pharmacokinetic parameters of all four VCD stereoisomers was made by ANOVA. 10 Results:

[0013] The stereoisomers of VCD were identified as A, B, C and D based on their respective retention times as shown in Figure 1. As a step towards characterization, enantiomeric pairs of diastereisomers were separated by re- 15 peated crystallization of racemic VCD. After eight re crystallizations, a fraction of one pair of enantiomers (A and C) contained only 12% of the other enantiomeric pair (B and D). Intra-day and inter-day coefficients of variation for each analyte were below 10%. Figure 2 shows a typical GC-MS chromatogram depicting the high resolution of the four stereoisomers (peaks A, B, C,. and D) of VCD. The mass spectrum of each of the four VCD stereoisomers was identical. The mean plasma con- 20 centrations of the four stereoisomers following single oral dosing of racemic VCD to healthy subjects and epileptic patients are shown in Figures 3 and 4, while Figure 5 depicts the mean plasma concentrations of the stereoisomers during the last day after eight days of repeated oral dosing of racemic VCD in epileptic patients. Figure 5 exhibits primary and secondary peaks due to the fact that on the last day only 2 hours elapsed between the morning and the midday doses. The individual and mean pharmacokinetic parameters of the stereoisomers in healthy subjects and 25 epileptic patients are given in Tables 1 to 3. In healthy subjects stereoisomer B had the largest oral clearance (CL/F = 8.7+_0.9 L/h) and its half-life (t1/2) and mean residence time (MRT) were the shortest of all VCD stereoisomers. Following single doses of VCD in epileptic patients, stereoisomer B also showed the largest oral clearance (80+_20L/ h) and the largest volume of distribution (Vss/F = 349+_ 60 L) compared to all the other stereoisomers. For all stere- oisomers, CL/F values in epileptic patients were about 10-fold greater than those determined in healthy subjects who 30 received the same dose under identical conditions. Vss/F values were also much larger in patients than in healthy subjects. Because of their short half-lives, all stereoisomers showed negligible accumulation during repeated dosing (Figure 5 and Table 3). After 7-day dosing, however, CL/F values at steady-state were lower than those determined in the same patients after a single dose. [0014] This study provides the first PK characterization of the four stereoisomers of VCD in man. When the concen- 35 tration of individual isomers in plasma was added up at each sampling time, the resulting values were in close agree- ment with those determined previously in the same samples by using a different non-stereoselective GC assay (Pisani et al., Epilepsia 34:954-959, 1993). This provides confirmatory evidence for the reliability of the assay used in this study and for the stability of VCD under the indicated storage conditions. Evaluation of the kinetic properties of individual isomers in healthy subjects and in epileptic patients receiving concom- 40 itant anticonvulsant therapy clearly demonstrated that VCD pharmacokinetics is stereoselective, with one isomer (ster- eoisomer B) exhibiting a much higher clearance and a shorter half-life compared to other isomers. In fact, a pronounced difference in PK behavior was observed not only between stereoisomer B and its diastereoisomers A and C, but also between individual enantiomers B and D. Previous studies have shown that VCD is completely absorbed and is eliminated predominantly by metabolism. There- 45 fore, the difference in PK properties between individual stereoisomers must involve a stereoselective process in the metabolism of the drug. In this respect, the larger apparent volume of distribution (Vss/F) of stereoisomers B, which was most clearly evident in epileptic patients, could be related to higher first -pass metabolism of this isomer, resulting in reduced oral . Compared with healthy control subjects, epileptic patients exhibited an approximately 10-fold greater apparent oral 50 clearance (CL/F) for each of the four stereoisomers. Epileptic patients also showed shorter half lives compared with control (about 3 h vs. 10 h for stereoisomers A,C and D, and about 3 h vs. 5.8 h for stereoisomer B). The prominent VCD elimination between healthy subjects and epileptic patients could be explained by induction of the oxidative me- tabolism of VCD stereoisomers by carbamazepine which is a potent inducer of cytochrome P450. Indeed the minimal urinary excretion of unchanged VCD and 55 its minor metabolic hydrolysis to its corresponding acid VCA suggest that oxidative metabolic pathways play an im- portant role in VCD elimination. Induction of first-pass metabolism by carbamazepine may also explain the larger vol- umes of distribution (Vss/F) of VCD stereoisomers in patients with epilepsy. The decrease in apparent oral clearance of VCD isomers which was observed during repeated dosing in epileptic patients, on the other hand, is suggestive of

5 EP 0 966 429 B1

time-dependent changes in VCD disposition, possibly related to concentration-dependent metabolism of the drug. Although stereoselective pharmacodynamics is not necessarily associated with PK stereoselectivity, the differences in PK behavior of VCD stereoisomers increase the likelihood that differences also exist in their anticonvulsant activity or undesired properties such as teratogenicity, hepatotoxicity and epoxide hydrolase inhibition. 5 A unique method for the synthesis of individual stereoisomer was developed and their absolute configuration was resolved by X-RAY diffraction analysis as shown in figure 9. The method for the asymetric synthesis of the 2S, 3S and 2R,3S VCD stereoisomers will be further described by the following examples:.

