Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CHIRAL RESOLUTION METHOD FOR PRODUCING COMPOUNDS USEFUL IN THE SYNTHESIS OF TAXANES
Document Type and Number:
WIPO Patent Application WO/2003/002509
Kind Code:
A1
Abstract:
A method is provided for processing a solution having optical isomers .to obtain a (2R,38) target isomer: wherein P¿1? is H or a hydroxyl protecting group, R¿1? is H, an alkyl group, an olefinic group or an aromatic group, and R¿2? is H or R¿3?CO, where R¿3? is an alkyl group, an olefinic group, an aromatic group, an O-alkyl group, an 0- olefinic group or an O-aromatic group, provided that R?1¿ is not H when R?3¿ is Ph and P?1¿ is H. The method includes passing the solution through a chromatographic stationary phase, such as S,S Whelk-O, that has a greater affinity for one of the target isomer and an optical isomer thereof. A portion of the solution with the target isomer is then collected. The solution may be a racemic mixture of (±)-N-CBZ-3-phenylisoserine ethyl ester.

Inventors:
MCCHESNEY JAMES D
BRINKMAN HERBERT R
ZEGAR SIEAD
BAEHR DAVID
Application Number:
PCT/US2002/020733
Publication Date:
January 09, 2003
Filing Date:
June 27, 2002
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NAPRO BIOTHERAPEUTICS INC (US)
International Classes:
B01J20/281; B01J20/283; G01N30/26; C07C227/34; C07C229/34; C07C269/08; C07C271/22; G01N30/88; (IPC1-7): C07C69/76; C07B57/00
Foreign References:
US5686298A1997-11-11
Other References:
HYUN ET AL.: "Liquid chromatographic resolution of 2-hydroxycarboxylic acids on a new chiral stationary phase derived from (S)-leucine", JOURNAL OF CHROMATOGRAPHY A, vol. 868, 2000, pages 31 - 39, XP004185053
PIRKLE ET AL.: "Use of a simultaneous face to face and face to edge pi-pi interactions to facilitate chiral recognition", TETRAHEDRON ASYMMETRY, vol. 5, no. 5, 1994, pages 777 - 780, XP002955212
STILL ET AL.: "Rapid chromatographic technique for preparative separations with moederate resolution", JOURNAL OF ORGANIC CHEMISTRY, vol. 43, 1978, pages 2923 - 2925, XP002955213
Attorney, Agent or Firm:
Martin, Timothy J. (Suite 200 Lakewood, CO, US)
Download PDF:
Claims:
We claim :
1. A method of processing a solution having a plurality of optical isomers, thereby to obtain a (2R, 3S) target isomer having a formula: wherein P1 is selected from H and a hydroxyl protecting group, R1 is selected from H, an alkyl group, an olefinic group and an aromatic group, and R2 is selected from H and R3CO, where R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0alkyl group, an Oolefinic group and an 0aromatic group, provided that R1 is not H when R3 is Ph and p1 is H, comprising the steps of: (a) passing the solution through a chromatographic stationary phase that has a greater affinity for one of the target isomer and an optical isomer thereof, such that the target isomer passes through said stationary phase at a different rate than does said optical isomer thereof; and (b) collecting a portion of the solution containing the target isomer.
2. A method according to claim 1 wherein said stationary phase is loaded into a chromatography column having a first opening and a second opening, and wherein the step of passing the solution through said stationary phase is accomplished by injecting the solution into said first opening, and wherein said portion of the solution containing the target isomer is collected from said second opening.
3. A method according to claim 1 wherein the solution contains the target isomer and said optical isomer thereof in a first ratio to one another, and wherein said portion of the solution contains the target isomer and said optical isomer thereof in a second ratio to one another that is greater than said first ratio.
4. A method according to claim 1 wherein the solution is a racemic solution of the target isomer and said optical isomer thereof.
5. A method according to claim 1 wherein the solution includes a diastereomer of the target isomer.
6. A method according to claim 1 wherein said stationary phase comprises a chromatographic media covalently bonded to silica.
7. A method according to claim 1 wherein said stationary phase is selected from Regis Technologies, Inc. product numbers 786101,786102, 786103,786104, 786615,786625, 786635, 786645, 786415,786425, 786435 and 786445.
8. A method according to claim 1 wherein said stationary phase comprises a chromatographic media that includes binding sites selected from tetrahydrophenanthrene n systems, dinitrobenzamide n systems and amido proton hydrogen bond donors.
9. A method according to claim 1 wherein said stationary phase comprises S, S Whelk0.
10. A method according to claim 1 wherein said stationary phase comprises S, S Whelk0 bonded to 10, u kromasil silica.
11. A method according to claim 1 wherein the solution includes a solvent selected from ethanol, isopropanol and hexane.
12. A method according to claim 1 wherein the solution includes 20% isopropanol/80% hexane.
13. A method according to claim 12 wherein the solution contains about 0.025g of said optical isomers per 1 mL isopropanol.
14. A method according to claim 1 wherein Ri is an alkyl group.
15. A method according to claim 1 wherein R2 is R3CO and wherein R3 is PhCH20.
16. A method according to claim 1 wherein P1 is H.
17. A method according to claim 1 wherein the target isomer is a (2R, 3S) ethyl ester of the formula :.
18. A method according to claim 17 wherein said stationary phase comprises S, S Whelk0.
19. A method according to claim 18 wherein said S, S Whelk0 is bonded to silica.
20. A method of separating a target isomer from an optical isomer thereof, wherein the target isomer has a formula: wherein P1 is selected from H and a hydroxyl protecting group, R1 is selected from H, an alkyl group, an olefinic group and an aromatic group, and R2 is selected from H and R3CO, where R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0alkyl group, an Oolefinic group and an 0aromatic group, provided that R'is not H when R3 is Ph and p1 is H, the method comprising the steps of: (a) passing the solution from an upstream location to a downstream location through a chromatographic stationary phase that has a greater affinity for one of the target isomer and the optical isomer thereof; and (b) collecting the solution in a plurality of stages, each stage correlating to a portion of the solution collected over a respective interval of time thereby to form a respective collection solution, wherein the target isomer and the optical isomer thereof are in a first ratio to one another in a first collection solution and a second ratio to one another in a second collection solution, said first ratio being different than said second ratio.
21. A method of processing a racemic mixture of ()NCBZ3 phenylisoserine ethyl ester of the formula: thereby to separate a (2R, 3S) isomer from an enantiomer thereof, comprising chromatographing said racemic mixture by HPLC using a stationary phase comprising S, S Whelk0 and a mobile phase including a solvent selected from ethanol, isopropanol and hexane.
22. A method according to claim 21 wherein said stationary phase comprises S, S Whelk0 covalently bonded to silica.
23. A method according to claim 21 wherein said mobile phase is 20% isopropanol/80% hexane.
24. A method according to claim 21 wherein said mobile phase includes about 0.025g of the ()NCBZ3phenylisoserine ethyl ester per 1 mL of solvent.
25. A method according to claim 21 wherein an HPLC peak for the (2R, 3S) isomer is resolved to a baseline from an HPLC peak for the enantiomer thereof.
26. A method of processing a mixture including optical isomers of the general formula : wherein P1 is selected from H and a hydroxyl protecting group, R1 is selected from H, an alkyl group, an olefinic group and an aromatic group, and R2 is selected from H and R3CO, where R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0alkyl group, an Oolefinic group and an 0aromatic group, provided that R'is not H when R3 is Ph and p1 is H, thereby to separate the optical isomers from one another, comprising chromatographing the mixture by HPLC using a stationary phase that has a different affinity for each of the optical isomers.
27. A method according to claim 26 wherein the mixture is a racemic mixture of enantiomers of the formula :.
28. A method according to claim 26 wherein said stationary phase comprises S, S Whelk0.
29. A method according to claim 26 including using a mobile phase including a solvent selected from ethanol, isopropanol and hexane.
30. A method according to claim 26 wherein Ri is an alkyl group.
31. A method according to claim 26 wherein R2 is R3CO and wherein R3 is PhCH20.
32. A method according to claim 26 wherein P1 is H.
33. A method of processing a mixture that includes a (2R, 3S) isomer of the formula : where R1 is an alkyl group and R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0alkyl group, an Oolefinic group and an O aromatic group, and that includes a (2S, 3R) enantiomer thereof of the formula : comprising chromatographing the mixture using a stationary phase that has a greater affinity for one of the (2R, 3S) isomer and the (2S, 3R) enantiomer thereof.
34. A method according to claim 33 wherein Pi is an ethyl group.
35. A method according to claim 33 wherein R3 is PhCH20.
36. A method according to claim 33 wherein R1 is an ethyl group and wherein R3 is PhCH20.
37. A method according to claim 33 wherein said stationary phase comprises S, S Whelk0.
38. A method according to claim 33 wherein R1 is an ethyl group, R3 is PhCH20, and wherein said stationary phase comprises S, S Whelk0.
Description:
CHIRAL RESOLUTION METHOD FOR PRODUCING COMPOUNDS USEFUL IN THE SYNTHESIS OF TAXANES FIELD OF THE INVENTION The present invention generally relates to the preparation of compounds useful in the semi-synthesis of taxanes. More particularly, the present invention is directed to the chiral resolution of mixtures of optical isomers to provide a target chiral compound that can be used as a C-13 side chain precursor to produce paclitaxel and other taxanes. The present invention specifically provides a chromatographic process for separating a target chiral compound from its enantiomer in a racemic mixture thereof.

