Sei sulla pagina 1di 7

JBA-06566; No of Pages 7

Biotechnology Advances xxx (2012) xxx–xxx

Contents lists available at SciVerse ScienceDirect

Biotechnology Advances
journal homepage: www.elsevier.com/locate/biotechadv

Research review paper

Production of shikimic acid


Saptarshi Ghosh a, Yusuf Chisti b, Uttam C. Banerjee a,⁎
a
Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar-160 062, Punjab, India
b
School of Engineering, Massey University, Private Bag 11 222, Palmerston North, New Zealand

a r t i c l e i n f o a b s t r a c t

Article history: Shikimic acid is a key intermediate for the synthesis of the antiviral drug oseltamivir (Tamiflu®). Shikimic
Received 23 November 2011 acid can be produced via chemical synthesis, microbial fermentation and extraction from certain plants. An
Received in revised form 1 March 2012 alternative production route is via biotransformation of the more readily available quinic acid. Much of the
Accepted 5 March 2012
current supply of shikimic acid is sourced from the seeds of Chinese star anise (Illicium verum). Supply
Available online xxxx
from star anise seeds has experienced difficulties and is susceptible to vagaries of weather. Star anise tree
Keywords:
takes around six-years from planting to bear fruit, but remains productive for long. Extraction and purifica-
Shikimic acid tion from seeds are expensive. Production via fermentation is increasing. Other production methods are
Oseltamivir too expensive, or insufficiently developed. In the future, production in recombinant microorganisms via fer-
Quinic acid mentation may become established as the preferred route. Methods for producing shikimic acid are
reviewed.
© 2012 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. Production methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.1. Extraction from plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2. Fermentation processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2.1. The shikimic acid pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2.2. Use of recombinant and engineered strains . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3. Chemical synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4. Microbial biotransformation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction from which it was first isolated. Shikimic acid is an intermediate of


the shikimic acid pathway (Herrmann and Weaver, 1999) that is in-
This review is focussed on the methods of producing shikimic acid volved in the synthesis of aromatic metabolites in plants and micro-
(3,4,5-trihydroxy-1-cyclohexene-1-carboxylic acid), a chemical organisms (Ganem, 1978; Herrmann, 1995; Pittard, 1996; Wilson et
building block for the antiviral drug oseltamivir (Tamiflu®). Shikimic al., 1998). Metabolically essential products of the shikimic acid path-
acid is named after the Japanese shikimi (Illicium anisatum) flower way include the three aromatic amino acids L-phenylalanine,
L-tryptophan and L-tyrosine. Shikimic acid pathway is not normally
associated with animals, but genes for coding some of the enzymes
Abbreviations: DAHP, 3-Deoxy-D-arabino-heptulosonate-7-phosphate; DHQ,
3-Dehydroquinate or 3-dehydroquinic acid; DHS, 3-Dehydroshikimate or 3- of the pathway have been found in certain animals (Starcevic et al.,
dehyroshikimic acid; DQD, 3-Dehydroquinate dehydrogenase; EPSP, 5-Enolpyruvylshiki- 2008). Properties and toxicology of shikimic acid have been discussed
mate-3-phosphate; E4P, Erythrose-4-phosphate; GDH, Glucose dehydrogenase; NADP, elsewhere (Stavric and Stoltz, 1976). An insufficiency of shikimic acid
Nicotinamide adenine dinucleotide phosphate; NADPH, Reduced form of nicotinamide ad- has in the past affected the supply of oseltamivir (Farina and Brown,
enine dinucleotide phosphate; PEP, Phosphoenolpyruvate; QDH, Quinic acid dehydroge-
nase; SKDH, Shikimic acid dehydrogenase.
2006).
⁎ Corresponding author. Tel.: + 91 172 2214682 87; fax: + 91 172 2214692. As a highly functionalised, six-carbon ring with three chiral car-
E-mail address: ucbanerjee@niper.ac.in (U.C. Banerjee). bons and a carboxylic acid functional group, shikimic acid (Fig. 1) is

0734-9750/$ – see front matter © 2012 Elsevier Inc. All rights reserved.
doi:10.1016/j.biotechadv.2012.03.001

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001
2 S. Ghosh et al. / Biotechnology Advances xxx (2012) xxx–xxx

extraction and purification processes are expensive. Use of recombi-


nant bacteria for commercial production of shikimic acid is develop-
ing. Newer routes for producing oseltamivir without the use of
shikimic or quinic acids have appeared (Fukuta et al., 2006; Yeung
et al., 2006), but not been commercialized. Here we review the existing
and emerging methods for producing shikimic acid.

