CurrPharmDesign2007 DXR Inhibitors

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Current Pharmaceutical Design, 2007, 13, 1161-1177 1161

Targeting the Methyl Erythritol Phosphate (MEP) Pathway for Novel


Antimalarial, Antibacterial and Herbicidal Drug Discovery: Inhibition of
1-Deoxy-D-Xylulose-5-Phosphate Reductoisomerase (DXR) Enzyme

Nidhi Singh1, Gweneal Chevé1, Mitchell A. Avery1,3,4 and Christopher R. McCurdy1,2,4,*

1
Department of Medicinal Chemistry and Laboratory for Applied Drug Design and Synthesis, School of Pharmacy
2
Department of Pharmacology, School of Pharmacy 3Department of Chemistry and Biochemistry 4National Center for
Natural Products Research, University of Mississippi, University, MS 38677-1848, USA

Abstract: The 2-C-methyl-D-erythritol-4-phosphate (MEP) pathway for isoprenoid biosynthesis has come under in-
creased scrutiny as a target for novel antimalarial, antibacterial and herbicidal agents. 1-Deoxy-D-xylulose 5-phosphate
reductoisomerase (DXR) is a key enzyme of the pathway that catalyzes the rearrangement and nicotinamide adenine
dinucleotide phosphate (NADPH)-dependent reduction of 1-deoxy-D-xylulose 5-phosphate (DXP) to MEP. The unique
properties of DXR make it a remarkable and rational target for drug design. First, it is a vital enzyme for synthesis of iso-
prenoids in algae, plants, several eubacteria including the pathogenic bacteria like Bacillus anthracis, Helicobacter pylori,
Yersinia pestis, Mycobacterium tuberculosis and the malarial parasite, Plasmodium falciparum. Second, there are no func-
tional equivalents to DXR in humans, making it an attractive target for therapeutic intervention. Third, DXR appears to be
a valid target and the results from fosmidomycin (1), the only available DXR inhibitor under clinical trials, suggests syn-
ergistic effects with the lincosamide antibiotics, lincomycin and clindamycin. Despite drug design efforts in this area, no
successful drug specifically designed to inhibit DXR has emerged yet. This review summarizes the recent and promising
developments with respect to the current knowledge of the MEP pathway with emphasis on the understanding of the struc-
ture and the catalytic mechanism of the DXR enzyme and the global quest for therapeutically useful inhibitors of DXR.
Key Words: Isoprenoids, MEP pathway, mevalonate, fosmidomycin, FR900098, DXP reductoisomerase, DXR.

INTRODUCTION units, isopentenyl diphosphate (IPP) and its isomer dimethy-


lallyl diphosphate (DMAPP). Since the initial discovery of
Isoprenoids constitute the largest single class of natural
the Acetate/Mevalonate (MVA) pathway in the early 1950’s,
products, [1] with over 35,000 compounds identified to date it was assumed that the isoprenoids are biosynthesized
[2]. They represent a heterogeneous group of biologically
through this route in all organisms (Fig. 2) [14-16]. This
active molecules that constitute a major class of primary and
pathway includes condensation of two molecules of acetyl-
secondary metabolites. Compounds derived from the path-
CoA to form acetoacetyl-CoA, which is then converted to 3-
way carry out biological functions fundamental for the nor-
hydroxy-3-methylglutaryl CoA (HMG-CoA). HMG-CoA in
mal growth and developmental processes in all living organ-
the next step is reduced to MVA, the reaction being cata-
isms (Fig. 1) [3-5]. These include functioning as modulators lyzed by HMG-CoA reductase, a key regulatory enzyme of
of membrane properties (prenyllipids in archaebacteria [6]
this pathway that has been extensively studied [17]. MVA
and sterols in eubacteria and eukaryotes [7]), as carriers for
then undergoes two successive phosphorylation steps to give
electron transport (menaquinone, plastoquinone, phylloqui-
mevalonic-5-monophosphate (MVA-P) that is converted to
none and ubiquinone), [8] as light harvesting and photo-
mevalonic-5-diphosphate (MVA-PP). An adenosine triphos-
protecting pigments (carotenoids, phytol side chain of chlo-
phate (ATP)-dependent decarboxylation of MVA-PP results
rophyll), as regulators of growth and development (steroid in formation of IPP that is subsequently transformed to
hormones, gibberellins, cytokinins, abscisic acid) [9] and in
DMAPP by IPP isomerase. In due time, however, several
signal transduction (prenylated proteins) [10]. Isoprenoids
observations emerged in the literature that were not consis-
also serve as attractants for pollinators and seed dispersers
tent with this pathway. For example, [13C] acetate was not
[11] and as antibiotics, and herbivore repellents as well as
incorporated into ubiquinone of Escherichia coli [18] or into
toxins in plants [12, 13].
pentalenolactone produced by Streptomyces chromofuscus
Despite the diversity of structure and function, all isopre- [19]. It was also found that mevinolin, a potent and highly
noids originate from the common five-carbon (C5) building specific inhibitor of HMG-CoA reductase, that blocks sterol
biosynthesis in the cytosol, did not affect carotenoid biosyn-
thesis in plastids [20]. These results provided strong support
*Address correspondence to this author at the National Center for Natural for the existence of a different biosynthetic pathway for IPP
Products Research, University of Mississippi, University, MS 38677-1848, formation.
USA; Tel: +1-662-915-5882; Fax: +1-662-915-5638;
E-mail: cmccurdy@olemiss.edu