BACKGROUND OF THE INVENTION Various taxane compounds are known to exhibit anti-tumor activity. As a result of this activity, taxanes have received increasing attention in the scientific and medical community. Primary among these is a compound known as"paclitaxel"which is also referred to in the literature as"taxol". Paclitaxel has been approved for the chemotherapeutic treatment of several different varieties of tumors, and the clinical trials indicate that paclitaxel promises a broad range of potent anti-leukemic and tumor-inhibiting activity.

Paclitaxel is a naturally occurring taxane diterpenoid which is found in several species of the yew (genus Taxus, family Taxaceae). Unfortunately, the concentration of this compound in the yew is very low, and the species of evergreen are also slow growing. Even though the bark of the yew trees typically exhibits the highest concentration of paclitaxel, the production of one kilogram of paclitaxel requires approximately 16,000 pounds of bark. Thus, the long-term prospects for the availability of paclitaxel through isolation are discouraging.

While the presence of paclitaxel in the yew tree is in extremely low concentrations, there are a variety of other taxane compounds, such as baccatin III, cephalomanine, 10-deacetylbaccatin III, etc. , which are also able to be extracted from the yew bark and leaves. Some of these other taxane compounds are more readily extracted in higher yields. Indeed, a relatively high concentration of 10-deacetylbaccatin III can be extracted from the leaves of the yew as a renewable resource.

Accordingly, attention has turned to the semi-synthesis of paclitaxel, which has the formula : as well as other related taxanes, such as docetaxel, which has the formula : from precursor compounds. In order to successfully synthesize paclitaxel and other taxanes, convenient access to a chiral, non-racemic side chain acid is desired, as well as an abundant natural source of a usable baccatin III backbone. Various approaches have been developed for esterifying such a side chain acid at the 13-hydroxyl of baccatin III or 10-deacetyl baccatin tit, or derivatives thereof, which respectively have the formulas: baccatin III 10-deacetyl baccatin Iil The coupled ester product may then be converted to paclitaxel, docetaxel or other taxanes. For example, U. S. Patent No. 4,924, 011 to Denis et al. describes a process for preparing paclitaxel using a (2R, 3S) side chain acid of the general formula : where R2 is a hydroxy-protecting group. Additionally, U. S. Patent No.

4,924, 012 to Colin et al. describes a process for preparing taxanes, such as docetaxel, that uses an acid of formula : where R1 is a hydroxy-protecting group. In the syntheses described by Denis et al. and Colin et al., the side chain acid is esterified with a baccatin III or 10- deacetyl baccatin III derivative, and the coupled product is thereafter deprotected, such as by replacing any protecting groups, including R2 or Ri in the above formulas, respectively, with hydrogen. It should be noted that the C6H5CONH-group of Denis et al. and the (CH3) 3COCONH- group of Colin et al. are the final desired groups for the resulting paclitaxel and docetaxel products, respectively, such that no further chemical transformation at the nitrogen position is performed in the chemical syntheses disclosed therein.

U. S. Patent No. 5,770, 745 to Swindell et al. describes another early synthetic route in the semi-synthesis of paclitaxel, wherein the use of protecting groups to protect various positions of the taxane backbone and the side chain acid was investigated as a means of improving the chemical process to form paclitaxel, and of improving the esterification step in particular. Specifically, a side chain acid of the general formula: is described, wherein R1 can be an alkyl, olefinic or aromatic group (such as PhCH2), R3 can be hydrogen or Ph, and P1 can be a hydroxyl protecting group.

More recently, attention has been focused on the use of a 3-N-CBZ-2-0- protected (2R, 3S)-3-phenylisoserine acid of the formula : wherein P, is a hydrogenatable protecting group, such as benzyloxymethyl (BOM) or benzyl. Synthetic routes to produce taxanes such as paclitaxel or docetaxel using such a side chain are described, for example, in U. S. Patent Nos. 5,675, 025; 5,684, 175; 5,688, 977; 5,750, 737; 5,939, 566; 5,948, 919; 5,973, 170; 6,048, 990; 6,066, 749; 6,072, 060; 6,107, 497; 6,133, 462; 6,136, 999; and 6,143, 902, and the teachings thereof are incorporated herein by reference.

As taught, for example, in U. S. Patent No. 5,684, 175 to Sisti et al., the 3- N-CBZ-2-O-protected (2R, 3S)-3-phenylisoserine may be produced from a (2R, 3S)-3-phenylisoserine ethyl ester starting compound of the formula: which may be protected at the 3'-N and 2'-O positions and saponified to the corresponding acid for use in the esterification step.

