Sei sulla pagina 1di 7

A two-step enzymatic resolution of glycidyl butyrate

Dahai Yu
a
, Lei Wang
a
, Qiang Gu
b
, Peng Chen
a
,
Yan Li
a
, Zhi Wang
a,
*
, Shugui Cao
a,
*
a
Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, Jilin University, Changchun 130023, PR China
b
Department of Chemical Technology and Applied Chemistry, College of Chemistry, Jilin University, Changchun 130023, PR China
Received 29 January 2007; received in revised form 22 June 2007; accepted 25 June 2007
Abstract
A two-step enzymatic resolution process for production of (R)- and (S)-glycidyl butyrate was investigated and the lipases were screened. The
rst step involved a hydrolysis of (R,S)-glycidyl butyrate catalyzed by porcine pancreatic lipase (S-favored) with an E of 21 for production of (R)-
glycidyl butyrate (13.2 mmol, 98% ee, 36% yield) under the optimal conditions (pH 7.4, 30 8C, 30 mg/ml CTAB). Then, the recovered (R)-
enriched glycidol (19.8 mmol, 65%ee, 56%yield) was used for transesterication catalyzed by Novozym435 (R-favored) with an E of 69 to obtain
(S)-glycidyl butyrate (15.1 mmol, 98% ee, 42% yield) under the optimum conditions (a
W
= 0.24, n-heptane, 80 min).
# 2007 Elsevier Ltd. All rights reserved.
Keywords: Two-step; Resolution; Glycidyl butyrate; Porcine pancreatic lipase; Novozym 435
1. Introduction
Optically pure 2,3-epoxy-1-propanol (glycidol) and its
derivatives are versatile intermediates in organic synthesis
because the epoxide ring is reactive towards nucleophiles for the
synthesis of asymmetric alcohols [13]. Particularly, both
enantiomers of the (R)- and (S)-glycidyl butyrate are chiral
synthesis units for the production of biologically active
compounds of commercial interest. (R)-Glycidyl butyrate has
been used to introduce a stereogenic center in the synthesis of
Linezolid [4,5], which is currently marketed for the treatment of
multidrugresistant Gram-positive infections such as nosocomial,
community-acquired pneumonia, and skin infections. (S)-
Glycidyl butyrate is also very useful as starting material in the
synthesis of many drugs, such as (R)-argentilactone [6] which
exhibits both antileishmanial activity and cytotoxic activity
against mouse leukemia cells [7]. Recently, lipases have been
widely used in the resolution of (R,S)-glycidyl butyrate [810].
Due to the existence of a lid in most lipases [1114], the open
close status of the conformation may be changed dramatically
under various experimental conditions, which in turn strongly
affects the activity and enantioselectivity of lipases [15,16].
Many methods (such as medium engineering, immobilization
techniques et al.) are often used to improve the activity and the
enantioselectivity of the lipase. However, the enantioselectivity
of most lipases toward (R,S)-glycidyl butyrate is still low with a
poor yield [17,18]. Although the (R)-glycidyl butyrate can be
obtained from hydrolysis of (R,S)-glycidyl butyrate in ton
quantities, it is necessary to reach high conversions for obtaining
adequate enantiomeric excesses (ee). It is more difcult to
prepare (S)-glycidyl butyrate on a large scale because most
lipases present (S)-stereochemical preference in hydrolysis of
(R,S)-glycidyl butyrate, producing (R)-glycidyl butyrate and (R)-
glycidol. Palomo et al. [19] have reported the enzymatic
hydrolytic resolution of (R,S)-glycidyl butyrate by using
Novozym 525L, which presents an enantioselectivity towards
the (R)-glycidyl butyrate, and obtained the (S)-glycidyl butyrate.
However, the (S)-glycidyl butyrate in their reports is only
obtained at 90% ee and less than 36% yield for its low
enantioselectivity of Novozym 525L.
Contrary to the transformations based on the classic kinetic
resolution (KR) catalyzed by enzymes, where one enantiomer
can be obtained with high enantiomeric excess [20], two-step
enzymatic resolution is a method to obtain both enantionmers
with high enantiomeric excess by subjecting the isolated
enantiomerically enriched product of the rst enzymatic
resolution to the second [21], which results in the increase
www.elsevier.com/locate/procbio
Process Biochemistry 42 (2007) 13191325
* Corresponding authors. Tel.: +86 431 88498972; fax: +86 431 88980440.
E-mail addresses: wangzhi@jlu.edu.cn (Z. Wang), caosg@jlu.edu.cn
(S. Cao).
