WO2009004350A1 - Methods for preparing bortezomib and intermediates used in its manufacture - Google Patents

Methods for preparing bortezomib and intermediates used in its manufacture Download PDF

Info

Publication number
WO2009004350A1
WO2009004350A1 PCT/GB2008/002302 GB2008002302W WO2009004350A1 WO 2009004350 A1 WO2009004350 A1 WO 2009004350A1 GB 2008002302 W GB2008002302 W GB 2008002302W WO 2009004350 A1 WO2009004350 A1 WO 2009004350A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
acid
lithium
bortezomib
Prior art date
Application number
PCT/GB2008/002302
Other languages
French (fr)
Inventor
Michal Janca
Petr Dobrovolny
Original Assignee
Pliva Hrvatska D.O.O.
Bucks, Teresa Anne
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pliva Hrvatska D.O.O., Bucks, Teresa Anne filed Critical Pliva Hrvatska D.O.O.
Publication of WO2009004350A1 publication Critical patent/WO2009004350A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds

Definitions

  • the present invention relates generally to the synthesis of boronic acid and ester compounds.
  • it relates to the preparation of bortezomib and intermediates for the preparation of bortezomib.
  • Bortezomib also known as N-(pyrazin-2-yl)carbonyl-L-phenylalanine-L-leucine boronic acid, is a biologically active compound having the following structure.
  • Bortezomib is an N-acylated dipeptide analog of phenylalanyl-leucine in which
  • B(OH) 2 replaces the C-terminal carboxylic acid.
  • bortezomib acts to inhibit proteasome and has been clinically approved for use in treating mantle cell lymphoma and multiple myeloma. Bortezomib has been previously prepared as disclosed in U.S. Patent Publication 2005/020047 ('047 application).
  • FIGURE 1 shows a process for preparation of bortezomib as described herein.
  • Figure I shows a synthetic route to bortezomib from a compound of formula I using methods of the invention. Briefly, the known boronic acid ester having formula I is halogenated by exposure to a lithium amide base, dichloromethane, and a Lewis acid in THF to give a compound of formula II.
  • the latter compound is converted to the silyl amine (formula III) using a salt of hexamethyldisilazane and then to the ammonium salt (formula IV) upon exposure to a suitable acid.
  • a salt of the N-pyrazinyl-phenylalanine (formula V) is then coupled to the compound of formula IV without the use of any additional base to give the boronate ester of formula VI.
  • the pinanediol ester group is removed under acidic conditions to give the boronic acid, bortezomib (formula VII).
  • the product obtained from the present method improves not only the yield and purity of the compound of formula II, but also the yield and purities of the final product, bortezomib, compared to the use of other solvents (such as tert-butyldimethylether) or lower percentages of THF.
  • the invention provides methods for preparing a compound of formula II comprising exposing a compound of formula I
  • the solvent comprises about 95.5 % tetrahydrofuran and about 4.5% dichloromethane by volume.
  • the compound of formula II was prepared in 91-95% yield and contained only 3-5% starting material.
  • the method used in the '047 application provided the compound of formula II in only 77-85% yield and contained 10-15% starting material.
  • lithium amide bases for such reactions include, but are not limited to, lithium diisopropyl amide, lithium diethylamide, and lithium dimethylamide. Typical concentrations of the lithium amide base used in the present reaction may range from about 0.5 M to about 2 M.
  • suitable Lewis acids for use in the present reaction include transition metal halide such as, but not limited to, ZnCl 2 , ZnBr 2 , FeBr 3 , FeCl 3 , or a mixture of any two or more thereof.
  • the present transformation may be carried out a temperature ranging from about -70 °C to about 10 0 C.
  • the present halogenation is carried out, at least initially, at a temperature ranging from about -70 0 C to about -60 0 C.
  • the present methods of synthesizing bortezomib may further include exposing the compound of formula II to the salt of a silylamine such as, e.g., lithium hexamethyldisilazane, sodium hexamethyldisilazane, or potassium hexamethyldisilazane, under conditions suitable to provide a compound of formula III
  • a silylamine such as, e.g., lithium hexamethyldisilazane, sodium hexamethyldisilazane, or potassium hexamethyldisilazane
  • Suitable conditions for the formation of the compound of formula III include carrying out the reaction in a solvent comprising tetrahydrofuran or a mixture of tetrahydrofuran and methylcyclohexane. Formation of the silylamine may be carried out at temperatures ranging from -60°C to 30°C or from -30°C to 30°C. In some embodiments, the temperature ranges from -20 0 C to about 25 0 C.
  • the compound of formula III was obtained in 89-93% yield and containing 0.6-0.8% of an isomer of the compound of formula III. In contrast, preparation of the compound of formula III according to the method disclosed in the '047 patent resulted in only 75-80% yield and contained 8-10% of the second isomer.
  • Methods of preparing bortezomib may further include contacting the compound of formula III with a suitable acid to provide a compound of formula IV
  • Suitable acids for desilylation of silylamines are known in the art and include those strong enough to remove the silyl groups without substantially degrading the reactant (formula III) or product (formula IV).
  • suitable acids include but are not limited to C 1-10 alkanoic acids such as formic and acetic acids, C 2-10 perhaloalkanoic acids such as trifluoroacetic acid and trichloroacetic acid, HCl, HBr, or C 1-6 sulfonic acids such as methanesulfonic acid and triflic acid.
  • trifluoroacetic acid is used.
  • Formation of the compound of formula IV may be carried out at temperatures ranging from about -30°C to about 25°C over the course of 0.5 to 2.5 hours.
  • the purity of the compound of formula IV produced by present methods was 99.8% or greater with only 0.04-0.1% of a second isomer of the compound produced.
  • the process disclosed in the '047 application yielded a purity of only 87-94% and a content of 5-6% of a second isomer of the compound of formula IV.
  • bortezomib is a convergent synthesis in which the ammonium compound of formula IV is coupled to a phenylalanine derivative of formula V.
  • the latter compound may be readily made using standard techniques.
  • a suitably C- protected derivative of phenylalanine e.g. the phenylalanine methyl ester may be coupled to pyrazine carboxylic acid using any standard technique for the formation of amide bonds, particularly those used in peptide synthesis.
  • the resulting ester of N-(pyrazine-2-ylcarbonyl)-L- phenylalanine may be hydrolyzed to give the corresponding salt (formula V), which may be used for coupling with the compound of formula IV.
  • Methods of hydrolysis are well known in the art and typically include exposing an ester to a solution of metal hydroxide in water, alcohol, or mixtures of water and various alcohols or other organic solvents.
  • the present methods may further include contacting a compound of formula V
  • Z is a lithium, sodium, potassium, rubidium, calcium, or magnesium cation
  • the coupling may be effected with any suitable agent for amide bond formation such as coupling agents used for peptide synthesis.
  • suitable agent for amide bond formation such as coupling agents used for peptide synthesis.
  • Such coupling agents include but are not limited to TBTU, DIP, DCC, EDC, HBTU, HCTU, TCTU 5 HATU, PyBOP, and PyABOP.
  • Use of the compound of formula V, i.e., a salt of the carboxylic acid advantageously avoids the use of an organic amine as a base and provides superior yields to known methods of forming the compound of formula VI.
  • the present methods of synthesis of bortezomib may include exposing a compound of formula VI to a sufficient amount of acid to produce a compound of formula VII or its cyclic anhydride
  • HCl, HBr, or combinations of HCl and/or HBr with a boronic acid such as 2- methylpropylboronic acid may be used.
  • the reaction is conducted in the presence of a non-polar solvent in which pinanediol is soluble but the product boronic acid is not.
  • Step 2 (IS)-(S) -Pinanediol l-chloro-3-methylbutane-l-boronate
  • reaction mixture was warmed up to -45 °C and stirred at this temperature for 30 minutes.
  • the mixture was further warmed to 10°C over a period of 90 minutes.
  • 10% H 2 SO4 33.0 ml was added and mixture warmed up to 25 0 C.
  • tert-Buthylmethyl ether (36.0 ml) was added and mixture was transferred to separatory funnel.
  • the jacketed (250 ml) flask was washed with 20.0 ml of water and layers were separated.
  • the organic layer was extracted with 90.0 ml H 2 O; 90.0 ml H 2 O + 0.5ml 10% H 2 SO 4 , 90.0 ml H 2 O + 1.5ml 10% H 2 SO 4 ; 90.0 ml +1.5ml 10% H 2 SO 4 ; 90.0 ml 10% NaCl.
  • Step 3 (1R ⁇ )-(S * )-Pinanediol 1 bis(trimethylsilyl ' )amino-3-butane-l-boronate
  • the mixture was warmed up to -15°C and stirred for 65 minutes. The mixture was further warmed up to 25°C over a period of 25 minutes. The resulting mixture was transferred to a 250 ml flask and the jacketed (250 ml) flask was washed 2 x 5 ml hexane + 30 ml hexane and the resultant solution was concentrated to dryness under vacuum at 50 0 C. 10 ml of hexane was used for the preparation of a suspension of 0.78 g CELITE which was transferred to a filtration apparatus with a PTFE filter (45 ⁇ m). The product was dissolved in 30 ml of hexane and filtered through CELITE.
  • Step 5 Recrystallization of (lR " )-(SVPinanediol-l-ammoniumtrifluoroacetate-3-methylbutane- 1-boronate
  • the mixture was then cooled down to -5°C and stirred for 150 minutes. From - 5 0 C the mixture was warmed to 20°C and filtered. The jacketed flask was washed with 2 x 27.8 ml of isopropylether, filtered, and the filter cake was washed with 27.8 ml of isopropylether. The title product was dried in vacuo at 45 0 C to provide white woolly crystals (4.0 g, yield 74.4 %, under analytical evaluation).
  • Step 1 Methylester ofN-(pyrazin-2-yl-carbonyl)-L-phenylalanine
  • Step 2 N-(Pyrazine-2-ylcarbonyl)-L-phenylalanine sodium salt
  • Step 1 (IR)-(S)- Pinanediol N-(pyrazine-2-ylcarbonyl)-L-phenylalanine-L-leucine boronate
  • N-(Pyrazine-2-ylcarbonyl)-L-phenylalanine sodium salt (1.8 g) and TBTU (2.2 g) were suspended in dichloromethane (115 ml ) in a beaker. The suspension was charged to a jacketed flask and the beaker was washed with dichloromethane (115 ml). The suspension was cooled to 0°C and stirred for 30 minutes.
  • the mixture was extracted with 2 x 230 ml H 2 O, 2 x 230 ml 1% H 3 PO 4, 1 x 230 ml and 1 x 195 ml 2% K 2 CO 3, 2 x 230 ml 10% NaCl , 2 x 230 ml H 2 O.
  • the organic layer was transferred to a 500 ml flask and dried over Na 2 SO 4 (26.5 g) which was subsequently filtered off and washed with 100 ml dichloromethane.

