PROCESS FOR PREPARING AN ENANTIOMERICALLY ENRICHED, DEUTERATED SECONDARY ALCOHOL FROM A CORRESPONDING KETONE WITHOUT REDUCING DEUTERIUM INCORPORATION

The present invention provides a process for the preparation of enantiomerically enriched, deuterated secondary alcohols of Formula 1-A by employing ketoreductases or carbonyl reductases without reducing deuterium incorporation.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application is claims the benefit of U.S. Provisional Patent Application No. 61/379,187, filed Sep. 1, 2010 and U.S. Provisional Patent Application No. 61/379,586, filed Sep. 2, 2010, the entire contents of which are hereby incorporated by reference.

BACKGROUND OF THE INVENTION

Deuterated (S) M-1 metabolites of pentoxifylline and related compounds have been reported as having therapeutic utility in WO2009/108383. There is a need for commercially feasible methods for producing such deuterated (S)-M-1 metabolites, as well as other deuterated (S)-alcohols.

SUMMARY OF THE INVENTION

Applicants have solved this problem by employing certain commercially available ketoreductases and carbonyl reductases to produce deuterated (S)-alcohols from the corresponding prochiral deuterated ketone in a highly stereoselective manner without loss of deuterium incorporation.

The present invention provides a process for the preparation of a compound of Formula I:

comprising reacting a compound of formula II:

with a hydride source or a deuteride source in the presence of a ketoreductase or carbonyl reductase to form a compound of formula I with an enantiomeric excess of at least 80%, wherein:

Y is H when a compound of Formula II is reacted with a hydride source; or

Y is D when a compound of Formula II is reacted with a deuteride source;

R1 is —CH3 or —CD3;

R2 is a C2-C10 alkylene-X wherein X is H, D, or R3 and the C2-C10 alkylene portion of R2 is optionally substituted with a group independently selected from the group consisting of (i) one or more deuterium, and (ii) one R3; and

R3 is (i) C6-C10 aryl, 5-10-membered heteroaryl, C3-C8 cycloalkyl, or saturated heterocyclyl, wherein R3 is optionally substituted with one or more substituents independently selected from deuterium, C1-C2 alkyl optionally substituted with deuterium, and —OH; or (ii) a tautomer thereof;

wherein at least one of R1 and the C2-C10 alkylene portion of R2 is substituted with deuterium; and

wherein the amount of deuterium incorporation at each deuterium in R1 and the C2-C10 alkylene portion R2 in the compound of formula I is substantially equal to the amount of deuterium incorporation at corresponding deuterium atoms in R1 and R2 in the compound of formula II.

The process of this invention is particularly useful to reduce deuterated forms of pentoxifylline to their corresponding deuterated alcohols.

DETAILED DESCRIPTION OF THE INVENTION Definitions

“The term “alkyl” refers to a monovalent, saturated hydrocarbon group having the indicated number or range of carbon atoms. For example, C2-C10 alkyl is an alkyl having from 2 to 10 carbon atoms. An alkyl may be linear or branched. Examples of alkyl groups include methyl; ethyl; propyl, including n-propyl and isopropyl; butyl, including n-butyl, isobutyl, sec-butyl, and t-butyl; pentyl, including, for example, n-pentyl, isopentyl, and neopentyl; and hexyl, including, for example, n-hexyl, 2-methylpentyl and heptyl.

The term “cycloalkyl” refers to a monovalent monocyclic or bicyclic saturated group containing only carbon ring atoms. The term “C3-C8 cycloalkyl” refers to a monocyclic saturated group containing between 3 and 7 carbon ring atoms. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, cycloheptyl, cis- and trans-decalinyl, and norbornyl.

The term “aryl” refers to an aromatic carbocycle. The term “C6-C10 aryl” refers to a monocyclic or bicyclic, aromatic carbocycle containing between 6 and 10 ring carbon atoms. Examples of aryl are phenyl and naphthyl.

The term “saturated heterocyclyl” refers to a monovalent monocyclic or bicyclic saturated group containing between 3 and 8 ring atoms, wherein one or more ring atoms is a heteroatom independently selected from N, O, and S. Examples of saturated heterocycles include azepanyl, azetidinyl, aziridinyl, imidazolidinyl, morpholinyl, oxazolidinyl, piperazinyl, piperidinyl, pyrazolidinyl, pyrrolidinyl, tetrahydrofuranyl, and thiomorpholinyl.

The term “heteroaryl” refers to a monovalent monocyclic or bicyclic aromatic group, wherein one or more ring atoms is a heteroatom independently selected from N, O, and S. A 5-10 membered heteroaryl is a monocyclic or bicyclic heteroaryl that contains between 5 and 10 ring atoms. Examples of heteroaryl groups include furanyl, thiazolyl, isothiazolyl, isoxazolyl, oxazolyl, pyrimidinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrrolyl, thiadiazolyl, thiophenyl, triazinyl, triazolyl, quinolinyl, quinazolinyl, indolyl, isoindolyl, 3,7-dihydro-1H-purine-2,6-dion-yl; xanthinyl, hypoxanthinyl, theobrominyl, uric acid, isoguaninyl, thymine, and uracilyl.

The term “ketoreductase or carbonyl reductase” refers to an enzyme belonging to Enzyme Classification Class 1.1.1.184, which, in the presence of which a hydride source, is capable of converting a methyl ketone into a secondary alcohol. The term “methyl ketone” refers to a ketone of the formula:

wherein RA is CnH2n+1 and n is an integer between 2 and 10.

The term “substituted” refers to the replacement of one or more hydrogen atoms with the indicated substituent. For avoidance of doubt, substitutions may occur on the terminus of a moiety. For example, the terminal —CH3 group on R2 may be substituted with an R3. “Substituted with deuterium” refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.

When a position is designated specifically as “D” or deuterium, the position is understood to have deuterium at an abundance that is at least 1000 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 15% incorporation of deuterium). In certain embodiments, when a position is designated as “D” or deuterium that position has at least 50.1% incorporation of deuterium, at least 75% incorporation of deuterium; at least 80% incorporation of deuterium, at least 85% incorporation of deuterium; at least 90% incorporation of deuterium; at least 95% incorporation of deuterium; at least 98% incorporation of deuterium; at least 99% incorporation of deuterium; or at least 99.5% incorporation of deuterium.

When a position is designated specifically as “H” or hydrogen, the position is understood to have hydrogen at its natural isotopic abundance.

The amount of deuterium incorporation at a deuterium atom in a compound of Formula I is said to be “substantially equal” to the amount of deuterium incorporation at the corresponding deuterium atom in a compound of Formula II if the difference in the amount of deuterium incorporation between them is no more than about 5%, as an example no more than about 3%; no more than about 2%; no more than about 1%; or no more than about 0.5%.

