WO2009039361A2 - Process for the preparation of (s)-2-(3-tert-butylureido)-3,3-dimethylbutanoic acid - Google Patents

Process for the preparation of (s)-2-(3-tert-butylureido)-3,3-dimethylbutanoic acid Download PDF

Info

Publication number
WO2009039361A2
WO2009039361A2 PCT/US2008/077001 US2008077001W WO2009039361A2 WO 2009039361 A2 WO2009039361 A2 WO 2009039361A2 US 2008077001 W US2008077001 W US 2008077001W WO 2009039361 A2 WO2009039361 A2 WO 2009039361A2
Authority
WO
WIPO (PCT)
Prior art keywords
accordance
therapeutic agent
tert
dimethylbutanoic acid
silyl ester
Prior art date
Application number
PCT/US2008/077001
Other languages
French (fr)
Other versions
WO2009039361A3 (en
Inventor
Jeffrey M. Dener
Original Assignee
Virobay, Inc.
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 Virobay, Inc. filed Critical Virobay, Inc.
Publication of WO2009039361A2 publication Critical patent/WO2009039361A2/en
Publication of WO2009039361A3 publication Critical patent/WO2009039361A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals

Definitions

  • the compound of formula I is identified as a hepatitis C virus ("HCV") protease inhibitor, useful for treating hepatitis C and related disorders. Specifically, the compound of formula I is identified as an inhibitor of the HCV NS3/NS4a serine protease.
  • HCV hepatitis C virus
  • HCV hepatitis C virus
  • the present invention provides a process for the preparation of (S)-2- (3-ter?-Butylureido)-3,3-dimethylbutanoic acid, comprising:
  • step (c) working up the reaction of step (b) to provide ( ⁇ S)-2-(3-tert-Butylureido)-3,3- dimethylbutanoic acid as a solid.
  • reaction workup in step (c) involves
  • step (cl) contacting the mixture produced in step (b) with H 2 O to quench unreacted tert- butylisocyanate;
  • step (c2) acidifying the mixture produced in step (cl) to a pH of from 1 to 4;
  • the process of the present invention can be, and is preferably, run in a single reaction vessel. Moreover, the process can be run on large scale, typically a kilogram or multi-kilogram scale.
  • the present invention provides a method for the preparation of a therapeutic agent, said method comprising preparing a (>S)-2-(3-tert-Butylureido)-3,3- dimethylbutanoic acid intermediate by the process described herein and coupling the intermediate with a suitable proline derivative to produce said therapeutic agent or a therapeutic agent precursor.
  • Figure 1 provides a reaction scheme for the preparation of (S)-2-(3-tert- Butylureido)-3,3-dimethylbutanoic acid using tert-butylisocyanate.
  • Figure 2 provides a reaction scheme for the preparation of (S)-2-(3-tert- Butylureido)-3,3-dimethylbutanoic acid using a fert-butylisocyanate equivalent.
  • Figure 3 illustrates the coupling of (iS)-2-(3-tert-Butylureido)-3, 3 -dimethylbutanoic acid to a proline derivative to produce a therapeutic agent precursor, which on oxidation is converted to a therapeutic agent.
  • the present invention provides a process for the preparation of a key intermediate in the production of HCV protease inhibitors.
  • the compound (S)- 2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid had been prepared via a multistep process involving isolation and, in some cases, purification of intermediates.
  • the present invention provides a process that is conveniently scalable and can be run in a single reaction vessel without isolation of intermediates.
  • the present invention provides a for the preparation of (5)-2-(3-tert-Butylureido)-3, 3 -dimethylbutanoic acid, comprising:
  • step (b) contacting said in situ silyl ester of (5)-2-amino-3, 3 -dimethylbutanoic acid with tert-butylisocyanate or a synthetic equivalent thereof; and (c) working up the reaction of step (b) to provide (S)-2-(3-f ⁇ rMButylureido)-3,3- dimethylbutanoic acid as a solid.
  • the starting material, (5)-2-amino-3,3-dimethylbutanoic acid (also referred to as tert-butyl-L-glycine or L-tert- Leucine) can be obtained from commercial sources such as Aldrich (Milwaukee, WI).
