Acido Shikimico-2

7
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 abstract article info Available online 14 March 2012 Keywords: Shikimic acid Oseltamivir Quinic acid Shikimic acid is a key intermediate for the synthesis of the antiviral drug oseltamivir (Tamiu®). Shikimic acid can be produced via chemical synthesis, microbial fermentation and extraction from certain plants. An alternative production route is via biotransformation of the more readily available quinic acid. Much of the current supply of shikimic acid is sourced from the seeds of Chinese star anise (Illicium verum). Supply from star anise seeds has experienced difculties and is susceptible to vagaries of weather. Star anise tree takes around six-years from planting to bear fruit, but remains productive for long. Extraction and purica- tion from seeds are expensive. Production via fermentation is increasing. Other production methods are too expensive, or insufciently developed. In the future, production in recombinant microorganisms via fer- mentation may become established as the preferred route. Methods for producing shikimic acid are reviewed. © 2012 Elsevier Inc. All rights reserved. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1425 2. Production methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1426 2.1. Extraction from plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1426 2.2. Fermentation processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1427 2.2.1. The shikimic acid pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1427 2.2.2. Use of recombinant and engineered strains . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1428 2.3. Chemical synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1429 2.4. Microbial biotransformation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1429 3. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1430 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1430 1. Introduction This review is focussed on the methods of producing shikimic acid (3,4,5-trihydroxy-1-cyclohexene-1-carboxylic acid), a chemical building block for the antiviral drug oseltamivir (Tamiu®). Shikimic acid is named after the Japanese shikimi (Illicium anisatum) ower from which it was rst isolated. Shikimic acid is an intermediate of the shikimic acid pathway (Herrmann and Weaver, 1999) that is in- volved in the synthesis of aromatic metabolites in plants and micro- organisms (Ganem, 1978; Herrmann, 1995; Pittard, 1996; Wilson et al., 1998). Metabolically essential products of the shikimic acid path- 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 of the pathway have been found in certain animals (Starcevic et al., 2008). Properties and toxicology of shikimic acid have been discussed elsewhere (Stavric and Stoltz, 1976). An insufciency of shikimic acid has in the past affected the supply of oseltamivir (Farina and Brown, 2006). As a highly functionalised, six-carbon ring with three chiral car- bons and a carboxylic acid functional group, shikimic acid (Fig. 1) is Biotechnology Advances 30 (2012) 14251431 Abbreviations: DAHP, 3-Deoxy-D-arabino-heptulosonate-7-phosphate; DHQ, 3-Dehydroquinate or 3-dehydroquinic acid; DHS, 3-Dehydroshikimate or 3- dehyroshikimic acid; DQD, 3-Dehydroquinate dehydrogenase; EPSP, 5-Enolpyruvylshiki- mate-3-phosphate; E4P, Erythrose-4-phosphate; GDH, Glucose dehydrogenase; NADP, Nicotinamide adenine dinucleotide phosphate; NADPH, Reduced form of nicotinamide ad- enine dinucleotide phosphate; PEP, Phosphoenolpyruvate; QDH, Quinic acid dehydroge- nase; SKDH, Shikimic acid dehydrogenase. Corresponding author. Tel.: +91 172 2214682 87; fax: +91 172 2214692. E-mail address: [email protected] (U.C. Banerjee). 0734-9750/$ see front matter © 2012 Elsevier Inc. All rights reserved. doi:10.1016/j.biotechadv.2012.03.001 Contents lists available at SciVerse ScienceDirect Biotechnology Advances journal homepage: www.elsevier.com/locate/biotechadv

description

articulo sobre el acido shikimico

Transcript of Acido Shikimico-2

  • Research review paper

    ,

    ceutland

    1. Introduction . . . . . . . . . . . . . .2. Production methods . . . . . . . . . .

    2.1. Extraction from plants . . . . . .2.2. Fermentation processes . . . . .

    pathwat and e. . . .n . . .. . . .. . . .

    Biotechnology Advances 30 (2012) 14251431

    Contents lists available at SciVerse ScienceDirect

    Biotechnolog

    j ourna l homepage: www.e lsev1. Introduction

    This review is focussed on the methods of producing shikimic acid(3,4,5-trihydroxy-1-cyclohexene-1-carboxylic acid), a chemicalbuilding block for the antiviral drug oseltamivir (Tamiu). Shikimicacid is named after the Japanese shikimi (Illicium anisatum) ower

    from which it was rst isolated. Shikimic acid is an intermediate ofthe shikimic acid pathway (Herrmann and Weaver, 1999) that is in-volved in the synthesis of aromatic metabolites in plants and micro-organisms (Ganem, 1978; Herrmann, 1995; Pittard, 1996; Wilson etal., 1998). Metabolically essential products of the shikimic acid path-way include the three aromatic amino acids L-phenylalanine,L-tryptophan and L-tyrosine. Shikimic acid pathway is not normallyassociated 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,dehyroshikimic acid; DQD, 3-Dehydroquinate dehydrogenmate-3-phosphate; E4P, Erythrose-4-phosphate; GDH, GNicotinamide adenine dinucleotide phosphate; NADPH, Reenine dinucleotide phosphate; PEP, Phosphoenolpyruvatenase; SKDH, Shikimic acid dehydrogenase. Corresponding author. Tel.: +91 172 2214682 87; f

    E-mail address: [email protected] (U.C. Banerje

    0734-9750/$ see front matter 2012 Elsevier Inc. Alldoi:10.1016/j.biotechadv.2012.03.001References . . . . . . . . . . . . .2.2.1. The shikimic acid2.2.2. Use of recombinan

    2.3. Chemical synthesis . . .2.4. Microbial biotransformatio

    3. Concluding remarks . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1425

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1426

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1426

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1427y . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1427ngineered strains . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1428. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1429. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1429. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1430. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1430Contentsa b s t r a c ta r t i c l e i n f o

    Available online 14 March 2012

    Keywords:Shikimic acidOseltamivirQuinic acid

    Shikimic acid is a key intermediate for the synthesis of the antiviral drug oseltamivir (Tamiu). Shikimicacid can be produced via chemical synthesis, microbial fermentation and extraction from certain plants. Analternative production route is via biotransformation of the more readily available quinic acid. Much of thecurrent supply of shikimic acid is sourced from the seeds of Chinese star anise (Illicium verum). Supplyfrom star anise seeds has experienced difculties and is susceptible to vagaries of weather. Star anise treetakes around six-years from planting to bear fruit, but remains productive for long. Extraction and purica-tion from seeds are expensive. Production via fermentation is increasing. Other production methods aretoo expensive, or insufciently developed. In the future, production in recombinant microorganisms via fer-mentation may become established as the preferred route. Methods for producing shikimic acid arereviewed.