Abstract

Provided herein are methods for preparing bortezomib and intermediates useful in the preparation of bortezomib. The present methods unexpectedly provide superior yields and purities, including optical purities, of bortezomib and the intermediates for the preparation of bortezomib.

Description

METHODS FOR PREPARING BORTEZOMIB AND INTERMEDIATES USED IN ITS
MANUFACTURE
FIELD OF THE INVENTION
[0001] The present invention relates generally to the synthesis of boronic acid and ester compounds. In particular it relates to the preparation of bortezomib and intermediates for the preparation of bortezomib.
BACKGROUND
[0002] Bortezomib, also known as N-(pyrazin-2-yl)carbonyl-L-phenylalanine-L-leucine boronic acid, is a biologically active compound having the following structure.
Figure imgf000002_0001
[0003] Bortezomib is an N-acylated dipeptide analog of phenylalanyl-leucine in which
B(OH)2 replaces the C-terminal carboxylic acid. Physiologically, bortezomib acts to inhibit proteasome and has been clinically approved for use in treating mantle cell lymphoma and multiple myeloma. Bortezomib has been previously prepared as disclosed in U.S. Patent Publication 2005/020047 ('047 application).
BRIEF DESCRIPTION OF THE FIGURES
[0004] FIGURE 1 shows a process for preparation of bortezomib as described herein.
DETAILED DESCRIPTION
[0005] It has been found that the methods of producing bortezomib described herein provide surprising and unexpectedly improved yields and purities of bortezomib and intermediates for the synthesis of bortezomib. Figure I shows a synthetic route to bortezomib from a compound of formula I using methods of the invention. Briefly, the known boronic acid ester having formula I is halogenated by exposure to a lithium amide base, dichloromethane, and a Lewis acid in THF to give a compound of formula II. The latter compound is converted to the silyl amine (formula III) using a salt of hexamethyldisilazane and then to the ammonium salt (formula IV) upon exposure to a suitable acid. A salt of the N-pyrazinyl-phenylalanine (formula V) is then coupled to the compound of formula IV without the use of any additional base to give the boronate ester of formula VI. The pinanediol ester group is removed under acidic conditions to give the boronic acid, bortezomib (formula VII). This synthetic route takes advantage of several unexpected discoveries.
[0006] First, a key improvement to previous methods of bortezomib synthesis includes the use of a very high percentage of tetrahydrofuran in the halogenation of the compound of formula I to provide the compound of formula II. The use of tetrahydrofuran has previously been criticized as resulting in dramatic reductions in diastereomeric ratios (see e.g. U.S. Patent Publication 2005/020047, paragraph 44) unless rigorously dried equipment, solvents and reagents are used. Yet, using only routine procedures to exclude moisture, it has been found that the product obtained from the present method improves not only the yield and purity of the compound of formula II, but also the yield and purities of the final product, bortezomib, compared to the use of other solvents (such as tert-butyldimethylether) or lower percentages of THF.
[0007] Thus, in one embodiment, the invention provides methods for preparing a compound of formula II comprising exposing a compound of formula I
Figure imgf000003_0001
to a lithium amide base and a transition metal halide in a solvent comprising dichloromethane (e.g., 2-5%) and at least 95 % tetrahydrofuran by volume, under conditions suitable to provide the compound of formula II
Figure imgf000004_0001
II
[0008] Preferably, the solvent comprises about 95.5 % tetrahydrofuran and about 4.5% dichloromethane by volume. By use of the present methods, the compound of formula II was prepared in 91-95% yield and contained only 3-5% starting material. By contrast, the method used in the '047 application provided the compound of formula II in only 77-85% yield and contained 10-15% starting material.
[0009] Without wishing to be bound by theory, it is believed that formation of the compound of formula II proceeds via a Lewis acid promoted rearrangement of a boron "ate" complex. Suitable lithium amide bases for such reactions are known in the art and include, but are not limited to, lithium diisopropyl amide, lithium diethylamide, and lithium dimethylamide. Typical concentrations of the lithium amide base used in the present reaction may range from about 0.5 M to about 2 M. Likewise, suitable Lewis acids for use in the present reaction include transition metal halide such as, but not limited to, ZnCl2, ZnBr2, FeBr3, FeCl3, or a mixture of any two or more thereof. The present transformation may be carried out a temperature ranging from about -70 °C to about 10 0C. Preferably, the present halogenation is carried out, at least initially, at a temperature ranging from about -70 0C to about -60 0C.
[0010] The present methods of synthesizing bortezomib may further include exposing the compound of formula II to the salt of a silylamine such as, e.g., lithium hexamethyldisilazane, sodium hexamethyldisilazane, or potassium hexamethyldisilazane, under conditions suitable to provide a compound of formula III
Figure imgf000005_0001
III
[0011] Suitable conditions for the formation of the compound of formula III include carrying out the reaction in a solvent comprising tetrahydrofuran or a mixture of tetrahydrofuran and methylcyclohexane. Formation of the silylamine may be carried out at temperatures ranging from -60°C to 30°C or from -30°C to 30°C. In some embodiments, the temperature ranges from -200C to about 250C. Prepared in accordance with the present methods, the compound of formula III was obtained in 89-93% yield and containing 0.6-0.8% of an isomer of the compound of formula III. In contrast, preparation of the compound of formula III according to the method disclosed in the '047 patent resulted in only 75-80% yield and contained 8-10% of the second isomer.