1381-6128/07 $50.00+.00 © 2007 Bentham Science Publishers Ltd.


1162 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

Fig. (1). A schematic representation showing branching of various isoprenic compounds at different steps of the pathway in diverse systems.

More recently, however, 13C-incorporation into various step, is converted into 2-C-methyl-D-erythritol-2,4-cyclodi-
isoprenic compounds in bacteria, algae and plants proved phosphate (MECP) and cytidine monophosphate (CMP) in a
useful for the elucidation of a novel pathway to IPP in these reaction catalyzed by MECP synthase [37]. 1-Hydroxy-2-
organisms [21-26]. With mutants of Escherichia coli defi- methyl-2-(E)-butenyl-4-diphosphate synthase (HMBPP syn-
cient in the enzymes of the triose phosphate pathway, incor- thase) catalyzes the conversion of MECP into HMBPP [38,
poration of 13C-labeled glycerol or pyruvate into the prenyl 39]. Finally, HMBPP is converted into a mixture of IPP and
chain of ubiquinone Q8 revealed that the isoprenic units DMAPP (usually in 5:1/3:1 ratio), [40] the reaction being
were formed in the new metabolic route from pyruvate and catalyzed by HMBPP reductase (also called as IPP/DMAPP
glyceraldehyde-3-phosphate [21]. To distinguish this alterna- synthase), seemingly replicating IPP isomerase activity [41].
tive pathway for the biosynthesis of the isoprenoid building By contrast, IPP isomerase favors an equilibrium ratio of 3:7
units from the MVA pathway, it was named as the non- between IPP and DMAPP [42].
mevalonate route. This pathway is also known as the 1- The MEP pathway is found operative in most eubacteria,
deoxy-D-xylulose-5-phosphate (DXP) pathway, where DXP
[43] algae, [26, 44-46] cyanobacteria and diatoms [47] but is
is the first intermediate; or 2C-methyl-D-erythritol-4-phos-
absent in animals, fungi, archaebacteria and certain bacteria
phate (MEP) pathway since MEP is considered to be the first
like streptococci, staphylococci and lactobacilli [48]. Al-
committed precursor [27, 28].
though higher plants possess both the MEP pathway as well
The MEP pathway is comprised of seven enzymatic steps as the MVA pathway for isoprenoid synthesis, [46, 49, 50]
(Fig. 2). The first step is the condensation of glyceraldehyde- distinct compartmentalization occurs; [51-57] the MVA
3-phosphate and pyruvate to produce DXP [29] in a transke- pathway occurs in the cytosol and mitochondria (e.g. for
tolase-type reaction catalyzed by DXP synthase. DXP is a sterols and ubiquinone) while the MEP pathway, for synthe-
precursor not only for IPP, the common metabolic precursor sis of both IPP and DMAPP, occurs in plastids (e.g. for iso-
of all isoprenoids, but also for thiamine (vitamin B1) [29-32] prene, plastoquinone-9, carotenoids and phytol) [58, 59].
and pyridoxal (vitamin B6), two important cofactors for Data from labeling and inhibitory studies from several labo-
many metabolic enzymes [29, 30, 33]. The next step is the ratories indicated the existence of substantial cross talk be-
conversion of DXP into MEP [34] in a reaction mediated by tween both cellular pathways [60-64]. Even though the
the DXR enzyme in a single step by intramolecular rear- transportation of isoprenoid building units, synthesized in
rangement and reduction, the exact mechanism of which is plastids through the MEP pathway, into cytosol has been
not yet clear. The transformation of MEP to 4-diphospho- reported, the inverse cooperation is known to occur to a rela-
cytidyl-2C-methyl-D-erythritol (CDP-ME) [35] is catalyzed tively lesser extent depending on the physiological state and
in a cytidine triphosphate (CTP)-dependent reaction by the developmental stage of the plant cells. In spite of the
CDP-ME synthase. The adenosine triphosphate (ATP)- evidence suggesting the existence of some cooperation be-
dependent CDP-ME kinase then phosphorylates CDP-ME at tween both IPP producing pathways, lack of one can never
the 2- hydroxy group, forming 4-diphosphocytidyl-2-C- be fully compensated by the other [65]. Nonetheless, the
methyl-D-erythritol-2-phosphate (CDP-ME2P) and adeno- existence of two different pathways for IPP biosynthesis in
sine diphosphate (ADP) [36]. This CDP-ME2P, in the next plants is fascinating and perhaps, is of evolutionary signifi-
MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1163