  • a silyl ester is formed in situ by contacting the amino acid with a silyl ester- forming reagent.
  • suitable silyl ester- forming reagents include trimethylsilylchloride, triethylsilylchloride, triisopropylsilylchloride, tert- butyldimethylsilylchloride, isopropyldimethylsilylchloride, hexamethyldisilazane, phenyldimethylsilylchloride, diisopropylmethylsilylchloride and combinations thereof.
  • the amount of silyl ester- forming reagent can vary, typically from an excess on a mole % basis (e.g., 110 mole %, 200 mole %, 300 mole %, or more) to a catalytic amount on a mole % basis (e.g., 0.5 mole %, 1.0 mole %, 2.0 mole %, 3-10 mole %). In one group of embodiments an amount of more than 100 mole % of silyl ester-forming reagent is used. In another group of embodiments, hexamethyldisilaze is used in combination with an amount of trimethylsilyl chloride that is less than 10 mole %.
  • a preferred silyl ester- forming reagent is trimethylsilyl chloride (or chlorotrimethylsilane).
  • silyl ester forming reagents can be used so long as the ester can be conveniently removed during workup.
  • the contacting step (a) is performed in the presence of a solvent such as methylene chloride, chloroform, dichloroethane, or tetrahydrofuran; with an amine base such as triethylamine, N,N-diisopropylethylamine, N-methylmorpholine, N- ethylpiperidine, N-methylpiperidine, N-ethylmorpholine, pyridine, 2,6-dimethylpyridine and N-methylpyrrolidine.
  • Preferred reagents include trimethylsilylchloride or triethylsilylchloride, in combination with triethylamine or N-methylmorpholine.
  • Preferred solvents are the chlorinated hydrocarbons such as methylene chloride and chloroform.
  • the reaction conditions for the formation of the silyl ester will vary, depending on the solvent used, the base used, the silylating reagent used, and other factors such as the concentration of all reactants. Generally, the reaction is run at temperatures of from O 0 C to the refluxing temperature of the solvent. For those instances in which an elevated temperature is used (relative to room temperature of about 25-28°C), the mixture can be held at the elevated temperature for about two to ten hours, generally about two to four hours. [0017] Following formation of the in situ silyl ester of (5)-2-amino-3,3-dimethylbutanoic acid, the ester is contacted with tert-butylisocyanate or a synthetic equivalent thereof.
  • the contacting is conveniently performed by addition of tert-butylisocyanate to the reaction mixture containing the in situ silyl ester, so that only a single reaction vessel need be employed.
  • the mixture will typically be cooled to room temperature or lower before the addition of tert-butylisocyanate.
  • the temperature of the reaction mixture is adjusted to 20-25°C prior to isocyanate addition.
  • the tert-butylisocyanate can be added neat (without solvent), or in a solvent or mixture of solvents. Generally, a single equivalent is used, or a small excess. In some embodiments, a synthetic equivalent of fert-butylisocyanate is used.
  • a synthetic equivalent of tert-butylisocyanate can be derived (generally in situ) from, for example, tert-butylamine and an aryl chloroformate (e.g., phenyl chloroformate or 4-nitrophenyl chloroformate).
  • Procedures for the in situ formation of an isocyanate equivalent are known to the skilled artisan and can be found in, for example, REAGENTS FOR ORGANIC SYNTHESIS, M. Fieser, ed., John Wiley & Sons, New York, NY (1967-2000). Additional guidance for the preparation of ureas from a carbamate (a stable isocyanate equivalent) can be found in Thavonekham, B. Synthesis 1997, 1189-1194.
  • reaction can be monitored by thin layer chromatography. Upon complete reaction, as evidenced by disappearance of starting material (tert-butyl-L-glycine), the reaction mixture is worked up in a conventional manner.
  • reaction workup in step (c) can be accomplished using a variety of techniques known to those of skill in the art. Generally, the workup involves:
  • step (cl) contacting the mixture produced in step (b) with H 2 O to quench unreacted tert- butylisocyanate; and (c2) acidifying the mixture produced in step (cl) to a pH of from 1 to 4; and in some embodiments, (c3) filtering and drying the product.
  • the mixture will generally be cooled, water will be added and the aqueous mixture will be acidified to a pH of from about 1 to 4.
  • the mixture will be cooled to about 0- 20°C, more preferably about 10-20°C, before acid is added to remove the silyl ester and provide (5)-2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid as a solid.
  • a number of different acids can be used in the present invention, although HCl, HBr, H 3 PO 4 , KHSO 4 and H 2 SO 4 are preferred. In the most preferred embodiments, HCl is used to acidify the reaction mixture.
  • the product is often obtained as a solid and can be isolated by, for example, filtration, and then dried to removed traces of solvent and/or moisture.
  • the present invention provides methods for the preparation of a therapeutic agent, the method comprising preparing a (5)-2-(3-terMButylureido)-3,3- dimethylbutanoic acid according to the processes as described above, and coupling the acid with a suitable proline derivative to produce said therapeutic agent or a therapeutic agent precursor.
  • the coupling process can use any of the known methods for amide formation.
  • the target therapeutic agents are generally selected from:
  • a proline derivative having a fused gem- dimethylcyclopropane at the 3- and 4-positions of the proline can be coupled with ( ⁇ S)-2-(3- /erM3utylureido)-3,3-dimethylbutanoic acid, and optionally oxidized to produce A.
  • ( ⁇ S)-2-(3- /erM3utylureido)-3,3-dimethylbutanoic acid can be coupled with ( ⁇ S)-2-(3- /erM3utylureido)-3,3-dimethylbutanoic acid, and optionally oxidized to produce A.
  • reaction mixture was cooled to 20-25 0 C and tert-butyl isocyanate (0.76 kg; 7.67 mol; 101 mole %) was added to the reaction mass at 20- 25 0 C
  • the reaction temperature was adjusted to 25-30 0 C and the resulting mixture stirred for 14-15 hours at 25-30 0 C.
  • the reaction mixture was monitored by thin layer chromatography (TLC) for the complete disappearance of tert-butyl-L-glycine. Once TLC confirmed the absence of the starting material, the reaction mixture was treated with water (4 L) at 25-30 °C.
  • the reaction was cooled to 15-20 0 C and the reaction mixture was acidified to a pH of 1.0 to 2.0 by the addition of 6N aqueous hydrochloric acid (1.0 L; 6.0 mol) at 15-20 0 C. Precipitation of the product was observed after the addition of the acid was complete.
  • the mixture was stirred for 20-30 minutes and the slurry was filtered. The filter cake was washed with water (1.00 L) and allowed to dry under vacuum on the filter until no additional solvent was collected in the receiver. The solid was transferred to polypropylene trays and dried at 60-70 0 C until loss on drying was below 0.50%. Yields of compound 3 (see Figure 1) were 1.30-1.56 kg (73.9-88.6%) for three runs. HPLC Purity of these lots ranged from 99.00 to 99.78%.
  • tert-Butyl-L-leucine 100 g, 0.76 mol
  • DCM dichloromethane
  • HMDS Hexamethyldisilazane
  • a catalytic amount of chlorotrimethylsilane 2.4 mL; 0.018 mol; 2 mole%).
  • the contents were reflux ed for 3 h, and then cooled to RT.
  • tert-Butylisocyanate (86.76 mL; 0.76 mol; 100 mole%) was added at ambient temperature over 30 min. and then stirred at ambient temperature for about 16 h, monitoring the progress of the reaction by TLC (elution solvent: 20% methanol in DCM; visualization by dipping the dry plate in a ninhydrin solution, then heating until the spots appear).
  • TLC elution solvent: 20% methanol in DCM; visualization by dipping the dry plate in a ninhydrin solution, then heating until the spots appear.
  • water 1.0 L
  • the reaction mass was stirred for 1 h at ambient temperature then the DCM was distilled under vacuum.
  • the reaction mass was cooled to 10-15 0 C and stirred for 30 minutes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