[0012] Methods of preparing bortezomib may further include contacting the compound of formula III with a suitable acid to provide a compound of formula IV
Figure imgf000005_0002
Suitable acids for desilylation of silylamines are known in the art and include those strong enough to remove the silyl groups without substantially degrading the reactant (formula III) or product (formula IV). For example, suitable acids include but are not limited to C1-10 alkanoic acids such as formic and acetic acids, C2-10 perhaloalkanoic acids such as trifluoroacetic acid and trichloroacetic acid, HCl, HBr, or C1-6 sulfonic acids such as methanesulfonic acid and triflic acid. In some embodiments, trifluoroacetic acid is used. Formation of the compound of formula IV may be carried out at temperatures ranging from about -30°C to about 25°C over the course of 0.5 to 2.5 hours. The purity of the compound of formula IV produced by present methods was 99.8% or greater with only 0.04-0.1% of a second isomer of the compound produced. In contrast, the process disclosed in the '047 application yielded a purity of only 87-94% and a content of 5-6% of a second isomer of the compound of formula IV.
[0013] The present synthesis of bortezomib is a convergent synthesis in which the ammonium compound of formula IV is coupled to a phenylalanine derivative of formula V. The latter compound may be readily made using standard techniques. For example, a suitably C- protected derivative of phenylalanine, e.g. the phenylalanine methyl ester may be coupled to pyrazine carboxylic acid using any standard technique for the formation of amide bonds, particularly those used in peptide synthesis. The resulting ester of N-(pyrazine-2-ylcarbonyl)-L- phenylalanine may be hydrolyzed to give the corresponding salt (formula V), which may be used for coupling with the compound of formula IV. Methods of hydrolysis are well known in the art and typically include exposing an ester to a solution of metal hydroxide in water, alcohol, or mixtures of water and various alcohols or other organic solvents.
[0014] The present methods may further include contacting a compound of formula V
Figure imgf000006_0001
V
wherein Z is a lithium, sodium, potassium, rubidium, calcium, or magnesium cation,
with a coupling agent and the compound of formula IV
Figure imgf000006_0002
under conditions suitable to provide a compound of formula VI
Figure imgf000007_0001
VI
[0015] The coupling may be effected with any suitable agent for amide bond formation such as coupling agents used for peptide synthesis. Such coupling agents include but are not limited to TBTU, DIP, DCC, EDC, HBTU, HCTU, TCTU5 HATU, PyBOP, and PyABOP. Use of the compound of formula V, i.e., a salt of the carboxylic acid, advantageously avoids the use of an organic amine as a base and provides superior yields to known methods of forming the compound of formula VI. While not wishing to be bound by theory, it is believed that the use of a salt of the N-(pyrazine-2-ylcarbonyl)-L-phenylalanine significantly reduces cleavage of the boronic acid group that results in formation of the byproduct, N-(pyrazine-2-ylcarbonyl)-L- phenylalanine-L-leucine. Thus, the present methods produced only about 1.5% of the byproduct compared to 10% using the method of the '047 application.
[0016] The present methods of synthesis of bortezomib may include exposing a compound of formula VI to a sufficient amount of acid to produce a compound of formula VII or its cyclic anhydride
Figure imgf000008_0001
VII
For example, HCl, HBr, or combinations of HCl and/or HBr with a boronic acid such as 2- methylpropylboronic acid may be used. In some embodiments, the reaction is conducted in the presence of a non-polar solvent in which pinanediol is soluble but the product boronic acid is not.
[0017] Improved methods of isolating bortezomib after deprotection are also provided.
The procedure disclosed in the '047 application requires the use of sodium hydroxide, which can cause decomposition of the bortezomib. In the present methods, the non-polar solvent is removed and the remaining residue is taken up in another solvent (e.g., dichloromethane) and precipitated with, e.g., diisopropylether to give bortezomib with enhanced final purity.
[0018] All publications, patent applications, issued patents, and other documents referred to in this disclosure are herein incorporated by reference as if each individual publication, patent application, issued patent, or other document were specifically and individually indicated to be incorporated by reference in its entirety. Definitions that are contained in text incorporated by reference are excluded to the extent that they contradict definitions in this disclosure.
[0019] The present invention, thus generally described, will be understood more readily by reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present invention.
EXAMPLES
[0020] The following abbreviations are used in the Examples and throughout this disclosure. DCC Dicyclohexylcarbodiimide
DIP Diisopropylcarbodiimide
EDC 1 -(3-Dimethylaminopropyl)-3 -ethyl-carbodiimide hydrochloride g Gram(s) GC Gas Chromatography
HATU 2-(7-Aza-lH-benzotriazole-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate HBTU O-(Benzotriazole-N,N,N',N'-tetramethyluronium hexafluorophosphate HCTU 1H-Benzotriazolium 1-
[δz5'(dimethylamino)methylene]
-5chloro-,hexafluorophosphate (l-),3-oxide
LDA Lithium diisopropylamide ml Milliliter(s)
PyABOP 7-Aza-benzotriazole- 1 -yl-oxy-tris- pyrrolidinophosphonium hexafluorphosphate