Ketones Useful as Compounds of Formula II

It will be understood that each of R1 and R2 in a compound of Formula I,

is structurally identical to the corresponding R1 and R2 in a compound of Formula II,

Notwithstanding this, according to the present invention, the amount of deuterium incorporation at any deuterium present in R1 and R2 of a compound of Formula I is substantially equal to the amount of deuterium incorporation at the corresponding deuterium atoms in a compound of Formula II.

In one embodiment, R2 is substituted with one or two R3.

In one embodiment, at least one of R1 and R2 comprises a deuterium bound to the carbon atom adjacent the carbonyl carbon.

In one embodiment, each of R1 and R2 is substituted with one or more deuterium. In one aspect of this embodiment at least one of R1 and R2 comprises a deuterium bound to the carbon atom adjacent the carbonyl carbon.

In one embodiment, R1 is CH3.

In another embodiment, R1 is CD3.

In one embodiment, R2 is C2-C6 alkyl optionally substituted with one or more deuterium and optionally substituted with one or two R3. In one aspect of this embodiment, R2 is —CH2—(C1-C5 alkyl optionally substituted with one or more deuterium and optionally substituted with one R3. In another aspect of this embodiment, R2 is —CH2—(C1-C5 alkyl optionally substituted with one or more deuterium and optionally substituted with one R3).

In one embodiment, R3 is (i) 5-10-membered heteroaryl optionally substituted with one or more substituents independently selected from deuterium, C1-C2 alkyl optionally substituted with deuterium, and —OH; or (ii) a tautomer thereof.

In one embodiment, a compound of Formula II has structural Formula II-A:

or a salt thereof, wherein:

each of R4, R6 and R7 is independently selected from —CH3 and —CD3;

R5 is hydrogen or deuterium or a combination thereof;

each Z2 is the same and is hydrogen or deuterium;

each Z3 is the same and is hydrogen or deuterium;

each Z4 is the same and is hydrogen or deuterium;

each Z5 is the same and is hydrogen or deuterium; and

either R7 is —CD3 or at least one of Z2, Z3, Z4 and Z5 is deuterium. In such an embodiment, the corresponding compound of Formula I has structural Formula I-A:

wherein R4, R5, R6, R7, Z2, Z3, Z4 and Z5 are as defined for Formula II-A; and Y is as defined for Formula I.

In one embodiment of Formula I-A and II-A, each R7 is —CD3.

In one embodiment of Formula I-A and II-A, each Z2 is deuterium.

In another embodiment of Formula I-A and II-A, each Z2 is hydrogen.

In one embodiment of Formula I-A and II-A, each R5 is deuterium.

In one embodiment of Formula I-A and II-A, each R5 is hydrogen.

In one embodiment of Formula I-A and II-A, each Z3, Z4 and Z5 is hydrogen. In one aspect of this embodiment each R6 is —CH3 and each R7 is —CD3. In a more specific aspect, each R4 is —CH3, each R6 is —CH3; and each R7 is —CD3. In another aspect of this embodiment each R6 is —CD3 and each R7 is —CD3.

In one embodiment of Formula I-A and II-A, each Z3, Z4 and Z5 is deuterium.

In one embodiment of Formula I-A and II-A, each R6 and each R4 is —CD3.

In one embodiment of Formula II-A, the compound is selected from any one of:

In one embodiment of Formula I-A, the compound is selected from the following:

In one specific aspect, the compound of Formula II-A is

and the compound of Formula I-A is

Typically, the preparation of Compound 407 from pentoxifylline results in partial deuteration at the 8-position in the 3,4,5,7-tetrahydro-1H-purine-2,6-dione ring (<70%). In one embodiment, Compound 421(5) is further converted to Compound 121(S)

by treatment with K2CO3 and water. Thus, maintenance of the deuteration at that position during the enzymatic conversion of Compound 407 to Compound 421(S) is unimportant. In one embodiment, compound 121(S) is substantially free of 421(S). “Substantially free” means that the amount of 421(S) is equal to or less than 5%, more preferably equal to or less than 1%, or more preferably equal to or less than 0.1%, of the amount of 121(S).

In one embodiment, any compound of formula I-A having a group

may be further converted to a compound having the same structure except for having a group

by treating with a suitable base and a proton source, such as water.

Applicants have discovered that the use of a ketoreductase or carbonyl reductase to reduce a deuterium-containing ketone compound of Formula II, in particular a compound of Formula II-A, allows for both high enantiomeric enrichment and substantially no loss of deuterium incorporation. In particular, the amount of deuterium incorporation at each deuterium in R1 and R2 in the compound of formula I is substantially equal to the amount of deuterium incorporation at corresponding deuterium atoms in R1 and R2 in the compound of formula II. This is particularly surprising in that it was unexpected that one could provide buffer conditions that (1) allowed the ketoreductase or carbonyl reductase to efficiently reduce the ketone without also allowing the enzyme to catalyze deuterium-to-hydrogen exchange on the existing deuterium atoms; (2) would not result in an acidic pH which would be expected to cause deuterium-to-hydrogen exchange on the existing deuterium atoms; and (3) would not require for activity sufficiently basic or acidic conditions that would also be expected to cause deuterium-to-hydrogen exchange.

In a related embodiment the invention provides a method of making Compound 133(S)

comprising the step of reacting pentoxifylline:

with a deuteride source in the presence of a ketoreductase or carbonyl reductase and an appropriate catalytic co-factor at a pH between 6.5 and 7.3 to form a compound of formula I with an enantiomeric excess of at least 90% and deuterium incorporation of at the hydroxy carbon of Compound 133(S) of least 90%.

Any ketoreductase or carbonyl reductase that produces a 90% or greater enantiomeric excess of the (S)—OH stereoisomer of Formula II may be utilized in the method of this invention. Commercially available kits containing different ketoreductases or carbonyl reductases are available from multiple vendors. A determination of whether or not a particular ketoreductase or carbonyl reductase produces a 90% or greater enantiomeric excess of the (S)—OH stereoisomer of Formula II may be achieved by standard techniques well-known in the art. For example, a compound of Formula II may be incubated with the ketoreductase or carbonyl reductase to be screened in the presence of a hydride or deuteride source.

In one embodiment, the ketoreductase is a naturally occurring ketoreductase selected from the group consisting of Candida magnoliae ketoreductase, Candida parapsilosis ketoreductase, and Sporobolomyces salmicolor ketoreductase.