    2012 Elsevier Inc. All rights reserved.Production of shikimic acid

    Saptarshi Ghosh a, Yusuf Chisti b, Uttam C. Banerjee a

    a Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmab School of Engineering, Massey University, Private Bag 11 222, Palmerston North, New Zea3-Dehydroshikimate or 3-ase; EPSP, 5-Enolpyruvylshiki-lucose dehydrogenase; NADP,duced form of nicotinamide ad-; QDH, Quinic acid dehydroge-

    ax: +91 172 2214692.e).

    rights reserved.ical Education and Research, Sector-67, S.A.S. Nagar-160 062, Punjab, India

    y Advances

    i e r .com/ locate /b iotechadvof the pathway have been found in certain animals (Starcevic et al.,2008). Properties and toxicology of shikimic acid have been discussedelsewhere (Stavric and Stoltz, 1976). An insufciency of shikimic acidhas in the past affected the supply of oseltamivir (Farina and Brown,2006).

    As a highly functionalised, six-carbon ring with three chiral car-bons and a carboxylic acid functional group, shikimic acid (Fig. 1) is

  • a versatile enantiomerically pure precursor for making potentiallyuseful products. Interest in shikimic acid has been rekindled as it isrequired for producing the avian u drug oseltamivir (Fig. 1). Inview of the potential impact of a u pandemic (Horimoto andKawaoka, 2001) and the limited usefulness of vaccines against rapidlyevolving u viruses, stockpiles of effective drugs are necessary formanaging a major outbreak. Oseltamivir is an effective treatmentfor inuenza (Widmer et al., 2010), especially if administered early.Oseltamivir is effective against both Type A and Type B inuenzaand it is used also in prophylaxis.

    Oseltamivir is a viral neuraminidase inhibitor. Its synthesis fromquinic acid via shikimic acid has been described (Kim et al., 1997,1998; Rohloff et al., 1998). Oseltamivir produced for early clinical trialshad been made from chemically synthesized shikimic acid (Federspielet al., 2001), but this method proved impractical for commercial pro-duction of the drug. Chemical and microbial methods for inexpensiveproduction of shikimic acid are being developed, but most of thiscompound is currently extracted from the seeds of Chinese staranise (Illicium verum) (Payne and Edmonds, 2005). The multistep

    extraction and purication 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 ofshikimic or quinic acids have appeared (Fukuta et al., 2006; Yeunget al., 2006), but not been commercialized. Herewe review the existingand emerging methods for producing shikimic acid.

    2. Production methods

    2.1. Extraction from plants

    As an intermediate of the shikimic acid pathway, shikimic acidis found widely in plants (Bohm, 1965; Dell and Frost, 1993;Gurib-Fakim, 2006; Herrmann and Weaver, 1999), but generally oc-curs in low concentration. Seeds of Chinese star anise (Illicium verum)are the main commercial source of shikimic acid. The botany and phar-macology of Chinese star anise have been reviewed (Wang et al., 2011).

    Shikimic acid was rst isolated from owers of the highly toxicJapanese star anise. Subsequently, it was reported in the leaves ofthe sweetgum tree (Liquidambar styraciua) (Plouvier, 1961). Theseeds of the American sweetgum tree have been found to containup to 3.7% (w/w) shikimic acid (Enrich et al., 2008). Extraction of shi-kimic acid from bark and wood of the sweetgum has been reported(Martin et al., 2010). A simple extraction method based on hotwater was used. Several varieties of pine, r and spruce are knownto produce shikimic acid. A hot water (4575 C) extraction of theneedles of Scots pine (Pinus sylvestris) yielded around 1.6% (w/w)shikimic acid (Sui, 2008). Extraction from the needles of the pinePinus elliottii has been described (Xie et al., 2010). Attempts arebeing made to identify other natural plant sources of shikimic acid(Raghavendra et al., 2009).

    Fig. 1. Structure of shikimic acid and oseltamivir.

    1426 S. Ghosh et al. / Biotechnology Advances 30 (2012) 14251431Fig. 2. The shikimic acid or shikimate pathway.

  • Plants of the genus Illicium appear to the best source of shikimicacid, but many Illicium species are poisonous (Lederer et al., 2006;Yamada et al., 1968) and therefore not suitable for producing com-mercial shikimic acid. Japanese star anise (I. anisatum) is highlytoxic, but the Chinese star anise (I. verum) is edible. Nevertheless,compounds found in Chinese star anise have been shown to betoxic to infants (Ize-Ludlow et al., 2004).

    Shikimic acid is highly soluble in water (180 g/L at 20 C), but notin non-polar solvents. Therefore, a hot water extraction of the planttissue is used as the primary extraction step (Ohira et al., 2009; Sui,2008; Xie et al., 2010; Ye et al., 2007). Shikimic acid content in theplant tissue varies depending on the source of the tissue, the time ofharvest and other possible factors. Roughly 1 kg of shikimic acid can

    duces DHS to shikimic acid. The rate limiting enzymes of the shikimicacid pathway of E. coli are DHQ synthase and shikimate kinase (Delland Frost, 1993) (Fig. 2). Shikimate kinase (aroL and aroK) is responsiblefor converting shikimic acid to shikimate-3-phosphate (Fig. 2).