(S)-2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid is conveniently prepared in a single reaction vessel from commercially available starting materials in a process that can be run on a kilogram or larger scale.

Description

PROCESS FOR THE PREPARATION OF (S)-2-(3-TERT- BUTYLUREIDO)-3,3-DIMETHYLBUTANOIC ACID
CROSS-REFERENCES TO RELATED APPLICATIONS [0001] This application claims the benefit of US Provisional Application, Serial No. 60/974,012, filed September 20, 2007, the content of which is incorporated herein by reference.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0002] NOT APPLICABLE
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK. [0003] NOT APPLICABLE
BACKGROUND OF THE INVENTION
[0004] Compounds having a (<S)-2-(3-tert-Butylureido)-3,3-dimethylbutanoyl group have been identified as useful for the treatment of hepatitis C and related disorders, hi particular, (Ii?, 2S, 55)-3-Azabicyclo[3,l,0]hexane-2-carboxamide, N-[3-amino-l-(cyclobutylmethyl)- 2,3-dioxopropyl]-3-[(25)-2-[[[l,l-dimethylethyl]amino]carbonylamino]-3,3-dimethyl-l- oxobutyl]-6,6-dimethyl is disclosed in U.S. Ser. No. 09/908,955 which was filed JuI. 19, 2001, and Ser. No. 10/052,386 which was filed Jan. 18, 2002, which are each incorporated herein by reference. The compound of formula I is identified as a hepatitis C virus ("HCV") protease inhibitor, useful for treating hepatitis C and related disorders. Specifically, the compound of formula I is identified as an inhibitor of the HCV NS3/NS4a serine protease.
Figure imgf000003_0001
(I)
In view of the importance of hepatitis C virus ("HCV") protease inhibitors, new, novel methods and intermediates for preparing such inhibitors are of interest.
BRIEF SUMMARY OF THE INVENTION
[0005] In one aspect, the present invention provides a process for the preparation of (S)-2- (3-ter?-Butylureido)-3,3-dimethylbutanoic acid, comprising:
(a) contacting (5)-2-amino-3,3-dimethylbutanoic acid with a silyl ester forming reagent under conditions sufficient to form an in situ silyl ester of (S^-amino-S, 3- dimethylbutanoic acid;
(b) contacting said in situ silyl ester of (5)-2-amino-3,3-dimethylbutanoic acid with tert-butylisocyanate or a synthetic equivalent thereof; and
(c) working up the reaction of step (b) to provide (<S)-2-(3-tert-Butylureido)-3,3- dimethylbutanoic acid as a solid.
[0006] Typically, the reaction workup in step (c) involves
(cl) contacting the mixture produced in step (b) with H2O to quench unreacted tert- butylisocyanate;
(c2) acidifying the mixture produced in step (cl) to a pH of from 1 to 4; and in some embodiments,
(c3) filtering and drying the product.
[0007] The process of the present invention can be, and is preferably, run in a single reaction vessel. Moreover, the process can be run on large scale, typically a kilogram or multi-kilogram scale.
[0008] In a related aspect, the present invention provides a method for the preparation of a therapeutic agent, said method comprising preparing a (>S)-2-(3-tert-Butylureido)-3,3- dimethylbutanoic acid intermediate by the process described herein and coupling the intermediate with a suitable proline derivative to produce said therapeutic agent or a therapeutic agent precursor.
BRIEF DESCRIPTION OF THE DRAWINGS
[0009] Figure 1 provides a reaction scheme for the preparation of (S)-2-(3-tert- Butylureido)-3,3-dimethylbutanoic acid using tert-butylisocyanate.
[0010] Figure 2 provides a reaction scheme for the preparation of (S)-2-(3-tert- Butylureido)-3,3-dimethylbutanoic acid using a fert-butylisocyanate equivalent.
[0011] Figure 3 illustrates the coupling of (iS)-2-(3-tert-Butylureido)-3, 3 -dimethylbutanoic acid to a proline derivative to produce a therapeutic agent precursor, which on oxidation is converted to a therapeutic agent.
DETAILED DESCRIPTION OF THE INVENTION
[0012] The present invention provides a process for the preparation of a key intermediate in the production of HCV protease inhibitors. Prior to the present invention, the compound (S)- 2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid had been prepared via a multistep process involving isolation and, in some cases, purification of intermediates. The present invention provides a process that is conveniently scalable and can be run in a single reaction vessel without isolation of intermediates.
[0013] Accordingly, in one aspect, the present invention provides a for the preparation of (5)-2-(3-tert-Butylureido)-3, 3 -dimethylbutanoic acid, comprising:
(a) contacting (5)-2-amino-3,3-dimethylbutanoic acid with a silyl ester forming reagent under conditions sufficient to form an in situ silyl ester of (5)-2-amino- 3, 3 -dimethylbutanoic acid;
(b) contacting said in situ silyl ester of (5)-2-amino-3, 3 -dimethylbutanoic acid with tert-butylisocyanate or a synthetic equivalent thereof; and (c) working up the reaction of step (b) to provide (S)-2-(3-førMButylureido)-3,3- dimethylbutanoic acid as a solid.
[0014] The starting material, (5)-2-amino-3,3-dimethylbutanoic acid (also referred to as tert-butyl-L-glycine or L-tert- Leucine) can be obtained from commercial sources such as Aldrich (Milwaukee, WI).