10 Synthetic procedure of (2S,3S)-valnoctamide

1. (2S,3S)-2-bromo-3-methyl valeric acid

[0015] To a cooled (0°C) solution of L-Isoleucine (66 g) dissolved in HBr (6N, 500 ml) was added dropwise a solution 15 of sodium nitrite (35 g in 100 ml water). The reaction was stirred overnight at room temperature and then extracted with EtOAc (3x150 ml). The combined organic extracts washed with 10% sodium bisulfit solution, water, saturated brine, dried over MgSO4, filtered and concentrated. The product a yellowish oil (53.9 g) was obtained in 55% yield. (The product can be purified by distillation and crystallization from PE). (see figure 6)

20 2. (3S)-methyl valeric acid

[0016] To a solution of (3S)-methyl valeric acid (53.9 g) dissolved in sulfuric acid (1N, 800 ml) was added slowly zinc powder (36 g). The reaction was stirred for two hours, filtered and extracted with Et2O (3x200 ml). The combined organic extracts washed with water, saturated brine, dried over MgSO4 and concentrated. The yellowish oil obtained 25 was purified by distillation under reduced pressure (5 torr, bp=55°C). The product a colorless oil (18.5 g) was obtained in 58% yield. (see figure 6).

3. (2S,3S)-2-chloro-3-methyl valeric acid

30 [0017] To a cooled (0°C) solution of L-Isoleucine (40 g) dissolved in HCl (5N, 750 ml) was added dropwise a solution of sodium nitrite (35 g in 100 ml water). The reaction was stirred overnight at room temperature (ϳ23°C), Na3CO3 (31 g) slowly added and the reaction mixture extracted with EtOAc (3x150 ml). The combined organic extracts washed with water, half saturated brine, dried over MgSO4, filtered and concentrated. The product a yellowish oil was distilled under reduced pressure (5 torr, bp=111°C) to afford a colorless oil (32 g) obtained in 70% yield. (see figure 6). 35 4. (3S)-methyl pentanol

[0018] To a solution of(2S,3S)-2-chloro-3-methyl valeric acid (66 g) dissolved in 2500 ml of dry Et2O was carefully added powdered LAH (50 g). The reaction was refluxed for 7-10 days. Excess LAH was neutralized by careful addition 40 of EtAOc (250 ml), methanol (100 ml), DDW (100 ml) and a 1:1 sulfuric acid and water mixture (100 ml). The etheral phase separated and the aqueous phase extracted with EtOAc (3x200 ml). The combined organic extracts washed with a 10% NaHCO3 solution, water, half saturated brine, dried over MgSO4 and concentrated. The product, a green oil, was purified by vacuum distillation (5 torr, bp=42°C) to afford a colorless oil (27.3 g), 61% yield, (see figure 6).

45 5. (3S)-methyl valeric acid

[0019] To a cool (0°C) solution of (3S)-methyl pentanol (37.9 g) dissolved in 1500 ml Et2O was slowly added 1200 ml of the oxidation reagent (240 g Na2Cr2O7, 177 ml H2SO4 97% and DDW to make 1200 ml). The reaction was stirred overnight at room temperature (ϳ23°C) and extracted with Et2O (3x150 ml). The combined organic extracts washed 50 with 5% HCl solution (3x50 ml) followed by alkalization with 2N NaOH solution. The alkaline aqueous phase separated, acidified with concentrated HCl (PH=2) and extracted with EtOAc (3x100 ml). The combined organic extracts dried over MgSO4 and concentrated to give a dark oil which was purified by vacuum distillation (5 torr, bp=110°C) to afford a colorless oil (25.2 g), 58% yield. (see figure 6).

55 6. (3S)-methyl valeryol chloride

[0020] To a cooled (0°C) solution of (3S)-methyl valeric acid (10 g) and dry DMF (6.7 ml) dissolved in dry DCM (50 ml) was added dropwise oxalyl chloride (7.3 ml in 50 ml dry DCM). After stirring one hour the DCM and excess oxalyl

6 EP 0 966 429 B1

chloride were evaporated by N2 stream. The crude product was rinsed with dry DCM (2x20 ml) which was evaporated by N2 stream. The reaction mixture was dissolved in cooled (0°C) dry THE (20 ml) and used without further purification at the next step. (see figure 7).

5 7. (4S,3'S)-3-(3'-methyl-1'-oxopentyl)-4-benzyl-2-oxazolidinone

[0021] To a cooled (-78°C) solution of (4S)-benzyl-2-oxazolidinone (3.65 g) dissolved in dry THE (50 ml) was added dropwise a solution of n-BuLi (13 ml, 1.6 M in hexane). After stirring for 30 min a cooled (-78°) solution of (3S)-methyl valeryol chloride (ϳ2.8 g) dissolved in dry THF (20 ml) was added via cannula at once. The reaction mixture was slowly 10 warmed to 0°C, stirred for 2 hours and quenched by addition of saturated NH4Cl solution. The THF evaporated and the aqueous phase extracted with DCM (3X50 ml). The combined organic extracts washed with water, half saturated brine, dried over MgSO4 and concentrated. The product (3.9 g) was crystallized from 5% EtOAc in PE, yield 69%. (see figure 7).