The formation of a 3-phenylisoserine alkyl ester is known in the art and is described, for example, in U. S. Patent No. 4,924, 012 to Colin et al. In particular, Colin et al. describes an epoxide of the formula : where R4 denotes alkyl containing 1 to 4 carbon atoms, and preferably ethyl, which may be obtained under the conditions described by F. W. Bachelor and R. K. Bansal, J. Org. Chem. , 34,3600-04 (1969). This epoxide is converted to an azide of general formula : according to known methods for opening an epoxide by means of sodium azide in ethanol in the heated state. The azide is thereafter reduced to a 3- phenylisoserine alkyl ester of the formula : However, because this compound has two chiral centers (C-2 and C-3, respectively), processes to produce this compound may produce both the cis (2R, 3S) and (2S, 3R) enantiomers respectively, such as of the formulas : as well as their trans (2R, 3R) and (2S, 3S) diastereomers respectively of the formulas : It should be appreciated that the above optical isomers are shown using an ethyl group for R4 in the formulas above, although other alkyl esters as described in Colin et al. may be formed. Additionally, various other derivatives or analogs of these optical isomers may be formed in the processes to produce the paclitaxel side chain, in view of the teachings of the above-identified patents, as would be understood by the ordinarily skilled person.

While baccatin III and derivatives thereof may be used as a resolving agent to selectively recover products having the desired (2R, 3S) configuration, such an approach is quite costly and results in a substantial yield reduction of the desired product. Accordingly, efficient and economical methods are needed for recovering a desired (2R, 3S) isomer or a derivative thereof at one or more points in the chemical processes for synthesizing taxanes.

One procedure to prepare the chiral (2R, 3S) C-13 side chain of paclitaxel is described by Srivastava and McChesney,"A Practical and Inexpensive Synthesis of the Taxol C-13 Side Chain; N-Benzoyl- (2R, 3S) -3- Phenylisoserine", Natural Products Letters, Vol. 6, p. 147 (1995). This procedure involves the classical resolution of cis-3-phenylglycidic acid using D- (+)-ephedrine as the resolving agent. In particular, an optically pure cis- (2R, 3R)-3-phenylglycidic acid- (+)-ephedrine salt of the formula: Ph 0 C02-)-ephodrine salt H H is recovered from a mixture of diastereomeric salts by fractional crystallization with acetone. The optically pure intermediate is then converted to a 3-phenylisoserine derivative. Additional classical resolutions of side chain derivatives useful in paclitaxel synthesis have been described in U. S. Patent No. 6,025, 516 to Ramaswamy et al. and in U. S. Patent No.

5,817, 867 to Li.

Several other procedures are reported in the literature to prepare chiral, cis-3-phenylglycidate derived intermediates useful in the partial synthesis of paclitaxel. Some examples include methodologies utilizing Sharpless asymmetric dihydroxylation (Denis et al., J. Org. Chem. , 55, p. 1957,1990 ; Koskinen et al., J. Chem. Soc., Chem. Commun. , p. 21,1994 ; and Bonini et al., J. Chem. Soc. , Chem. Commun. , p. 2767,1994) and some utilizing Jacobsen's asymmetric epoxidation (Deng et al., J. Org. Chem. , 57, p. 4323,1992). Other references have reported chemoenzymatic resolution methodologies (Chen et al., J. Org. Chem. , 58, p. 1287 ; U. S. Patent No.

6,020, 174 to Chen et al. ; Honig et al., Tetrahedron Letters, 46, p. 3841,1990 ; U. S. Patent No. 5,773, 629 to Yang et al. ; and U. S. Patent Nos. 5,811, 292, 5,567, 614 and 5,686, 298 to Patel et al).

However, there remains a need to provide new and improved methods for the efficient and economical preparation of chiral compounds useful in taxane semi-synthesis. In particular, there remains a need for simple methods for producing chiral compounds useful as precursors for the C-13 side-chain esterification with a baccatin III backbone. The present invention is directed to meeting these needs.

SUMMARY OF THE INVENTION It is an object of the present invention to provide a new and useful method for preparing chiral compounds useful in taxane semi-synthesis.

It is another object to provide methods for preparing optically pure derivatives of 3-phenylisoserine.

It is yet another object to provide for simple and efficient chiral chromatographic separation of optical isomers useful in the synthesis of taxanes such as paclitaxel and docetaxel.

A still further object is to provide chromatography media useful in chromatographically separating optical isomers of 3-phenylisoserine derivatives.

Yet another object is to synthesize an N-CBZ- (2R, 3S) -3- phenylisoserine side chain derivative in optically pure form that is suitable for coupling with a suitable baccatin III derivative to provide paclitaxel after various synthetic transformations.

According to the present invention, a method is provided for processing a solution having a plurality of optical isomers, such as a racemic solution, thereby to obtain a (2R, 3S) target isomer having a formula : wherein P1 is selected from H and a hydroxyl protecting group, Pi is selected from H, an alkyl group, an olefinic group and an aromatic group, and R2 is selected from H and R3CO, where R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0-alkyl group, an O-olefinic group and an 0-aromatic group. The method comprises the steps of passing the solution through a chromatographic stationary phase that has a greater affinity for either the target isomer or an optical isomer thereof, such that the target isomer passes through the stationary phase at a different rate than does the optical isomer thereof, and thereafter collecting a portion of the solution containing the target isomer.

The stationary phase may be loaded into a chromatography column having a first opening and a second opening, and the step of passing the solution through the stationary phase may be accomplished by injecting the solution into the first opening. The portion of the solution containing the target isomer may be collected from the second opening. Preferably, the solution contains the target isomer and the optical isomer thereof in a first ratio to one another, and the collected portion of the solution contains the target isomer and the optical isomer thereof in a second ratio to one another that is greater than the first ratio.

The stationary phase may include tetrahydrophenanthrene systems, dinitrobenzamide n systems and amido-proton hydrogen bond donors, and preferably comprises S, S Whelk-0 which may be covalently bonded to silica.

The solution may include ethanol, isopropanol, hexane or the like, and preferably includes 20% isopropanol/80% hexane, and may contain about 0.025g of the optical isomers per 1 mL isopropanol.

Various optical isomers are contemplated, including ones wherein Ri is an alkyl group, R2 is R3CO where R3 is PhCH20, and wherein P1 is hydrogen, and particularly a (2R, 3S)-N-CBZ-3-phenylisoserine ethyl ester of the formula: The present invention particularly contemplates a method of processing a racemic mixture of ()-N-CBZ-3-phenylisoserine ethyl ester thereby to separate the (2R, 3S) isomer from the enantiomer thereof, comprising chromatographing the racemic mixture by HPLC using a stationary phase comprising S, S Whelk-0 and a mobile phase including ethanol, isopropanol or hexane. The HPLC peak for the (2R, 3S) isomer is preferably resolved to the baseline from the HPLC peak for the enantiomer thereof.

Further, the invention may broadly include a method of processing optical isomers of the general formula : wherein Pi is H or a hydroxyl protecting group, R1 is selected from H, an alkyl group, an olefinic group and an aromatic group, and R2 is selected from H and R3CO, where R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0-alkyl group, an O-olefinic group and an 0-aromatic group, thereby to separate the optical isomers from one another, comprising chromatographing the racemic mixture by HPLC using a stationary phase that has a different affinity for each of the optical isomers.