In one embodiment, the ketoreductase or carbonyl reductase is selected from any one or ALMAC Carbonyl Reductases CRED A131, CRED A801, CRED A901, CRED A251, or CRED A271 (each commercially available from ALMAC Group Ltd, Craigavon, England), any one of CODEXIS Ketoreductases KRED-119, KRED-137, KRED-148, KRED-169, KRED-174, KRED-NAD 101, KRED-NAD 102, KRED-NAD112, or KRED-NAD 126 (each commercially available from Codexis Inc., Redwood City, Calif.), or SYNCORE Ketoreductases ES-KRED-121, ES-KRED-128, ES-KRED-130, ES-KRED-142, ES-KRED-175, ES-KRED-169, or ES-KRED-171 (each commercially available from Syncore Labs, Shanghai, China). In one aspect of this embodiment, the enzyme is selected from CRED A131, CRED A251, KRED-NAD 101, KRED-NAD 102, KRED-NAD112, KRED-NAD 126, ES-KRED-121, ES-KRED-128, ES-KRED-130, ES-KRED-142, ES-KRED-169, or ES-KRED-171. In a more specific aspect, the enzyme is selected from CRED A131, CRED A251, and KRED-NAD 101.

In one embodiment, the amount of enzyme used in the reaction ranges from 0.05 wt % to 10 wt % as a percentage of the weight of the substrate, such as 0.5 wt % to 5 wt %. In one embodiment, the amount of enzyme is between 1.0 wt % and 2.0 wt %. In a more specific aspect, the amount of enzyme is about 1.0 wt %.

In one embodiment, the compound of formula I is formed with an enantiomeric excess of at least 90%. In one aspect of this embodiment, the enantiomeric excess is at least 94%. In a more particular aspect of this embodiment, the enantiomeric excess is at least 96%. In a more particular aspect of this embodiment, the enantiomeric excess is at least 98%.

The process of this invention requires the presence of a hydride source or a deuteride source. The term “hydride source” refers to a compound or mixture that is capable of providing a hydride anion or a synthetic equivalent of a hydride anion. Similarly, the term “deuteride source” refers to a compound or mixture that is capable of providing a deuteride anion or a synthetic equivalent of a deuteride anion. A hydride or deuteride source comprises a co-factor, which may be in catalytic or stoichiometric amounts. When the co-factor is in a catalytic amount, the hydride or deuteride source comprises a co-factor regeneration system.

A co-factor used with the ketone reductase or carbonyl reductase in the process of this invention is selected from NAD, NADP, NADH, NADPH, NAD2H and NADP2H. The choice of co-factor may be based upon (a) the presence or absence of a co-factor regeneration system; (b) the requirement for a hydride versus a deuteride source; and (c) compatibility with the specific ketone reductase or carbonyl reductase employed. In embodiments where the hydride or deuteride source does not comprise a co-factor regeneration system, the co-factor is in a stoichiometric amount and is a reduced co-factor which is therefore selected from NADH and NADPH for a hydride source, or NAD2H and NADP2H for a deuteride source. It is well known in the art—or information is available from the commercial supplier of the specific ketone reductase or carbonyl reductase—whether NADH or NADPH is the appropriate co-factor for a given ketone reductase or carbonyl reductase. In this embodiment, the reduced co-factor is present in stoichiometric amounts as compared to the compound of Formula II.

In another embodiment, the hydride or deuteride source additionally comprises a co-factor regeneration system. The high cost of co-factors, in particular the deuterated co-factors, makes their use on a stoichiometric basis impractical. A low-cost co-factor regeneration system continually produces and regenerates the reduced form of the co-factor, requiring the co-factor to be present in only catalytic amounts. Moreover, the use of a co-factor regeneration system eliminates the need to use a reduced co-factor or a deuterated co-factor. The co-factor regeneration system produces the required reduced or reduced and deuterated co-factor in situ. Accordingly, any cofactor or combinations of cofactors compatible with the chosen ketone reductase or carbonyl reductase can be employed with a co-factor regeneration system. In this embodiment, therefore, NAD is interchangeable with NADH and NAD2H; and NADP is interchangeable with NADPH and NADP2H. Similarly, the designations “-NAD” and “-NADH”, and “-NADP” and “-NADPH”, respectively, are used interchangeably herein in conjunction with enzymes that use, respectively, NADH and NADPH as co-factors.

When used in conjunction with a co-factor regeneration system the amount of co-factor can range from 0.1 wt % to 5 wt %. In one aspect of this embodiment, the amount of co-factor is between 1 wt % and 3 wt %. In an alternate aspect of this embodiment, the amount of cofactor is between 0.1 wt % and 1.1 wt %.

A typical co-factor regeneration system consists of a dehydrogenase and a substrate for that dehydrogenase. Without being bound by theory or mechanism, Applicants believe that upon catalysis by the dehydrogenase, its substrate provides a hydride or deuteride anion to regenerate (reduce) the cofactor. The newly reduced cofactor can then subsequently donate a hydride or deuteride atom to the compound of Formula II to provide a compound of Formula I. In certain embodiments, the substrate for the dehydrogenase may be generated in situ from the corresponding ketone and a reducing agent. A second co-factor regeneration system takes advantage of the fact that certain ketoreductases and carbonyl reductases both possess alcohol dehydrogenase activity. In this system an alcohol dehydrogenase substrate is used and upon catalysis donates the hydride or deuteride ion to the co-factor.

Examples of hydride cofactor regeneration systems include, but are not limited to, reducing sugars and their corresponding hydrogenase , e.g., glucose and glucose dehydrogenase (“GDH”), glucose-6-phosphate and glucose-6-phosphate dehydrogenase, etc.; formate and formate dehydrogenase; a secondary (e.g., isopropanol) alcohol and a secondary alcohol dehydrogenase; phosphite and phosphite dehydrogenase; molecular hydrogen and hydrogenase; and ethanol, aldehyde dehydrogenase and an alcohol dehydrogenase.

Examples of deuteride co-factor regeneration systems include, but are not limited to, deuterated reducing sugars and their corresponding dehydrogenase, e.g., deuterated glucose and GDH, deuterated glucose-6-phosphate and glucose-6-phosphate dehydrogenase, etc.; deuterated formate and formate dehydrogenase; a secondary deuterated (e.g., deuterated isopropanol) alcohol alone or together with a secondary alcohol dehydrogenase; deuterated phosphite and phosphite dehydrogenase; molecular deuterium and hydrogenase; and deuterated ethanol and aldehyde dehydrogenase optionally together with an alcohol dehydrogenase.