15 8. (4S,2'S,3'S)-3-(2'-ethyl-3'-methyl-1'-oxopentyl)-4-benzyl-2-oxasolidinone

[0022] Synthesis by the same procedure as (4S,2'R)-3-(2'-isopropyl-1'-oxopentyl)-4-benzyl- 2-oxazolidinone using (4S,3'S)-3-(3'-methyl-1'-oxopentyl)-4-benzyl-2-oxazolidinone and ethyl trifluoromethane sulphonate. (see figure 7).

20 9. (2S,3S)-valnoctic acid ((2S,3S)-VCA)

[0023] (4R,5S,2'S)-3-(2'-isopropyl-1'-oxopentyl)-4-methyl-5-phenyl-2-oxazolidinone was synthesized from (4R,5S)- 3-(1'-oxopentyl)-4-methyl-4-phenyl-2-oxazolidinone and isopropyl triflate by the same procedure as (4S,2'S)-3-(2'-iso- propyl-1'-oxopentyl)-4-benzyl-2-oxazolidinone. The product (5.53 g) was obtained in 32% yield. (see figure 8). 25 10. (2S,3S)-valnoctamide((2S,3S)-VCD)

[0024] Under N2 to a cooled (0°C) solution of(2R)-PIA (3.3 g) dissolved in dry DCM (100 ml) and dry DMF (1.77 ml) was added dropwise a solution of oxalyl chloride (34.3 ml, 2.0 M solution in DCM). After stirring one hour the DCM and 30 excess oxalyl chloride were evaporated by N2 stream. In order to remove traces of oxalyl chloride the crude product was treated with dry DCM (2x20 ml) which was evaporated by N2 stream. To the crude reaction mixture dissolved in cooled (0°C) dry DCM (100 ml) was added NH4OH (20 ml, 25% solution in water) and the reaction mixture stirred for an hour. The organic phase washed with water and half saturated brine, dried over MgSO4, filtered and concentrated. The product was crystallized from 20% EtOAc in PE to afford 2.14 g, 65% yield. 35 Synthetic procedure of (2R,3S)-valnoctamide

[0025] Synthesis of (2R,3S)-VCD is performed by using the same procedures as for the synthesis of (2S,3S)-VCD except for the chiral auxiliary which is (4R,5S)-4-methyl-5- phenyl-2-oxazolidinone. 40 Table I: Pharmacokinetic parameters of the stereoisomers of valnoctamide (VCD) following single oral administration (400 mg) of the commercially available racemic drug (Nirvanil) to seven healthy subjects.

45 Subjects CP CG ML GM VL BV GL Mean ± SD Stereoisomer A t1/2 (h) 8.8 13.8 8.1 10.7 9.5 8.5 12.5 10.2 ± 2.1 50 Cmax (mg/L) 2.0 1.5 1.8 1.8 1.7 1.8 1.5 1.7 ± 0.2 tmax (h) 1 0.5 1 1 0.5 0.5 0.5 0.7 ± 0.3 CL/F (L/h) 3.6 4.5 4.1 3.7 4.6 4.2 3.5 4.2 ± 0.6 Vss/F (L) 46 89 48 55 62 52 63 59 ± 15 MRT (h) 13 20 12 15 13 12 18 55 15 ± 3

7 EP 0 966 429 B1

Table I: (continued) Pharmacokinetic parameters of the stereoisomers of valnoctamide (VCD) following single oral administration (400 mg) of the commercially available racemic drug (Nirvanil) to seven healthy subjects.

5 Subjects CP CG ML GM VL BV GL Mean ± SD Stereoisomer B t1/2 (h) 4.7 7.3 4.6 6.0 5.8 5.2 7.1 5.8 ± 1.1* 10 Cmax (mg/L) 1.5 1.1 1.4 1.4 1.4 1.5 1.1 1.3 ± 0.2 tmax (h) 1 0.5 1 1 0.5 0.5 0.5 0.7 ± 0.3 CL/F (L/ti) 7.9 9.2 7.9 8.4 10.2 8.1 9.3 8.7 ± 0.9* Vss/F (L) 57 94 58 73 83 62 91 74 ± 16 15 MRT (h) 7.0 10 7 9 8 8 10 8 ± 1 Subjects CP CG ML GM VL BV GL Mean ± SD Stereoisomer C 20 t1/2 (h) 8.4 13 8.0 10 11.1 9.2 12.4 10.3 ± 1.9 Cmax (mg/L) 1.8 1.4 1.7 1.7 1.5 1.8 1.5 1.8 ± 0.2 tmax (h) 1 0.5 1 1 0.5 0.5 0.5 0.7 ± 0.3 CL/F (L/h) 4.0 5.1 4.3 4.1 4.6 4.1 3.9 4.3 ± 0.4 25 Vss/F (L) 49 93 50 58 72 55 70 64 ± 16 MRT (h) 12 18 12 14 16 13 18 15 ± 3 Subjects CP CG ML GM VL BV GL Mean ± SD 30 Stereoisomer D t1/2 (h) 8.7 12.9 8.0 13.0 9.5 7.9 13 10 ± 2 Cmax (mg/L) 1.6 1.3 1.6 1.5 1.4 1.8 1.5 1.5 ± 0.2 tmax (h) 1 0.5 1 1 0.5 0.5 0.5 0.7 ± 0.3 35 CL/F (Lh) 4.3 5.6 4.6 3.8 5.4 4.7 4.1 4.6 ± 0.7 Vss/F (L) 54 102 53 70 72 55 76 69 ± 17 MRT (h) 13 18 12 18 13 12 19 15 ± 3