These and other objects of the present invention will become more readily appreciated and understood from a consideration of the following detailed description of the exemplary embodiments of the present invention when taken together with the accompanying drawings, in which: BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a diagram showing an exemplary chemical process according to the present invention for the preparation of racemic 3- phenylisoserine.

Figure 2 is a diagram showing an exemplary chemical process for converting the racemic 3-phenylisoserine formed in Figure 1 to a racemic 3- N-CBZ ethyl ester; Figure 3 is a diagram showing an exemplary chiral chromatographic separation to provide a chiral 3-N-CBZ- (2R, 3S)-ethyl ester from the racemic ethyl ester formed in Figure 2; Figure 4 is a diagram showing an exemplary chemical process for the protection and saponification of the chiral ethyl ester formed in Figure 3; and Figure 5 is a diagram showing a generalized semi-synthesis of paclitaxel using the chiral 3-N-CBZ-2-O-BOM-(2R, 3S)-3-phenylisoserine compound formed in Figure 4.

DETAILED DESCRIPTION OF THE EXEMPLARY EMBODIMENTS The N-benzoyl- (2R, 3S)-3-phenylisoserine side chain of the formula : is crucial to the antitumor activity of paclitaxel. This side chain must be available in highly enantiomerically pure form for use in the partial synthesis of paclitaxel and its analogs. Analogs and derivatives of this side chain are also useful in forming other taxanes, such as docetaxel, which may be useful in the treatment of cancer.

The present invention provides a method for the chiral chromatographic resolution of racemic mixtures of enantiomers, such as ()- N-CBZ-3-phenylisoserine ethyl ester, which may be employed as side chain precursors in the semi-synthesis of paclitaxel and its analogs that bear a C- 13 side chain. The chiral resolution is achieved by preparative chiral column chromatography using columns packed with a stationary phase comprising an appropriate chromatographic media, such as S, S Whelk-0, 1OR FEC, which is operative to separate the enantiomers of the racemic mixture.

In the exemplary process, a racemic N-CBZ-3-phenylisoserine ethyl ester was synthesized according to the sequence shown with respect to Figures 1 and 2, and as discussed below. It should be appreciated, though, that the present invention contemplates the chiral chromatographic resolution of mixtures of other taxane side-chain precursor compounds, such as compounds having a 3-amino group, compounds utilizing 3-N-protecting groups other than CBZ, compounds protected at the 2-0-position, compounds having ester substituents other than an ethyl group, such as other alkyl groups and the like, or carboxylic acid derivatives of the side-chain ester compounds, as discussed more fully below.

A. Formation of racemic phenylisoserine Racemic phenylisoserine of the formula : is first formed as described below. It should be appreciated that the starting materials and reagents used in the processes described herein are readily available from chemical suppliers, as would be understood by the ordinarily skilled artisan. Further, as apparent from the description of the various chemical steps herein, it should be appreciated that certain chemical steps in the processes were repeated at various scales to accumulate quantities of intermediate product for use in subsequent steps, while other steps used less than the total quantity of intermediate product produced in a previous step.

However, it should be understood that adjustments in the quantities of materials may be made without departing from the general inventive concept, and in particular the individual chemical steps described herein may be scaled to accommodate quantities of materials produced in previous steps.

First, a Darzen's condensation of benzaldehyde with methylchloroacetate in the presence of sodium methoxide in methanol provides the trans-methyl 3-phenylglycidate (Formula 1), as follows : Formula 1 Here, benzaldehyde (1.76 Kg, 1.0 eq. ) and methylchloroacetate (2.70 Kg, 1.5 eq. ) were added to methanol (2. 55UKg benzaldehyde) in a dry 22 liter 3-necked round bottom flask equipped with a mechanical stirrer, condenser, thermometer, addition funnel, nitrogen purge, and ice/methanol bath. While stirring, the reaction mixture was cooled to between 0°C and- 10°C, and 1.5 eq. sodium methoxide (25% in methanol) was slowly titrated into the reaction mixture at the cooled temperature. The cooling bath was then removed and the flask was allowed to warm to room temperature naturally, via an overnight stir. The reaction was monitored by TLC and was deemed complete by the disappearance of starting materials. The reaction was worked-up by concentrating the mixture to approximately 20% of the starting volume under reduced pressure and at a temperature of : 5400C. The concentrate was then quenched with purified water (10UKg benzaldehyde) and extracted into ethyl ether (1OUKg benzaldehyde), and the extracts were dried over sodium sulfate (1. 1Kg/Kg benzaldehyde), treated with activated carbon, filtered, and concentrated to an oil, resulting in 81 % yield of the trans isomer of Formula 1.

The trans epoxide of Formula 1 is next converted to the cis isomer (Formula 3) via oxirane opening with gaseous HCI in chloroform to form a chlorohydrin derivative (Formula 2) followed by ring closure with sodium methoxide in the presence of a phase transfer catalyst such as tetrabutylammonium bromide, in chloroform and water, as follows : H O C02Me T CO2Me Ph O CO2Me e ; HCI gas NaOMe, H Ph H CHCI3 Ph H CHC, 3 H H R) i M Rr Formula 1 Formula 2 Bu4NBr Formula 3 Here, a solvent, preferably chloroform (10UKg trans epoxide), was added to a 3-necked, round bottom flask equipped with a mechanical stirrer, reflux condenser, nitrogen purge, gas dispersion tube and thermometer.

Various solvents are contemplated, including chloroform, chloroform/hexane (10: 90), toluene and benzene. While stirring, the methylphenyloxirane carboxylate of Formula 1 (2.4 Kg, 1. 0 eq. ) was added, and after dissolving, constant stirring continued and HCI gas was bubbled into the solution for at least 3 hours, after which the reaction mixture was allowed to stir overnight at a temperature of room temperature to 50°C. The reaction mixture was concentrated to an oil under reduced pressure at a temperature of : 5400C.

The oil was titrated with ethyl ether (1.25 Ukg trans epoxide) and cooled to 0°C to force crystallization of the product of Formula 2 as a white solid. The solid was collected by filtration and washed with a hexane: ethyl ether (4: 1) solution (6. 25Ukg trans epoxide), and the mother liquor was cooled to 0°C for over 24 hours to recover further product. Further experiments indicate that use of a trans-ethyl 3-phenylglycidate significantly increased the yield of this step while maintaining the same level of purity.

Next, a solvent, preferably chloroform (8UKg chlorohydrin), and purified water (2L/Kg chlorohydrin) were added to a 3-necked round bottom flask equipped with a mechanical stirrer, reflux condenser, nitrogen purge, addition funnel and thermometer. The use of other solvents, such as THF or methanol, is contemplated. With stirring, the chlorohydrin derivative of Formula 2 (0. 81 Kg, 1.0 eq. ) was added, along with a phase transfer catalyst, such as between 0.1 and 0.3 eq. tetrabutyl-N-bromide (Bu4NBr). These reactants were allowed to dissolve, followed by the addition of 1.5 to 2.0 eq. sodium methoxide (25% in methanol) at room temperature over a 15 minute period. It should be appreciated that other bases, such as NaOH or Amberlite, may be used in this step, such as with THF solvent. It should additionally be noted that the use of a phase transfer catalyst may not be necessary when weaker bases are used having increased solubility. The reaction progress was monitored by TLC and was deemed complete after 30 minutes of stirring at room temperature. The product (Formula 3) was worked up by phase separation, and the organic phase was washed twice with purified water, followed by drying over sodium sulfate, filtration, and concentration to an oil. Notably, the cis product of Formula 3 was found to be unstable after isolation, even under nitrogen at <10°C, such that it is desirable to minimize reaction and work-up time related to this step.