    In E. coli, the enzymes DHQ synthase, DHQ dehydratase andshikimate dehydrogenase (Fig. 2) are constitutively expressedwhereas the production of the DAHP synthases and one of the

    Table 2Modied E. coli strains with 3-dehydroshikimic acid yield and titer (Li et al., 1999).

    Number Strain Modication DHS yield(mol/mol)

    DHS titer(g/L)

    1 KL3/pKL4.33B

    aroFFBR expression controlled byParoF in absence of amplied tktA

    0.17 20.3

    2 KL3/pKL4.66A

    aroFFBR expression controlled byParoF in absence of amplied tktA

    0.16 38.5

    3 KL3/pKL4.130B

    aroFFBR expression controlled byParoF in the presence of ampliedtktA

    0.30 69.0

    4 KL3/ aroFFBR expression is controlled 0.18 41.2

    Table 3Effect of carbon sources on the production of 3-dehydroshikimic acid by recombinantE. coli strains (Li et al., 1999).

    Strain KL3/pKL4.124A(tktA over expression)

    KL3/pKL4.79B (tktA)

    Carbon source DHS titer(g/L)

    DHS yield(mol/mol)

    Carbonsource

    DHS titer(g/L)

    DHS yield(mol/mol)

    Glucose 46.0 0.28 Glucose 36.4 0.22Xylose 43.1 0.33 Xylose 41.7 0.32Mixa 64.0 0.41 Mixa 53.0 0.36

    a

    1427S. Ghosh et al. / Biotechnology Advances 30 (2012) 14251431be recovered from 30 kg of dry seed pods of Chinese star anise(Ohira et al., 2009; Roche, 2006). Recovery is nearly complete (>95%)within 10 min of water extraction at 70 C from seed pods ground to aparticle size of 355600 m (Ohira et al., 2009). The rate of recoverycan be further enhanced by using a higher extraction temperature.

    Other extraction methods have been described, involving the useof acids (Harring et al., 1998; Mueller, 2003), alcohols (Anderson etal., 2001; Jaroszynska, 2003), complex formation (Miles et al., 1994;Sadaka and Garcia, 1999) and microwave-assisted extraction(Matallo et al., 2009). The crude water extract contains numerousother water-soluble plant metabolites and requires extensive furtherprocessing to yield pure shikimic acid. The recovery and puricationprocesses used in commercial production from Chinese star aniseare proprietary and involve multiple steps.

    2.2. Fermentation processes

    Microbial production of shikimic acid involves the shikimic acid (orshikimate) pathway. Shikimic acid pathway occurs commonly inmicro-organisms and therefore they can be used to overproduce this com-pound from carbon sources such as glucose (Draths et al., 1999;Simonart and Wiaux, 1960). Shikimic acid is a precursor for essentialamino acids and other metabolites in microorganisms. Its overproduc-tion can be achieved for example by metabolic engineering to partlyblock some of the biochemical pathways that consume shikimic acidand by overexpression of the enzymes responsible for its synthesis. Pro-duction by fermentation using engineered microorganisms is alreadysupplementing the commercial supply of shikimic acid.

    2.2.1. The shikimic acid pathwayThe shikimic acid pathway is shown in Fig. 2. Thismetabolic pathway

    is used to produce the aromatic amino acids that are essential to growth.Shikimic acid is an intermediate in the pathway. This pathway has beenpreviously reviewed (Herrmann andWeaver, 1999).

    The shikimic acid pathway begins with phosphoenolpyruvate(PEP) and erythrose-4-phosphate (E4P) (Fig. 2). These two com-pounds must rst be generated from metabolism of glucose, orother carbohydrate. PEP is produced through the glycolysis pathway(Fig. 2) whereas the completely independent pentose phosphate

    Table 1Modied E.coli strains with shikimic acid (SA) yield and titer.

    Strain Modication SA yield(mol/mol)

    SAtiter (g/L)

    SP1.1/pKD15.071B(Chandran et al., 2003)

    Over expression of ppsA 0.23 66

    SP1.1pts/pSC6.090B(Chandran et al., 2003)

    PTS/glf+/tktA overexpression

    0.27 71

    SP1.1/pKD12.138(Knop et al., 2001)

    Over expression of tktA 0.18 52

    SP1.1/pKD12.112(Draths et al., 1999)

    Insertion of aroB into theserA locus of E.coli anddisruption of aroL and aroK

    0.15 27.2pathway is used to generate E4P (Fig. 2). Thus, the shikimic acid path-way is dependent on the glycolytic pathway and the pentose phos-phate pathway to provide the two starting materials it requires.Therefore, metabolic engineering of the shikimic acid pathway alonemay be insufcient for increasing the yield of shikimic acid from thecarbon source used in a fermentation. Furthermore, because shikimicacid is produced far downstream in the metabolic pathway relative tothe point where glucose rst enters the metabolism, channelling theow of carbon to production of shikimic acid can be difcult.

    Once E4P and PEP have been generated through carbohydrate me-tabolism in other independent pathways, they are condensed in theshikimic acid pathway through the action of the enzyme DAHPsynthase to produce DAHP. Action of three other enzymes then con-verts DAHP to shikimic acid. Chorismic acid is the nal product ofthe SA pathway and this compound is the common precursor forthe biosynthesis of other aromatic products such as aromatic aminoacids, as it is shown in Fig. 2.

    The shikimic acid pathway engineering has most commonly in-volved the bacterium Escherichia coli. This microorganism has threeisoforms of the DAHP synthase enzyme encoded by aroF, aroG, aroHshow feedback inhibition by the three aromatic amino acids, i.e.L-tyrosine, L-phenylalanine and L-tryptophan respectively. TheDAHP synthase (aroG) of Bacillus subtilis is inhibited by the pathwayintermediate chorismate (Jensen and Nester, 1966a,b) (Fig. 2).