2. Production methods

2.1. Extraction from plants


Fig. 1. Structure of shikimic acid and oseltamivir.
As an intermediate of the shikimic acid pathway, shikimic acid
a versatile enantiomerically pure precursor for making potentially is found widely in plants (Bohm, 1965; Dell and Frost, 1993;
useful products. Interest in shikimic acid has been rekindled as it is Gurib-Fakim, 2006; Herrmann and Weaver, 1999), but generally oc-
required for producing the avian flu drug oseltamivir (Fig. 1). In curs in low concentration. Seeds of Chinese star anise (Illicium verum)
view of the potential impact of a flu pandemic (Horimoto and are the main commercial source of shikimic acid. The botany and phar-
Kawaoka, 2001) and the limited usefulness of vaccines against rapidly macology of Chinese star anise have been reviewed (Wang et al., 2011).
evolving flu viruses, stockpiles of effective drugs are necessary for Shikimic acid was first isolated from flowers of the highly toxic
managing a major outbreak. Oseltamivir is an effective treatment Japanese star anise. Subsequently, it was reported in the leaves of
for influenza (Widmer et al., 2010), especially if administered early. the sweetgum tree (Liquidambar styraciflua) (Plouvier, 1961). The
Oseltamivir is effective against both Type A and Type B influenza seeds of the American sweetgum tree have been found to contain
and it is used also in prophylaxis. up to 3.7% (w/w) shikimic acid (Enrich et al., 2008). Extraction of shi-
Oseltamivir is a viral neuraminidase inhibitor. Its synthesis from kimic acid from bark and wood of the sweetgum has been reported
quinic acid via shikimic acid has been described (Kim et al., 1997, (Martin et al., 2010). A simple extraction method based on hot
1998; Rohloff et al., 1998). Oseltamivir produced for early clinical trials water was used. Several varieties of pine, fir and spruce are known
had been made from chemically synthesized shikimic acid (Federspiel to produce shikimic acid. A hot water (45–75 °C) extraction of the
et al., 2001), but this method proved impractical for commercial pro- needles of Scots pine (Pinus sylvestris) yielded around 1.6% (w/w)
duction of the drug. Chemical and microbial methods for inexpensive shikimic acid (Sui, 2008). Extraction from the needles of the pine
production of shikimic acid are being developed, but most of this Pinus elliottii has been described (Xie et al., 2010). Attempts are
compound is currently extracted from the seeds of Chinese star being made to identify other natural plant sources of shikimic acid
anise (Illicium verum) (Payne and Edmonds, 2005). The multistep (Raghavendra et al., 2009).

Fig. 2. The shikimic acid or shikimate pathway.

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001
S. Ghosh et al. / Biotechnology Advances xxx (2012) xxx–xxx 3

Plants of the genus Illicium appear to the best source of shikimic Table 2
acid, but many Illicium species are poisonous (Lederer et al., 2006; Modified E. coli strains with 3-dehydroshikimic acid yield and titer (Li et al., 1999).

Yamada et al., 1968) and therefore not suitable for producing com- Number Strain Modification DHS yield DHS titer
mercial shikimic acid. Japanese star anise (I. anisatum) is highly (mol/mol) (g/L)
toxic, but the Chinese star anise (I. verum) is edible. Nevertheless, 1 KL3/ aroFFBR expression controlled by 0.17 20.3
compounds found in Chinese star anise have been shown to be pKL4.33B ParoF in absence of amplified tktA
toxic to infants (Ize-Ludlow et al., 2004). 2 KL3/ aroFFBR expression controlled by 0.16 38.5
pKL4.66A ParoF in absence of amplified tktA
Shikimic acid is highly soluble in water (180 g/L at 20 °C), but not
3 KL3/ aroFFBR expression controlled by 0.30 69.0
in non-polar solvents. Therefore, a hot water extraction of the plant pKL4.130B ParoF in the presence of amplified
tissue is used as the primary extraction step (Ohira et al., 2009; Sui, tktA
2008; Xie et al., 2010; Ye et al., 2007). Shikimic acid content in the 4 KL3/ aroFFBR expression is controlled 0.18 41.2
plant tissue varies depending on the source of the tissue, the time of pKD11.291A by ParoF in absence of amplified
tktA
harvest and other possible factors. Roughly 1 kg of shikimic acid can
5 KL3/ aroFFBR expression is controlled 0.24 58.1
be recovered from 30 kg of dry seed pods of Chinese star anise pKL5.17A by ParoF in presence of amplified
(Ohira et al., 2009; Roche, 2006). Recovery is nearly complete (>95%) tktA
within 10 min of water extraction at 70 °C from seed pods ground to a 6 KL3/ aroFFBR expression under the 0.28 66.0
pKL4.124A control of Ptac in presence of tktA
particle size of 355–600 μm (Ohira et al., 2009). The rate of recovery
can be further enhanced by using a higher extraction temperature.
Other extraction methods have been described, involving the use
of acids (Harring et al., 1998; Mueller, 2003), alcohols (Anderson et pathway is used to generate E4P (Fig. 2). Thus, the shikimic acid path-