[0015] As an initial step, a silyl ester is formed in situ by contacting the amino acid with a silyl ester- forming reagent. Examples of suitable silyl ester- forming reagents include trimethylsilylchloride, triethylsilylchloride, triisopropylsilylchloride, tert- butyldimethylsilylchloride, isopropyldimethylsilylchloride, hexamethyldisilazane, phenyldimethylsilylchloride, diisopropylmethylsilylchloride and combinations thereof. The amount of silyl ester- forming reagent can vary, typically from an excess on a mole % basis (e.g., 110 mole %, 200 mole %, 300 mole %, or more) to a catalytic amount on a mole % basis (e.g., 0.5 mole %, 1.0 mole %, 2.0 mole %, 3-10 mole %). In one group of embodiments an amount of more than 100 mole % of silyl ester-forming reagent is used. In another group of embodiments, hexamethyldisilaze is used in combination with an amount of trimethylsilyl chloride that is less than 10 mole %. A preferred silyl ester- forming reagent is trimethylsilyl chloride (or chlorotrimethylsilane). One of skill in the art will appreciate that other silyl ester forming reagents can be used so long as the ester can be conveniently removed during workup. Generally, the contacting step (a) is performed in the presence of a solvent such as methylene chloride, chloroform, dichloroethane, or tetrahydrofuran; with an amine base such as triethylamine, N,N-diisopropylethylamine, N-methylmorpholine, N- ethylpiperidine, N-methylpiperidine, N-ethylmorpholine, pyridine, 2,6-dimethylpyridine and N-methylpyrrolidine. Preferred reagents include trimethylsilylchloride or triethylsilylchloride, in combination with triethylamine or N-methylmorpholine. Preferred solvents are the chlorinated hydrocarbons such as methylene chloride and chloroform.
[0016] The reaction conditions for the formation of the silyl ester will vary, depending on the solvent used, the base used, the silylating reagent used, and other factors such as the concentration of all reactants. Generally, the reaction is run at temperatures of from O0C to the refluxing temperature of the solvent. For those instances in which an elevated temperature is used (relative to room temperature of about 25-28°C), the mixture can be held at the elevated temperature for about two to ten hours, generally about two to four hours. [0017] Following formation of the in situ silyl ester of (5)-2-amino-3,3-dimethylbutanoic acid, the ester is contacted with tert-butylisocyanate or a synthetic equivalent thereof. The contacting is conveniently performed by addition of tert-butylisocyanate to the reaction mixture containing the in situ silyl ester, so that only a single reaction vessel need be employed. For those embodiments in which elevated temperatures are used for the in situ silyl ester formation, the mixture will typically be cooled to room temperature or lower before the addition of tert-butylisocyanate. In one embodiment, the temperature of the reaction mixture is adjusted to 20-25°C prior to isocyanate addition. The tert-butylisocyanate can be added neat (without solvent), or in a solvent or mixture of solvents. Generally, a single equivalent is used, or a small excess. In some embodiments, a synthetic equivalent of fert-butylisocyanate is used. A synthetic equivalent of tert-butylisocyanate can be derived (generally in situ) from, for example, tert-butylamine and an aryl chloroformate (e.g., phenyl chloroformate or 4-nitrophenyl chloroformate). Procedures for the in situ formation of an isocyanate equivalent are known to the skilled artisan and can be found in, for example, REAGENTS FOR ORGANIC SYNTHESIS, M. Fieser, ed., John Wiley & Sons, New York, NY (1967-2000). Additional guidance for the preparation of ureas from a carbamate (a stable isocyanate equivalent) can be found in Thavonekham, B. Synthesis 1997, 1189-1194.
[0018] The reaction can be monitored by thin layer chromatography. Upon complete reaction, as evidenced by disappearance of starting material (tert-butyl-L-glycine), the reaction mixture is worked up in a conventional manner.
[0019] The reaction workup in step (c) can be accomplished using a variety of techniques known to those of skill in the art. Generally, the workup involves:
(cl) contacting the mixture produced in step (b) with H2O to quench unreacted tert- butylisocyanate; and (c2) acidifying the mixture produced in step (cl) to a pH of from 1 to 4; and in some embodiments, (c3) filtering and drying the product.
[0020] For those embodiments in which water is added to the resulting mixture (from step (b)), the mixture will generally be cooled, water will be added and the aqueous mixture will be acidified to a pH of from about 1 to 4. Generally, the mixture will be cooled to about 0- 20°C, more preferably about 10-20°C, before acid is added to remove the silyl ester and provide (5)-2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid as a solid. Additionally, a number of different acids can be used in the present invention, although HCl, HBr, H3PO4, KHSO4 and H2SO4 are preferred. In the most preferred embodiments, HCl is used to acidify the reaction mixture.
[0021] Once the mixture has been acidified to a pH of about 1 to 4, the product is often obtained as a solid and can be isolated by, for example, filtration, and then dried to removed traces of solvent and/or moisture.
[0022] In a related aspect, the present invention provides methods for the preparation of a therapeutic agent, the method comprising preparing a (5)-2-(3-terMButylureido)-3,3- dimethylbutanoic acid according to the processes as described above, and coupling the acid with a suitable proline derivative to produce said therapeutic agent or a therapeutic agent precursor. The coupling process can use any of the known methods for amide formation.