PyBOP Benzotriazole- 1 -ly-oxy-tris- pyrrolidinophosphonium hexafluorphosphate
THF Tetrahydrofuran TBTU 2-(lH-Benzotriazole-l-yl)-l, 1,3,3- tetramethyluronium tetrafluoroborate
TCTU 2-(lH-6-Chloro-benzotriazole-l-yl)-l,l,3,3- tetramethyluronium tetrafluoroborate
Example 1: Preparation of (lR)-(S)-Pinanediol-l-ammonium trifluoroacetate-3-methyIbutane-l-boronate
Step 1: (S)-Pinanediol 2-methylpropane-l-boronate
Figure imgf000010_0001
[0021] A reaction mixture containing (+)-pinanediol (6.9 g) and 2-methylpropylboronic acid (4.4 g) and diethylether (49.3 ml) was stirred for 23 hours at laboratory temperature, than dried over Na2SO4 (16.5 g). The sulfate was filtered off and washed with diethylether (69.4 ml), and the resulting solution was concentrated. The product was obtained as colorless oil (9.9 g, purity 100 % (GC), yield 97.2 %). MS (m/z): 236, 221, 195, 167, 140, 134, 83, 67, 55, 43; 1H- NMR (DMSO-d6): δ= 4.27 (IH, dd, J=2.1 Hz, 8.7 Hz); 2.30 (IH, m); 2.18 (IH, m); 1.96 (IH, t, J=5.4 Hz); 1.86 (IH, m); 1.79 (IH, sx, J-6.8 Hz); 1.69 (IH, m); 1.30 (3H, s); 1.25 (3H, s); 1.02 (IH, d, J=I 0.6 Hz); 0.9 (3H, d, J-6.6 Hz); 0.81 (3H, s); 0.69 (2H, m)).
Step 2: (IS)-(S) -Pinanediol l-chloro-3-methylbutane-l-boronate
Figure imgf000010_0002
[0022] Under an inert atmosphere of argon, 6.0 g (S)-pinanediol-2-methylpropane-l- boronate was charged to jacketed flask (250 ml) together with 36.0 ml THF and 9 ml dichloromethane. The mixture was cooled to -65°C. A 1.5 M LDA solution in THF (22.08 ml) was added to the mixture over a period of 5 minutes and the mixture stirred for additional 20 minutes at -650C. A 0.5M ZnCl2 solution in THF (135.0 ml ) was added drop wise to the reaction over a period of 18 minutes. The reaction mixture was warmed up to -45 °C and stirred at this temperature for 30 minutes. The mixture was further warmed to 10°C over a period of 90 minutes. 10% H2SO4 (33.0 ml) was added and mixture warmed up to 250C. tert-Buthylmethyl ether (36.0 ml) was added and mixture was transferred to separatory funnel. The jacketed (250 ml) flask was washed with 20.0 ml of water and layers were separated. The organic layer was extracted with 90.0 ml H2O; 90.0 ml H2O + 0.5ml 10% H2SO4 , 90.0 ml H2O + 1.5ml 10% H2SO4; 90.0 ml +1.5ml 10% H2SO4; 90.0 ml 10% NaCl. The organic layer was poured to 250 ml flask and dried over Na2SO4 (57.56 g) and stored in fridge over night. The next day the Na2SO4 was filtered off and washed with 2 x 50.0 ml tetrahydrofuran and residue evaporated to dryness (50°C, pmm= 6 mbar ). The product was isolated as oil (7.7 g, 97.0 % yield) with purity 91.7 % (GC) and content of starting material 4.9 %. MS (m/z): 284, 283, 269, 248, 214, 199, 173, 158, 145, 134, 118, 117, 83, 67, 55, 43)
Step 3 : (1R~)-(S*)-Pinanediol 1 bis(trimethylsilyl')amino-3-butane-l-boronate
Figure imgf000011_0001
[0023] Under an inert atmosphere of argon, 25.61 ml of a 1.0 M LiHMDS solution in tetrahydrofuran was charged to a jacketed flask (250 ml) and cooled down to -20°C. (IS)-(S) - Pinanediol 1-chloro —3- methylbutane — 1- boronate from the previous step (7.7 g) was dissolved in methylcyclohexane (20.97 ml) in another flask and added to the LiHMDS solution at -200C. The flask was washed 2 x 4.62 ml methylcyclohexane and the washings added to the LiHMDS solution. The mixture was warmed up to -15°C and stirred for 65 minutes. The mixture was further warmed up to 25°C over a period of 25 minutes. The resulting mixture was transferred to a 250 ml flask and the jacketed (250 ml) flask was washed 2 x 5 ml hexane + 30 ml hexane and the resultant solution was concentrated to dryness under vacuum at 500C. 10 ml of hexane was used for the preparation of a suspension of 0.78 g CELITE which was transferred to a filtration apparatus with a PTFE filter (45 μm). The product was dissolved in 30 ml of hexane and filtered through CELITE. The CELITE was washed with hexane (50 ml). The filtrate was transferred to 100 ml flask and concentrated in vacuum to dryness (500C, pmm=5 mbar). The product was isolated as oil (10.1 g, 91.62 % yield, purity 92.5 %), content of second isomer was 0.65 % (GC) and content of starting material 3.3 %; MS (m/z): 409, 394, 352, 336, 242, 202, 147, 135, 100, 93, 73, 43). Step 4 : (1 R")-(S)-Pinanediol-1 -ammonium trifluoroacetate-3-methylbutane-l-boronate
Figure imgf000012_0001
[0024] Under inert atmosphere of argon, trifluoroacetic acid (4.17 ml) was added to a jacketed flask and addition was completed by isopropylether (54.83 ml). The resulting solution was cooled to -10°C and a solution of (lR)-(S)-pinanediol-l-bis(trimethylsilyl)arnino-3-butane- 1-boronate from previous step (10.1 g) in methylcyclohexane (23.17 ml ) and isopropylether (in total 22.8 ml, 4.26 ml for washing) was added over a period of 10 minutes. The resulting mixture was stirred for 150 minutes at - 10 0C then warmed up to 20°C. The suspension was filtered, the jacketed flask was washed with 2 x 71.36 ml of isopropylether, and the isolated product was washed with 3 x 17.41 ml of isopropylether and dried in vaccuo at 45°C. The title product was obtained as white crystals (5.9 g , purity 99.8 % (GC), [α]D 25: +13.0°, impurity: 0.04 % of second isomer, yield 68.3 %)
Step 5: Recrystallization of (lR")-(SVPinanediol-l-ammoniumtrifluoroacetate-3-methylbutane- 1-boronate