al., 2001; Jaroszynska, 2003), complex formation (Miles et al., 1994; way is dependent on the glycolytic pathway and the pentose phos-
Sadaka and Garcia, 1999) and microwave-assisted extraction phate pathway to provide the two starting materials it requires.
(Matallo et al., 2009). The crude water extract contains numerous Therefore, metabolic engineering of the shikimic acid pathway alone
other water-soluble plant metabolites and requires extensive further may be insufficient for increasing the yield of shikimic acid from the
processing to yield pure shikimic acid. The recovery and purification carbon source used in a fermentation. Furthermore, because shikimic
processes used in commercial production from Chinese star anise acid is produced far downstream in the metabolic pathway relative to
are proprietary and involve multiple steps. the point where glucose first enters the metabolism, channelling the
flow of carbon to production of shikimic acid can be difficult.
2.2. Fermentation processes Once E4P and PEP have been generated through carbohydrate me-
tabolism in other independent pathways, they are condensed in the
Microbial production of shikimic acid involves the shikimic acid (or shikimic acid pathway through the action of the enzyme DAHP
shikimate) pathway. Shikimic acid pathway occurs commonly in micro- synthase to produce DAHP. Action of three other enzymes then con-
organisms and therefore they can be used to overproduce this com- verts DAHP to shikimic acid. Chorismic acid is the final product of
pound from carbon sources such as glucose (Draths et al., 1999; the SA pathway and this compound is the common precursor for
Simonart and Wiaux, 1960). Shikimic acid is a precursor for essential the biosynthesis of other aromatic products such as aromatic amino
amino acids and other metabolites in microorganisms. Its overproduc- acids, as it is shown in Fig. 2.
tion can be achieved for example by metabolic engineering to partly The shikimic acid pathway engineering has most commonly in-
block some of the biochemical pathways that consume shikimic acid volved the bacterium Escherichia coli. This microorganism has three
and by overexpression of the enzymes responsible for its synthesis. Pro- isoforms of the DAHP synthase enzyme encoded by aroF, aroG, aroH
duction by fermentation using engineered microorganisms is already show feedback inhibition by the three aromatic amino acids, i.e.
supplementing the commercial supply of shikimic acid. L-tyrosine, L-phenylalanine and L-tryptophan respectively. The
DAHP synthase (aroG) of Bacillus subtilis is inhibited by the pathway
2.2.1. The shikimic acid pathway intermediate chorismate (Jensen and Nester, 1966a,b) (Fig. 2).
The shikimic acid pathway is shown in Fig. 2. This metabolic pathway The enzyme DHQ synthase (aroB) in E. coli converts DAHP into
is used to produce the aromatic amino acids that are essential to growth. 3-dehydroquinate (DHQ). The enzyme DHQ dehydratase (aroD) then
Shikimic acid is an intermediate in the pathway. This pathway has been converts DHQ into 3-dehydroshikimate (DHS) by eliminating water.
previously reviewed (Herrmann and Weaver, 1999). Subsequently, NADPH-dependent shikimate dehydrogenase (aroE) re-
The shikimic acid pathway begins with phosphoenolpyruvate duces DHS to shikimic acid. The rate limiting enzymes of the shikimic
(PEP) and erythrose-4-phosphate (E4P) (Fig. 2). These two com- acid pathway of E. coli are DHQ synthase and shikimate kinase (Dell
pounds must first be generated from metabolism of glucose, or and Frost, 1993) (Fig. 2). Shikimate kinase (aroL and aroK) is responsible
other carbohydrate. PEP is produced through the glycolysis pathway for converting shikimic acid to shikimate-3-phosphate (Fig. 2).
(Fig. 2) whereas the completely independent pentose phosphate In E. coli, the enzymes DHQ synthase, DHQ dehydratase and
shikimate dehydrogenase (Fig. 2) are constitutively expressed
whereas the production of the DAHP synthases and one of the
Table 1
Modified E.coli strains with shikimic acid (SA) yield and titer.
Table 3
Strain Modification SA yield SA Effect of carbon sources on the production of 3-dehydroshikimic acid by recombinant
(mol/mol) titer (g/L) E. coli strains (Li et al., 1999).
SP1.1/pKD15.071B Over expression of ppsA 0.23 66
Strain KL3/pKL4.124A KL3/pKL4.79B (tktA−)
(Chandran et al., 2003)
(tktA over expression)
SP1.1pts/pSC6.090B PTS−/glf+/tktA over 0.27 71
(Chandran et al., 2003) expression Carbon source DHS titer DHS yield Carbon DHS titer DHS yield
SP1.1/pKD12.138 Over expression of tktA 0.18 52 (g/L) (mol/mol) source (g/L) (mol/mol)
(Knop et al., 2001) Glucose 46.0 0.28 Glucose 36.4 0.22
SP1.1/pKD12.112 Insertion of aroB into the 0.15 27.2 Xylose 43.1 0.33 Xylose 41.7 0.32
(Draths et al., 1999) serA locus of E.coli and Mixa 64.0 0.41 Mixa 53.0 0.36
disruption of aroL and aroK a
Molar ratio of glucose/xylose/arabinose: 3:3:2.