(*p < 0.05 vs. stereoisomers A, C and D)

40

Table II: Pharmacokinetic parameters of the stereoisomers of valnoctamide (VCD) following single oral administration (400 mg) of the commercially available racemic VCD (Nirvanil) 45 to six epileptic patients. Patients CA CC DFR PA SG CCA Mean ± SD Stereoisomer A 50 t1/2 (h) 2.8 3.6 2.5 3.6 8.3 3.8 4.1 ± 2.1 Cmax (mg/L) 0.71 0.94 0.71 0.82 0.88 0.67 0.78 ± 0.10 tmax (h) 1.5 0.5 1.5 0.5 0.5 0.5 0.8 ± 0.5 CL/F (L/h) 40 27 35 32 20 35 32 ± 7 55 Vss/F (L) 175 138 148 172 169 182 164 ± 17 MRT (h) 4.4 5.1 4.3 5.3 8.3 5.1 5.4 ± 1.5

8 EP 0 966 429 B1

Table II: (continued) Pharmacokinetic parameters of the stereoisomers of valnoctamide (VCD) following single oral administration (400 mg) of the commercially available racemic VCD (Nirvanil) to six epileptic patients. 5 Patients CA CC DFR PA SG CCA Mean ± SD Stereoisomer B 10 t1/2 (h) 1.4 4.0 1.3 3.0 4.0 4.1 3.0 ± 1.3 Cmax (mg/L) 0.31 0.56 0.3 0.48 0.51 0.32 0.41 ± 0.12 tmax (h) 1 0.5 1.5 0.5 0.5 0.5 0.75 ± 0.41 CL/F (L/h) 108 60 94 84 56 79 80 ± 20*

15 Vss/F (L) 386 307 307 365 287 444 349 ± 60* MRT (h) 3.6 5.2 3.3 1.3 5.1 5.6 4.0 ± 1.6 Patients CA CC ML PA SG CCA Mean ± SD 20 Stereoisomer C t1/2 (h) 2.8 3.4 2.6 3.7 3.5 3.7 3.3 ± 0.5 Cmax (mg/L) 0.55 0.82 0.56 0.76 0.8 0.7 0.7 ± 0.12 tmax (h) 1.5 0.5 1.5 0.5 0.5 0.5 0.83 ± 0.5 25 CL/F (L/h) 50 31 44 38 28 41 39 ± 8 Vss/F (L) 215 147 186 208 141 197 181 ± 31 MRT (h) 4.4 4.8 4.2 5.4 5.0 4.8 4.8 ± 0.4 Patients

30 CA CC DFR PA SG CCA Mean ± SD Stereoisomer D t1/2 (h) 1.2 3.8 2.7 3.8 3.9 3.8 3.3 ± 1.1 Cmax (mg/L) 0.51 0.72 0.51 0.69 0.72 0.51 0.61 ± 0.11 35 tmax (h) 1.5 0.5 1.5 0.5 0.5 0.5 0.83 ± 0.51 CL/F (L/h) 60 35 45 42 30 42 46 ± 11 Vss/F (L) 195 183 198 233 161 226 202 ± 22 MRT (h) 3.3 5.2 4.5 5.6 5.4 5.3 4.7 ± 1.0

40 available racemic VCD (Nirvanil) to six epileptic patients during the last dosing interval and the last day of the study. Patients CA CC DFR PA SG CCA Mean ± SD Stereoisomer A 45 (1/2 (h) 2.2 2.7 3.8 2.4 4.8 3.8 2.8 ± 0.7 Cmax (mg/L) 0.9 1.1 0.72 0.86 0.8 1.2 0.9 ± 0.2 tmax (h) 2.0 1.5 1.5 1.0 1.75 1.5 1.5 ± 0.3 CL/F (L/h) 23 19 20 29 19 19 21 ± 4 50 Patients CA CC DFR PA SG CCA Mean ± SD Stereoisomer B t1/2 (h) 2.0 1.9 2.9 2.1 4.3 4.3 2.2 0.5 55 ± Cmax (mg/L) 0.4 0.5 0.3 0.3 0.4 0.5 0.4 ± 0.1

(*p < 0.05 vs. stereoisomers A, C and D)

9 EP 0 966 429 B1

Table II: (continued) Pharmacokinetic parameters of the stereoisomers of valnoctamide (VCD) following single oral administration (400 mg) of the commercially available racemic VCD (Nirvanil) to six epileptic patients. 5 Patients CA CC DFR PA SG CCA Mean ± SD Stereoisomer B 10 tmax (h) 2.0 1.5 1.5 1.0 1.25 1.5 1.4 ± 0.3 CL/P (L/h) 79 50 52 71 46 50 58 ± 14* Patients