[0023] The target therapeutic agents are generally selected from:
Figure imgf000007_0001
B
Figure imgf000007_0002
D. [0024] Routes for the conversion of (5)-2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid to the agents above can be found in, for example, PCT/US2006/25996, PCT/US2008/50033, and PCT/US2004/018914. Additionally, Figure 3 provides an illustration of the coupling of (S)-2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid to a 4-(aryloxy)- or A- (heteroaryloxy)pyrrolidine-2-carboxamide derivative which produces a therapeutic agent precursor. Oxidation of the hydroxy group produces target therapeutic agents of the type provided above as B, C and D. Similarly, a proline derivative having a fused gem- dimethylcyclopropane at the 3- and 4-positions of the proline, can be coupled with (<S)-2-(3- /erM3utylureido)-3,3-dimethylbutanoic acid, and optionally oxidized to produce A. One of skill in the art will appreciate that coupling of (5)-2-(3-tert-Butylureido)-3,3- dimethylbutanoic acid to other pyrrolidine-2-carboxamides followed by elaboration of the 2-carboxamide portion is also an aspect of the present invention.
EXAMPLES
Experimental Section
(S)-2-(3-te^Butylureido)-3,3-dimethyIbutanoic acid (3) - Method A
[0025] A 20-L glass flask was charged with dichloromethane (7.00 L) under a nitrogen atmosphere at 25-35 0C followed by tert-butyl-L-glycine (1.00 kg; 7.62 mol) with stirring. To the suspension was added triethylamine (0.94 kg; 9.29 mol; 122 mole %) at 25-35 0C. The reaction mixture was then treated with chlorotrimethylsilane (1.15 L; 9.00 mol; 118 mole %) with stirring at 25-35 0C. The reaction mixture was then heated to reflux temperature (39- 41 0C) and maintained under reflux for 3 h. The reaction mixture was cooled to 20-25 0C and tert-butyl isocyanate (0.76 kg; 7.67 mol; 101 mole %) was added to the reaction mass at 20- 25 0C The reaction temperature was adjusted to 25-30 0C and the resulting mixture stirred for 14-15 hours at 25-30 0C. The reaction mixture was monitored by thin layer chromatography (TLC) for the complete disappearance of tert-butyl-L-glycine. Once TLC confirmed the absence of the starting material, the reaction mixture was treated with water (4 L) at 25-30 °C. The reaction was cooled to 15-20 0C and the reaction mixture was acidified to a pH of 1.0 to 2.0 by the addition of 6N aqueous hydrochloric acid (1.0 L; 6.0 mol) at 15-20 0C. Precipitation of the product was observed after the addition of the acid was complete. The mixture was stirred for 20-30 minutes and the slurry was filtered. The filter cake was washed with water (1.00 L) and allowed to dry under vacuum on the filter until no additional solvent was collected in the receiver. The solid was transferred to polypropylene trays and dried at 60-70 0C until loss on drying was below 0.50%. Yields of compound 3 (see Figure 1) were 1.30-1.56 kg (73.9-88.6%) for three runs. HPLC Purity of these lots ranged from 99.00 to 99.78%.
(S)-2-(3-te^-ButyIureido)-3,3-dimethyIbutanoic acid (3) - Method B
Synthetic Scheme - Use of Hexamethyldisilazane and Catalytic Chlorotrimethylsilane
Figure imgf000009_0001
H H 4) (-Bu-N=C=O, 20-250C ^ XNV N^CO2H
5) H2O O
6) 5N aq HCI
7) Filter and dry (50-600C) 3
[0026] tert-Butyl-L-leucine (100 g, 0.76 mol) and dichloromethane (DCM; 1000 mL) were charged to a clean, dry round-bottom flask at ambient temperature. Hexamethyldisilazane (HMDS; 319.6 mL, 1.52 mol; 200 mole%) was added slowly to the flask with stirring at room temperature over 10 to 15 minutes followed by a catalytic amount of chlorotrimethylsilane (2.4 mL; 0.018 mol; 2 mole%). The contents were reflux ed for 3 h, and then cooled to RT. tert-Butylisocyanate (86.76 mL; 0.76 mol; 100 mole%) was added at ambient temperature over 30 min. and then stirred at ambient temperature for about 16 h, monitoring the progress of the reaction by TLC (elution solvent: 20% methanol in DCM; visualization by dipping the dry plate in a ninhydrin solution, then heating until the spots appear). Once the TLC assay indicated that the reaction was complete, water (1.0 L) was added and the reaction mass was acidified to pH = 2 with 5 N aqueous hydrochloric acid. The reaction mass was stirred for 1 h at ambient temperature then the DCM was distilled under vacuum. The reaction mass was cooled to 10-15 0C and stirred for 30 minutes. The precipitated solids were filtered, washed with hexanes and dried at 50-60 0C under vacuum for 4-5 h to provide 120 g (68%) of (5)-2-(3-tert-butylureido)-3,3-dimethylbutanoic acid. HPLC: 98.8%; Chiral HPLC: 97.8%; 1H NMR (DMSO-D6, 200 MHz): δ 12.3 (br s, IH), 5.9- 5.95 (d, J= 10 Hz, 2NH), 3.85-3.9 (d, J= 10 Hz, IH), 1.2 (s, 9H), 0.84 (s, 9H). [0027] The foregoing invention has been described in some detail by way of illustration and example, for purposes of clarity and understanding. It will be obvious to one of skill in the art that changes and modifications may be practiced within the scope of the appended claims. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the following appended claims, along with the full scope of equivalents to which such claims are entitled.