In one embodiment of the process, the deuteride co-factor regeneration system comprises a substrate having a —C(D)OH functional group and its associated dehydrogenase. In a related embodiment, the substrate having a —C(D)OH functional group is generated in situ. In this related embodiment, the deuteride co-factor regeneration system comprises (a) a compound having a C═O functional group; (b) a metal deuteride or a mixed metal deuteride, such as a borodeuteride or aluminum deuteride of a metal such as sodium or lithium, capable of reducing the C═O functional group to a —C(D)OH functional group; and (c) a dehydrogenase that acts upon the —C(D)OH functional group. As an example, the substrate having a —C(D)OH functional group is a C1-C6 alcohol such as CH3C(D)(OH)CH3. As another example, the substrate having a —C(D)OH functional group is carbohydrate of the formula C6H11DO6, such as deuterated glucose (shown below in its open chain and pyranose forms):

As yet another example the deuterated glucose is generated in situ from D-glucono-δ-lactone and NaBD4. This embodiment is advantageous in that an otherwise expensive deuterated glucose substrate is generated in situ from relatively inexpensive D-glucono-δ-lactone and NaBD4.

Moreover, the inventors have discovered that the use of deuterated glucose (or D-glucono-δ-lactone and NaBD4) as part of the deuteride source consistently produced high deuterium incorporation (>90%) at the Y position of a compound of Formula I and in particular a compound of Formula I-A. The use of deuterated glucose in the deuteride source in the production of

from pentoxifylline:

in accordance with this invention will also result in a surprisingly high deuterium incorporation at the indicated position of Compound 133(S).

When a combination of (a) GDH, glucose and a co-factor; (b)(i) GDH, deuterated glucose and a cofactor; or (b)(ii) GDH, D-glucono-δ-lactone, a metal deuteride or a mixed metal deuteride, and a cofactor is used as (a) the hydride source or (b) the deuteride source, respectively, the amount of GDH in the reaction can range from 0.01 wt % to 5 wt %. The term “wt %” means the amount of substance that is the recited percent of the amount of substrate present on a wt/wt basis. In one embodiment, the amount of GDH is between 0.05 wt % and 0.15 wt %. In an alternate embodiment, the amount of GDH is between 0.1 wt % and 0.2 wt %.

An appropriate pH to perform the method according to the present invention means buffer conditions that maintain the pH at between 6.0 and 7.5 throughout the reaction. In one embodiment, the pH of the reaction was maintained at between 6.5 and 7.3. In another embodiment, the pH of the reaction was maintained between 6.0 and 7.0. Typically dropwise addition of KOH is used to maintain the desired pH because the enzymatic reaction generates acid. In one aspect, the pH of the reaction is maintained between 6.90 and 7.05. If the pH of the reaction is allowed to drop below the desired range, the enzyme will typically become irreversibly inactivated and the compound of Formulae I and II subject to acid-catalyzed deuterium-to-hydrogen exchanged.

In one embodiment of the process of the invention, the process is performed at a temperature of about 20° C. to 37° C. In one aspect of this embodiment, the temperature is about 29° C. to 32° C.

In one embodiment of the process of the invention, the process is performed over a time period of about 12 hours to about 24 hours. In one embodiment, the time period is about 24 hours to about 40 hours. In one embodiment, the time period is about 40 hours to about 72 hours. In one embodiment, the time period is a time period sufficient for less than about 5% of the initial amount of compound of formula (II) to be present.

EXAMPLE 1 Preparation of Compound 407

In a 50-L Jacket Glass Reactor, pentoxifylline (900 g) was reacted with deuterium oxide (99% of “D”, 2.7 L) in the presence of potassium carbonate (0.25 equiv) in toluene (18 L) at 85-87° C. (refluxing) over four hours. The reaction mixture was cooled to 55° C. and the agitation was stopped to allow the layers to separate. The reaction mixture was held overnight at 55° C. 1H NMR analysis of an IPC sample of the organic layer showed the deuterium incorporation was 94.4% “D” at the methyl position. The bottom aqueous layer was collected. Following the same exchange conditions, a second exchange was conducted with a solution of potassium carbonate (0.25 equiv) in deuterium oxide (99% of “D”, 2.7 L). After separation, a third exchange was conducted with a solution of potassium carbonate (0.25 equiv) in deuterium oxide (99.8% of “D”, 2.7 L). 1H NMR analysis of an IPC sample of the organic layer showed the deuterium incorporation was 99.6% “D” at the methyl position after three exchanges. The organic layer was concentrated to ca. 5.5 L (6 vol) at 60° C. (Batch temperature) and cooled slowly. The solids were formed at 36° C. and n-heptane (1.8 L) was added to form a thin slurry. The slurry was stirred at 25° C. over the weekend and filtered to provide white solids (825 g, wet). The solids were dried in vacuum oven (28.5 inch Hg) at 45-48° C. over night to afford Compound 407 (778.5 g, 84.6% yield). 1H NMR analysis of Compound 407 showed that the deuterium incorporation was 99.7% “D” at the methyl position. An HPLC purity check by area showed that the purity was 99.96%.

EXAMPLE 2 Screening of Carbonyl Reductases and Ketoreductases ALMAC Carbonyl Reductases

The ability of the 40 individual carbonyl reductases in the ALMAC Carbonyl Reductase (CRED) Screening Kit to convert Compound 407 to Compound 421(S)

was tested as follows:

Into a vial was added 1 mL of a 15 mg/mL solution of the CRED to be tested in 0.1M K2HPO4, pH 7.0. To that vial was added 100 μL of a 300 mg/mL solution of glucose; 100 μL of a 10 mg/mL solution of the appropriate co-factor NAD or NADP (as indicated in the table below) in 0.1M K2HPO4, pH 7.0; 100 μL of a 20 mg/mL solution of glucose dehydrogenase in 0.1M K2HPO4, pH 7.0; and approximately 20 mg of Compound 407 in 50-150 μL of DMSO or MTBE. The sample was shaken or stirred overnight at 30° C. The sample was then extracted with METE or EtOAc and analyzed by TLC and certain select samples by chiral GC/HPLC to determine conversion to Compound 421(5) and enantiomeric enrichment. The results are set forth in Table 1.