    The enzyme DHQ synthase (aroB) in E. coli converts DAHP into3-dehydroquinate (DHQ). The enzyme DHQ dehydratase (aroD) thenconverts DHQ into 3-dehydroshikimate (DHS) by eliminating water.Subsequently, NADPH-dependent shikimate dehydrogenase (aroE) re-

    pKD11.291A by ParoF in absence of ampliedtktA

    5 KL3/pKL5.17A

    aroFFBR expression is controlledby ParoF in presence of ampliedtktA

    0.24 58.1

    6 KL3/pKL4.124A

    aroFFBR expression under thecontrol of Ptac in presence of tktA

    0.28 66.0Molar ratio of glucose/xylose/arabinose: 3:3:2.

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

    1428 S. Ghosh et al. / Biotechnology Advances 30 (2012) 14251431shikimate kinases is transcriptionally regulated. Shikimic acid in-hibits shikimate dehydrogenase (Dell and Frost, 1993).

    An alternative microbial route to shikimic acid is through bio-transformation of quinic acid. Certain microorganisms (Pseudomonas,Achromobacter, Aspergillus and Neurospora crassa) can use quinic acid(or its salt, quinate) as the sole carbon source (Case et al., 1978; DaSilva et al., 1986; Rogoff, 1958) to produce aromatic amino acids viathe shikimic acid pathway. Quinate enters the pathway at the pointshown in Fig. 2. E. coli strains specically engineered for overprodu-cing quinic acid from glucose have also been developed (Ran et al.,2001).

    2.2.2. Use of recombinant and engineered strainsMetabolically engineered bacteria provide an important emerging

    route to production of shikimic acid via fermentation (Campbell et al.,1993; Krmer et al., 2003). The bacterium E. coli has been the focus ofmost metabolic engineering effort (Ahn et al., 2008; Escalante et al.,2010; Johansson and Liden, 2006; Johansson et al., 2005, 2006;Knop et al., 2001; Yi et al., 2002, 2003), but studies in other bacteriahave also been reported.

    Several metabolic engineering approaches have been developed tooverproduce shikimic acid in E. coli (Ahn et al., 2008; Chandran et al.,2003; Escalante et al., 2010; Gibson et al., 2001; Johansson et al.,2005; Knop et al., 2001; Krmer et al., 2003). All these are based ongenetic modications to alter the central carbon metabolism andthe shikimic acid pathway (Table 1).

    The shikimic acid pathway requires PEP and E4P (Fig. 2). The sup-ply of PEP and E4P can be enhanced via metabolic engineering of theglycolytic pathway and the pentose phosphate pathway, respectively.Both these approaches have been demonstrated. Over expression oftransketolase (tktA) resulted in the increase of shikimic acid yieldfrom 0.12 to 0.18 mol/mol and titer from 38 to 52 g/L by enhancingthe concentration of E4P (Knop et al., 2001). In recombinant E. coli,Fig. 4. Shikimic acid synthesis of Koreeda and Ciufolian increased availability of PEP has enhanced production of shikimicacid (Chandran et al., 2003; Yi et al., 2002). The modication of theglycolytic pathway involved over expression of PEP synthase (ppsA)leading to increased shikimic acid titer to 66 g/L and yield on glucoseof 0.23 mol/mol (Chandran et al., 2003). Inactivation of the PTS operon(PTS), expression of non-PTS glucose transporters like glucose facilita-tors (glf), glucokinase (glk) in combinationwith over expression of tktAgene was reported to increase the shikimic acid titer to 71 g/L(Chandran et al., 2003; Gibson et al., 2001). Using the engineered E.coli strain, the shikimic acid concentration could be further raised to84 g/L by supplementing the minimal medium with yeast extract(Chandran et al., 2003). Shikimic acid could be produced during expo-nential growth on glucose to a nal concentration of 87 g/L (Chandranet al., 2003). The nature of the carbon source affected the productivityof shikimic acid (Ahn et al., 2008; Li et al., 1999).

    In earlier studies, the shikimic acid titers from metabolically engi-neered E. coli strains were low partly due to simultaneous productionof quinic acid (Knop et al., 2001). Production of quinic acid during bio-synthesis of shikimic acid is a consequence of the microbe-catalyzedequilibration of the initially synthesized shikimic acid (Knop et al.,2001). This problem appears to have been resolved in view of the afore-mentioned high titers.

    Shikimic acid can be produced readily from DHS (Fig. 2). There-fore, some metabolic engineering studies have focussed on overpro-duction of DHS. Substantial overproduction of DHS in metabolicallyengineered microorganisms has been achieved (Draths and Frost,1990; Li and Frost, 1999; Li et al., 1999; Yi et al., 2002, 2003)(Table 2). For example, E. coli constructs have produced as much as60 g/L DHS from glucose in 60 h (Yi et al., 2003). A comparative analysisof D-xylose, D-glucose and D-arabinose as carbon source for microbialsynthesis of DHSwas also reported (Li and Frost, 1999) (Table 3). Differ-ent recombinant E. coli strains were used with various carbon sourcesfor DHS production.ni (1982) as summarized by Ambhaikar (2005).

  • nate supplied in the medium could be converted to DHQ (Adachi et al.,

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

    1429S. Ghosh et al. / Biotechnology Advances 30 (2012) 14251431The activity of DAHP synthase (Fig. 2) controls the amount of cel-lular carbon directed into DHS synthesis. Transcriptional repressionand feedback inhibition of DAHP synthase by aromatic amino acidsare believed to control the activity of this enzyme. The ampliedexpression of a mutant DAHP synthase, which is insensitive tofeedback inhibition by aromatic amino acids, has been used to en-hance production of DHS. Metabolic engineering approaches havebeen employed to produce shikimic acid in E. coli strains derivedfrom an evolved strain PB 12 lacking the PTS system but with abilityto grow on glucose. The double aroK-, aroL- mutant of strain PB12.SA22 showed shikimic acid titer of 7 g/L and yield of 0.29 mol/mol(Escalante et al., 2010).