[0025] ( 1R)-(S)-Pinanediol- 1 -ammonium trifluoroacetate-3 -methylbutane-1 -boronate from the previous step was dissolved in a beaker using trifluoroacetic acid (8.75 ml), and the resulting solution was transferred to a jacketed flask; the beaker was rinsed with isopropylether (16.8 ml) and the rinsing added to the jacketed flask. The resulting mixture was cooled down to 4°C and stirred for 10 minutes. The mixture was warmed up to 160C, and isopropylether (72.7 ml) was added over a period 6 minutes and the mixture stirred an additional 10 minutes. The mixture was then cooled down to -5°C and stirred for 150 minutes. From - 5 0C the mixture was warmed to 20°C and filtered. The jacketed flask was washed with 2 x 27.8 ml of isopropylether, filtered, and the filter cake was washed with 27.8 ml of isopropylether. The title product was dried in vacuo at 45 0C to provide white woolly crystals (4.0 g, yield 74.4 %, under analytical evaluation). MS (m/z): 265, 222, 208, 135, 93, 74, 55, 43; IR (cm"1): 3049, 2987, 2961, 2872, 1678, 1420, 1392, 1203, 1178, 1141, 721; 1H-NMR (DMSOd6): δ= 7.78 (3H, b); 4.44 (IH, dd, J=2 Hz, 8.9 Hz); 2.80 (IH, m); 2.34 (IH, m); 2.20 (IH, m); 2.01 (IH, t, J=5.4 Hz); 1.89 (IH, m); 1.75 (IH, m); 1.72 (IH5 sx, J=6.6 Hz); 1.48 (2H, m); 1.37 (3H, s); 1.26 (3H, s); 1.10 (IH, d, J=10.8 Hz); 0.87 (6H, d, J=6.6 Hz) 0.83 (3H,s); DSC (0C): 175.2, 177.2, 182.4, 187.8, 196.3; TGA: -0.05% (28-110°C), -2.81% (110-1680C); XRPD (° 2Θ): 8.08, 8.87, 11.72, 16.10, 17.69, 19.47, 20.15, 23.49.
Example 2: Preparation of N-(Pyrazine-2-ylcarbonyI)-L-phenylalanine sodium salt
Step 1: Methylester ofN-(pyrazin-2-yl-carbonyl)-L-phenylalanine
Figure imgf000013_0001
[0026] The methyl ester of phenylalanine (7.5 g), pyrazinecarboxylic acid (4.4 g), TBTU
(12.8 g) and dichloromethane (200 ml) were charged to a 250 ml jacketed flask. The mixture was cooled down to 00C and 18.2 ml of diisopropylethylamine were added drop wise. After 30 minutes of stirring at 0 °C, the mixture was warmed to 25 0C over a period of 90 minutes and then transferred to a separatory funnel. The jacketed flask was rinsed with dichloromethane (10 ml) and liquid transfered to the separatory funnel. The reaction mixture was extracted with 3* 225 ml of water and dried over Na2SO4 (14.5 g). The sulfate was filtered off and washed with 3χ 10 ml of dichloromethane. The filtrate was concentrated (40 °C, pmm = 30 mbar) to an oily residue (13.3 g, purity 90.0 %, yield 76.0 %). IR (cm4): 3443, 3308, 1753, 1705, 1605, 1497, 1454, 1440, 1405, 1368, 765, 749, 736, 697, 627).
Step 2 : N-(Pyrazine-2-ylcarbonyl)-L-phenylalanine sodium salt
Figure imgf000013_0002
[0027] The methyl ester of N-(pyrazin-2-yl-carbonyl)-L-phenylalanine (13.3 g) from the previous step was dissolved in methyl alcohol (23.3 ml) and added to a 100 ml flask. 2M solution of NaOH in water was added (23.3 ml). The resulting solution was stirred for 150 minutes, than 200 ml of methanol was added followed by addition of 1064 ml of diisopropylether. The resulting suspension was filtered, and the product was washed with 2χ 100.0 ml of diisopropylether. The title product was isolated as a white wool and dried in vacuo at 450C (8.6 g, purity 95.7 %, yield 63,2 %; MS (m/z): 565 [2M+Na]+; IR (cnf1): 3367, 1655, 1616, 1522, 1454, 1402, 1022, 699; 294 [M+Na]+; 1H-NMR (DMSO-d6): δ= 9.18 (IH, d, J=1.5 Hz); 8.83 (IH, d, J=2.5 Hz); 8.65 (IH, dd, J=1.5 Hz, 2.5 Hz); 8.63 (IH, ABMX, J=6.4 Hz); 7.14- 7.06 (5H, m); 4.24-4.20 (IH, ABMX); 3.25-3.13 (2H, ABMX)).
Example 3: Preparation of Bortezomib anhydride
Step 1: (IR)-(S)- Pinanediol N-(pyrazine-2-ylcarbonyl)-L-phenylalanine-L-leucine boronate
Figure imgf000014_0001
[0028] N-(Pyrazine-2-ylcarbonyl)-L-phenylalanine sodium salt (1.8 g) and TBTU (2.2 g) were suspended in dichloromethane (115 ml ) in a beaker. The suspension was charged to a jacketed flask and the beaker was washed with dichloromethane (115 ml). The suspension was cooled to 0°C and stirred for 30 minutes. (1R)-(S)-Pinanediol 1-ammonium trifluoroacetate-3- methylbutane-1 -boronate (2.3 g) in dichloromethane (115 ml) was charged to the jacketed flask, the beaker was washed with dichloromethane (115 ml), and the washing added to the jacketed flask. The resulting mixture was stirred for 30 minutes at 00C. The mixture was then warmed from O0C to 25°C over a period of 150 minutes and stirred over night (21 hours). The mixture was extracted with 2 x 230 ml H2O, 2 x 230 ml 1% H3PO4, 1 x 230 ml and 1 x 195 ml 2% K2CO3, 2 x 230 ml 10% NaCl , 2 x 230 ml H2O. The organic layer was transferred to a 500 ml flask and dried over Na2SO4 (26.5 g) which was subsequently filtered off and washed with 100 ml dichloromethane. The title product was isolated by evaporation to provide an oil (2.77 g, purity 84.0 %, yield 87.1 %); MS (m/z): 1053 βM+NEUf, 1036 [2M+H]+, 519 [M+H]+.
Step 2: Bortezomib anhydride
Figure imgf000015_0001
[0029] (IR)-(S)- Pinanediol N-(pyrazine-2-ylcarbonyl)-L-phenylalanine-L-leucine boronate (2.8 g) prepared in previous step was charged to a 250 ml flask along with methanol (23.0 ml), hexane (21.0 ml), IN HCl (11.6 ml) and 2-methylρroρylboronic acid (0.92 g). The reaction mixture was stirred over night, separated and extracted with 2 x 21.0 ml hexane. The hexane layers were discarded and the residue extracted with 2 x 25 ml of dichloromethane. The combined dichloromethane fractions were evaporated to dryness. The resulting residue was subsequently dissolved in ethyl acetate and precipitated with diisopropyl ether to yield the product, bortezomib. (1.58 g, purity 98.5-101 %; yield 80 %; MS (m/z): 1099 [MfH]+, 733 [2/3M+H]+, 367 [1/3M+H-H2O]+; IR (cm^1): 3389, 2954, 2929, 1674, 1521, 1466, 1398, 1386, 1366, 1331, 1280, 1201, 1020, 700; 1H-NMR (DMSO-d6): δ= 9.28-8.41 (2H, m); 9.11-8.60 (IH5 m); 8.67 (IH, ABMX, J=10.1 Hz); 7.75 (IH, ABMX, J=6.5 Hz, 6.7 Hz, 17.6 Hz); 7.35-6.96 (IH, m); 7.29-7.04 (2H, m); 7.23-7.10 (2H, m); 4.80 (IH, ABMX, J-16 Hz); 3.17 (IH, ABMX, J=14.3 Hz, 14.4 Hz, 16 Hz); 3.10 (IH, ABMX, J=13.8 Hz, 17.6 Hz, 18.5 Hz, 19.5 Hz); 3.07 (IH, ABMX5 J=14.3 Hz, 14.4 Hz, 16 Hz); 1.56 (IH, ABMX, J=13.05 Hz); 1.39 (IH, ABMX, J=13.1 Hz, 13.8 Hz); 1.32 (IH, ABMX, J=13.1 Hz, 13.8 Hz); 0.84 (6H, AX3, J=12.7 Hz).