1359-5113/$ see front matter # 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.procbio.2007.06.012
of the enantiomeric excess of the product in the second step. A
good example has been proposed by Fishman et al. [22], who
use CALB as catalyst for a two-step enzymatic resolution of
(R,S)-ethyl-3-hydroxybutyrate (HEB) and both enantiomers of
(R)- and (S)-HEB are obtained at over 96% enantiomeric
excess, with a total process yield of 73%. Similarly, Cong et al.
[23] have reported the double kinetic resolution of the (R,S)-2-
octanol through transesterication by Pseudomonas cp. lipase.
The produced (S)-2-octanol and (R)-2-octanol in their results
have been obtained with 98% ee and 95% ee, respectively and
80% racemic substrate are converted to enantiopure products.
Zada et al. [24] also reported the production of (R)-lavandulol
with 96.7% ee and (S)-lavandulol with 92.6% ee by combining
the two-cycle enzymatic transesterication of racemic lavan-
dulol with vinyl acetate as acyl donor.
In the present study, an efcient method to produce (R)- and
(S)-glycidyl butyrate with high enantiomeric purity is performed
by using a two-step enzymatic resolution with sequential
hydrolysis and transesterication by porcine pancreatic lipase
(PPL) and the immobilized Candida antarctica lipase B
(Novozym435) withthe opposite enantioselectivity, respectively
(Scheme 1). The reaction conditions for the hydrolysis and
transesterication are also optimized.
2. Materials and methods
2.1. Materials
(R,S)-Glycidyl butyrate was prepared according to the procedure previously
described [25]. Lipase from Pseudomonas sp. (PSL) and Candida cylindracea
A.Y. lipase (AYL) was purchased fromAmano Pharmaceutical Co. Ltd. (Japan).
Novozym 435 was purchased from Novo (Bagsvaerd, Denmark). Candida
rugosa lipase (CRL) was purchased from Sigma (St. Louis, MO). Porcine
pancreatic lipase (PPL) was purchased from Shanghai Dongfeng Biochemical
Reagent Co. Ltd. (China). Candida lipolytic lipase (CLL) was provided by
Wuxi enzyme preparation plant (China). (R)-Glycidol, (S)-glycidol, (R)-gly-
cidyl butyrate and (S)-glycidyl butyrate were purchased from Sigma (St. Louis,
MO) to determine the retention times. Tween-80 was purchased from Tianjin
Chemical Reagent Company (China). Bis-(2-ethylhexyl) sodiumsulfosuccinate
(AOT), hexadecyltrimethyl ammonium bromide (CTAB), (R,S)-glycidol, vinyl
butyrate and other reagents of analytical grade were purchased from Shanghai
Chemical Reagent Company (China).
2.2. Enantioselective hydrolysis of (R,S)-glycidyl butyrate
The hydrolysis was performed by adding (R,S)-glycidyl butyrate (5 ml,
36 mmol) and the lipase (100 mg) into sodium phosphate buffer (5 ml, pH 7.4,
10 mmol) containing surfactant CTAB (30 mg/ml), with magnetically stirred at
200 rpm and 30 8C. And the pH was kept constant by automatic titration of
aqueous sodium hydroxide (NaOH, 4 M) by using a pH controller. The activity
(mmol/min mg
prot
) of the lipases in hydrolysis of (R,S)-glycidyl butyrate and the
conversion were calculated based on the consumption of NaOH. Samples were
recovered and analyzed by chiral high performance liquid chromatography
(HPLC) at various time intervals. The hydrolysis was stopped at about 60%
conversion by removing the lipase through centrifugation (11197 g, 5 min),
and then the reaction mixture was extracted with ether (10 ml) over three times.
The combined organic extracts were subsequently washed with 10% (v/v)
aqueous HCl (20 ml), saturated aqueous sodium bicarbonate (20 ml), brine
(20 ml) and nally dried with anhydrous magnesium sulfate. The remained
glycidyl butyrate was in ether layer while the generated glycidol was in the
aqueous layer after extraction. The remained glycidyl butyrate (13.2 mmol) was
obtained after the removal of ether by rotary evaporation (50 8C, 150 mbar).
2.3. Recovery of the generated glycidol
The generated glycidol in the rst step was isolated by extraction of the
aqueous layer with dichloromethane (20 ml) after prior removal of the glycidyl
butyrate with ether. The organic phase concentrated by rotary evaporation
(30 8C, 200 mbar) was analyzed by chiral gas chromatography (GC) and
19.8 mmol glycidol (65% ee) was recovered.