    In addition to recombinant E. coli, genetically modied or other-wise mutated Bacillus subtilis (Iomantas et al., 2002) and Citrobacterfreundii bacteria (Shirai et al., 2001) have been used to successfullyoverproduce shikimic acid although the titers have not exceededabout 20 g/L. It was reported that aroI (shikimate kinase) decientstrain of B. subtilis showed shikimic acid titer of 8.5 g/L with hightiter of DHS (9.5 g/L) as the by-product (Iomantas et al., 2002). Useof glyphosate, an inhibitor of the enzyme EPSP synthase (Fig. 2), inthe fermentation medium to enhance accumulation of shikimic acidby blocking its downstream consumption, has been described(Bogosian, 2011). Metabolic engineering of microorganisms for pro-ducing shikimic acid has been reviewed by Krmer et al. (2003).

    2.3. Chemical synthesis

    Chemical synthesis of shikimic acid was rst achieved during the1960s using the basic chemistry of Diels-Alder reaction (Fig. 3) toform six membered rings (McCrindle et al., 1960; Smissman et al.,1959), but the yield was low at 15%. Later attempts to increaseyield were not particularly successful (Grewe and Hinrichs, 1964).In 1982, the efciency of Diels-Alder synthesis could be improved(Fig. 4) to raise the yield to 29% (Koreeda and Ciufolini, 1982). Amore efcient synthesis further raised the yield to 55% (Koreeda etal., 1990).

    Synthesis of shikimic acid from benzene has been shown to bepossible (Birch et al., 1988). Synthesis via a palladiummediated elim-ination reaction was advanced by Yoshida and Ogasawara (2000).Synthesis of ()-shikimic acid has been reported from sugars suchas D-mannose (Dangschat and Fischer, 1938, 1950; Fleet et al., 1984;Jiang et al., 1994). Shikimic acid can be made also from ()-quinicacid and its derivatives by chemical transformations (Box et al., 2002;Cleophax et al., 1971, 1973; Federspiel et al., 2001; Kim et al., 1997,1998; Rohloff et al., 1998) (Fig. 5). Other synthetic methods haveFig. 6. Synthesis of oseltam2003). This transformation could be achieved also with dried cells ofG. oxydans (Adachi et al., 2003) and with fresh cells immobilized inalginate beads. The intermediate DHQ could be further transformedto DHS in a reasonable yield by both growing and immobilizedcells. Lactobacillus pastorianus is capable of converting quinic acid to aproduct that was claimed to be dihydroshikimic acid (Carr et al.,1957), but was more likely DHS.

    A two-step scheme for biotransformation of quinic acid to shikimicacid has been published (Fig. 7) (Adachi et al., 2006). The rst step ofthis scheme involves an oxidative fermentation with G. oxydans to es-sentially quantitatively convert quinic acid in the culture medium toDHS. The latter is subsequently converted to shikimic acid in anNADPH-dependent reaction using the enzyme shikimic acid dehy-drogenase (SKDH) (Fig. 7) puried from cells of G. oxydans. The en-zymatic biotransformation medium also contained glucose and theenzyme glucose dehydrogenase to continuously regenerate NADPH(Fig. 7).been described (Kancharla et al., 2009; Snchez-Abella et al., 2006).Chemical synthesis of shikimic acid and its analogs is further reviewedelsewhere (Campbell et al., 1993; Jiang and Singh, 1998).

    Although several routes have been described for chemical synthesis,production of shikimic acid by these methods is too expensive to beof commercial use. Consequently, isolation from Chinese star anisefruit remains the principal source of commercial shikimic acid(Raghavendra et al., 2009).

    The ten step commercial route of oseltamivir (Tamiu) synthesisuses ()-shikimic acid as the starting material (Abrecht et al., 2004).This precursor is converted into a diethyl ketal intermediate, which isreductively opened to give 1,2-epoxide. This epoxide is then con-verted into oseltamivir via a ve step reaction involving 3 potentiallytoxic and explosive azide intermediates (Fig. 6). This route gives 35%yield of oseltamivir. Future methods for making oseltamivir maycompletely circumvent the need for shikimic or quinic acids (Fukutaet al., 2006; Yeung et al., 2006).

    2.4. Microbial biotransformation

    Many microorganisms are able to convert quinic acid to shikimicacid (Adachi et al., 2003, 2006). Quinic acid (or quinate) is convertedto 3-dehydroquinate (DHQ) (Fig. 2) through the action of quinate de-hydrogenase. DHQ is an intermediate of the shikimic acid pathwayand is transformed to shikimic acid in a two-step process (Fig. 2).

    During exponential growth of Gluconobacter oxydans, nearly all qui-ivir from shikimic acid.

  • pro

    1430 S. Ghosh et al. / Biotechnology Advances 30 (2012) 142514313. Concluding remarks

    Shikimic acid is a precursor for the synthesis of the important anti-viral drug oseltamivir. Extraction from Chinese star anise is the mainsource for shikimic acid, but fermentation processes based on the re-combinant bacterium E. coli have been shown to be a viable alternativesource. Chemical synthesis of shikimic acid is possible, but apparentlynot commercially viable. Chemical methods of making oseltamivirmay entirely circumvent the need for shikimic acids, but are not com-mercially used. Bioconversion of quinic acid to shikimic acid is anotheroption for production, but appears not to have been developed to theextent of the fermentation route from glucose.

    References

    Abrecht S, Harrington P, Iding H, Karpf M, Trussardi R, Wirz B, et al. The synthetic de-

    Fig. 7. Overall reaction for shikimic acidvelopment of the anti-inuenza neuraminidase inhibitor oseltamivir phosphate(Tamiu): a challenge for synthesis and process research. Chimia 2004;58:6219.

    Adachi O, Tanasupawat S, Yoshihara N, Toyama H, Matsushita M. 3-Dehydroquinate pro-duction by oxidative fermentation and further conversion of 3-dehydroquinate to theintermediates in the shikimate pathway. Biosci Biotechnol Biochem 2003;67:212431.

    Adachi O, Ano Y, Toyama H, Matsushita K. High shikimate production from quinatewith two enzymatic systems of acetic acid bacteria. Biosci Biotechnol Biochem2006;70:257982.

    Ahn JO, Lee HW, Saha R, Park MS, Jung JK, Lee DY. Exploring the effects of carbonsources on the metabolic capacity for shikimic acid production in Escherichia coliusing in silico metabolic predictions. J Microbiol Biotechnol 2008;18:177384.