Claims

CLAIMSWHAT IS CLAIMED IS:
1. A method comprising exposing a compound of formula I
Figure imgf000016_0001
I
to a lithium amide base and a transition metal halide in a solvent comprising dichloromethane and at least 95 % tetrahydrofuran by volume, under conditions suitable to provide a compound of formula II
Figure imgf000016_0002
II
2. The method of claim 3, wherein the solvent comprises about 95.5 % tetrahydrofuran and about 4.5% dichloromethane by volume.
3. The method of claim 1 wherein the lithium amide base is lithium diisopropyl amide, lithium diethylamide, or lithium dimethylamide.
4. The method of claim 4 wherein the concentration of the lithium amide base ranges from about 0.5 M to about 2 M.
5. The method of claim 1 wherein the transition metal halide is ZnCl2, ZnBr2, FeBr3, FeCl3, or a mixture of any two or more thereof.
6. The method of claim 1 wherein the method is carried out a temperature ranging from about -70 0C to about 10 °C.
7. The method of claim 1 wherein the method is carried out a temperature ranging from about -70 0C to about -60 °C.
8. The method of claim 1 further comprising exposing the compound of formula II to lithium hexamethyldisilazane, sodium hexamethyldisilazane, or potassium hexamethyldisilazane under conditions suitable to provide a compound of formula III
Figure imgf000017_0001
III
9. The method of claim 8 wherein the formation of the compound of formula III is carried out in a solvent comprising tetrahydrofuran.
10. The method of claim 8 wherein the formation of the compound of formula III is carried out in a solvent comprising methylcyclohexane and tetrahydrofuran.
11. The method of claim 8 further comprising contacting the compound of formula III with a suitable acid to provide a compound of formula IV
Figure imgf000017_0002
IV
12. The method of claim 11 wherein the acid is a Ci-10 alkanoic acid, C2-10 perhaloalkanoic acid, HCl, HBr, or C1-6 sulfonic acid.
13. The method of claim 11 wherein the acid is trifluoroacetic acid.
14. A method comprising contacting a compound of formula V
Figure imgf000018_0001
V wherein Z is a lithium, sodium, potassium, rubidium, calcium, or magnesium cation, with a coupling agent and the compound of formula IV
Figure imgf000018_0002
IV under conditions suitable to provide a compound of formula VI
Figure imgf000018_0003
VI
15. The method of claim 12 wherein the coupling agent is TBTU, DIP, DCC, EDC, HBTU, HCTU, TCTU, HATU, PyBOP, or PyABOP.
16. The method of claim 12 further comprising exposing a compound of formula VI to a sufficient amount of acid to produce a compound of formula VII
Figure imgf000019_0001
VII
17. The method of claim 16 wherein the acid is HCl or HBr.
18. The method of claim 16 wherein the exposure of the compound of formula VI to acid is carried out in the presence of a non-polar solvent in which pinanediol is soluble but the compound of formula VII is not.
PCT/GB2008/002302 2007-07-03 2008-07-02 Methods for preparing bortezomib and intermediates used in its manufacture WO2009004350A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US94783807P 2007-07-03 2007-07-03
US60/947,838 2007-07-03