2.4. Enantioselective transesterication of the recovered glycidol
The transesterication were performed by using the recovered glycidol
(19.8 mmol, 65% ee), vinyl butyrate (40 mmol), n-heptane (5 ml), water
activity (a
W
= 0.24) and lipase (10 mg) with magnetically stirred at 40 8C
and 200 rpm. The transesterication was stopped at around 80% conversion by
removing the lipase through centrifugation (11197 g, 5 min). After the
removal of the vinyl butyrate by rotary evaporation (50 8C, 100 mbar), the
mixture was extracted with saturated aqueous sodium bicarbonate (10 ml) over
three times to wash out the remaining glycidol and dried with anhydrous
magnesium sulfate. (S)-Glycidyl butyrate (15.1 mmol) was obtained. All
experiments were repeated three times with the measurement errors less than
5%, and the illustrative graphs were based on the average values.
2.5. Water activity setting
All the reaction mixture components were pre-equilibrated to the water
activity (a
W
) of the experiment through the vapor phase with saturated salt
solutions at 25 8C [26,27]: LiBr (a
W
= 0.06), LiCl (a
W
= 0.11), KCH
3
COO
(a
W
= 0.24), MgCl
2
(a
W
= 0.33), Mg(NO
3
)
2
(a
W
= 0.53), NaCl (a
W
= 0.75),
K
2
SO
4
(a
W
= 0.97). Equilibration was performed overnight.
2.6. Analytical methods
The extracted glycidyl butyrate were diluted by the mobile phase of n-
hexane and ethanol (90:10, v/v) at a ow rate of 0.4 ml/min and analyzed by
chiral HPLC using a Chiralpak AD column. UV detection was performed at
254 nm. The retention time of the (R)- and (S)-glycidyl butyrate were 14.3 min
and 13.6 min, respectively.
The recovered glycidol was analyzed directly by GC on an Agilent 6890
instrument equipped with a ame ionization detector (FID) and a chiral column
(HP-Chiral Cap. 30 m 0.25 mm 0.25 mm). The temperatures of the injec-
tor and the detector were 200 and 280 8C, respectively. Nitrogen was used as the
Scheme 1. Outline of the process developed for the production of (R)- and (S)-
glycidyl butyrate.
D. Yu et al. / Process Biochemistry 42 (2007) 13191325 1320
carrier gas at a ow rate of 60 ml/min. Temperature programming was
performed between 60 and 240 8C with the increment of 12 8C/min. The
retention time of the (R)-glycidol and (S)-glycidol was 12.4 min and
12.9 min, respectively.
The enantiomeric excess of glycidyl butyrate and glycidol were determined
by calculating the peak areas of the enantiomers, and the enantioselectivity (E
value) was calculated according to Chen et al. [28]:
enantiomeric excesses; ee
s
%
R S
R S
100 (1)
enantioselectivity; E
ln1 C1 ee
s

ln1 C1 ee
s

(2)
E
ln1 C1 ee
0
=1 ee
o

ln1 C1 ee
0
=1 ee
o

(3)
In Eq. (1), R and S represent the concentrations of the (R)-glycidyl butyrate and
(S)-glycidyl butyrate, respectively and the enantioselectivity for the rst step
was calculated by using Eq. (2).
The enantioselectivity for the second step was calculated by using equation
(3), where ee
o
and ee
0
represent the enantiomeric excess of the initial substrate
and the enantiomeric excess of the recycled product fraction, respectively.
3. Results
3.1. Screening lipases for hydrolysis of (R,S)-glycidyl
butyrate
Lipase catalytic resolutions depended mainly on the type
and origin of the enzyme. Some commercially available lipases
from various sources had been selected for the further study as
the catalyst (Table 1). It could be found that almost all the
selected lipases possessed (S)-stereoselectivity while Novozym
435 exhibited opposite stereoselectivity with E = 7. In the
present study, our aim was to obtain two enantiomers with high
enantiomeric purity, PPL which exhibited highest activity and
enantioselectivity was selected as the biocatalyst for the
hydrolysis. Notably, there was a conformation conversion
between the glycidyl butyrate and the as-produced glycidol
(Scheme 2). Crude PPL was used directly in the experiments
concerning the expense and difculty for purication although
it had been reported that 25 kDa protein, among three proteins
in PPL, was responsible for the resolution of glycidyl butyrate
[29]. As for Novozym 435, it may be the best candidate for the
following transesterication due to its (R)-stereoselectivity.
3.2. Screening optimum reaction conditions for hydrolysis
catalyzed by PPL
It was important to screen the optimum conditions because
the enantioselectivity of PPL in hydrolysis of (R,S)-glycidyl
butyrate was moderate.