    Ambhaikar N. Shikimic acid. A seminar presentation dated 12 January 2005. La Jolla,California, USA: The Scripps Research Institute; 2005http://www.scripps.edu/chem/baran/images/grpmtgpdf/Ambhaikar_Jan_05.pdf.

    Anderson KA, Cobb WT, Loper BR. Analytical method for determination of shikimicacid: Shikimic acid proportional to glyphosate application rates. Commun Soil SciPlant Anal 2001;32:283140.

    Birch AJ, Kelly LF, Weerasuria DV. Facile synthesis of (+)- and ()-shikimic acid withasymmetric deuterium labelling, using tricarbonyliron as a lateral control group. JOrg Chem 1988;53:27881.

    Bogosian, G., Frantz, J.P. Use of glyphosate to produce shikimic acid in microorganisms.United States Patent Application Pub. No.: US 2011/0020885 Al; 2011.

    Bohm BA. Shikimic acid (3,4,5-trihydroxy-1-cyclohexene-1-carboxylic acid). Chem Rev1965;65:43566.

    Box JM, Harwood LM, Humphreys JL, Morris GA, Redon PM, Whitehead RC. Dehydrationof quinate derivatives: synthesis of a diuoromethylene homologue of shikimic acid.Synlett 2002;2:35860.

    Campbell MM, Sainsbury M, Searle PA. The biosynthesis and synthesis of shikimic acid,chorismic acid, and related-compounds. Synth Stuttg 1993;2:17993.

    Carr JG, Pollard A, Whiting GC, Williams AH. The reduction of quinic acid to dihydroshi-kimic acid by certain lactic acid bacteria. Biochem J 1957;66:2835.Case ME, Pueyo C, Barea JL, Giles NH. Genetical and biochemical characterization ofQA-3 mutants and revertants in the QA gene cluster of Neurospora crassa. Genetics1978;90:6984.

    Chandran SS, Yi J, Draths KM, Von Daeniken R, Weber W, Frost JW. Phosphoenolpyr-uvate availability and the biosynthesis of shikimic acid. Biotechnol Prog 2003;19:80814.

    Cleophax J, Mercier D, Gro SD. A stereospecic conversion of ()-methyltri-O-benzoylquinate to the corresponding ()-methyl shikimate. Angew ChemInt Ed 1971;10:6523.

    Cleophax J, Leboul J, Mercier D, Gaudemer A, Gero SD. New and easy synthetic route toshikimic and 4-epishikimic acid. Bull Soc Chim Fr Partie II 1973:29925.

    Da Silva AJ, Whittington H, Clements J, Roberts C, Hawkins AR. Sequence analysis andtransformation by the catabolic 3-dehydroquinase (QUTE) gene from Aspergillusnidulans. Biochem J 1986;240:4818.

    Dangschat G, Fischer HOL. bergang der Chinasure in shikimisure. Naturwissenschaften1938;26:5623.

    Dangschat G, Fischer HOL. Congurational relationships between naturally occurringcyclic plant acids and glucose transformation of quinic acid into shikimic acid. Bio-chem Biophys Acta 1950;4:199204.

    Dell KA, Frost JW. Identication and removal of impediments to biocatalytic synthesisof aromatics from D-glucose: rate limiting enzymes in the common pathway of

    duction (based on Adachi et al., 2006).aromatic amino acid biosynthesis. J Am Chem Soc 1993;115:115819.Draths KM, Frost JW. Genomic direction of synthesis during plasmid-based biocatalysis. J

    Am Chem Soc 1990;112:96302.Draths KM, Knop DR, Frost JW. Shikimic acid and quinic acid: replacing isolation from

    plant sources with recombinant microbial biocatalysis. J Am Chem Soc 1999;121:16034.

    Enrich LB, Scheuermann ML, Mohadjer A, Matthias KR, Eller CF, Newman MS, et al.Liquidambar styraciua: a renewable source of shikimic acid. Tetrahedron Lett2008;49:25035.

    Escalante A, Caldern R, Valdivia A, de Anda R, Hernndez G, Ramrez OT, et al. Meta-bolic engineering for the production of shikimic acid in an evolved Escherichiacoli strain lacking the phosphoenolpyruvate: carbohydrate phosphotransferasesystem. Microb Cell Fact 2010;9:21.

    Farina V, Brown JD. Tamiu: the supply problem. Angew Chem Int Ed 2006;45:73304.Federspiel M, Fischer R, HennigM,Mair HJ, Oberhauser T, Rimmler G, et al. Industrial syn-

    thesis of the key precursor in the synthesis of the anti-inuenza drug oseltamivirphosphate (Ro 64-0796/002, GS-4104-02): Ethyl (3R, 4S, 5S)-4, 5-epoxy-3-(1-ethyl-propoxy)-cyclohex-1-ene-1-carboxylate. Org Process Res Dev 2001;3:26674.

    Fleet GWJ, Shing TKM, Warr SM. Enantiospecic synthesis of shikimic acid fromD-mannose: formation of a chiral cyclohexene by intramolecular olenation of acarbohydrate-derived intermediate. J Chem Soc Perkin Trans 1 1984:9058.

    Fukuta Y, Mita T, Fukuda N, Kanai M, Shibasaki M. De novo synthesis of Tamiu via acatalytic asymmetric ring-opening of meso-aziridines with TMSN3. J Am ChemSoc 2006;128:63123.

    Ganem B. Shikimate-derivedmetabolites. 4. From glucose to aromatics recent develop-ments in natural-products of shikimic acid pathway. Tetrahedron 1978;34:335383.

    Gibson JM, Thomas PS, Thomas JD, Barker JL, Chandran SS, Harrup MK, et al. Benzene-free synthesis of phenol. Angew Chem Int Ed 2001;40:19458.

    Grewe R, Hinrichs I. Eine neue synthese der shikimisaure. Chem Ber 1964;97:443.Gurib-Fakim A. Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol

    Aspects Med 2006;27:1-93.Harring T, Streibig JC, Husted S. Accumulation of shikimic acid: a technique for screening

    glyphosate efcacy. J Agric Food Chem 1998;46:440612.