The resulting cis-methyl 3-phenylglycidate (Formula 3) was converted to an ammonium salt by saponification with KOH in ethanol followed by reaction with ammonia gas in the presence of aqueous HCI, water and ether: Ph 0 C02Me KOH, ETOH 1 2 4 Et20, H20, H H H H HCI, NH3 H H Formula 3 Formula 4 Here, a solvent, preferably ethanol (3UKg of cis epoxide), was added to a 3-necked round bottom flask, equipped with a mechanical stirrer, reflux condenser, addition funnel, thermometer, ice-water bath and nitrogen purge.

The use of other solvents, such as methanol, is contemplated. KOH (1.3 to 1.8 eq) was added with stirring, while maintaining the temperature between 0-5 °C, followed by the slow addition of the oxirane derivative of Formula 3 in ethanol (0.66 Kg, 1.0 eq dissolved in 2UKg of cis epoxide), over a time of 15- 30 minutes. The temperature was maintained between 0-5 °C during the addition process and slowly raised to ambient temperatures by removing the ice-water bath.

The resulting reaction mixture was stirred for 3 hours and monitored by TLC for completion. The reaction mixture was cooled to 0-5 °C for complete precipitation of the intermediate product, ()-potassium- (+)-3- phenyloxirane carboxylate as a white solid, which was filtered, washed with cold ethanol and dried in the vacuum oven at 35 °C.

This potassium salt was added to a 3-necked round bottom flask, equipped with a mechanical stirrer, reflux condenser, addition funnel, thermometer, ice-water bath and nitrogen purge. Water (6UKg of potassium salt) was added followed by cooling to < 10 °C. To this solution was added ether (5UKg of potassium salt) and 1 N HCI (5UKg of potassium salt) while stirring vigorously at : 5 10 °C. Temperature was slowly raised to ambient temperatures by removing the ice-water bath and stirring continued for 30 min. The organic phase was separated and the aqueous phase was extracted with ether. The organic layers were combined and transferred to a 3-necked round bottom flask into which ammonia gas was bubbled at ambient temperatures for 30 minutes while continuously stirring. The reaction mixture was cooled to 0-5 °C and stirring continued for an additional 30 minutes. The resulting solid was filtered, washed with cold ether and dried to afford the ammonium salt of formula 4, in 69% yield.

The resulting ()-ammonium- (+)-3-phenyloxirane carboxylate (Formula 4) was refluxed with ammonium hydroxide in methanol to provide the racemic phenylisoserine (Formula 5): NH2 0 Ph O CO+NH4 NH40H, CH30H Reflux H H Reflux- Formula4 Formula 5 Here, the ammonium salt of Formula 4 (0.15 Kg, 1.0 eq. ) was added to methanol (45 L/Kg ammonium salt), while stirring, in a 3-necked round bottom flask equipped with a mechanical stirrer, reflux condenser, thermometer, and heating mantle. Ammonium hydroxide (0.26 Kg/Kg ammonium salt) was added in 3 increments over a 16 hour period, in order to replenish lost ammonia. The reaction was allowed to reflux for 24 hours, and the mixture was concentrated to approximately 15% of starting volume, cooled to S1 0°C to form a precipitate, filtered, and washed with ethyl ether to provide the racemic product of Formula 5 in greater than 90% yield.

It should be understood that the present invention contemplates the use of racemic phenylisoserine formed according to the method described herein or according to other methods, to the extent understood by the ordinarily skilled artisan. Further, it should be understood that the present invention contemplates the use of racemic phenylisoserine mixtures including one or more of the (2R, 3R) and (2S, 3S) diastereomers of the desired (2R, 3S) isomer, in addition to one or more of the enantiomers thereof.

B. Conversion to N-protected ester precursor The ()-phenylisoserine (Formula 5) was next protected as its N-CBZ derivative using Schotten-Baumann conditions in the presence of benzylchloroformate, sodium bicarbonate and water to furnish ()-N-CBZ-3- phenylisoserine (Formula 6), as follows : NH2 0 CBZ-NH 0 () Ph, OH Bl. () Ph OH OH N aH CO3, H20 OH Formula 5 Formula 6 Here, the racemic 3-phenylisoserine of Formula 5 (0.08 Kg, 1.0 eq. ) was added with agitation to a reaction flask containing a 10% aqueous solution of sodium bicarbonate (135 UKg phenylisoserine) under a nitrogen blanket, which was stirred until dissolved. The reaction mixture was then cooled to 0-5°C and approximately 3 eq. benzyl chloroformate was added dropwise while maintaining the temperature of the mixture. The resulting solution was allowed to stir for 6 hours at <5°C or until the disappearance of the racemic 3-phenylisoserine reactant was confirmed by TLC. Hydrochloric acid was added dropwise, with stirring, at room temperature to bring the mixture to a pH of <4. The mixture was extracted into ethyl acetate (200 L/Kg phenylisoserine), dried over sodium sulfate, filtered, and concentrated at <45°C under reduced pressure to yield the crude product of Formula 6, which was purified by slurrying in 50: 50 hexane/chloroform.

Although Formula 6 relates to the preferred embodiment of the present invention, it should be appreciated that other acylating agents may be used in place of benzyl chloroformate, to the extent understood by the ordinarily skilled artisan, to provide compounds having different chemical groups at the 3-N-position, such as compounds of the general formula: Formula 6a wherein R may be an alkyl group, olefinic group, aromatic group such as Ph, O-alkyl, O-olefinic, or 0-aromatic group (such as PhCH20-in the case of the CBZ group). Alternatively, the present invention contemplates that the 3- amino group may remain unprotected during the chiral separation step discussed below.

The racemic N-CBZ-3-phenylisoserine of Formula 6 was then esterified by refluxing in ethanol with methane sulfonic acid to provide the ()- N-CBZ-3-phenylisoserine ethyl ester (Formula 7) ready for chiral chromatographic separation: CBZ-NH O CBZ-NH 0 CH3S03H () Ph OH > Ph OC2H5 OH OH Formula 6 Reflux Formula 7 Here, Formula 6 (0.072 Kg, 1.0 eq. ) was added to an excess of ethanol (230 UKg phenylisoserine) and 0.5 eq. methyl sulfonic acid in a 3- necked round bottom flask equipped with a mechanical stirrer, nitrogen purge, heating mantle, and thermometer. The reaction mixture was refluxed for 3 hours and was then concentrated at <45°C under reduced pressure to yield an oil. The oil was dissolved in chloroform and the organics were washed once with 5% NaHCO3, twice with purified water, dried over sodium sulfate, filtered and then concentrated to an oil, which solidified to an off- white residue while standing. The solid was slurried in hexane, filtered, and dried to provide purified racemic 3-N-CBZ-3-phenylisoserine ethyl ester of Formula 7 in greater than 90% yield.