TABLE 1 Screening of ALMAC Carbonyl Reductases Estimated Conversion Conversion S/R Ratio of Enzyme Cofactor By TLC By HPLC Alcohol A101 NADP 10%  N/A N/A A201 NADP 0% N/A N/A A301 NADP 5% N/A N/A A401 NADP 10%  N/A N/A A501 NADP 0% N/A N/A A601 NADP 100%  99.90%  0.2/99.8 A701 NADP 0% N/A N/A A801 NADP 20%   8.20% 92.4/7.6 A901 NADP 50%  32.10% 96.5/3.5 A121 NADP 10%  N/A N/A A131 NAD 100%  99.70% 99.7/0.3 A141 NADP 5% N/A N/A A151 NAD 35%  20.10%  35.0/65.0 A161 NAD 100%  99.90%  2.6/97.4 A171 NAD 0% N/A N/A A181 NADP 10%  N/A N/A A191 NAD 0% N/A N/A A211 NADP 0% N/A N/A A221 NAD 0% N/A N/A A231 NADP 5% N/A N/A A241 NADP 0% N/A N/A A251 NAD 100%  99.90% 99.9/0.1 A261 NADP 0% N/A N/A A271 NADP 45%  36.50% 99.4/0.6 A281 NADP 40%  36.70%  73.2/26.8 A291 NADP 90%  97.20%  0.1/99.9 A311 NADP 100%  99.80%  0.1/99.9 A321 NADP 0% N/A N/A A331 NADP 5% N/A N/A A341 NADP 10%  N/A N/A A351 NADP 0% N/A N/A A361 NADP 0% N/A N/A A371 NADP 50%  51.60%  73.0/27.0 A381 NADP 0% N/A N/A A391 NADP 0% N/A N/A N501 NADP 0% N/A N/A N701 NADP 0% N/A N/A N121 NADP 0% N/A N/A N131 NADP 5% N/A N/A N151 NADP 10%  N/A N/A

The results of this experiment demonstrated that ALMAC Carbonyl Reductases CRED A131, CRED A801, CRED A901, CRED A251, or CRED A271 were useful in the process of this invention.

Codexis Ketoreductases

The CODEXIS Ketoreductase (KRED) Enzyme Screening Kit contained 34 enzymes that used either NADP (Kit KRED-22000) or NAD (Kit KRED-12000) as a co-factor. The screening Kit KRED-22000 contained 22 KRED-NADP enzymes and their screening test results were carried out according to manufacturer' s directions using approximately 5 mg of KRED-NADP enzyme and 71 mg of Compound 407 and the appropriate CODEXIS buffer system (KRED-NADPH Recycle Mix A or KRED-NADH Recycle Mix A) for each reaction. Reactions were tested by TLC (data not shown) and selected reactions were tested by chiral HPLC after 40 hours. These results are set forth in Tables 2 and 3. Results indicated by a dash (“-”) indicate an insufficient amount of Compound 421(S) or its stereoisomer were produced by TLC to warrant detection by HPLC.

TABLE 2 Screening of Codexis Ketoreductases in Kit KRED-22000. KRED-NADP Conversion S/R Ratio of Enzyme By HPLC Alcohol KRED-101 26.30%  29.8/70.2 KRED-102 KRED-103 KRED-107 KRED-112 23.80%  27.2/72.8 KRED-113 KRED-118 52.80%  7.8/92.2 KRED-119 99.70% 91.6/8.4 KRED-121 KRED-128 41.80%  72.0/28.0 KRED-129 KRED-130 99.50%  78.7/21.3 KRED-131 KRED-137 25.60% 96.4/3.6 KRED-140 KRED-142 KRED-147 KRED-148 76.70% 99.5/0.5 KRED-149 KRED-164 KRED-169 65.10% 98.9/1.1 KRED-174 73.90% 99.2/0.8

TABLE 3 Screening of Codexis Ketoreductases in Kit KRED-12000. KRED- NADH Conversion S/R Ratio of Enzyme By HPLC Alcohol 101 100% 100/0 102 100% 100/0 107 108 109 110 100%   1.5/98.5 112 99.90%  99.8/0.2 113 119 121 124 126 100% >99.9/0.1

The above results indicated that CODEXIS Ketoreductases KRED-119, KRED-137, KRED-148, KRED-169, KRED-174, KRED-NADH 101, KRED-NADH 102, KRED-NADH 112, and KRED-NADH 126 were useful in the process of this invention.

Syncore Ketoreductases

SYNCORE Ketoreductase (ES-KRED) Enzyme Screening Kit contained 75 enzymes. Twenty-one ketoreductases (NADH dependent) were tested for the reduction of Compound 407 on 100 mg scale. The CODEXIS buffer system (KRED-NADH Recycle Mix A) was used for these screening tests. The reduction was conducted with 5 wt % of enzyme loading in 50 vol of buffer at 30° C. The reactions were checked by TLC (data not shown) and selected reaction mixtures were checked by chiral HPLC and worked up. Results indicated by a dash (“-”) indicate an insufficient amount of Compound 421(S) or its stereoisomer were produced by TLC to warrant detection by HPLC. The results are summarized in Table 4.

TABLE 4 Screening of Syncore NADH-dependent Ketoreductases. Conversion S/R Ratio of Enzyme By HPLC Alcohol ES-KRED-121  99.62% 100/0 ES-KRED-122 ES-KRED-125 ES-KRED-126 100.00%   0/100 ES-KRED-128  99.42%  99.83/0.17 ES-KRED-129 ES-KRED-130 100.00% 100/0 ES-KRED-132 ES-KRED-133 ES-KRED-134 ES-KRED-137 ES-KRED-138 ES-KRED-141 ES-KRED-142 100.00% 100/0 ES-KRED-143 ES-KRED-144 ES-KRED-155 ES-KRED-159 ES-KRED-165 ES-KRED-166 ES-KRED-175  22.71% 100/0

The remaining 54 NADPH dependent ketoreductases from Syncore were also tested for the reduction of Compound 407 (100 mg) using the CODEXIS buffer system (KRED-NADPH Recycle Mix A). The reduction was conducted with 5 wt % of enzyme loading in 50 vol of buffer at 30° C. The reactions were checked by TLC (data not shown) and selected reaction mixtures were checked by chiral HPLC and worked up. Results indicated by a dash (“-”) indicate an insufficient amount of Compound 421(S) or its stereoisomer were produced by TLC to warrant detection by HPLC. The results are summarized in Table 5.