  • Herrmann KM. The shikimate pathway: early steps in the biosynthesis of aromaticcompounds. Plant Cell 1995;7:90719.

    Herrmann KM, Weaver LM. The shikimate pathway. Annu Rev Plant Physiol Plant MolBiol 1999;50:473503.

    Horimoto T, Kawaoka Y. Pandemic threat posed by avian inuenza A viruses. ClinMicrobiol Rev 2001;14:12949.

    Iomantas, Y., Abalakina, E.G., Polanuer, B., Yampolskaya, T.A., Bachina, T.A., Kozlov, J.I.Method for producing shikimic acid. US Patent 6436664; 2002.

    Ize-Ludlow D, Ragone S, Bernstein JN, Bruck IS, Duchowny M, Pena BMG. Chemicalcomposition of Chinese Star anise (Illicium verum) and neurotoxicity in infants. JAm Med Assoc 2004;291:5623.

    Jaroszynska J. Isolation of free phenolic compounds from arboreal leaves by use of the

    Ohira H, Torii N, Aida TM, Watanabe M, Smith Jr RL. Rapid separation of shikimic acidfrom Chinese star anise (Illicium verum Hook. f.) with hot water extraction. SepPurif Technol 2009;69:1028.

    Payne R, Edmonds M. Isolation of shikimic acid from star anise seeds. J Chem Educ2005;82:599600.

    Pittard AJ. Biosynthesis of aromatic amino acids. In: Neidhardt FC, Curtiss III R,Ingraham JL, Lin ECC, Low KB, Magasanic B, Reznikoff WS, Riley M, Schaechter M,Umbarger HE, editors. Escherichia coli and Salmonella. Cellular and Molecular Biolo-gyWashington, DC: American Society of Microbiology; 1996. p. 45884.

    Plouvier V. Sur la recherche des acides quinique et shikimique, de la bergenine et desheterosides chez quelques hamamelidacees. CR Hebd Seances Acad Sci 1961;252:599.

    Raghavendra TR, Vaidyanathan P, Swathi HK, Ramesha BT, Ravikanth G, GaneshaiahKN, et al. Prospecting for alternate sources of shikimic acid, a precursor of Tamiu,a bird-u drug. Curr Sci 2009;96(6):7712.

    Ran N, Knop DR, Draths KM, Frost JW. Benzene-free synthesis of hydroquinone. J Am

    1431S. Ghosh et al. / Biotechnology Advances 30 (2012) 14251431Jensen RA, Nester EW. Regulatory enzymes of aromatic amino acid biosynthesis inBacillus subtilis. J Biol Chem 1966a;241:336572.

    Jensen RA, Nester EW. The enzymology of feedback inhibition of 3-deoxy-D-arabino-heptulosonate-7-phosphate synthase. J Biol Chem 1966b;241:337380.

    Jiang SD, Singh G. Chemical synthesis of shikimic acid and its analogues. Tetrahedron1998;54:4697753.

    Jiang S, Mekki B, Singh G, Wightman H. Enantiospecic synthesis of ()-5-epi-shikimicacid and a new route to ()-shikimic acid. Tetrahedron Lett 1994;35:55058.

    Johansson L, Liden G. Transcriptome analysis of a shikimic acid producing strain ofEscherichia coli W3110 grown under carbon- and phosphate-limited conditions. JBiotechnol 2006;126:52845.

    Johansson L, Lindskog A, Silfversparre G, Cimander C, Nielsen KF, Lidn G. Shikimic acidproduction by a modied strain of E. coli (W3110.shik1) under phosphate-limitedand carbon-limited conditions. Biotechnol Bioeng 2005;92:54152.

    Johansson, L., Metabolic analysis of shikimic acid producing Escherichia coli. PhD thesis,Lund University, Sweden, 2006.

    Kancharla PK, Doddi VR, Kokatla H, Vankar YD. A concise route to ()-shikimic acidand ()-5-epi-shikimic acid, and their enantiomers via Barbier reaction andring-closing metathesis. Tetrahedron Lett 2009;50:69514.

    Kim CU, Lew W, Williams MA, Liu H, Zhang L, Swaminathan S, et al. Inuenza neur-aminidase inhibitor possessing a novel hydrophobic interaction in the enzyme ac-tive site: design, synthesis, and structural analysis of carbocyclic sialic acidanalogues with potent anti-inuenza activity. J Am Chem Soc 1997;119:68190.

    Kim CU, LewW, Williams MA, Wu H, Zhang L, Chen X, et al. Structure activity relation-ship studies of novel carbocyclic inuenza neuraminidase inhibitors. J Med Chem1998;41:245160.

    Knop DR, Draths KM, Chandran SS, Barker JL, Frost JW. Hydroaromatic equilibrium dur-ing biosynthesis of shikimic acid. J Am Chem Soc 2001;123:1017382.

    Koreeda M, Ciufolini MA. Natural product synthesis via allylsilanes. 1. Synthesis and re-actions of (1E, 3E)-4-acetoxy-l-(trimethylsilyl)1,3-butadiene and its use in thetotal synthesis of ()-shikimic acid. J Am Chem Soc 1982;104:230810.

    Koreeda M, Teng K, Murata T. (1E,3E)-4-acetoxy-1-phenyldimethylsilyl-1,3-butadieneas a surrogate for (1E,3E)-1,4-diacetoxy-1,3-butadiene: a highly efcient synthesisof ()-shikimic acid. Tetrahedron Lett 1990;31:59976000.

    Krmer M, Bongaerts J, Bovenberg R, Kremer S, Mller U, Orf S, et al. Metabolic engi-neering for microbial production of shikimic acid. Metab Eng 2003;5:27783.

    Lederer I, Schulzki G, Gross J, Steffen JP. Combination of TLC and HPLC-MS/MS methods.Approach to a rational quality control of Chinese star anise. J Agric Food Chem2006;54:19704.