Claims

WHAT IS CLAIMED IS:
1. A process for the preparation of (5)-2-(3-tert-Butylureido)-3, 3- dimethylbutanoic acid, comprising: (a) contacting (5)-2-amino-3, 3 -dimethylbutanoic acid with a silyl ester forming reagent under conditions sufficient to form an in situ silyl ester of (5)-2-amino-3,3- dimethylbutanoic acid; (b) contacting said in situ silyl ester of (5)-2-amino-3,3-dimethylbutanoic acid with tert-butylisocyanate or a synthetic equivalent thereof; and (c) working up the reaction of step (b) to provide (5)-2-(3-tert-Butylureido)- 3, 3 -dimethylbutanoic acid as a solid.
2. A process in accordance with claim 1, wherein said silyl ester forming reagent is selected from the group consisting of trimethylsilyl chloride, triethylsilylchloride, triisopropylsilylchloride, fert-butyldimethylsilylchloride, isopropyldimethylsilylchloride, hexamethyldisilazane, phenyldimethylsilylchloride and diisopropylmethylsilylchloride.
3. A process in accordance with claim 1, wherein said silyl ester forming reagent is trimethylsilyl chloride.
4. A process in accordance with claim 1, wherein said silyl ester forming reagent is hexamethyldisilazane in combination with trimethylsilyl chloride, wherein an amount of less than 10 mole % of trimethylsilyl chloride is used.
5. A process in accordance with claim 1, wherein said contacting in step (a) is performed in the presence of a solvent selected from the group consisting of methylene chloride, chloroform, dichloroethane, and tetrahydrofuran; and an amine base selected from the group consisting of triethylamine, N,N-diisopropylethylamine, N-methylmorpholine, N- ethylpiperidine, N-methylpiperidine, N-ethylmorpholine, pyridine, 2,6-dimethylpyridine and N-methylpyrrolidine.
6. A process in accordance with claim 5, wherein step (a) is carried out at a temperature of from 0°C to the refluxing temperature of the solvent.
7. A process in accordance with claim 1, wherein step (b) is carried out at a temperature of from 10°C to 350C.
8. A process in accordance with claim 1, wherein step (c) comprises, (cl) contacting the mixture produced in step (b) with H2O to quench unreacted ter/-butylisocyanate; and (c2) acidifying the mixture produced in step (cl) to a pH of from 1 to 4.
9. A process in accordance with claim 8, wherein step(c) further comprises,
(c3) filtering and drying the product.
10. A process in accordance with claim 1, wherein said process is run in a single reaction vessel.
11. A process in accordance with claim 1, wherein said process is run on a kilogram or multi-kilogram scale.
12. A method for the preparation of a therapeutic agent, said method comprising preparing a (5)-2-(3-tert-Butylureido)-3,3-dimethylbutanoic acid according to the process of any of claims 1 - 11, and coupling said acid with a suitable proline derivative to produce said therapeutic agent or a therapeutic agent precursor.
13. A method in accordance with claim 12, wherein said proline derivative is a 4-(heteroaryloxy)pyrrolidine-2-carboxyamide component, and said coupling produces a therapeutic agent precursor that is further oxidized to said therapeutic agent.
14. A method in accordance with claim 12 or 13, wherein said therapeutic agent has a structure selected from the group consisting of:
Figure imgf000012_0001
Figure imgf000013_0001
15. A method in accordance with claim 12 or 13, wherein said therapeutic agent has the structure:
Figure imgf000013_0002
16. A method in accordance with claim 12 or 13, wherein said therapeutic agent has the structure:
Figure imgf000013_0003
17. A method in accordance with claim 12 or 13, wherein said therapeutic agent has the structure:
Figure imgf000014_0001
18. A method in accordance with claim 12 or 13, wherein said therapeutic agent has the structure:
Figure imgf000014_0002
PCT/US2008/077001 2007-09-20 2008-09-19 Process for the preparation of (s)-2-(3-tert-butylureido)-3,3-dimethylbutanoic acid WO2009039361A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US97401207P 2007-09-20 2007-09-20
US60/974,012 2007-09-20

Publications (2)

Publication Number Publication Date
WO2009039361A2 true WO2009039361A2 (en) 2009-03-26
WO2009039361A3 WO2009039361A3 (en) 2009-05-14

Family

ID=40468782

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/077001 WO2009039361A2 (en) 2007-09-20 2008-09-19 Process for the preparation of (s)-2-(3-tert-butylureido)-3,3-dimethylbutanoic acid

Country Status (1)