Again, it should be appreciated that other esters of the acid of Formula 6 may be formed for use in the present invention, as known in the art. For example, other alkyl esters, such as methyl esters and the like, as well as aromatic and olefinic esters of the acid of Formula 6 are contemplated.

Additionally, as indicated above, various other acyl groups may be utilized at the 3-N-position in place of the CBZ group, or alternatively the compound may remain free as the amine or amine salt for the chiral separation step discussed below. Furthermore, the present invention contemplates that the 2-hydroxyl may alternatively be protected, such as in the manner discussed below, prior to the chiral separation step.

Accordingly, it should be appreciated that the present invention contemplates the formation of various compounds for use in the chiral separation of the present invention, such as compounds of the general formula : Formula 7a wherein P1 is selected from H and a hydroxyl protecting group, R1 is selected from H, an alkyl group, an olefinic group and an aromatic group, and R2 is selected from H and R3CO, where R3 is selected from an alkyl group, an olefinic group, an aromatic group, an 0-alkyl group, an O-olefinic group and an 0-aromatic group. Table A below demonstrates, without limitation, various compounds of general Formula 7a contemplated by the present invention.

Table A Compound P1 R1 R2 R3 1HPiH 2 H H R3CO alkyl 3 H H R3CO olefinic 4 H H R3CO aromatic 5 H H R3CO Ph 6 H H R3CO O-alkyl 7 H H R3CO O-olefinic 8 H H R3CO O-aromatic 9 H H R3CO O-CH2Ph 10 H alkyl H --- 11 H alkyl R3CO alkyl 12 H alkyl R3CO olefinic 13 alkyl R3CO aromatic 14 H alkyl R3CO Ph 15 H alkyl R3CO O-alkyl 16 H alkyl R3CO O-olefinic 17 H alkyl R3CO O-aromatic 18 H alkyl R3CO O-CH2Ph 19 H ethyl H --- 20 H ethyl R3CO alkyl 21 H ethyl R3CO olefinic 22 H ethyl R3CO aromatic 23 H ethyl R3CO Ph 24 H ethyl R3CO O-alkyl 25 H ethyl R3CO O-olefinic 26 ethyl R3C O O-aromatic 27 H ethyl R3CO O-CH2Ph 28 H methyl H--- 29 H methyl R3CO alkyl 30 H methyl R3CO olefinic 31 H methyl R3CO aromatic 32 H methyl R3CO Ph 33 H methyl R3CO O-alkyl 34 H methyl R3CO O-olefinic 35 H methyl R3CO O-aromatic 36 H methyl R3CO O-CH2Ph 37 H olefinic H --- 38 H olefinic R3CO alkyl 39 H olefinic R3CO olefinic 40 H olefinic R3CO aromatic 41 H olefinic R3CO Ph 42 H olefinic R3CO O-alkyl 43 H olefinic R3CO O-olefinic 44 H olefinic RsCO 0-aromatic 45 H olefinic R3CO O-CH2Ph 46 H aromatic H --- 47 H aromatic R3CO alkyl 48 H aromatic R3CO olefinic 49 H aromatic R3CO aromatic 50 H aromatic R3CO Ph 51 H aromatic R3CO O-alkyl 52 H aromatic R3CO O-olefinic 53 H aromatic R3CO 0-aromatic 54 H aromatic R3CO O-CH2Ph 55 hydroxyl protecting group H H 56 hydroxyl protecting group H R3CO alkyl 57 hydroxyl protecting group H R3CO olefinic 58 hydroxyl protecting group H R3CO aromatic 59 hydroxyl protecting group H R3CO Ph 60 hydroxyl protecting group H R3CO 0-alkyl 61 hydroxyl protecting group H R3CO O-olefinic 62 hydroxyl protecting group H R3CO 0-aromatic 63 hydroxyl protecting group H R3CO O-CH2Ph 64 hydroxyl protecting group alkyl H 65 hydroxyl protecting group alkyl R3CO alkyl 66 hydroxyl protecting group alkyl R3CO olefinic 67 hydroxyl protecting group alkyl R3CO aromatic 68 hydroxyl protecting group alkyl R3CO Ph 69 hydroxyl protecting group alkyl R3CO 0-alkyl 70 hydroxyl protecting group alkyl R3CO O-olefinic 71 hydroxyl protecting group alkyl R3CO O-aromatic 72 hydroxyl protecting group alkyl R3CO O-CH2Ph 73 hydroxyl protecting group ethyl H 74 hydroxyl protecting group ethyl R3CO alkyl 75 hydroxyl protecting group ethyl R3CO olefinic 76 hydroxyl protecting group ethyl R3CO aromatic 77 hydroxyl protecting group ethyl R3CO Ph 78 hydroxyl protecting group ethyl R3CO 0-alkyl 79 hydroxyl protecting group ethyl R3CO O-olefinic 80 hydroxyl protecting group ethyl R3CO 0-aromatic 81 hydroxyl protecting group ethyl R3CO O-CH2Ph 82 hydroxyl protecting group methyl H 83 hydroxyl protecting group methyl R3CO alkyl 84 hydroxyl protecting group methyl R3CO olefinic 85 hydroxyl protecting group methyl R3CO aromatic 86 hydroxyl protecting group methyl R3CO Ph 87 hydroxyl protecting group methyl R3CO 0-alkyl 88 hydroxyl protecting group methyl R3CO 0-olefinic 89 hydroxyl protecting group methyl RsCO 0-aromatic 90 hydroxyl protecting group methyl R3CO O-CH2Ph 91 hydroxyl protecting group olefinic H 92 hydroxyl protecting group olefinic R3CO alkyl 93 hydroxyl protecting group olefinic R3CO olefinic 94 hydroxyl protecting group olefinic R3CO aromatic 95 hydroxyl protecting group olefinic R3CO Ph 96 hydroxyl protecting group olefinic R3C O O-alkyl 97 hydroxyl protecting group olefinic R3CO 0-olefinic 98 hydroxyl protecting group olefinic RsCO 0-aromatic 99 hydroxyl protecting group olefinic R3CO O-CH2Ph 100 hydroxyl protecting group aromatic H 101 hydroxyl protecting group aromatic R3CO alkyl 102 hydroxyl protecting group aromatic R3CO olefinic 103 hydroxyl protecting group aromatic R3CO aromatic 104 hydroxyl protecting group aromatic R3CO Ph 105 hydroxyl protecting group aromatic R3CO 0-alkyl 106 hydroxyl protecting group aromatic R3CO O-olefinic 107 hydroxyl protecting group aromatic R3CO 0-aromatic 108 hydroxyl protecting group aromatic R3CO O-CH2Ph It should be appreciated that various hydroxyl protecting groups contemplated for P1 in Table A above include, without limitation, benzyloxymethyl (BOM), benzyl, substituted benzyl, benzoyl, and the like.