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001
4 S. Ghosh et al. / Biotechnology Advances xxx (2012) xxx–xxx

Fig. 3. Synthesis of shikimic acid via Diels-Alder reaction (McCrindle et al., 1960; Smissman et al., 1959). Based on Ambhaikar (2005).

shikimate kinases is transcriptionally regulated. Shikimic acid in- an increased availability of PEP has enhanced production of shikimic
hibits shikimate dehydrogenase (Dell and Frost, 1993). acid (Chandran et al., 2003; Yi et al., 2002). The modification of the
An alternative microbial route to shikimic acid is through bio- glycolytic pathway involved over expression of PEP synthase (ppsA)
transformation of quinic acid. Certain microorganisms (Pseudomonas, leading to increased shikimic acid titer to 66 g/L and yield on glucose
Achromobacter, Aspergillus and Neurospora crassa) can use quinic acid of 0.23 mol/mol (Chandran et al., 2003). Inactivation of the PTS operon
(or its salt, quinate) as the sole carbon source (Case et al., 1978; Da (PTS−), expression of non-PTS glucose transporters like glucose facilita-
Silva et al., 1986; Rogoff, 1958) to produce aromatic amino acids via tors (glf), glucokinase (glk) in combination with over expression of tktA
the shikimic acid pathway. Quinate enters the pathway at the point gene was reported to increase the shikimic acid titer to 71 g/L
shown in Fig. 2. E. coli strains specifically engineered for overprodu- (Chandran et al., 2003; Gibson et al., 2001). Using the engineered E.
cing quinic acid from glucose have also been developed (Ran et al., coli strain, the shikimic acid concentration could be further raised to
2001). 84 g/L by supplementing the minimal medium with yeast extract
(Chandran et al., 2003). Shikimic acid could be produced during expo-
2.2.2. Use of recombinant and engineered strains nential growth on glucose to a final concentration of 87 g/L (Chandran
Metabolically engineered bacteria provide an important emerging et al., 2003). The nature of the carbon source affected the productivity
route to production of shikimic acid via fermentation (Campbell et al., of shikimic acid (Ahn et al., 2008; Li et al., 1999).
1993; Krämer et al., 2003). The bacterium E. coli has been the focus of In earlier studies, the shikimic acid titers from metabolically engi-
most metabolic engineering effort (Ahn et al., 2008; Escalante et al., neered E. coli strains were low partly due to simultaneous production
2010; Johansson and Liden, 2006; Johansson et al., 2005, 2006; of quinic acid (Knop et al., 2001). Production of quinic acid during bio-
Knop et al., 2001; Yi et al., 2002, 2003), but studies in other bacteria synthesis of shikimic acid is a consequence of the microbe-catalyzed
have also been reported. equilibration of the initially synthesized shikimic acid (Knop et al.,
Several metabolic engineering approaches have been developed to 2001). This problem appears to have been resolved in view of the afore-
overproduce shikimic acid in E. coli (Ahn et al., 2008; Chandran et al., mentioned high titers.
2003; Escalante et al., 2010; Gibson et al., 2001; Johansson et al., Shikimic acid can be produced readily from DHS (Fig. 2). There-
2005; Knop et al., 2001; Krämer et al., 2003). All these are based on fore, some metabolic engineering studies have focussed on overpro-
genetic modifications to alter the central carbon metabolism and duction of DHS. Substantial overproduction of DHS in metabolically
the shikimic acid pathway (Table 1). engineered microorganisms has been achieved (Draths and Frost,
The shikimic acid pathway requires PEP and E4P (Fig. 2). The sup- 1990; Li and Frost, 1999; Li et al., 1999; Yi et al., 2002, 2003)
ply of PEP and E4P can be enhanced via metabolic engineering of the (Table 2). For example, E. coli constructs have produced as much as
glycolytic pathway and the pentose phosphate pathway, respectively. 60 g/L DHS from glucose in 60 h (Yi et al., 2003). A comparative analysis
Both these approaches have been demonstrated. Over expression of of D-xylose, D-glucose and D-arabinose as carbon source for microbial
transketolase (tktA) resulted in the increase of shikimic acid yield synthesis of DHS was also reported (Li and Frost, 1999) (Table 3). Differ-
from 0.12 to 0.18 mol/mol and titer from 38 to 52 g/L by enhancing ent recombinant E. coli strains were used with various carbon sources
the concentration of E4P (Knop et al., 2001). In recombinant E. coli, for DHS production.

Fig. 4. Shikimic acid synthesis of Koreeda and Ciufolini (1982) as summarized by Ambhaikar (2005).

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001
S. Ghosh et al. / Biotechnology Advances xxx (2012) xxx–xxx 5

Fig. 5. Chemical conversion of (−)-Quinic acid to (−)-Shikimic acid (Dangschat and Fischer, 1938, 1950). Based on Ambhaikar (2005).

The activity of DAHP synthase (Fig. 2) controls the amount of cel- have been described (Kancharla et al., 2009; Sánchez-Abella et al.,
lular carbon directed into DHS synthesis. Transcriptional repression 2006). Chemical synthesis of shikimic acid and its analogs is further