Country Link
WO (1) WO2009039361A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103396344A (en) * 2013-08-08 2013-11-20 苏州永健生物医药有限公司 One-pot synthetic method for N-t-butyl-aminocarbonyl-3-methyl-L-valine
WO2013190509A2 (en) * 2012-06-22 2013-12-27 Dr.Reddys Laboratories Limited Preparation of intermediates of boceprevir

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004113294A1 (en) * 2003-06-17 2004-12-29 Schering Corporation Process and intermediates for the preparation of (1r, 2s, 5s)-3-azabicyclo[3, 1, 0]hexane-2-carboxamide, n-[3-amino-1-(cyclobutylmethyl)-2, 3-dioxopropyl] ]-3-[(2s)-2-[[[1, 1-dimethylethyl]amino]carbonylamino]-3, 3-dimethyl-1-oxobutyl]-6, 6-dimethyl

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS60155967A (en) * 1984-01-25 1985-08-16 Sumitomo Chem Co Ltd Gas chromatography stationary phase and analysis of enantiomer mixture using the same
JPH11349552A (en) * 1998-06-08 1999-12-21 Ricoh Co Ltd New ester compound with urea bond

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004113294A1 (en) * 2003-06-17 2004-12-29 Schering Corporation Process and intermediates for the preparation of (1r, 2s, 5s)-3-azabicyclo[3, 1, 0]hexane-2-carboxamide, n-[3-amino-1-(cyclobutylmethyl)-2, 3-dioxopropyl] ]-3-[(2s)-2-[[[1, 1-dimethylethyl]amino]carbonylamino]-3, 3-dimethyl-1-oxobutyl]-6, 6-dimethyl

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013190509A2 (en) * 2012-06-22 2013-12-27 Dr.Reddys Laboratories Limited Preparation of intermediates of boceprevir
WO2013190509A3 (en) * 2012-06-22 2014-03-13 Dr.Reddys Laboratories Limited Preparation of intermediates of boceprevir
CN103396344A (en) * 2013-08-08 2013-11-20 苏州永健生物医药有限公司 One-pot synthetic method for N-t-butyl-aminocarbonyl-3-methyl-L-valine

Also Published As

Publication number Publication date
WO2009039361A3 (en) 2009-05-14

Similar Documents

Publication Publication Date Title
KR20090033183A (en) Rosuvastatin intermediates and process for the preparation of rosuvastatin
CN111491941B (en) Novel alkyl diphenyl methane protective agent
JP2007508293A (en) Method for producing O-carbamoyl compound in the presence of active amine group
CA2258712C (en) Asymetric synthesis of chiral beta-amino acids
JPWO2020175472A1 (en) Peptide compound production method, protecting group forming reagent, and condensed polycyclic aromatic hydrocarbon compound
JP7505498B2 (en) Method for producing peptide compounds
WO2009039361A2 (en) Process for the preparation of (s)-2-(3-tert-butylureido)-3,3-dimethylbutanoic acid
TW200827335A (en) Efficient method for producing mugineic acids
CN107428784B (en) Process for the preparation of tert-butyl 4- ((2S,5R) -6- (benzyloxy) -7-oxo-1, 6-diazabicyclo [3.2.1] octane-2-carboxamido) piperidine-1-carboxylate and analogues thereof
JP3165698B2 (en) Thioacylating reagents and intermediates, thiopeptides, and their preparation and use
CN107793351B (en) Beta-amino acid synthesis method and beta-amino acid synthesized by same
Babu et al. N-Silylation of amines and amino acid esters under neutral conditions employing TMS-Cl in the presence of zinc dust
Katritzky et al. DBU-Catalyzed transprotection of N-Fmoc-cysteine di-and tripeptides into S-Fm-cysteine di-and tripeptides
US8987493B2 (en) Process for synthesis of silane dipeptide analogs
TWI386395B (en) Stereoselective synthesis of piperidine derivatives
JP2010508334A (en) Aminobenzocycloheptene derivatives, methods for their preparation and their use in therapy
JP2019167317A (en) Method for producing l-carnosine derivative and l-carnosine
NZ523217A (en) Chemical process
JPH11503713A (en) Guanine synthon for peptide nucleic acid synthesis and method for preparing the same
WO2023058645A1 (en) Method for producing compound or pharmaceutically acceptable salt thereof
JP4178378B2 (en) Method for producing chlorosilane compound
KR101621754B1 (en) PROCESS OF PREPARING α-KETO (CYANOMETHYLENE)TRIPHENYLPHOSPHORANES USING XANTHATE COMPOUNDS
EP3596097A1 (en) A process for preparing ketolide compounds
JP3054704B1 (en) Deprotection method of amino group and hydroxyl group protected by alkynylmethyloxycarbonyl group
JP4214229B2 (en) Novel N-sulfenylamino acid ester compound and method for producing the same

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: 08831821

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08831821

Country of ref document: EP

Kind code of ref document: A2