TABLE 5 Screening of Syncore NADPH-dependent Ketoreductases. Conversion S/R Ratio of Enzyme By HPLC Alcohol ES-KRED-101 ES-KRED-102 ES-KRED-103 ES-KRED-104 ES-KRED-105 ES-KRED-106 ES-KRED-107 ES-KRED-108 ES-KRED-109 ES-KRED-110 ES-KRED-111 ES-KRED-112 ES-KRED-113 ES-KRED-114 ES-KRED-115 ES-KRED-116 ES-KRED-117 ES-KRED-118 ES-KRED-119 ES-KRED-120 99.30% 0.18/99.82 ES-KRED-123 ES-KRED-124 ES-KRED-127 ES-KRED-131 99.90% 0.06/99.94 ES-KRED-135 ES-KRED-136 ES-KRED-139 ES-KRED-140 ES-KRED-145 ES-KRED-146 ES-KRED-147 ES-KRED-148 ES-KRED-149 ES-KRED-150 ES-KRED-151 ES-KRED-152 ES-KRED-153 ES-KRED-154 ES-KRED-156 ES-KRED-157 ES-KRED-158 ES-KRED-160 ES-KRED-161 ES-KRED-162 ES-KRED-163 ES-KRED-164 ES-KRED-167 ES-KRED-168 ES-KRED-169 99.96% 99.94/0.06  ES-KRED-170 ES-KRED-171 100% 98.70/1.30  ES-KRED-172 ES-KRED-173 ES-KRED-174

From the above experiments, it was determined that SYNCORE Ketoreductases ES-KRED-121, ES-KRED-128, ES-KRED-130, ES-KRED-142, ES-KRED-175, ES-KRED-169, and ES-KRED-171 were suitable for use in the present invention.

EXAMPLE 3 Effect of pH on Enzymatic Reduction of Pentoxifylline CRED A131

To determine the pH effect on the enzymatic reduction using CRED A131, reductions of pentoxifylline (100 mg) were conducted using different buffers with different initial pH values between 6.7 and 7.3 using glucose/glucose dehydrogenase as the hydride source. The reactions were checked by HPLC after 18-20 hours. The results demonstrated that the conversion of pentoxifylline to its corresponding (S)-alcohol increased with increased initial pH of the buffers. However, no reactions were complete even after two days. Moreover, the final pH of the reactions was between 4.1 and 5.66 suggesting that the decrease in the pH during the course of the reaction decreased the enzymatic activity.

To further investigate the buffer pH effect on the reduction, three reactions of pentoxifylline (100 mg) using 1 mg of CRED A131 in 20 mL of buffer (pH: 7.0, 7.2, and 7.3) were conducted. The results are summarized in Table 5.

TABLE 5 Effect of pH on CRED A131 Reduction of Pentoxifylline. Buffer Conversion Mixture Conversion Mixture Conversion Mixture pH (16 hrs) pH (40 hrs) pH (100 hrs) pH 7 43.60% 6.71 60.40% 6.42 67.50% 6.15 7.2 46.80% 6.91 61.90% 6.71 73.20% 6.44 7.3 51.20% 6.98 69.40% 6.8 80.30% 6.51

After 16 hours, the reaction went to 40-50% conversion and the pH of the solution went down 0.3 pH units. After 40 hours, the reaction conversion increased to 60-70% and pH of the reaction mixtures went down another 0.2-0.3 pH units. After 100 hours, the reaction conversion increased only 7-10% and the pH of the reaction mixtures went down further. The highest conversion with 1 wt % of CRED A131 loading was 80%. This indicated that if the pH of the reaction mixture were kept at 6.5 to 7.3, the enzymatic activity could be prolonged. This would enable minimization of the enzyme use.

Based on the above results, we conducted a reaction with 1.0 g of Compound 407 and 50 mg of CRED A131 at a constant pH in buffer solution (pH at 6.95 to 7.05). The results are summarized in Table 6. The reaction conversion followed a first order kinetic curve fit with a half time of about 1.5 hours. The reaction went to 96.8% conversion after 8 hours and to completion overnight (100% conversion). A total of 3.5 mL of 1M KOH (1.0 equiv) solution was added during first 5 hours. The reaction was run overnight and the final pH went to 7.00 from 7.04. The reaction mixture was worked up. The first DCM extraction (50 mL) provided 0.88 g of Compound 421(S) as a single S-enantiomer by chiral HPLC. 1H NMR showed that deuteration of the methyl group was maintained at 99.3% “D”. This result demonstrated that these reduction conditions were suitable for the reduction of deuterated ketone Compound 407.

TABLE 6 Time Course of CRED A131 Conversion of Compound 407 to Compound 421(S). Reaction Reaction Conversion First Order Kinetic Predicted Time (hour) by HPLC Conversion (t1/2 = 1.5 hrs) 1 35.90% 37.00% 2 60.70% 60.30% 3 74.80% 75.00% 4 83.20% 84.70% 5 90.30% 90.10% 6 93.50% 93.70% 7 95.80% 96.10% 8 97.80% 97.50% 20 100% 99.99%

KRED-101, -102, -112 and -126

We tested each of KRED-101, -102, -112 and -126 for the effect of pH on their ability to reduce Compound 407 to Compound 421(5). The manufacturer's recommended starting pH for reactions using KRED-101, -112, and -126 is 7.0. For KRED-102 the recommended starting pH is 7.5. Reactions used 100 mg of Compound 407, 2.5 mg KRED enzyme, 5 mg of NAD, 150 mg of glucose, and 0.35 mg of GDH in 5 mL of buffer. As with the CRED A131, the reactions did not go to completion after 24 hours and the final pH after that time was below 5. This again suggested control of pH during the reaction was necessary.

In a second experiment we used 667 mg of Compound 407, 10 mg KRED enzyme, 20 mg of NAD, 1.0 g of glucose, and1.4 mg of GDH in 30 mL of buffer. For KRED-101, -112, and -126, the pH was maintained at between 6.90 and 7.05. For KRED-102, the pH was maintained between 7.40 and 7.55. After 24 hours, each of the reactions using KRED-101, -112, and -126 demonstrated at least 99.98% completion. The reaction with KRED-102 demonstrated 59.91% completion.

The completed reactions were worked up and the crude products were isolated. The results are shown in Table 7, below.

TABLE 7 Analytical Results of Crude Products Produced from Reduction of Compound 407 by KRED Enzymes. Isolated 1H NMR at KRED-NADH Crude methyl HPLC Chiral Enzyme Yield position Purity HPLC 101 100% 98.9% “D” 99.86%   100:0 (S/R) 126  92% 98.8% “D” 99.62% 99.88:0.12 (S/R) 112 107% 99.0% “D” 99.86% 99.52:0.48 (S/R)

Enzyme KRED-NADH 101 provided the best chiral selectivity (100/0 of S/R) and purity of the crude product (99.86%). The purity of crude product isolated from KRED-NADH 101 reduction was 99.86% and no single impurity was higher than 0.05% by HPLC. The crude product isolated from KRED-NADH 126 reduction contained 0.26% of Compound 407. The deuteration at the methyl group of these crude products was checked by 1H NMR and found to be between 98.8% “D” and 99.0% “D”. All tested KRED showed sufficient chiral selectivity of at least 99.52% (greater than 99% ee).