15 CA CC DFR PA SG CCA Mean ± SD Stereoisomer C t1/2(h) 2.2 3.0 4.1 2.3 5.0 4.1 2.9 ± 0.9 Cmax (mg/L) 0.81 0.9 0.62 0.7 0.6 1.0 0.8 ± 0.2

20 tmax (h) 2.0 1.5 1.5 1.0 1.25 1.5 1.4 ± 0.3 CL/F (L/h) 26 23 24 36 28 23 27± 5 Patients CA CC DFR PA SG CCA Mean±SD 25 Stereoisomer D t1/2 (h) 2.1 2.8 5.2 2.5 5.3 4.6 3.2 ± 1.4 Cmax (mg/L) 0.7 0.8 0.6 0.6 0.6 0.5 0.6 ± 0.1 tmax (h) 2.0 1.5 1.5 1.0 1.25 1.5 1.4 ± 0.3 30 CLF (L/h) 36 25 26 38 24 25 29 ± 6

(*p < 0.05 vs. stereoisomers A, C and D)

Claims 35 1. Valnoctamide stereoisomer (2R) (3S) useful in the treatment of neurological and psychotic disorders, and affective disorders and useful as tranquilizers and to treat pain obtainable by a process comprising:

(a) synthesizing (35)-methyl valeric acid (1) from L-isoleucine; 40 (b) subsequently synthesizing of (3S)-methyl valeroyl chloride (II); and (c) subsequently synthesizing (2R,3S) valnoctic acid (III); (d) subsequently synthesizing (2R,3S) valnoctamide (V) .

2. Valnoctamide stereoisomer (2R) (3S) according to claim 1 wherein said affective disorder is convulsions or epilepsy. 45 3. A pharmaceutical composition containing as an active ingredient at least valnoctamide stereoisomer (2R) (3S) obtainable by the process as defined in claim 1.

4. Pharmaceutical compositions according to claim 3 wherein the compositions are useful as anticonvulsant drugs 50 or tranquilizers and in the treatment of neurological and psychotic disorders and affective disorders and to treat pain.

5. A method for baseline separation of the four valnoctamide isomers comprising subjecting a mixture of racemic valnoctamide to gas chromatography and separating the isomers on a chiral stationary phase column character- ized in that said chiral stationary phase column comprises Chirasil-γ-Dex (octakis (3-O-butanoyl-2, 6-di-o-pentyl)- 55 γ-cyclodextrin) chemically linked via a 6-mono-octamethylene spacer to a dimethylpolysiloxane and said Chirasil- γ-Dex is coated on a fused silica capillary column.

10 EP 0 966 429 B1

6. A method according to claim 5 wherein the gas chromatography has oven temperature programming conditions; 50°C for 1 minute, increasing at a rate of 8°C / minute until 100°C, holding for a minute then reaching final tem- perature of 250°C at a rate of 4°C / minute; the injector temperature is 240°C and the GCMS transfer line temper- ature is 250°C; inlet pressure is 0.41 bars (5 psi) with a linear flow rate 20 cm / second and the GC carrier gas is 5 helium.

7. A method according to claim 5 wherein the fused silica capillary column is 25 m x 250µm coated by 0.25 µm Chirasil-γ-Dex (octakis (3-O-butanoyl-2, 6-di-o-pentyl)-γ-cyclodextrin).

10 8. A method according to claim 5 wherein the mixture of racemic valnoctamide is obtained from blood plasma from patients treated with valnoctamide or the drug valnoctamide itself.

9. A method for the stereoselective synthesis of the valnoctamide stereoisomers (2S,3S) and (2R,3S) comprising;

15 (a) synthesizing (3S)-methyl valeric acid (1) from L-isoleucine; (b) subsequently synthesizing of (3S)-methyl valeryl chloride (II); and (c) subsequently synthesizing (2S,3S) valnoctic acid (III') or subsequently synthesizing (2R,3S) valnoctic acid (III); (d) subsequently synthesizing (2S,3S) valnoctamide (V') or (2R,3S) valnoctamide (V). 20 10. A method for the stereoselective synthesis of the valnoctamide according to claim 9 wherein