C. Chiral resolution of the ester precursor As shown with reference to Figure 3 and as discussed below, the racemic ()-N-CBZ-3-phenylisoserine ethyl ester of Formula 7 (or a compound of the general formula 7a) is next purified by chiral chromatographic purification to separate the desired (2R, 3S)-3-N-CBZ-3- phenylisoserine ethyl ester (Formula 8) from its enantiomer (and diastereomers if present), by using a chromatographic stationary phase that has a different affinity for the optical isomers, such that they pass through the stationary phase at different rates and are accordingly chromatographically separated, wholly or partially, from one another. CBZ-NH O CBZ-NH O Chiral separation -5, OC2H5 S, S Whelk-0 Column- OU Formula 7 Formula 8 A number of columns and conditions were first evaluated using a racemic mixture of an N-benzoyl ethyl ester of the formula : The results of this evaluation are shown in Table 1, below : Table 1 Column Mobile K'of first a Baseline Phase eluted Resolved ? S, S Whelk-0, 50% MTBE/1. 9 1. 86 Yes 10µ FEC 48% Hexane/ 2% EtOH S, S Whelk-0, 30% EtOH/0. 786 1. 32 No 10µ FEC 70% Hexane S, S Whelk-0, 20% EtOH/2. 4 1. 25 No 10µ FEC 80% Hexane S, S Whelk-0, 10% EtOH/3. 8 1. 35 Barely 10 ! 1 FEC 90% Hexane S, S Whelk-0, 30% n-BuOH/0. 974 1. 4 No 10p FEC 70% Hexane Naproxen various------n. s. Napthyl Leucine various------n. s. ChiraIcel OD IPA/Hexane,------n. s. EtOH/Hexane ChiralPak AD 10% IPA/5. 0 1. 6 Barely 90% Hexane IPA : isopropanol EtOH : ethanol BuOH : butanol MTBE: methyl t-butyl ether n. s.: no separation k' : relative retention time of the compound a : selectivity of the column Baseline resolved : complete separation of the enantiomers to the HPLC baseline Based on these results, a number of conditions, including various mobile phases, were then evaluated to separate enantiomers of N-CBZ phenylisoserine ethyl ester of Formula 7, as shown in Table 2, below : Table 2 Column Mobile K'of first α Baseline Phase eluted Resolved ? S, S Whelk-0, 20% EtOH/2. 13 1. 45 Yes 10µ FEC 80% Hexane S, S Whelk-0, 7. 5% IPA/3. 253 1. 54 Yes 10µ FEC 7.5% EtOH/ 85% Hexane S, S Whelk-0, 10% IPA/2. 549 1. 54 Yes 10µ FEC 10% EtOH/ 80% Hexane S, S Whelk-O, 5% IPA/4. 603 1. 56 Yes 10µ FEC 5% EtOH/ 90% Hexane S, S Whelk-0, 20% IPA/2. 69 1. 63 Yes 1 Opt FEC 80% Hexane S, S Whelk-0, 50% MTBE/2. 28 1. 64 Yes 10O, FEC 50% Hexane S, S Whelk-0, 50% MTBE/1. 2 1. 64 Yes 10µ FEC 48% Hexane/ 2% EtOH IPA : isopropanol EtOH : ethanol MTBE: methyl t-butyl ether k' : relative retention time of the compound a : selectivity of the column Baseline resolved : complete separation of the enantiomers to the HPLC baseline Where a positive indication is given in the final column for"Baseline Resolved ? ", the chromatogram generally shows two distinct peaks, each rising from the approximate baseline and respectively corresponding to the two enantiomers, indicating virtually complete separation thereof. Where the final column indicates the baseline is"barely"resolved, the chromatogram generally shows two distinct peaks for which some overlap is observed above the baseline, suggesting partial but incomplete separation of the enantiomers. Where"n. s." is indicated in the final column, the chromatogram generally shows a single peak, suggesting little or no separation of the enantiomers. It should be noted that, under certain mixtures of alcohol/hexane mobile phases, two additional peaks appear on the chromatogram, which are believed to be the diastereomers. It should further be appreciated that additional minor peaks may be observed on the chromatogram, as generally understood in the art.

Interestingly, a racemic mixture of the benzoyl acid of the formula : was not separated under any of the conditions attempted, such as those set forth in Tables 1 and 2.

The best conditions achieved for the separation of the ethyl ester enantiomers of Formula 7 employed a stationary phase comprising S, S Whelk-0 bonded to 10t Kromasil silica. This S, S Whelk-0 stationary phase may be obtained from Regis Technologies, Inc., located in Morton Grove, IL.

In particular, various column lengths of the S, S Whelk-0 covalently bonded to spherical silica, irregular silica, or spherical Kromasil silica, in various sizes such as 101l and 5, are available from Regis Technologies. One version of this S, S Whelk-0 stationary phase, identified by Regis Technologies as S, S Whelk-0 1, has a formula as follows : A variation of the S, S Whelk-0 stationary phase, identified by Regis Technologies as S, S Whelk-0 2, has a covalent trifunctional linkage to the silica support. The present invention contemplates the use of either of these stationary phases, as well as other stationary phases/chromatographic media as discussed herein.

In the preferred process, the racemic CBZ-phenylisoserine ethyl ester was resolved into its respective enantiomers using a ProChrom HPLC system with an 8cm diameter column containing 800g of stationary phase, comprising fully endcapped S, S Whelk-O covalently bonded to 10/100 Kromasil silica. Whelk-O's key binding sites that affect the chromatographic separation are tetrahydrophenanthrene n systems and dinitrobenzamide z systems, in addition to amido-proton hydrogen bond donors. The mobile phase was 20% isopropanol/80% hexane; the flow rate was 300 mL per minute and detector wavelength was 220nm. A solution of 0.025 g of racemate per 1 mL of isopropanol was prepared and successive 60mL aliquots (equivalent to 1.5g of the racemic material) were injected. The desired enantiomer elutes first and was collected separately. Solvent was removed under reduced pressures to obtain the optically pure (2R, 3S)-N- CBZ-3-phenylisoserine ethyl ester product as white powder at 85% recovery.

It should be appreciated that other types of chromatographic media may be employed in the present invention, where such chromatographic media is operative to separate the enantiomers of racemic phenylisoserine ethyl ester and/or other compounds contemplated herein by chiral chromatographic resolution. For example, chromatographic media having as key binding sites one or more tetrahydrophenanthrene 7z systems, dinitrobenzamide z systems, or amido-proton hydrogen bond donors, or other similar binding sites, may be substituted for S, S Whelk-0 in the stationary phase of the present invention. Additionally, it should be appreciated that the silica substrate may be substituted with other appropriate chromatographic substrates, to the extent known in the art.

It should further be understood that while it is preferred that the enantiomers are completely separated, such that chromatographic peaks corresponding thereto are resolved to the HPLC baseline, the present invention appreciates that less efficient separations may occur wherein some overlap of the HPLC peaks is observed.

D. Use of the ester precursor in taxane synthesis After chiral chromatographic purification, the resulting optically pure, (2R, 3S)-3-N-CBZ-3-phenylisoserine ethyl ester may be used in the production of desired taxanes, such as by various methods known in the art.