and feedback inhibition of DAHP synthase by aromatic amino acids reviewed elsewhere (Campbell et al., 1993; Jiang and Singh, 1998).
are believed to control the activity of this enzyme. The amplified Although several routes have been described for chemical synthesis,
expression of a mutant DAHP synthase, which is insensitive to production of shikimic acid by these methods is too expensive to be
feedback inhibition by aromatic amino acids, has been used to en- of commercial use. Consequently, isolation from Chinese star anise
hance production of DHS. Metabolic engineering approaches have fruit remains the principal source of commercial shikimic acid
been employed to produce shikimic acid in E. coli strains derived (Raghavendra et al., 2009).
from an evolved strain PB 12 lacking the PTS system but with ability The ten step commercial route of oseltamivir (Tamiflu®) synthesis
to grow on glucose. The double aroK -, aroL- mutant of strain PB uses (−)-shikimic acid as the starting material (Abrecht et al., 2004).
12.SA22 showed shikimic acid titer of 7 g/L and yield of 0.29 mol/mol This precursor is converted into a diethyl ketal intermediate, which is
(Escalante et al., 2010). reductively opened to give 1,2-epoxide. This epoxide is then con-
In addition to recombinant E. coli, genetically modified or other- verted into oseltamivir via a five step reaction involving 3 potentially
wise mutated Bacillus subtilis (Iomantas et al., 2002) and Citrobacter toxic and explosive azide intermediates (Fig. 6). This route gives 35%
freundii bacteria (Shirai et al., 2001) have been used to successfully yield of oseltamivir. Future methods for making oseltamivir may
overproduce shikimic acid although the titers have not exceeded completely circumvent the need for shikimic or quinic acids (Fukuta
about 20 g/L. It was reported that aroI (shikimate kinase) deficient et al., 2006; Yeung et al., 2006).
strain of B. subtilis showed shikimic acid titer of 8.5 g/L with high
titer of DHS (9.5 g/L) as the by-product (Iomantas et al., 2002). Use
of glyphosate, an inhibitor of the enzyme EPSP synthase (Fig. 2), in 2.4. Microbial biotransformation
the fermentation medium to enhance accumulation of shikimic acid
by blocking its downstream consumption, has been described Many microorganisms are able to convert quinic acid to shikimic
(Bogosian, 2011). Metabolic engineering of microorganisms for pro- acid (Adachi et al., 2003, 2006). Quinic acid (or quinate) is converted
ducing shikimic acid has been reviewed by Krämer et al. (2003). to 3-dehydroquinate (DHQ) (Fig. 2) through the action of quinate de-
hydrogenase. DHQ is an intermediate of the shikimic acid pathway
2.3. Chemical synthesis and is transformed to shikimic acid in a two-step process (Fig. 2).
During exponential growth of Gluconobacter oxydans, nearly all qui-
Chemical synthesis of shikimic acid was first achieved during the nate supplied in the medium could be converted to DHQ (Adachi et al.,
1960s using the basic chemistry of Diels-Alder reaction (Fig. 3) to 2003). This transformation could be achieved also with dried cells of G.
form six membered rings (McCrindle et al., 1960; Smissman et al., oxydans (Adachi et al., 2003) and with fresh cells immobilized in algi-
1959), but the yield was low at ≤15%. Later attempts to increase nate beads. The intermediate DHQ could be further transformed to
yield were not particularly successful (Grewe and Hinrichs, 1964). DHS in a reasonable yield by both growing and immobilized cells.
In 1982, the efficiency of Diels-Alder synthesis could be improved Lactobacillus pastorianus is capable of converting quinic acid to a
(Fig. 4) to raise the yield to 29% (Koreeda and Ciufolini, 1982). A product that was claimed to be dihydroshikimic acid (Carr et al.,
more efficient synthesis further raised the yield to 55% (Koreeda et 1957), but was more likely DHS.
al., 1990). A two-step scheme for biotransformation of quinic acid to shikimic
Synthesis of shikimic acid from benzene has been shown to be acid has been published (Fig. 7) (Adachi et al., 2006). The first step of
possible (Birch et al., 1988). Synthesis via a palladium mediated elim- this scheme involves an oxidative fermentation with G. oxydans to es-
ination reaction was advanced by Yoshida and Ogasawara (2000). sentially quantitatively convert quinic acid in the culture medium to
Synthesis of (−)-shikimic acid has been reported from sugars such DHS. The latter is subsequently converted to shikimic acid in an
as D-mannose (Dangschat and Fischer, 1938, 1950; Fleet et al., NADPH-dependent reaction using the enzyme shikimic acid dehy-
1984; Jiang et al., 1994). Shikimic acid can be made also from (−)- drogenase (SKDH) (Fig. 7) purified from cells of G. oxydans. The en-
quinic acid and its derivatives by chemical transformations (Box et zymatic biotransformation medium also contained glucose and the
al., 2002; Cleophax et al., 1971, 1973; Federspiel et al., 2001; Kim et enzyme glucose dehydrogenase to continuously regenerate NADPH
al., 1997, 1998; Rohloff et al., 1998) (Fig. 5). Other synthetic methods (Fig. 7).