step (a) comprises dissolving L-isoleucine in HBr or HCl and the dropwise adding of NaNO2 to obtain (2S)- bromo-(3S)-methyl valeric acid or (2S)-chloro(3S)-methyl valeric acid and dissolving the (2S)-bromo-(3S)- 25 methyl valeric acid in H2SO4 and adding Zn obtaining compound (I) by extraction or adding LiAlH4 to the (2S)- cloro-(3S)-methyl valeric acid and refluxing it obtaining (3S)-methyl pentanol and further oxidizing the (3S)- methyl pentanol obtaining compound (I) by extraction; and step (b) comprises dropwise adding oxalyl chloride to compound (I) and dry DMF, dissolving the reaction product in THF obtaining compound (II); and 30 step (c) comprises dropwise adding to cool (4S)-benzyl-3-(1-oxo-(2S)-ethyl-(3S)methyl valeroyl)-2-oxazolidi- none (IV'), n-BuLi and cooling compound (II) and adding it at once to compound (IV') obtaining (4S)-benzyl- 3-(oxo-(2S)-ethyl(3S)-methyl valeroyl)-2-oxazolidinone or dropwise adding to cool (4R)-methyl(5S)-phenyl- 3-(1-oxo-(3S)-methyl valeroyl)-2-oxazolidinone, n-BuLi (IV) and cooling compound (II) and adding it at once to (IV) obtaining (4R)-methyl-(5S)phenyl-3-(1-oxo-(3S)-methyl-(2R)-ethyl valeroyl)-2-oxazolidinone; and 35 step (d) comprises adding H2O2 and LiOH to cooled said (4S)-benzyl-3-(oxo(2S)-ethyl-(3S)-methyl valeroyl)- 2-oxazolidinone in a mixture of THF:DDW or adding H2O2 and LiOH to a cooled said (4R)-methyl-(5S)-phenyl- 3-(1-oxo-(3S)methyl-(2R)-ethyl valeroyl)-2-oxazolidinone, warming the resulting mixture to room temperature, cooling to 0°C quenching by sodium sulfite, DCM extracting, obtaining the chiral auxiliary, (4S)-benzyl-2-ox- azolidinone or the chiral auxiliary (4R, 5S) -4-methyl-5-phenyl-2-oxazolidinone, and dropwise adding oxalyl 40 chloride to dissolved in dry DCM dry adding DCM and NH4OH, obtaining compound (V') or compound (V) by organic extraction.

Patentansprüche 45 1. Valnoctamid-Stereoisomer (2R) (3S) verwendbar bei der Behandlung von neurologischen und psychotischen Stö- rungen und Affektstörungen und verwendbar als Beruhigungsmittel. und zur Behandlung von Schmerzen, erhält- lich durch ein Verfahren mit:

50 (a) Synthetisieren von (3S)-Methylvaleriansäure (1) aus L-Isoleucin; (b) nachfolgendem Synthetisieren von (3S)-Methylvaleroylchlorid (II); und (c) nachfolgendem Synthetisieren von (2R,3S)-Valnoctinsäure (III); (d) nachfolgendem Synthetisieren von (2R,3S)-Valnoctamid (V).

55 2. Valnoctamid-Stereoisomer (2R) (3S) nach Anspruch 1, wobei die Affektstörung Krämpfe oder Epilepsie ist.

3. Pharmazeutische Zusammensetzung, welche als einen wirksamen Bestandteil wenigstens das durch das in An- spruch 1 definierte Verfahren erhältliche Valnoctamid-Stereoisomer (2R) (3S) enthält.

11 EP 0 966 429 B1

4. Pharmazeutische Zusammensetzungen nach Anspruch 3, wobei die Zusammensetzungen als krampflösende Wirkstoffe oder Beruhigungsmittel und in der Behandlung neurologischer und psychotischer Störungen und Af- fektstörungen und zur Behandlung von Schmerz verwendbar sind.

5 5. Verfahren zur Basislinientrennung der vier Valnocatmidisomere mit Gaschromatographie und Trennung der Iso- mere einer Mischung von racemischen Valnoctamid auf einer Säule mit chiraler stationärer Phase, dadurch ge- kennzeichnet, dass die chirale stationäre Phase Chirasil-γ-Dex (Octakis (3-O-Butanoyl-2,6-di-o-pentyl)-γ-cy- clodextrin chemisch über einen 6-Mono-Octamethylen-Spacer an ein Dimethylpolysiloxan gekoppelt, und das Chir- asil-γ-Dex auf eine Quarzglas-Kapillarsäule geschichtet ist. 10 6. Verfahren nach Anspruch 5, wobei die Gaschromatographie(GC) Ofentemperatur-Programmbedingungen hat; 50°C für 1 Minute, Ansteigen mit einer Rate von 8°C/Minute bis 100°C, Halten für eine Minute, dann Erreichen der Endtemperatur von 250°C mit einer Rate von 4°C/Minute; die Injektortemperatur ist 240°C und die Temperatur der GC-MS-Transferleitung ist 250°C; der Einlassdruck ist 0,41 bar (5 psi) mit einer linearen Flussgeschwindigkeit 15 von 20 cm/Sekunde und das Trägergas ist Helium.

7. Verfahren nach Anspruch 5, wobei die Quarzglas-Kapillarsäule 25 m x 250 um ist, beschichtet mit 0,25 µm Chirasil- γ-Dex 0,25 µm (Octakis (3-O-Butanoyl-2,6-di-opentyl)-γ-cyclodextrin).

20 8. Verfahren nach Anspruch 5, wobei die Mischung von racemischen Valnoctamid aus Blutplasma von mit Valnoc- tamid behandelten Patienten oder dem Wirkstoff Valnoctamid selbst erhalten wird.