3.2.1. Effect of pH
In general, enzyme activity and enantioselectivity were pH-
depended. In the present study, the effect of pH in the range of
6.09.0 was investigated. The maximum activity and enantios-
electivity of PPL was obtained at pH 7.8 and 7.4, respectively
(Fig. 1), while a further increase in the pH to 9.0 resulted in a
sharp decrease in both. The activity and enantioselectivity
might be signicantly affected by the breaking of weak ionic or
hydrogen bonds on enzyme caused by the increase of pH [21].
And 7.4 was selected as the optimum pH for further study.
3.2.2. Effect of temperature
The effect of temperature on the activity and enantioselec-
tivity of PPL was examined in the range of 1050 8C. The
results (Fig. 2) demonstrated that the maximum activity of PPL
was obtained at 30 8C with a higher enantioselectivity (E = 21),
and then 30 8C was selected as the optimum temperature for
further investigation.
3.2.3. Effect of surfactant
It was well described in the literature that the surfactants can
be applied in lipase assay to increase the lipidwater interfacial
area, which, in turn, enhanced the observed rates of lipase-
catalyzed reactions [30,31]. In order to assess the effect of
surfactants on hydrolysis, three surfactants including non-ionic,
cationic and anionic as additives were examined. As shown in
Fig. 3, the activity of PPL was drastically improved in the
presence of the cationic surfactant (CTAB), whereas the anionic
surfactant (AOT) and non-ionic surfactant (Tween-80) inhib-
ited the enzyme activity. Under the optimum additive condition
(30 mg/ml CTAB), the activity of PPL was about 1.8-fold than
that in pure buffer medium. It was well known that lipases
displayed a very complex mechanism of action, including the
process of interfacial activation [3234]. Lipases may exist
in two different forms. Where the active center of the lipase was
protected from the reaction medium by a polypeptide chain
lid, the enzyme was considered to be inactive (closed form).
When the lid was displaced and the active center exposed to the
reaction medium, the enzyme was active (open form). In
aqueous or homogeneous media, lipase molecules exist in
equilibrium between these two forms, with this equilibrium
shifting towards the closed form [35]. This dramatic
conformational change may make the activity of lipases
strongly depend on the experimental conditions. In the present
study, the open form of PPL might be stabilized by the addition
of CTAB, resulting in the high enzyme activity [36,37]. On the
other hand, the changes of E value were not observed with the
Table 1
Effect of lipase sources on the enantioselective hydrolysis of (R,S)-glycidyl
butyrate
Lipases Enzyme activity
(mmol/min mg)
Conversion
(%)
ee
(%)
a
E
value
b
Stereoselectivity
PPL 2.4 60 98 21 S
PSL 3.1 74 93 6 S
CRL 1.6 29 24 5 S
AYL 0.4 5 2 2 S
CLL 0.1 4 1 1 S
Novozym
435
1.3 18 16 7 R
Conditions: (R,S)-glycidyl butyrate (5 ml, 36 mmol), lipases (100 mg), sodium
phosphate buffer (5 ml, pH 7.4, 10 mM) and CTAB (30 mg/ml) were performed
at 30 8C and 200 rpm for 6 h.
a
ee of (R)-glycidyl butyrate.
b
Calculated according to Eq. (2).
D. Yu et al. / Process Biochemistry 42 (2007) 13191325 1321
addition of the surfactants. Under the optimum conditions (pH
7.4, 30 8C, 30 mg/ml CTAB), (R)-glycidyl butyrate with 98%
ee and 36% yield (13.2 mmol) was obtained. However, the
surfactants made the workup much difcult in the reaction, and
it would necessarily lead the products hard to be separated on a
larger scale. Further investigation on proper methods to
separate or substitute the surfactant is under way.
3.3. Screening lipases for transesterication of the
generated glycidol
The side-product glycidol was recycled and analyzed after
separating the (R)-glycidyl butyrate. The analysis showed that
the generated glycidol was (R)-enriched with 65% ee and
55.6% yield (19.8 mmol). In order to obtain the ideal product in
the following procedure, lipases favoring (R)-glycidol should
be selected. As shown in Table 2, Novozym 435 exhibited the
(R)-stereoselectivity whereas the other lipases exhibited (S)-
stereoselectivity. Considering the conguration of the substrate
and the stereoselectivity of the lipases, it was obvious that
Novozym 435 was the best source of lipase used in
transesterication.