Fig. 6. Synthesis of oseltamivir from shikimic acid.

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001
6 S. Ghosh et al. / Biotechnology Advances xxx (2012) xxx–xxx

Fig. 7. Overall reaction for shikimic acid production (based on Adachi et al., 2006).

3. Concluding remarks Case ME, Pueyo C, Barea JL, Giles NH. Genetical and biochemical characterization of QA-
3 mutants and revertants in the QA gene cluster of Neurospora crassa. Genetics
1978;90:69–84.
Shikimic acid is a precursor for the synthesis of the important anti- Chandran SS, Yi J, Draths KM, Von Daeniken R, Weber W, Frost JW. Phosphoenolpyr-
viral drug oseltamivir. Extraction from Chinese star anise is the main uvate availability and the biosynthesis of shikimic acid. Biotechnol Prog 2003;19:
808–14.
source for shikimic acid, but fermentation processes based on the re- Cleophax J, Mercier D, Géro SD. A stereospecific conversion of (−)-methyl tri-O-
combinant bacterium E. coli have been shown to be a viable alternative benzoylquinate to the corresponding (−)-methyl shikimate. Angew Chem Int Ed
source. Chemical synthesis of shikimic acid is possible, but apparently 1971;10:652–3.
Cleophax J, Leboul J, Mercier D, Gaudemer A, Gero SD. New and easy synthetic route to
not commercially viable. Chemical methods of making oseltamivir shikimic and 4-epishikimic acid. Bull Soc Chim Fr Partie II 1973:2992–5.
may entirely circumvent the need for shikimic acids, but are not com- Da Silva AJ, Whittington H, Clements J, Roberts C, Hawkins AR. Sequence analysis and
mercially used. Bioconversion of quinic acid to shikimic acid is another transformation by the catabolic 3-dehydroquinase (QUTE) gene from Aspergillus
nidulans. Biochem J 1986;240:481–8.
option for production, but appears not to have been developed to the
Dangschat G, Fischer HOL. übergang der Chinasäure in shikimisäure. Naturwissenschaften
extent of the fermentation route from glucose. 1938;26:562–3.
Dangschat G, Fischer HOL. Configurational relationships between naturally occurring
cyclic plant acids and glucose transformation of quinic acid into shikimic acid. Bio-
References chem Biophys Acta 1950;4:199–204.
Dell KA, Frost JW. Identification and removal of impediments to biocatalytic synthesis
Abrecht S, Harrington P, Iding H, Karpf M, Trussardi R, Wirz B, et al. The synthetic de- of aromatics from D-glucose: rate limiting enzymes in the common pathway of
velopment of the anti-influenza neuraminidase inhibitor oseltamivir phosphate aromatic amino acid biosynthesis. J Am Chem Soc 1993;115:11581–9.
(Tamiflu®): a challenge for synthesis and process research. Chimia 2004;58: Draths KM, Frost JW. Genomic direction of synthesis during plasmid-based biocatalysis. J
621–9. Am Chem Soc 1990;112:9630–2.
Adachi O, Tanasupawat S, Yoshihara N, Toyama H, Matsushita M. 3-Dehydroquinate pro- Draths KM, Knop DR, Frost JW. Shikimic acid and quinic acid: replacing isolation from
duction by oxidative fermentation and further conversion of 3-dehydroquinate to the plant sources with recombinant microbial biocatalysis. J Am Chem Soc 1999;121:
intermediates in the shikimate pathway. Biosci Biotechnol Biochem 2003;67: 1603–4.
2124–31. Enrich LB, Scheuermann ML, Mohadjer A, Matthias KR, Eller CF, Newman MS, et al. Liq-
Adachi O, Ano Y, Toyama H, Matsushita K. High shikimate production from quinate uidambar styraciflua: a renewable source of shikimic acid. Tetrahedron Lett
with two enzymatic systems of acetic acid bacteria. Biosci Biotechnol Biochem 2008;49:2503–5.
2006;70:2579–82. Escalante A, Calderón R, Valdivia A, de Anda R, Hernández G, Ramírez OT, et al. Meta-
Ahn JO, Lee HW, Saha R, Park MS, Jung JK, Lee DY. Exploring the effects of carbon bolic engineering for the production of shikimic acid in an evolved Escherichia
sources on the metabolic capacity for shikimic acid production in Escherichia coli coli strain lacking the phosphoenolpyruvate: carbohydrate phosphotransferase
using in silico metabolic predictions. J Microbiol Biotechnol 2008;18:1773–84. system. Microb Cell Fact 2010;9:21.
Ambhaikar N. Shikimic acid. A seminar presentation dated 12 January 2005. La Jolla, Farina V, Brown JD. Tamiflu: the supply problem. Angew Chem Int Ed 2006;45:7330–4.
California, USA: The Scripps Research Institute; 2005http://www.scripps.edu/ Federspiel M, Fischer R, Hennig M, Mair HJ, Oberhauser T, Rimmler G, et al. Industrial syn-
chem/baran/images/grpmtgpdf/Ambhaikar_Jan_05.pdf. thesis of the key precursor in the synthesis of the anti-influenza drug oseltamivir
Anderson KA, Cobb WT, Loper BR. Analytical method for determination of shikimic phosphate (Ro 64-0796/002, GS-4104-02): Ethyl (3R, 4S, 5S)-4, 5-epoxy-3-(1-
acid: Shikimic acid proportional to glyphosate application rates. Commun Soil Sci ethyl-propoxy)-cyclohex-1-ene-1-carboxylate. Org Process Res Dev 2001;3:266–74.
Plant Anal 2001;32:2831–40. Fleet GWJ, Shing TKM, Warr SM. Enantiospecific synthesis of shikimic acid from D-
Birch AJ, Kelly LF, Weerasuria DV. Facile synthesis of (+)- and (−)-shikimic acid with mannose: formation of a chiral cyclohexene by intramolecular olefination of a
asymmetric deuterium labelling, using tricarbonyliron as a lateral control group. J carbohydrate-derived intermediate. J Chem Soc Perkin Trans 1 1984:905–8.
Org Chem 1988;53:278–81. Fukuta Y, Mita T, Fukuda N, Kanai M, Shibasaki M. De novo synthesis of Tamiflu via a
Bogosian, G., Frantz, J.P. Use of glyphosate to produce shikimic acid in microorganisms. catalytic asymmetric ring-opening of meso-aziridines with TMSN3. J Am Chem
United States Patent Application Pub. No.: US 2011/0020885 Al; 2011. Soc 2006;128:6312–3.
Bohm BA. Shikimic acid (3,4,5-trihydroxy-1-cyclohexene-1-carboxylic acid). Chem Rev Ganem B. Shikimate-derived metabolites. 4. From glucose to aromatics — recent develop-
1965;65:435–66. ments in natural-products of shikimic acid pathway. Tetrahedron 1978;34:3353–83.
Box JM, Harwood LM, Humphreys JL, Morris GA, Redon PM, Whitehead RC. Dehydration Gibson JM, Thomas PS, Thomas JD, Barker JL, Chandran SS, Harrup MK, et al. Benzene-
of quinate derivatives: synthesis of a difluoromethylene homologue of shikimic acid. free synthesis of phenol. Angew Chem Int Ed 2001;40:1945–8.
Synlett 2002;2:358–60. Grewe R, Hinrichs I. Eine neue synthese der shikimisaure. Chem Ber 1964;97:443.
Campbell MM, Sainsbury M, Searle PA. The biosynthesis and synthesis of shikimic acid, Gurib-Fakim A. Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol
chorismic acid, and related-compounds. Synth Stuttg 1993;2:179–93. Aspects Med 2006;27:1-93.
Carr JG, Pollard A, Whiting GC, Williams AH. The reduction of quinic acid to dihydroshi- Harring T, Streibig JC, Husted S. Accumulation of shikimic acid: a technique for screening
kimic acid by certain lactic acid bacteria. Biochem J 1957;66:283–5. glyphosate efficacy. J Agric Food Chem 1998;46:4406–12.