9. Verfahren für die stereoselektive Synthese der Valnoctamid-Stereoisomere (2S,3S) und (2R,3S) mit;

25 (a) Synthetisieren von (3S)-Methylvaleriansäure (1) aus L-Isoleucin; (b) nachfolgendem Synthetisieren von (3S)-Methylvaleroylchlorid (II); und (c) nachfolgendem Synthetisieren von (2S,3S)-Valnoctinsäure (III') oder nachfolgendem Synthetisieren von (2R,3S)-Valnoctinsäure (III); (d) nachfolgendem Synthetisieren von (2S,3S)-Valnoctamid (V') oder (2R,3S)-Valnoctamid (V). 30 10. Verfahren für die stereoselektive Synthese von Valnoctamid nach Anspruch 9, wobei

Schritt (a) umfasst das Lösen des L-Isoleucine in HBr oder HCl und die tropfenweise Zugabe von NaNO2,um (2S)-Brom-(3S)-methylvaleriansäure oder (2S)-Chlor-(3S)-methylvaleriansäure zu erhalten, und Lösen der 35 (2S)-Brom-(3S)-methylvaleriansäure in H2SO4 und Zugabe von Zn, um Verbindung (I) durch Extraktion zu erhalten, oder Zugabe von LiAlH4 zu der (2S)-Chlor-(3S)-methylvaleriansäure und Halten unter Reflux, um (3S)-Methylpentanol zu erhalten und ferner Oxidation des (3S)-Methylpentanol, um Verbindung (I) durch Ex- traktion zu erhalten; und Schritt (b) umfasst die tropfenweise Zugabe von Oxalylchlorid zur Verbindung (I) und trockenem DMF, Lösen 40 des Reaktionsproduktes in THF zum Erhalt von Verbindung (II); und Schritt (c) umfasst die tropfenweise Zugabe von n-BuLi zu kaltem (4S)-Benzyl-3-(1-oxo-(2S)-ethyl-(3S)me- thylvaleroyl)-2-oxazolidon (IV') und Kühlen der Verbindung (II) und ihre Zugabe auf einmal zur Verbindung (IV'), um (4S)-Benzyl-3-(oxo-(2S)-ethyl-(3S)-methylvaleroyl)-2-oxazolidon zu erhalten, oder tropfenweise Zu- gabe von n-BuLi zu kaltem (4R)-Methyl(5S)-phenyl-3-(1-oxo-(3S)-methylvaleroyl)-2-oxazolidon (IV) und Küh- 45 len der Verbindung (II) und ihre Zugabe auf einmal zu (IV), um (4R)-Methyl-(55)phenyl-3-(1-oxo-(3S)-me- thyl-(2R)-ethylvaleroyl)-2-oxazolidon zu erhalten; und Schritt (d) umfasst die Zugabe von H2O2 und LiOH zu dem gekühlten (4S)-Benzyl-3-(oxo-(2S)-ethyl-(3S)- methylvaleroyl)-2-oxazolidon in einer Mischung aus THF:DDW, oder Zugabe von H2O2 und LiOH zu dem gekühlten (4R)-Methyl-(5S)phenyl-3-(1-oxo-(3S)methyl-(2R)-ethylvaleroyl)-2-oxazolidon, Erwärmen der re- 50 sultierenden Mischung auf Raumtemperatur, Abkühlen auf 0°C, Quenchen durch Natriumsulfit, DCM-Extrak- tion, Erhalten des chiralen Zusatzes (4S)-Benzyl-2-oxazolidinon oder des chiralen Zusatzes (4R, 5S)-4-Me- thyl-5-phenyl-2-oxazolidinon, und tropfenweise Zugabe von Oxalylchlorid gelöst in trockenem DCM, trockene Zugabe von DCM und NH4OH, Erhalt der Verbindung (V') oder Verbindung (V) durch organische Extraktion.

55 Revendications

1. Stéréoisomère de valnoctamide (2R) (3S) utile pour le traitement de troubles neurologiques et psychotiques, et

12 EP 0 966 429 B1

de troubles affectifs et utile comme tranquillisant et pour le traitement de la douleur, pouvant être obtenu par un procédé comprenant :

(a) la synthèse de l'acide (3S)-méthyle valérique (1) à partir de L-isoleucine ; 5 (b) la synthèse subséquente de chlorure de (3S)-méthyle valéroyle (II) ; et

(c) la synthèse subséquente d'acide valnoctique (2R, 3S) (III) ;

10 (d) la synthèse subséquente de valnoctamide (2R, 3S) (V).

2. Stéréoisomère de valnoctamide (2R) (3S) selon la revendication 1 dans lequel ledit trouble affectif est des con- vulsions ou de l'épilepsie.

15 3. Composition pharmaceutique contenant comme principe actif au moins un stéréoisomère de vainoctamide (2R) (3S) pouvant être obtenu par le procédé tel que défini dans la revendication 1.

4. Compositions pharmaceutiques selon la revendication 3 dans lesquelles les compositions sont utiles comme mé- dicaments anticonvulsifs ou tranquillisants et pour le traitement de troubles neurologiques et psychotiques et de 20 troubles affectifs et pour traiter la douleur.

5. Procédé pour la séparation à la ligne de base de quatre isomères de valnoctamide comprenant la soumission d'un mélange de valnoctamide racémique à la chromatographie gazeuse et la séparation des isomères sur une colonne contenant une phase stationnaire chirale, caractérisé en ce que ladite colonne contenant une phase stationnaire 25 chirale comprend du Chirasil-γ-Dex (octakis (3-O-butanoyl-2,6-di-o-pentyle)-γ-cyclodextrine) lié chimiquement via un espaceur 6-mono-octaméthylène à un diméthylpolysiloxane et ledit Chirasil-γ-Dex est déposé sur une colonne capillaire de silice fondue.