Publications (1)

Publication Number Publication Date
WO2009004350A1 true WO2009004350A1 (en) 2009-01-08

Family

ID=39791680

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2008/002302 WO2009004350A1 (en) 2007-07-03 2008-07-02 Methods for preparing bortezomib and intermediates used in its manufacture

Country Status (1)

Country Link
WO (1) WO2009004350A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2185159A2 (en) * 2007-09-12 2010-05-19 Dr. Reddy's Laboratories, Ltd. Bortezomib and process for producing same
CN101812026A (en) * 2010-04-12 2010-08-25 孙江涛 Method for synthesizing bortezomib
CN101899062A (en) * 2009-05-26 2010-12-01 上海威智医药科技有限公司 Synthesis technology of alpha-chiral boric acid and boric acid ester
WO2010146172A2 (en) 2009-06-19 2010-12-23 Lek Pharmaceuticals D.D. NEW SYNTHETIC ROUTE FOR THE PREPARATION OF α-AMINO BORONIC ACID DERIVATIVES VIA SUBSTITUTED ALK-1-YNES
EP2270019A1 (en) 2009-06-19 2011-01-05 LEK Pharmaceuticals d.d. New synthetic route for the preparation of alpha-amino boronic esters
EP2280016A1 (en) 2009-07-27 2011-02-02 LEK Pharmaceuticals d.d. New synthetic route for the preparation of alpha-amino boronic esters via substituted alk-1-ynes
WO2011098963A1 (en) 2010-02-09 2011-08-18 Ranbaxy Laboratories Limited Process for the preparation of bortezomib
EP2377868A1 (en) * 2004-03-30 2011-10-19 Millennium Pharmaceuticals, Inc. Synthesis of Bortezomib
WO2012048745A1 (en) 2010-10-14 2012-04-19 Synthon Bv Process for making bortezomib and intermediates for the process
CN102675415A (en) * 2012-05-11 2012-09-19 武汉人福医药集团股份有限公司 Method for preparing bortezomib
CN103012551A (en) * 2012-12-14 2013-04-03 江苏奥赛康药业股份有限公司 Synthetic method of high-purity bortezomib and intermediate thereof
CN103044468A (en) * 2012-11-28 2013-04-17 深圳万乐药业有限公司 Preparation method of N-(2-pyrazine carbonyl)-L-phenylalanine-L- leucine boracic acid
CN103204867A (en) * 2012-01-14 2013-07-17 成都爱群科技有限公司 chiral alpha-amino boric acid esters, a preparation method and an application in the synthesis of bortezomib thereof
US8497374B2 (en) 2011-05-12 2013-07-30 Scinopharm Taiwan, Ltd. Process for preparing and purifying bortezomib
WO2014170628A1 (en) 2013-04-16 2014-10-23 Cipla Limited Process for the preparation of bortezomib mannitol ester
CN104387409A (en) * 2012-05-11 2015-03-04 人福医药集团股份公司 Catalyst and applications thereof
KR20150066724A (en) * 2013-12-09 2015-06-17 주식회사 경보제약 Manufacturing method for Bortezomib and new intermediate thereof
US9505787B2 (en) 2012-09-11 2016-11-29 Cipla Limited Process for preparing of bortezomib
WO2018150386A1 (en) 2017-02-17 2018-08-23 Fresenius Kabi Oncology Ltd. An improved process for the preparation of boronic acid esters
CN117820347A (en) * 2024-03-06 2024-04-05 深圳智微通科技有限公司 Method for synthesizing bortezomib by continuous flow

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4525309A (en) * 1983-03-15 1985-06-25 Washington State University Research Foundation, Inc. Lewis acid catalysis of the homologation of boronic esters with haloalkylmetal reagents
WO2005097809A2 (en) * 2004-03-30 2005-10-20 Millennium Pharmaceuticals, Inc. Synthesis of boronic ester and acid compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4525309A (en) * 1983-03-15 1985-06-25 Washington State University Research Foundation, Inc. Lewis acid catalysis of the homologation of boronic esters with haloalkylmetal reagents
WO2005097809A2 (en) * 2004-03-30 2005-10-20 Millennium Pharmaceuticals, Inc. Synthesis of boronic ester and acid compounds
US20050240047A1 (en) * 2004-03-30 2005-10-27 Millennium Pharmaceuticals, Inc. Synthesis of boronic ester and acid compounds