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001
S. Ghosh et al. / Biotechnology Advances xxx (2012) xxx–xxx 7

Herrmann KM. The shikimate pathway: early steps in the biosynthesis of aromatic Ohira H, Torii N, Aida TM, Watanabe M, Smith Jr RL. Rapid separation of shikimic acid
compounds. Plant Cell 1995;7:907–19. from Chinese star anise (Illicium verum Hook. f.) with hot water extraction. Sep
Herrmann KM, Weaver LM. The shikimate pathway. Annu Rev Plant Physiol Plant Mol Purif Technol 2009;69:102–8.
Biol 1999;50:473–503. Payne R, Edmonds M. Isolation of shikimic acid from star anise seeds. J Chem Educ
Horimoto T, Kawaoka Y. Pandemic threat posed by avian influenza A viruses. Clin 2005;82:599–600.
Microbiol Rev 2001;14:129–49. Pittard AJ. Biosynthesis of aromatic amino acids. In: Neidhardt FC, Curtiss III R,
Iomantas, Y., Abalakina, E.G., Polanuer, B., Yampolskaya, T.A., Bachina, T.A., Kozlov, J.I. Ingraham JL, Lin ECC, Low KB, Magasanic B, Reznikoff WS, Riley M, Schaechter M,
Method for producing shikimic acid. US Patent 6436664; 2002. Umbarger HE, editors. Escherichia coli and Salmonella. Cellular and Molecular Biolo-
Ize-Ludlow D, Ragone S, Bernstein JN, Bruck IS, Duchowny M, Pena BMG. Chemical gyWashington, DC: American Society of Microbiology; 1996. p. 458–84.
composition of Chinese Star anise (Illicium verum) and neurotoxicity in infants. J Plouvier V. Sur la recherche des acides quinique et shikimique, de la bergenine et des
Am Med Assoc 2004;291:562–3. heterosides chez quelques hamamelidacees. CR Hebd Seances Acad Sci 1961;252:
Jaroszynska J. Isolation of free phenolic compounds from arboreal leaves by use of the 599.
Florisil C18 system. Anal Bioanal Chem 2003;377:702–8. Raghavendra TR, Vaidyanathan P, Swathi HK, Ramesha BT, Ravikanth G, Ganeshaiah
Jensen RA, Nester EW. Regulatory enzymes of aromatic amino acid biosynthesis in KN, et al. Prospecting for alternate sources of shikimic acid, a precursor of Tamiflu,
Bacillus subtilis. J Biol Chem 1966a;241:3365–72. a bird-flu drug. Curr Sci 2009;96(6):771–2.
Jensen RA, Nester EW. The enzymology of feedback inhibition of 3-deoxy-D-arabino- Ran N, Knop DR, Draths KM, Frost JW. Benzene-free synthesis of hydroquinone. J Am
heptulosonate-7-phosphate synthase. J Biol Chem 1966b;241:3373–80. Chem Soc 2001;123:10927–34.
Jiang SD, Singh G. Chemical synthesis of shikimic acid and its analogues. Tetrahedron Roche. Factsheet Tamiflu. http://www.roche.com/med_mbtamiflu05e.pdf2006.
1998;54:4697–753. Rogoff MH. An aromatic intermediate in the bacterial oxidation of quinic acid. J Gen
Jiang S, Mekki B, Singh G, Wightman H. Enantiospecific synthesis of (−)-5-epi-shikimic Microbiol 1958;19:330–9.
acid and a new route to (−)-shikimic acid. Tetrahedron Lett 1994;35:5505–8. Rohloff JC, Kenneth MK, Postich MJ, Becker MW, Chapman HH, Kelly DE, et al.
Johansson L, Liden G. Transcriptome analysis of a shikimic acid producing strain of Practical total synthesis of the anti-influenza drug GS-4104. J Org Chem
Escherichia coli W3110 grown under carbon- and phosphate-limited conditions. J 1998;63:4545–50.
Biotechnol 2006;126:528–45. Sadaka M, Garcia A. Extraction of shikimic and quinic acids. Chem Eng Commun
Johansson L, Lindskog A, Silfversparre G, Cimander C, Nielsen KF, Lidén G. Shikimic acid 1999;173:91-102.
production by a modified strain of E. coli (W3110.shik1) under phosphate-limited Sánchez-Abella L, Fernández S, Armesto N, Ferrero M, Gotor V. Novel and efficient syn-
and carbon-limited conditions. Biotechnol Bioeng 2005;92:541–52. thesis of (−)-methyl 4-epi-shikimate and 4,5-epoxy-quinic and (−)-shikimic acid
Johansson, L., Metabolic analysis of shikimic acid producing Escherichia coli. PhD thesis, derivatives as key precursors to prepare new analogues. J Org Chem 2006;71:
Lund University, Sweden, 2006. 5396–9.
Kancharla PK, Doddi VR, Kokatla H, Vankar YD. A concise route to (−)-shikimic acid Shirai, M., Miyata, R., Sasaki, S., Sakamoto, K., Yahanda, S., Shibayama, K., Yonehara, T.,
and (−)-5-epi-shikimic acid, and their enantiomers via Barbier reaction and Ogawa, K. Microorganism belonging to the genus Citrobacter and process for pro-
ring-closing metathesis. Tetrahedron Lett 2009;50:6951–4. ducing shikimic acid. European Patent 1092766; 2001.
Kim CU, Lew W, Williams MA, Liu H, Zhang L, Swaminathan S, et al. Influenza neur- Simonart P, Wiaux A. Production of shikimic acid by Penicillium griseofulvum Dierckx.
aminidase inhibitor possessing a novel hydrophobic interaction in the enzyme ac- Nature 1960;186(4718):78–9.
tive site: design, synthesis, and structural analysis of carbocyclic sialic acid Smissman EE, Suh JT, Oxman M, Daniels R. A stereospecific synthesis of DL-shikimic