EXAMPLE 4 Preparation of Compound 421(S) from Compound 407 Using CRED A131

A 12-L 3-necked RB flask equipped with a heating mantle, a J-Kem thermocouple, a mechanical stirrer, a reflux condenser, and a pH probe was charged with CRED A131 (9.5 g, ALMAC lot #IM-1311-061-1) and 2 L of buffer solution (0.1M KH2PO4, pH=7.0, same as below). The reaction mixture was stirred to dissolve all solids. A solution of glucose (558 g, Aldrich lot #088K0039) in buffer (2 L) was added in one portion followed by a solution of NAD (19.25 g, Spectrum lot #YA0655) in buffer (500 mL), and a solution of GDH (1.5 g, ALMAC lot #IM-1311-131-1) in buffer (500 mL). The initial reaction mixture was pH 6.98. A mixture of Compound 407 in buffer (3 L) at 30° C. was added to the reaction mixture and the container was rinsed with buffer (1.6 L). The rinse was charged to the reactor. The pH of the reaction mixture was 6.99. The reaction mixture was warmed to 30° C. and monitored by pH meter. The reaction temperature was kept at 29.0 to 31.5° C. and the pH of the reaction mixture was kept between pH 6.93 and pH 7.02 by adding 4M KOH solution drop-wise as needed. The reaction was complete after 22 hours with 99.96% conversion as determined by HPLC. The chiral HPLC analysis of the resulting product showed the chiral selectivity was 99.85% to the desired S-alcohol.

The reaction mixture was mixed with NaCl (2 kg) and extracted with EtOAc (1×4 L and 3×2 L). During the first extraction, a rag layer was formed and the reaction mixture was filtered through a celite pad. No further issues with phase separation were encountered after the filtration. The combined organic extracts were concentrated to about 1.5 L at 50-60° C. and n-heptane (2 L) was added to precipitate the solids. The slurry was cooled to 20° C. and filtered. The flask was rinsed with filtrate to complete the transfer. The filter cake was washed with n-heptane (2×500 mL) and dried over the weekend at 40-50° C. to afford Compound 421(S) (366 g, 94% yield). The product was analyzed by HPLC (99.95% purity), chiral HPLC (99.88/0.12 S/R), and 1H NMR (99.5% “D” incorporation at the methyl position).

EXAMPLE 4 Preparation of Compound 421(S) from Compound 407 Using KRED-NADH-101

A 3-necked 12-L RB flask equipped with a heating mantle, a J-Kem thermocouple, a mechanical stirrer, and a pH probe was charged with glucose (547.5 g, Aldrich lot #088K0039) followed by buffer (9.5 vol, 3.47 L). The reaction mixture was stirred to dissolve all solids. A mixture of Compound 407 (365 g) in buffer (2.92 L) was added and the container was rinsed with buffer (1.28 L). The rinse was added to the reactor. Initially, the reaction mixture was a very thin milky suspension. A solution of KRED-NADH-101 (3.65 g, CODEXIS lot #1021908WW), NAD (2.19 g, SPECTRUM lot #YA0655), GDH (365 mg, CODEXIS lot #22016700017) in buffer solution (1.46 L) was charged to the reactor. The container was rinsed with buffer (2×0.91 L) and the rinses were added to the reactor. The reaction mixture was warmed to 20-30° C. and monitored by a pH meter. The reaction mixture turned clear after 30 minutes. The pH of the reaction mixture was maintained between 6.50 and 6.90 by adding 4M KOH solution drop-wise as needed. The reaction was monitored by HPLC and was complete after 5 hours with 99.97% conversion by HPLC. The reaction mixture was stirred at 20-25° C. overnight and warmed to 30° C. for the work-up.

Sodium chloride (1.825 kg) was added to the reaction mixture and dissolved completely after stirring for 15 minutes. The batch was extracted with EtOAc (10 vol). The organic phase contained a thin solid gel, which collapsed into a slimy separate phase between the aqueous and organic layers immediately when agitated slightly. The slime could be retained on a paper filter but formed a thin impermeable layer that prevented flow through the filter. It was observed on a sample that a small amount of filter aid (celite) easily adsorbed the slime. The aqueous layer was charged back to the reactor and extracted with EtOAc (10 vol). Filter aid (100 g) was charged to the reactor to absorb the slime. The batch was filtered (less than one hour) and the organic layer was collected. The aqueous layer was then extracted with EtOAc (2×5 vol) without any problems (no further slime or emulsion was observed). The combined organic extracts were concentrated to ca. 10 volume and polish filtered to remove a small amount of the inorganic solids. The filtrate was concentrated further to ca.5 volumes and product solids were precipitated. n-heptane (8 vol) was added to the slurry (at 40-60° C.) over 30 minutes. The slurry was stirred overnight at 20-25° C. and filtered. The filter cake was washed with n-heptane (2×1 vol). The wet cake (370 g) was dried at 40-50° C. over the weekend to afford Compound 421(S) as a white solid (332.0 g, 90.0% yield). The filtrate was concentrated followed by precipitation with n-heptane to afford a second crop of Compound 421(S) (7.1 g, 1.9% yield). In order to check the mass balance of the product, the aqueous layer was extracted again with EtOAc (10 vol) and afforded only 4.8 g of Compound 421(S) (1.3% yield) of product as a white solid. The combined mother liquor was concentrated to afford 2.0 g of Compound 421(S) as a yellow solid (0.5% yield). The isolated product was a very high quality (100% purity by HPLC) and a single enantiomer (100/0 S/R % by chiral HPLC) from the main lot with 99.5% “D” incorporation at the methyl position by 1H NMR.

EXAMPLE 5 Preparation of Compound 437(S) from Compound 407 Using KRED-NAD-101 and Deuterated Glucose Generated In Situ

a) In situ Generation of Deuterated Glucose: Deuterated glucose (D-[1-2H1] Glucose) was prepared according to the procedure described in Liebigs Ann. Chem. 1992, 1201-1203. D-Glucono-δ-lactone (5 g, 28.09 mmoles) was added in one portion to ice-cold water (35 mL, 0-3° C.) and stirred for 10 min. A freshly prepared, ice-cold solution of NaBD4 (0.294 g, 7.02 mmoles, 99% D) in 10 mL of water was added slowly during 10 min. The reaction is slightly exothermic (2° to 10° C.) and the pH of the reaction was 7.42. Stirring was continued for 30 min, keeping the temperature by cooling to 0-3° C. Acetic acid (0.32 mL, 5.61 mmoles) was added and stirring was continued further 30 min.

b) Preparation of Compound 437(S) from Compound 407:

The reaction mixture obtained in step (a) was diluted with 18 ml mL of water and the solution was heated to 25-30° C. KH2PO4 (0.85 g) was added to the mixture and the pH was adjusted to 7 with 4M KOH solution. To this was added 2.5 g (8.8 mmoles) of 407. A solution of NAD (15 mg), GDH (2.5 mg), KRED 101 (25 mg) in 12.5 mL of 0.1 KH2PO4 buffer was added. The resulting solution was stirred at 25-30° C. The pH of the reaction mixture was maintained between 6 and 7 by adding 4M KOH solution drop-wise. The reaction was monitored by HPLC and was complete after 12 hours with 99.97% conversion by HPLC. Sodium chloride (12.5 g) was added and stirred for 30 min. The mixture was extracted with ethyl acetate (3×25 mL). The organic layer was separated, filtered through celite pad and concentrated to a small volume (−5 vol) and product solids were precipitated. Heptanes (20 mL) were added to the slurry (at 40-60° C.) over 10 minutes. The slurry was stirred overnight at 20-25° C. and filtered. The wet cake was dried at 50° C. for 12 hours to afford 437(S) as a white solid. (2.12 g, 85% yield). The isolated product purity was >99.5% by HPLC and as a single enantiomer by chiral HPLC. The deuterium incorporation at the methine position was ˜95% D.

EXAMPLE 6 Preparation of Compound 121(S) from Compound 421(S)

In a 3-L 3-necked RB flask, Compound 421(S) (100 g) was charged followed by water (1.0 L) and K2CO3 (0.25 equiv). The reaction mixture was heated to 80±5° C. and monitored by 1H NMR. The reaction was complete after 24 hours and worked up after 65 hours. The resulting product was extracted with three times with EtOAc and the solid products from the three extractions combined and re-dissolved in 5 volumes of EtOAc at 60-65° C. n-heptane (5.5 vol.) was added at 60-65° C. over 15 minutes and cooled to 20° C. over night (16 hrs). The slurry was filtered and the wet cake was washed with n-heptane (2×1 vol. to afford product Compound 121(S) after drying at 40-50° C. A total of 92.4 g of Compound 121(S) was isolated. HPLC purity was 99.92% (AUC) and chiral selectivity was 100% to “S” enantiomer. The 1H NMR analysis showed 99.2% of “H” at the 8-position in the 3,4,5,7-tetrahydro-1H-purine-2,6-dione ring and 99.4% of “D” at the methyl position.

Claims

1.-23. (canceled)

24. A process for the preparation of a compound of Formula I-A comprising the step of reacting a compound of Formula II-A: or a salt thereof, with a hydride source or a deuteride source in the presence of a ketoreductase or carbonyl reductase to form a compound of Formula I-A with an enantiomeric excess of at least 80%, wherein: wherein either R7 is —CD3 or at least one of Z2, Z3, Z4 and Z5 is deuterium, and wherein the amount of deuterium incorporation at each deuterium in R7, Z2, Z3, Z4 and Z5 in the compound of Formula I-A is substantially equal to the amount of deuterium incorporation at corresponding deuterium atoms in R7, Z2, Z3, Z4 and Z5 in the compound of Formula II-A.

for each position designated as deuterium in the compound of Formula I-A and Formula II-A, that position has at least 90% incorporation of deuterium;
the ketoreductase or carbonyl reductase is selected from CRED A131, CRED A801, CRED A901, CRED A251, CRED A271, KRED-119, KRED-137, KRED-148, KRED-169, KRED-174, KRED-NAD 101, KRED-NAD 102, KRED-NAD112, KRED-NAD 126, ES-KRED-121, ES-KRED-128, ES-KRED-130, ES-KRED-142, ES-KRED-175, ES-KRED-169, and ES-KRED-171;
each of R4, R6 and R7 is independently selected from —CH3 and —CD3;
each R5 is hydrogen or deuterium or a combination thereof;
each Z2 is the same and is hydrogen or deuterium;
each Z3 is the same and is hydrogen or deuterium;
each Z4 is the same and is hydrogen or deuterium;
each Z5 is the same and is hydrogen or deuterium; and
Y is H when a compound of Formula II-A is reacted with a hydride source; or
Y is D when a compound of Formula II-A is reacted with a deuteride source,

25. The process of claim 24, wherein each R7 is —CD3.

26. The process of claim 24, wherein each Z2 is deuterium.

27. The process of claim 24, wherein each Z2 is hydrogen.

28. The process of claim 24, wherein each R5 is deuterium.

29. The process of claim 24, wherein each R5 is hydrogen.

30. The process of claim 24, wherein each Z3, Z4 and Z5 is hydrogen.

31. The process of claim 30, wherein R6 is —CH3 and R7 is —CD3.

32. The process of claim 31, wherein R4 is —CH3.

33. The process of claim 30, wherein R6 is —CD3 and R7 is —CD3.

34. The process of claim 24, wherein each Z3, Z4 and Z5 is deuterium.

35. The process of claim 24, wherein: and

a. the compound of Formula II-A is selected from any one of the following:
b. the compound of Formula I-A is selected from any one of the following:

36. The process of claim 24, wherein: and the compound of Formula I-A

the compound of Formula II-A is

37. The process of claim 36, further comprising the step of converting Compound 421(S) to Compound 121(S):

38. The process of claim 24, wherein the hydride source or deuteride source is (i) a cofactor selected from NAD, NADH, NADP and NADPH, (ii) a reducing sugar and (iii) a dehydrogenase that dehydrogenates the reducing sugar.

39. The process of claim 38, wherein Y is H; and the hydride source is (i) a cofactor selected from NAD, NADH, NADP and NADPH, (ii) glucose and (iii) glucose dehydrogenase.

40. The process of claim 24, wherein the step of reacting a compound of Formula II-A or a salt thereof with a hydride source or a deuteride source is performed at a pH between 6.0 and 7.5.

41. The process of claim 40, wherein the pH is between 6.90 and 7.05.

42. The process of claim 24, wherein the ketoreductase or carbonyl reductase is selected from CRED A131, CRED A251, and KRED-NAD 101.

43. The compound of claim 24, wherein for each position designated as deuterium, that position has at least 95% deuterium incorporation.

Patent History
Publication number: 20150376665
Type: Application
Filed: Jul 1, 2015
Publication Date: Dec 31, 2015
Inventors: Ramanaiah C. Kanamarlapudi (Bridgewater, NJ), Steven A. Weissman (Short Hills, NJ), Emerich Eisenreich (Claremont, CA), Xuejun Liu (Arcadia, CA)
Application Number: 14/789,840
Classifications
International Classification: C12P 17/18 (20060101); C07D 473/10 (20060101);