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2377868A1 (en) * 2004-03-30 2011-10-19 Millennium Pharmaceuticals, Inc. Synthesis of Bortezomib
EP2185159A2 (en) * 2007-09-12 2010-05-19 Dr. Reddy's Laboratories, Ltd. Bortezomib and process for producing same
EP2185159A4 (en) * 2007-09-12 2012-02-08 Reddys Lab Ltd Dr Bortezomib and process for producing same
CN101899062A (en) * 2009-05-26 2010-12-01 上海威智医药科技有限公司 Synthesis technology of alpha-chiral boric acid and boric acid ester
US9315525B2 (en) 2009-06-19 2016-04-19 Lek Pharmaceuticals D.D. Synthetic route for the preparation of α-amino boronic acid derivatives via substituted alk-1-ynes
WO2010146172A2 (en) 2009-06-19 2010-12-23 Lek Pharmaceuticals D.D. NEW SYNTHETIC ROUTE FOR THE PREPARATION OF α-AMINO BORONIC ACID DERIVATIVES VIA SUBSTITUTED ALK-1-YNES
EP2270019A1 (en) 2009-06-19 2011-01-05 LEK Pharmaceuticals d.d. New synthetic route for the preparation of alpha-amino boronic esters
WO2010146172A3 (en) * 2009-06-19 2012-02-23 Lek Pharmaceuticals D.D. New synthetic route for the preparation of alpha-amino boronic esters
US8785674B2 (en) 2009-06-19 2014-07-22 Lek Pharmaceuticals D.D. Process for hydrogenation of halogenoalkenes without dehalogenation
EP2280016A1 (en) 2009-07-27 2011-02-02 LEK Pharmaceuticals d.d. New synthetic route for the preparation of alpha-amino boronic esters via substituted alk-1-ynes
WO2011098963A1 (en) 2010-02-09 2011-08-18 Ranbaxy Laboratories Limited Process for the preparation of bortezomib
CN101812026B (en) * 2010-04-12 2013-08-28 亚邦医药股份有限公司 Method for synthesizing bortezomib
CN101812026A (en) * 2010-04-12 2010-08-25 孙江涛 Method for synthesizing bortezomib
WO2012048745A1 (en) 2010-10-14 2012-04-19 Synthon Bv Process for making bortezomib and intermediates for the process
US8884009B2 (en) 2010-10-14 2014-11-11 Synthon Bv Process for making bortezomib and intermediates for the process
US8497374B2 (en) 2011-05-12 2013-07-30 Scinopharm Taiwan, Ltd. Process for preparing and purifying bortezomib
CN103204867A (en) * 2012-01-14 2013-07-17 成都爱群科技有限公司 chiral alpha-amino boric acid esters, a preparation method and an application in the synthesis of bortezomib thereof
CN104387409A (en) * 2012-05-11 2015-03-04 人福医药集团股份公司 Catalyst and applications thereof
CN102675415A (en) * 2012-05-11 2012-09-19 武汉人福医药集团股份有限公司 Method for preparing bortezomib
CN104387409B (en) * 2012-05-11 2017-09-29 人福医药集团股份公司 Catalyst and its application
US9505787B2 (en) 2012-09-11 2016-11-29 Cipla Limited Process for preparing of bortezomib
CN103044468A (en) * 2012-11-28 2013-04-17 深圳万乐药业有限公司 Preparation method of N-(2-pyrazine carbonyl)-L-phenylalanine-L- leucine boracic acid
CN103012551A (en) * 2012-12-14 2013-04-03 江苏奥赛康药业股份有限公司 Synthetic method of high-purity bortezomib and intermediate thereof
US10023611B2 (en) 2013-04-16 2018-07-17 Cipla Limited Process for the preparation of bortezomib mannitol ester
WO2014170628A1 (en) 2013-04-16 2014-10-23 Cipla Limited Process for the preparation of bortezomib mannitol ester
KR101691353B1 (en) 2013-12-09 2016-12-30 주식회사 경보제약 Manufacturing method for Bortezomib and new intermediate thereof
KR20150066724A (en) * 2013-12-09 2015-06-17 주식회사 경보제약 Manufacturing method for Bortezomib and new intermediate thereof
AU2018221670B2 (en) * 2017-02-17 2021-02-04 Fresenius Kabi Oncology Ltd. An improved process for the preparation of boronic acid esters
WO2018150386A1 (en) 2017-02-17 2018-08-23 Fresenius Kabi Oncology Ltd. An improved process for the preparation of boronic acid esters
CN110312727A (en) * 2017-02-17 2019-10-08 费森尤斯卡比肿瘤学有限公司 A kind of improved method for preparing borate
JP2020507615A (en) * 2017-02-17 2020-03-12 フレゼニウス・カビ・オンコロジー・リミテッド Improved process for the preparation of boronic esters
US11667654B2 (en) 2017-02-17 2023-06-06 Fresenius Kabi Oncology Ltd. Process for the preparation of boronic acid esters
CN117820347A (en) * 2024-03-06 2024-04-05 深圳智微通科技有限公司 Method for synthesizing bortezomib by continuous flow

Similar Documents

Publication Publication Date Title
WO2009004350A1 (en) Methods for preparing bortezomib and intermediates used in its manufacture
CN111925385B (en) Synthesis of esters and acid compounds of boron
US5384410A (en) Removal of boronic acid protecting groups by transesterification
JP4717117B2 (en) Method for producing glycerol derivative and intermediate thereof
CN102143967B (en) Purification method for adefovir dipivoxil
AU2013380572B2 (en) Sodium salt of (2S, 5R)-6-benzyloxy-7-oxo-1,6-diaza-bicyclo [3.2.1 ] octane-2-carboxylic acid and its preparation
KR19980018088A (en) Stereoselective Preparation of 4-acetoxyazetidinone
WO2014141092A2 (en) Improved process for the preparation of tenofovir
EP2794632B1 (en) Process for alkynylating 16-substituted-17-keto steroids
KR20040108717A (en) Method for preparing combretastatins
JP3089037B2 (en) Method for preparing specific azacyclohexapeptides
CA2575280C (en) Novel processes for stereoselective synthesis of trans isatx 247
CN1678560A (en) Process for preparing nitrooxyderivatives of naproxen
CN106554301A (en) A kind of preparation method of BMS-477118 key intermediate
JPH10505077A (en) Protecting or anchoring groups and their use
RU2402564C2 (en) Method of obtaining pyrazinoyl-(l)-phenylalanyl-(l)-leucine boronic acid or its anhydride
CN1197056A (en) Chlorination of substituted alkenes using trichloroisocyanuric acid
JPH08245612A (en) Production of 2-deoxy-2,2-difluoro-beta-d-libopentopyranose
EP4201952A1 (en) Process for the controlled synthesis of voclosporin
JP4220658B2 (en) Method for producing tertiary alcohol having adamantane skeleton
EP1426356A1 (en) Intermediate compounds for the preparation of mirtazapine and the production methods thereof
JP3888914B2 (en) Highly fluorinated carboxylic acid derivatives and processes and intermediates thereof
JPS6254104B2 (en)
RU2450011C1 (en) Method of producing n-pyrazinoyl-l-phenylalanyl-l-leucine boronic acid or anhydride thereof
WO2020206367A1 (en) Processes for the preparation of deuterated d-serine

Legal Events

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

Ref document number: 08775850

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08775850

Country of ref document: EP

Kind code of ref document: A1