analogues with potent anti-influenza activity. J Am Chem Soc 1997;119:681–90. acid. J Am Chem Soc 1959;81:2909–10.
Kim CU, Lew W, Williams MA, Wu H, Zhang L, Chen X, et al. Structure activity relation- Starcevic A, Akthar S, Dunlap WC, Shick JM, Hranueli D, Cullum J, et al. Enzymes of
ship studies of novel carbocyclic influenza neuraminidase inhibitors. J Med Chem the shikimic acid pathway encoded in the genome of a basal metazoan, Nema-
1998;41:2451–60. tostella vectensis, have microbial origins. Proc Natl Acad Sci U S A 2008;105:
Knop DR, Draths KM, Chandran SS, Barker JL, Frost JW. Hydroaromatic equilibrium dur- 2533–7.
ing biosynthesis of shikimic acid. J Am Chem Soc 2001;123:10173–82. Stavric B, Stoltz DR. Shikimic acid. Food Cosmet Toxicol 1976;14:141–5.
Koreeda M, Ciufolini MA. Natural product synthesis via allylsilanes. 1. Synthesis and re- Sui RH. Separation of shikimic acid from Pine needles. Chem Eng Technol 2008;31:
actions of (1E, 3E)-4-acetoxy-l-(trimethylsilyl)1,3-butadiene and its use in the 469–73.
total synthesis of (±)-shikimic acid. J Am Chem Soc 1982;104:2308–10. Wang G-W, Hu W-T, Huang B-K, Qin L-P. Illicium verum: a review on its botany,
Koreeda M, Teng K, Murata T. (1E,3E)-4-acetoxy-1-phenyldimethylsilyl-1,3-butadiene traditional use, chemistry and pharmacology. J Ethnopharmacol 2011;136:
as a surrogate for (1E,3E)-1,4-diacetoxy-1,3-butadiene: a highly efficient synthesis 10–20.
of (±)-shikimic acid. Tetrahedron Lett 1990;31:5997–6000. Widmer N, Meylan P, Ivanyuk A, Aouri M, Decosterd LA, Buclin T. Oseltamivir in
Krämer M, Bongaerts J, Bovenberg R, Kremer S, Müller U, Orf S, et al. Metabolic engi- seasonal, avian H5N1 and pandemic 2009 A/H1N1 influenza pharmacokinetic
neering for microbial production of shikimic acid. Metab Eng 2003;5:277–83. and pharmacodynamic characteristics. Clin Pharmacokinet 2010;49:741–65.
Lederer I, Schulzki G, Gross J, Steffen JP. Combination of TLC and HPLC-MS/MS methods. Wilson DJ, Patton S, Florova G, Hale V, Reynolds KA. The shikimic acid pathway and
Approach to a rational quality control of Chinese star anise. J Agric Food Chem polyketide biosynthesis. J Ind Microbiol Biotechnol 1998;20:299–303.
2006;54:1970–4. Xie JY, Chen XP, Chen F. Study on extraction of shikimic acid from pine needles of Pinus
Li K, Frost JW. Microbial synthesis of 3-dehydroshikimic acid: a comparative analysis of elliottii by decompressing inner ebullition. Zhong Yao Cai 2010;33:1480–4. [In
D-xylose, D-arabinose, and D-glucose carbon sources. Biotechnol Prog 1999;15: Chinese].
876–83. Yamada K, Takada S, Nakamura S, Hirata Y. Structures of anisatin and neoanisatin — toxic
Li K, Mikola MR, Draths KM, Worden RM, Frost JW. Fed-batch fermenter synthesis of 3- sesquiterpenes from Illicium anisatum L. Tetrahedron 1968;24:199–229.
dehydroshikimic acid using recombinant Escherichia coli. Biotechnol Bioeng Ye, Y., Junyi, S., Tang, C., Sugimoto, K., Ke, C., Li, X., Tan, M., Ge, F., Yang, X., Ding, J. Meth-
1999;64:61–73. od for separating and purifying shikimic acid. China Patent 200510111303; 2007.
Martin E, Duke J, Pelkki M, Clausen EC, Carrier DJ. Sweetgum (Liquidambar styraciflua Yeung Y-Y, Hong S, Corey EJ. A short enantioselective pathway for the synthesis of the
L.): extraction of shikimic acid coupled to dilute acid pretreatment. Appl Biochem anti-influenza neuramidase inhibitor oseltamivir from 1, 3-butadiene and acrylic
Biotechnol 2010;162:1660–8. acid. J Am Chem Soc 2006;128:6310–1.
Matallo MB, Almeida SDB, Cerdeira AL, Franco DA, Blanco FMG, Menezes PTC, et al. Micro- Yi K, Li K, Draths KM, Frost JW. Modulation of phosphoenolpyruvate synthase expres-
wave-assisted solvent extraction and analysis of shikimic acid from plant tissues. sion increases shikimate pathway product yields in E. coli. Biotechnol Prog
Planta Daninha 2009;27:987–94. 2002;18:1141–8.
McCrindle R, Overton KH, Raphael RA. A stereospecific total synthesis of D-(−)-shikimic Yi J, Draths KM, Li K, Frost JW. Altered glucose transport and shikimate pathway product
acid. J Am Chem Soc 1960:1560–6. yields in E. coli. Biotechnol Prog 2003;19:1450–9.
Miles D, Garcia AA, Sadaka M, Whited G. Recovery of shikimic acid using temperature- Yoshida N, Ogasawara K. An enantioconvergent route to (−)-shikimic acid via a
swing complexation extraction and displacement back extraction. Isolation Purif palladium-mediated elimination reaction. Org Lett 2000;2:1461–3.
1994;2:75–82.
Mueller TC. Shikimate accumulates in both glyphosate-sensitive and glyphosate-
resistant horseweed (Conyza canadensis L. Cronq.). J Agric Food Chem 2003;51:
680–4.

Please cite this article as: Ghosh S, et al, Production of shikimic acid, Biotechnol Adv (2012), doi:10.1016/j.biotechadv.2012.03.001

Potrebbero piacerti anche