3.4. Screening optimum reaction conditions for
transesterication catalyzed by Novozym 435
3.4.1. Effect of organic media
Many reports demonstrated that organic media inuenced
not only the enzymatic activity but also the enantioselectivity
[38]. In this study, effects of the solvents with different log P
(logarithm of the partition coefcient of a given solvent
between n-octanol and water and was widely used to denote the
polarity or hydrophobicity of a solvent) were investigated
(Table 3). When polar solvents such as 1,4-dioxane or DMF
were used, the enzyme activity and E value were very low.
When the reaction was performed under polar solvents, water
exhibited higher afnity in solvent rather than bound to the
enzyme, so the hydrophilic solvent disrupts the functional
structure of enzyme by stripping off the essential water from
the enzyme whose function was to preserve the correct
Scheme 2. Conformational conversion of the (S)-glycidol butyrate to the (R)-glycidol.
Fig. 1. Effect of pH on the activity (*) and enantioselectivity (&) of PPL in
hydrolysis of (R,S)-glycidyl butyrate. The reaction ask contained (R,S)-
glycidyl butyrate (5 ml, 36 mmol), PPL (100 mg), sodium phosphate buffer
(5 ml, 10 mM) with pH varying from 6.0 to 9.0, and CTAB (30 mg/ml) was
performed at 30 8C and 200 rpm for 6 h.
Fig. 2. Effect of temperature on the activity (*) and enantioselectivity (&) of
PPL in hydrolysis of (R,S)-glycidyl butyrate. The reaction ask contained (R,S)-
glycidyl butyrate (5 ml, 36 mmol), PPL (100 mg), sodium phosphate buffer
(5 ml, pH 7.4, 10 mM), and CTAB (30 mg/ml) was performed at the reaction
temperature varied from 10 to 50 8C at 200 rpm for 6 h.
D. Yu et al. / Process Biochemistry 42 (2007) 13191325 1322
conformation of enzyme molecular [19]. In addition, the
strong interactions of polar solvent with enzyme resulted in
the variation of the conformation of the enzyme active site
and decreased the E values [39,40]. As shown in Table 3, there
was an increase in both of the enzyme activity and E value
with the hydrophobicity of solvents increased. The highest
enzyme activity and E value were achieved in n-heptane as the
solvent.
3.4.2. Effect of water activity
Water activity was a crucial parameter in nonaqueous
enzymology [4143]. In the present study, the a
W
was
controlled by the addition of salts/salt hydrates in the organic
solvent or substrate as described by Halling and the activity and
the enantioselectivity of Novozym 435 were investigated in n-
heptane in a wide range of initial a
W
values (0.060.97). At low
a
W
values (0.060.11), low enzyme activity was observed,
followed by a considerable increase with a summit at a
W
value
of 0.24, and a further increase in the initial a
W
value to 0.97
resulted in a strong decrease in enzyme activity (Fig. 4). It was
known that water in the reaction mixture could act as competing
nucleophile for acyl-enzyme thus suppressing the expected acyl
transfer. Furthermore, the decrease in enzyme activity at higher
initial a
W
values can also be ascribed to the observed enzyme
particle aggregation that may, in turn, had limited access of the
substrate to the enzyme active site. In contrast, the lower
enzyme activity obtained at lower a
W
values may be attributed
to a decrease in enzyme exibility [44]. As far as the effect of
a
W
on enantioselectivity was concerned, the results indicated
Fig. 3. Effect of CTAB concentration (*), Tween-80 concentration (&) and
AOT concentration (^) on the activity of PPL in hydrolysis of (R,S)-glycidyl
butyrate. The reaction ask contained (R,S)-glycidyl butyrate (5 ml, 36 mmol),
PPL (100 mg), sodiumphosphate buffer (5 ml, pH7.4, 10 mM) were performed
at 30 8C and 200 rpm for 6 h. The concentration of the surfactant was varied
from 0 to 50 mg/ml.
Table 2
Effect of lipase sources on the transesterication of the (R)-enriched glycidol
Lipases Enzyme activity
(mmol/min mg)
Conversion
(%)
ee
(%)
a
E
value
b
Stereoselectivity
PPL 1.1 23 97 16 S
PSL 1.4 35 99 54 S
CRL 0.5 11 95 9 S
AYL 0.3 5 93 6 S
CLL 0.1 2 90 4 S
Novozym
435
27.0 80 98 69 R
Conditions: (R)-enriched glycidol (19.8 mmol), vinyl butyrate (40 mmol), n-
heptane (5 ml), lipases (10 mg) were performed at 40 8C and 200 rpm for
80 min with a
W
of 0.24.
a
ee of (S)-glycidyl butyrate.
b
Calculated according to Eq. (3).