For example, as shown with reference to Figure 4 and as discussed below, Formula 8 may now be protected at the 2-0-position with a suitable protecting group, such as BOM, benzyl, substituted benzyl, benzoyl and the like, and may thereafter be saponified to the corresponding carboxylic acid thereby to provide a side chain precursor suitable for coupling with a baccatin III derivative to form desired taxanes, such as paclitaxel, docetaxel, or their analogs. Exemplary preparations of taxanes using a (2R, 3S)-3-N-CBZ-3- phenylisoserine ethyl ester precursor are taught, for example, in U. S. Patent Nos. 5,675, 025; 5,684, 175; 5,688, 977; 5,750, 737; 5,770, 745; 5,939, 566; 5,948, 919; 6,048, 990; 6,066, 749; 6,072, 060; 6,107, 497; 6,136, 999; and 6,143, 902, the teachings of which are incorporated herein by reference.

For example, as taught in U. S. Patent No. 5,750, 737 to Sisti et al., the (2R, 3S)-3-N-CBZ-3-phenylisoserine ethyl ester may be protected at the 2- hydroxy position with a benzyloxymethyl (BOM) group as follows : CBZ-NH O CBZ-NH O - BOM-CI. Ph OC2H5-- Ph OC2H5 n-BuLi OH THF,-78°C OBOM Formula 8 Formula 9 Here, the ethyl ester of Formula 8 is dissolved in anhydrous THF under a nitrogen atmosphere and cooled to a reduced temperature such as- 40°C or-78°C, for example, in a dry ice/acetone bath followed by the dropwise addition of an alkyllithium agent such as n-butyl lithium, although it is desirable that the alkyllithium agent be a straight chain alkyl. In any event, the reaction is best done at a temperature no greater than 0°C. The resulting mixture is stirred for about ten minutes. Benzyloxymethyl chloride (BOM-CI) is then added dropwise over an interval of about five minutes, and the mixture stirred for approximately two to five hours at the reduced temperature. Thereafter, the solution is warmed to 0°C and quenched with water to eliminate excess n-butyl lithium. The resulting mixture is concentrated under vacuum to residue, and this residue is thereafter taken up in ethyl acetate and washed with water and brine to remove unwanted salts. The organic layer may then be dried and concentrated under vacuum and the residue recrystallized from ethyl acetate: hexane or chromatographed with ethyl acetate: hexane to give the ethyl ester of Formula 9.

Formula 9 may then be saponified to its corresponding acid as follows : CBZ-NH 0 CBZ-NH O Li0OH- OC2H5 EtOH : H20- OBOM OBOM Formula 9 Formula 10 Here, Formula 9 is dissolved in ethanol/water (ratio 8: 1). Lithium hydroxide (or other suitable alkali hydroxide) is added to the solution and the resulting mixture stirred for approximately three hours in order to saponify the compound. The mixture is then acidified (1 N hydrochloric acid) and extracted with ethyl acetate. The resulting organic layer is separated, dried and concentrated under vacuum. The residue acid is then isolated for use without further purification. This produces the acid of Formula 10.

As shown with reference to Figure 5 and as discussed below, Formula 10 may then be coupled to a suitable taxane backbone, such as a protected baccatin III or 10-deacetylbaccatin III backbone, as known in the art, and the coupled product may thereafter be converted to paclitaxel or other taxanes.

For example, as taught in U. S. Patent No. 5,750, 737, Formula 10 may be esterified with 7-CBZ baccatin III as follows : CBZ-NH O 11 Ph OH OBOM AcO O OCBZ Formula 10 toluene DMAP, DCC cgZ-NH O \Hw1 I L \ 13 60°C-80°C Ph3 OH= OAc 30min-lh OBOM OCOPh AcO 0 OCBZ Formula 12 10 7 Zozo Oh= oye OCOPh Formula 11 Here, Formula 11 (1 equivalent) and the acid side chain of Formula 10 (6 equivalents) are dissolved in toluene. To this mixture, 0.5 equivalents of dimethylamino pyridine (DMAP) and preferably 6 equivalents of dicyclohexylcarbodiimide (DCC) are added, and the resulting mixture heated at 70°C for thirty (30) minutes to one (1) hour, although the range of temperature could be 60°C to 80°C. Other dialkyl carbodiimides may be substituted for the DCC, with one example being diisopropylcarbodiimide.

Next, the solution is cooled to room temperature and an equal volume of ethyl acetate or diethyl ether is added to the solution. The resulting mixture is then cooled to 0°C and held at this temperature for twenty-four (24) hours.

After this time it is filtered, and the residue is rinsed with either diethyl ether or ethyl acetate. The combined organics are then washed with hydrochloric acid (5%), water, and finally brine. The organic phase is separated, dried and concentrated under vacuum. The resulting residue is then dissolved in ethyl acetate: hexane and eluted over a silica gel plug. The eluent is then concentrated under vacuum to result in the esterified compound of Formula 12.

As further taught in U. S. Patent No. 5,750, 737, the 3'-N-CBZ and 7-O- CBZ protecting groups may be removed by hydrogenation, the 3'-N-amino group may be acylated using benzoyl chloride as the acylating agent to add a benzoyl group at the 3'-N-position, and the 2'-O-BOM protecting group may be removed by further hydrogenation, thereby to produce paclitaxel : AcO O OCBZ tu CBZ-NH O/ ='\ 13 2 0 OH :- : OAc OBOM OCOPh Formula 12 AcO 0 OH Ph NH 0 10 7 Ici 10 Ph3NH O/ '"="'OH= OAc OH= OAc OH OCOPh Paclitaxel Here, the coupled product of Formula 12 is dissolved in isopropanol to which Pearlman's catalyst is added. The resulting mixture is hydrogenated at 40 psi for twenty-four hours, although alternatively, the mixture can be stirred under one atmosphere of hydrogen for 24 hours. Thereafter, the mixture is filtered through diatomaceous earth and reduced under vacuum to residue.

Preferably, the residue is taken up in toluene and anhydrous potassium carbonate added. Alternatively, the residue may be taken up in ethyl acetate or toluene and a tertiary amine base, such as triethylamine, is added. In either case, benzoyl chloride is then added dropwise, and the mixture stirred for two hours. The resulting mixture is then washed with water and finally brine. The resulting organic phase is then separated, dried, and concentrated under vacuum to yield C2'-OBOM paclitaxel. The BOM group is removed by dissolving the C2'-OBOM paclitaxel in isopropanol to which Pearlman's catalyst is added. This mixture is hydrogenated for 24 hours under 40 psi hydrogen to yield paclitaxel.

Additional processes for the semi-synthesis of taxanes such as paclitaxel, docetaxel and the like using various suitable side chain precursors and baccatin III backbones are described, for example, in U. S. Patent Nos.

5,675, 025; 5,684, 175; 5,688, 977; 5,939, 566; 5,948, 919; 5,973, 170; 6,048, 990; 6,066, 749; 6,072, 060; 6,107, 497; 6,133, 462; 6,136, 999; and 6,143, 902, and the teachings thereof are incorporated herein by reference. It should be appreciated that the present invention is particularly adapted to provide suitable (2R, 3S) side chain precursors for use in such processes.

Accordingly, the present invention has been described with some degree of particularity directed to the exemplary embodiment of the present invention. It should be appreciated, though, that the present invention is defined by the following claims construed in light of the prior art so that modifications or changes may be made to the exemplary embodiment of the present invention without departing from the inventive concepts contained herein.