    Li K, Frost JW. Microbial synthesis of 3-dehydroshikimic acid: a comparative analysis ofD-xylose, D-arabinose, and D-glucose carbon sources. Biotechnol Prog 1999;15:87683.

    Li K, Mikola MR, Draths KM, Worden RM, Frost JW. Fed-batch fermenter synthesis of3-dehydroshikimic acid using recombinant Escherichia coli. Biotechnol Bioeng1999;64:6173.

    Martin E, Duke J, Pelkki M, Clausen EC, Carrier DJ. Sweetgum (Liquidambar styraciuaL.): extraction of shikimic acid coupled to dilute acid pretreatment. Appl BiochemBiotechnol 2010;162:16608.

    MatalloMB, Almeida SDB, Cerdeira AL, Franco DA, Blanco FMG,Menezes PTC, et al. Micro-wave-assisted solvent extraction and analysis of shikimic acid from plant tissues.Planta Daninha 2009;27:98794.

    McCrindle R, Overton KH, Raphael RA. A stereospecic total synthesis of D-()-shikimicacid. J Am Chem Soc 1960:15606.

    Miles D, Garcia AA, Sadaka M, Whited G. Recovery of shikimic acid using temperature-swing complexation extraction and displacement back extraction. Isolation Purif1994;2:7582.

    Mueller TC. Shikimate accumulates in both glyphosate-sensitive and glyphosate-resistant horseweed (Conyza canadensis L. Cronq.). J Agric Food Chem 2003;51:6804.Chem Soc 2001;123:1092734.Roche. Factsheet Tamiu. http://www.roche.com/med_mbtamiu05e.pdf2006.Rogoff MH. An aromatic intermediate in the bacterial oxidation of quinic acid. J Gen

    Microbiol 1958;19:3309.Rohloff JC, Kenneth MK, Postich MJ, Becker MW, Chapman HH, Kelly DE, et al.

    Practical total synthesis of the anti-inuenza drug GS-4104. J Org Chem1998;63:454550.

    Sadaka M, Garcia A. Extraction of shikimic and quinic acids. Chem Eng Commun1999;173:91-102.

    Snchez-Abella L, Fernndez S, Armesto N, Ferrero M, Gotor V. Novel and efcient syn-thesis of ()-methyl 4-epi-shikimate and 4,5-epoxy-quinic and ()-shikimic acidderivatives as key precursors to prepare new analogues. J Org Chem 2006;71:53969.

    Shirai, M., Miyata, R., Sasaki, S., Sakamoto, K., Yahanda, S., Shibayama, K., Yonehara, T.,Ogawa, K. Microorganism belonging to the genus Citrobacter and process for pro-ducing shikimic acid. European Patent 1092766; 2001.

    Simonart P, Wiaux A. Production of shikimic acid by Penicillium griseofulvum Dierckx.Nature 1960;186(4718):789.

    Smissman EE, Suh JT, Oxman M, Daniels R. A stereospecic synthesis of DL-shikimicacid. J Am Chem Soc 1959;81:290910.

    Starcevic A, Akthar S, Dunlap WC, Shick JM, Hranueli D, Cullum J, et al. Enzymes ofthe shikimic acid pathway encoded in the genome of a basal metazoan, Nema-tostella vectensis, have microbial origins. Proc Natl Acad Sci U S A 2008;105:25337.

    Stavric B, Stoltz DR. Shikimic acid. Food Cosmet Toxicol 1976;14:1415.Sui RH. Separation of shikimic acid from Pine needles. Chem Eng Technol 2008;31:

    46973.Wang G-W, Hu W-T, Huang B-K, Qin L-P. Illicium verum: a review on its botany,

    traditional use, chemistry and pharmacology. J Ethnopharmacol 2011;136:1020.

    Widmer N, Meylan P, Ivanyuk A, Aouri M, Decosterd LA, Buclin T. Oseltamivir inseasonal, avian H5N1 and pandemic 2009 A/H1N1 inuenza pharmacokineticand pharmacodynamic characteristics. Clin Pharmacokinet 2010;49:74165.

    Wilson DJ, Patton S, Florova G, Hale V, Reynolds KA. The shikimic acid pathway andpolyketide biosynthesis. J Ind Microbiol Biotechnol 1998;20:299303.

    Xie JY, Chen XP, Chen F. Study on extraction of shikimic acid from pine needles of Pinuselliottii by decompressing inner ebullition. Zhong Yao Cai 2010;33:14804. [InChinese].

    Yamada K, Takada S, Nakamura S, Hirata Y. Structures of anisatin and neoanisatin toxicsesquiterpenes from Illicium anisatum L. Tetrahedron 1968;24:199229.

    Ye, Y., Junyi, S., Tang, C., Sugimoto, K., Ke, C., Li, X., Tan, M., Ge, F., Yang, X., Ding, J. Meth-od for separating and purifying shikimic acid. China Patent 200510111303; 2007.

    Yeung Y-Y, Hong S, Corey EJ. A short enantioselective pathway for the synthesis of theanti-inuenza neuramidase inhibitor oseltamivir from 1, 3-butadiene and acrylicacid. J Am Chem Soc 2006;128:63101.

    Yi K, Li K, Draths KM, Frost JW. Modulation of phosphoenolpyruvate synthase expres-sion increases shikimate pathway product yields in E. coli. Biotechnol Prog2002;18:11418.

    Yi J, Draths KM, Li K, Frost JW. Altered glucose transport and shikimate pathway productyields in E. coli. Biotechnol Prog 2003;19:14509.

    Yoshida N, Ogasawara K. An enantioconvergent route to ()-shikimic acid via apalladium-mediated elimination reaction. Org Lett 2000;2:14613.Florisil C18 system. Anal Bioanal Chem 2003;377:7028.

    Production of shikimic acid1. Introduction2. Production methods2.1. Extraction from plants2.2. Fermentation processes2.2.1. The shikimic acid pathway2.2.2. Use of recombinant and engineered strains

    2.3. Chemical synthesis2.4. Microbial biotransformation

    3. Concluding remarksReferences