Table 3
Effect of solvent on the activity and enantioselectivity of Novozym 435 in transesterication
Solvent
1,4-Dioxane Acetonitrile n-Heptane n-Hexane Toluene solvent free
log P 1.1 0.33 4.6 3.5 2.5 -
Relative activity (%)* 46 50 100 89 78 63
E values 22 25 69 58 44 35
*The activity of Novozym 435 with free solvent was used as reference in this experiment (100%). Conditions: (R)-enriched glycidol (19.8 mmol), vinyl butyrate
(40 mmol), organic solvents (5 ml) and Novozym 435 (10 mg) were performed at 40 8C and 200 rpm for 80 min with a
W
of 0.24.
Fig. 4. Effect of water activity on the activity (*) and enantioselectivity (&) of
Novozym 435 in transesterication. The reaction ask contained (R)-enriched
glycidol (19.8 mmol), vinyl butyrate (40 mmol), n-heptane (5 ml) and Novo-
zym 435 (10 mg) were performed at 40 8C and 200 rpm for 80 min. The water
activity varied from 0.06 to 0.97.
D. Yu et al. / Process Biochemistry 42 (2007) 13191325 1323
that the E value remained almost the same with the variation of
a
W
values.
3.4.3. Effect of reaction time
The effect of reaction time on the transesterication
catalyzed by Novozym 435 was shown in Fig. 5. It could be
found that the enantiomeric excess of the (S)-glycidyl butyrate
decreased with the reaction time. In contrast, the conversion of
the (R)-enriched glycidol increased with the reaction time and
the increase slowed down after 80 min. The decrease of
enantiomeric excess depended on the changes of the relative
content of glycidol enantiomers with the evolution of the
reaction. With the consumption of (R)-glycidol in the reaction,
the relative concentration of (S)-glycidol became higher and
more competitive to react with vinyl butyrate, leading to a
decrease in the enantiomeric excess. Therefore, we chose
80 min as the optimum reaction time to achieve higher yield of
(S)-glycidyl butyrate and higher enantioselectivity. Under the
optimum conditions (a
W
= 0.24, n-heptane, 80 min), (S)-
glycidyl butyrate with 98% ee and 76% yield (15.1 mmol) in
the second step was obtained (41.9% yield based on the initial
(R,S)-glycidyl butyrate).
4. Conclusion
An efcient two-step enzymatic resolution process for
production of both enantiomers of glycidyl butyrate was
developed. The total yield of (R)- and (S)-glycidyl butyrate was
78%compared with the initial (R,S)-glycidyl butyrate added. In
the rst step, PPL was used to hydrolyze (R,S)-glycidyl butyrate
and the (R)-glycidyl butyrate was prepared with 98%
enantiomeric excess and 36% yield. Then, in the second step,
the produced (R)-enriched glycidol in hydrolysis was used as
substrate in transesterication and the (S)-glycidyl butyrate was
prepared with 98% enantiomeric excess and 41.9% yield based
on the initial (R,S)-glycidyl butyrate.
Acknowledgements
The authors are grateful for the nancial support from
National Natural Science Foundation of China (No. 20432010,
20672045) and the Foundation of Research Program of Jilin
University, China.
Reference
[1] Kloosterman M, Elferink VHM, Vaniersel J, Roskam JH, Meijer EM,
Hulshof LA, et al. Lipases in the preparation of b-blockers. Trends
Biotechnol 1988;6:2516.
[2] Kasai N, Suzuki T, Furukawa Y. Chiral C3 epoxides and halohydrins: their
preparation and synthetic application. J Mol Catal B: Enzym 1998;4:
23752.
[3] Palomo JM, Segura RL, Mateo C, Terreni M, Guisan JM, Fernandez-
Lafuente R. Synthesis of enantiomerically pure glycidol via a fully
enantioselective lipase-catalyzed resolution. Tetrahedron: Asymmetry
2005;16:86974.
[4] Brickner SJ, Hutchinson DK, Barbachyn MR, Manninen PR, Ulanowicz
DA, Garmon SA, et al. Synthesis and antibacterial activity of U-100592
and U-100766, two oxazolidinone antibacterial agents for the potential
treatment of multidrug-resistant Gram-positive bacterial infections. J Med
Chem 1996;39:6739.
[5] Weidner-Wells MA, Boggs CM, Foleno BD, Melton J, Bush K, Gold-
schmidt RM, et al. Novel piperidinyloxy oxazolidinone antibacterial
agents. diversication of the N-substituent. Bioorgan Med Chem
2002;10:234551.
[6] Matsuda M, Endo Y, Fushiya S, Endo T, Nozoe S. Cytotoxic 6-substituted
5,6-dihydro-2H-pyran-2-ones from a brazilian medicinal plant, chorisia-
crispiora. Heterocycles 1994;38:122932.