6. Procédé selon la revendication 5 dans lequel la chromatographie gazeuse a des conditions de programmation de 30 la température du four ; 50°C pendant 1 minute, augmentant par rampe de 8°C/minute jusqu'à 100 °C, se main- tenant à cette température pendant une minute puis atteignant la température finale de 250°C à une rampe de 4 °C/minute ; la température de l'injecteur est 240 °C et la température de la ligne de transfert CGSM est de 250°C, la pression d'entrée est 0,41 bar (5 psi) avec un débit linéaire de 20 cm/seconde et le gaz vecteur de la CG est l'hélium. 35 7. Procédé selon la revendication 5 dans lequel la colonne capillaire de silice fondue est de 25mx250µm recouverte de 0,25 µm de Chirasil-γ-Dex (octakis (3-O-butanoyle-2,6-di-o-pentyl)-γ-cyclodextrine).

8. Procédé selon la revendication 5 dans lequel le mélange de valnoctamide racémique est obtenu à partir de plasma 40 sanguin de patients traités avec du vainoctamide ou le médicament valnoctamide lui-même.

9. Procédé pour la synthèse stéréo-sélective des stéréoisomères de valnoctamide (2S, 3R) et (2R, 3S) comprenant:

(a) la synthèse (3S)-méthyle acide valérique (1) à partir de L-isoleucine ; 45 (b) la synthèse subséquente de (35)-méthyle chlorure de valéroyle (II); et

(c) la synthèse subséquente d'acide valnoctique (2S, 3S) (III') ou la synthèse subséquente d'acide valnoctique (2R, 3S) (III); 50 (d) la synthèse subséquente de valnoctamide (2R, 3S) (V') ou valnoctamide (2R, 3S) (V).

10. Procédé pour la synthèse stéréo-sélective du valnoctamide selon la revendication 9 dans lequel

55 l'étape (a) comprend la dissolution de L-isoleucine dans HBr ou HCl et l'addition goutte à goutte de NaNO2 pour obtenir de l'acide (2S)-bromo-(3S)-méthyl valérique ou de l'acide (2S)-chloro-(3S)-méthyl valérique et la dissolution de l'acide (2S)-bromo-(3S)-méthyle valérique dans H2SO4 et, l'addition de Zn donnant le composé (I) par extraction ou l'addition de l'acide LiAlH4 au (2S)-chloro-(3S)-méthyl valérique et son reflux donnant du

13 EP 0 966 429 B1

(3S)-méthyle pentanol puis l'oxydation ultérieure du (3S)-méthyl pentanol donnant le composé (I) par extraction ; et

l'étape (b) comprend l'addition par goutte de chlorure d'oxalyle au composé (I) et de DMF anhydre, la disso- 5 lution du produit réactionnel dans du THF donnant le composé (II); et

l'étape (c) comprend l'addition par goutte de n-BuLi pour refroidir le (4s)-benzyl-3-(1-oxo-(2S)-éthyl-(3S) mé- thyl valéroyle)-2-oxazolidinone (IV'), et le refroidissement du composé (II) et son addition en une seule fois au composé (IV') donnant du (4S)-benzyl-3-(oxo-(2S)-éthyl (3S)-méthyl valéroyle)-2-oxazolidinone ou l'addi- 10 tion par goutte de n-BuLi pour refroidir le (4R)-méthyl (5S)-phényl-3-(1-oxo-(3S)-méthyl valéroyl)-2-oxazolidi- none (IV) et le refroidissement du composé (II) et son addition en une seule fois à (IV) donnant du (4R)- méthyle-(5S) phényl-3-(1-oxo-(3S)-méthyl-(2R)-éthyl valéroyl)-2-oxazolidinone ; et

l'étape (d) comprend l'addition de H2O2 et de LiOH audit (4S)-benzyl-3-(oxo (2S)-éthyl-(3S)-méthyl valéroyle)- 15 2-oxazolidinone refroidi dans un mélange de THF/DDW ou l'addition de H2O2 et LiOH à un dit (4R)-mé- thyl-(5S)-phényl-3-(1-oxo-(3S) méthyl-(2R)-éthyle valéroyle)-2-oxazolidinone refroidi, le réchauffement du mélange résultant à température ambiante, le refroidissement à 0 °C, la trempe au sulfite de sodium, l'extrac- tion au DCM, l'obtention de l'auxiliaire chiral, ((4S)-benzyl-2-oxazohdinone ou l'auxiliaire chiral (4R, 5S)-4-mé- thyl-5-phényl-2-oxazolidinone, et l'addition par goutte de chlorure d'oxalyle pour la dissolution dans du DCM 20 anhydre, l'adition de DCM anhydre et de NH4OH, donnant le composé (V') ou le composé (V) par extraction organique.

25

30

35

40

45

50

55

14 EP 0 966 429 B1

15 EP 0 966 429 B1

16 EP 0 966 429 B1

17 EP 0 966 429 B1

18 EP 0 966 429 B1

19 EP 0 966 429 B1

20 EP 0 966 429 B1

21 EP 0 966 429 B1

22 EP 0 966 429 B1

23