[7] Waechter AI, Ferreira ME, Fournet A, Rojas de Arias A, Nakayama H,
Torres S, et al. Experimental treatment of cutaneous leishmaniasis with
argentilactone isolated from Annona haematantha. Planta Med 1997;63:
4335.
[8] Lozano P, De Diego T, Carrie D, Vaultier M, Iborra JL. Synthesis of
glycidyl esters catalyzed by lipases in ionic liquids and supercritical
carbon dioxide. J Mol Catal A-Chem 2004;214:1139.
[9] Li C, Liu Q, Song X, Ding D, Ji A, Qu Y. Epoxide hydrolase-catalyzed
resolution of ethyl 3-phenylglycidate using whole cells of Pseudomonas
sp. Biotechnol Lett 2003;25:21136.
[10] Jia S, Xu J, Li Q, Yu J. Isolation of lipase producer and its performance in
enantioselective hydrolysis of glycidyl butyrate. Appl Biochem Biotech
2003;104:6979.
[11] Schmid RD, Verger R. Lipases: interfacial enzymes with attractive
applications. Angew Chem Int Ed 1998;37:160933.
[12] Brady L, Brzozowski AM, Derewenda ZS, Dodson E, Dodson G, Tolley S,
et al. A serine protease triad forms the catalytic centre of a triacylglycerol
lipase. Nature 1990;343:76770.
[13] Sarda L, Desnuelle P. Action de la lipase pancreatique sur les esters en
emulsion. Biochim Biophys Acta 1958;30:51321.
[14] Magnusson A, Hult K, Holmquist M. Creation of an enantioselectivity
hydrolase by engineering substrate-assisted catalysis. J Am Chem Soc
2001;123:43545.
[15] Derewenda U, Brzozowski AM, Lawson DM, Derewenda ZS. Catalysis at
the interface: the anatomy of a conformational change in a triglyceride
lipase. Biochemistry 1992;31:153241.
[16] Pregnolato M, Terreni M, de Fuentes IE, Leon ARA, Sabuquillo P,
Fernandez-Lafuente R, et al. Enantioselective enzymatic hydrolysis of
racemic glycidyl esters by using immobilized porcine pancreas lipase with
improved catalytic properties. J Mol Catal B: Enzym 2001;11:75763.
[17] Ladner WE, Whitesides GM. Lipase-catalyzed hydrolysis as a route to
esters of chiral epoxy alcohols. J Am Chem Soc 1984;106:72501.
[18] Crosby J. Chirality in Industry. Chichester: Wiley; 1992. p. 1.
[19] Palomo JA, Segura RL, Fuentes M, Ortiz CC, Guisan JM, Fernandez-
Lafuente R. Unusual enzymatic resolution of (+/)-glycidyl-butyrate for
Fig. 5. Time course of (R)-glycidol conversion (*) and enantiomeric excess of
the generated (S)-glycidyl butyrate (&) catalyzed by the Novozym 435. The
reaction ask contained (R)-enriched glycidol (19.8 mmol), vinyl butyrate
(40 mmol), n-heptane (5 ml) and Novozym 435 (10 mg) were performed at
40 8C and 200 rpm for 120 min with a
W
of 0.24.
D. Yu et al. / Process Biochemistry 42 (2007) 13191325 1324
the production of (S)-glycidyl derivatives. Enzyme Microb Technol
2006;38:42935.
[20] Wandel U, Machado SS, Jongejan JA, Duine JA. Enzymes involved in the
glycidaldehyde (2,3-epoxy-propanal) oxidation step in the kinetic resolu-
tion of racemic glycidol (2,3-epoxy-1-propanol) by Acetobacter pasteur-
ianus. Enzyme Microb Technol 2001;28:2339.
[21] Vanttinen E, Kanerva LT. Optimized double kinetic resolution for the
preparation of (S)-solketal. Tetrahedron: Asymmetry 1997;8:92333.
[22] Fishman A, Eroshov M, Dee-Noor SS, van Mil J, Cogan U, Effenberger R.
A two-step enzymatic resolution process for large-scale production of (S)-
and (R)-ethyl-3-hydroxybutyrate. Biotechnol Bioeng 2001;74:25663.
[23] Cong FD, Wang YH, Ma CY, Yu HF, Han SP, Tao J, et al. A way for
resolution of (R,S)-2-octanol by combining dynamic kinetic resolution
with double kinetic resolution. Enzyme Microb Technol 2005;36:
5959.
[24] Zada A, Harel M. Enzymatic transesterication of racemic lavandulol:
preparation of the two enantiomeric alcohols and of the two enantiomers
of lavandulyl senecioate. Tetrahedron: Asymmetry 2004;15:233943.
[25] Yu DH, Wang Z, Zheng LY, Zhang GR, Cao SG. Screening the reaction
system for enzymatic resolution of glycidyl butyrate. Chin J Bioproc Eng
2004;2:403.
[26] Ma L, Persson M, Adlercreutz P. Water activity dependence of lipase
catalysis in organic media explains successful transesterication reac-
tions. Enzyme Microb Technol 2002;31:10249.
[27] Halling PJ. Salt hydrates for water activity control with biocatalyst in
organic media. Biotechnol Tech 1992;6:2716.
[28] Chen CS, Fujimoto Y, Girdaukas G, Sih CJ. Quantitative analyses of
biochemical kinetic resolutions of enantiomers. J Am Chem Soc
1982;104:72949.
[29] Segura RL, Betancor L, Palomo JM, Hidalgo A, Fernandez-Lorente G,
Terreni M, et al. Purication and identication of different lipases con-
tained in PPL commercial extracts: a minor contaminant is the main
responsible of most esterasic activity. Enzyme Microb Technol 2006;39:
81723.
[30] Veeraragavan K. A simple and sensitive method for the estimation of
microbial lipase activity. Anal Biochem 1990;186:3015.
[31] Liu YY, Xu JH, Hu Y. Enhancing effect of Tween-80 on lipase perfor-
mance in enantioselective hydrolysis of Ketoprofen ester. J Mol Catal B:
Enzym 2000;10:5239.
[32] Kamiya N, Kasagi H, Inoue M, Kusunoki K, Goto M. Enantioselective
recognition mechanism of secondary alcohol by surfactant-coated lipases
in nonaqueous media. Biotechnol Bioeng 1999;65:22732.
[33] Klibanov AM. Enzymes that work in organic solvents. Chemtech
1986;16:3549.
[34] Okazaki S, Kamiya N, Abe K, Goto M, Nakashio F. Novel preparation
method for surfactant-lipase complexes utilizing water in oil emulsions.
Biotechnol Bioeng 1997;55:45560.
[35] Fernandez-Lorente G, Palomo JM, Fuentes M, Mateo C, Guisan JM,
Fernandez-Lafuente R. Self-assembly of Pseudomonas uorescens lipase
into bimolecular aggregates dramatically affects functional properties.
Biotechnol Bioeng 2003;82:2327.
[36] Fernandez-Lorente G, Palomo JM, Mateo C, Munilla R, Ortiz C, Cabrera
Z, et al. Glutaraldehyde cross-linking of lipases adsorbed on aminated
supports in the presence of detergents leads to improved performance.
Biomacromolecules 2006;7:26105.
[37] Mingarro I, Abad C, Braco L. Interfacial activation-based molecular
bioimprinting of lipolytic enzymes. Proc Natl Acad Sci USA 1995;92:
330812.
[38] Laane C, Boeren S, Vos K, Veeger C. Rules for optimization of bioca-
talysis in organic solvents. Biotechnol Bioeng 1987;30:817.
[39] Giacometti J, Giacometti F. Study of organic solvent hydrophobicity on
lipase catalyzed reaction-esterication. Biotechnol Bioeng 2006;20:
26974.
[40] Janssen AEM, Sjursnes BJ, Vakurov AV, Halling PJ. Kinetics of lipase-
catalyzed esterication in organic media: correct model and solvent
effects on parameters. Enzyme Microb Technol 1999;24:46370.
[41] Halling PJ. Thermodynamic predictions for biocatalysis in nonconven-
tional media: theory, tests, and recommendations for experimental design
and analysis. Enzyme Microb Technol 1994;16:178206.
[42] Halling PJ. Enzymatic conversions in organic and other low-water media.
In: Drauz K, Waldmann H, editors. Handbook of Enzyme Catalysis in
Organic Synthesis.. Weinheim, Germany: WileyVCH Verlag GmbH;
2002. p. 25986.
[43] Wehjte E, Costes D, Adlercreutz P. Enantioselectivity of lipases: effects of
water activity. J Mol Catal B: Enzym 1997;3:22130.
[44] Persson M, Costes D, Wehtje E, Adlercreutz P. Effects of solvent, water
activity and temperature on lipase and hydroxynitrile lyase enantioselec-
tivity. Enzyme Microb Technol 2002;30:91623.
D. Yu et al. / Process Biochemistry 42 (2007) 13191325 1325

Potrebbero piacerti anche