MVA PATHWAY (CYTOSOL) MEP PATHWAY (PLASTID)

O O O
O
Acetyl CoA Glyceraldehyde-3-P
SCoA COO- H O P O-
+
Pyruvate OH O-
Acetyl CoA Acetyl transferase
CO2 DXP synthase (dxs)

O O OH
O
1-Deoxy-D-xylulose-5-P (DXP)
SCoA O P O-
Acetoacetyl-CoA O OH O-

Fosmidomycin NADPH
HSCoA
NADP+
O HMG-CoA synthase
DXP reductoisomerase (dxr/ispC)
OH O
SCoA
HO O P O- 2-C-methyl-D-erythritol-4-P (MEP)
Hydroxymethylglutaryl-CoA(HMG-CoA) OH OH O-
COOH COSCoA CTP
PPi
HMG-CoA reductase NH2
NADPH CDP-ME synthase (ispD/ygbP)
CoA
NADP+ OH N
Mevanolin O O
O P O P O N O
HO O
OH OH O- O-
Mevalonic acid (MVA)
COOH CH2OH CDP-methyl-D-erythritol(CDP-ME) HO OH
ATP
ATP
Mevalonate kinase ADP
ADP CDP-ME kinase (ispE/ychB) NH2
O
N
HO O- P O O O
O- O- O
N O
Mevalonic-5-monophosphate (MVA-P) O P O P O
COOH CH2O P O- O- O-
OH OH
O
ATP CDP-methyl-D-erythritol-2-P(CDP-ME-2P) HO OH
Mevalonate 5-phosphate
ADP kinase CMP
MECP synthase (ispF/ygbB)
HO
O-
O- O- O O
P O
COOH CH2O P O P O- O P
O-
O O O

Mevalonic-5-diphosphate (MVA-PP) 2-C-methyl-D-erythritol-2,4-diphosphate (MECP)


OH OH

Mevalonate 5-diphosphate HMBPP synthase (ispG/gcpE)


decarboxylase

O- O-
ATP CO2 O P O P O-
ADP + Pi OH O O
NADH
HMBPP reductase (ispH/lytB)
1-Hydroxy-2-methyl-2-(E)-butenyl-4-diphosphate (HMBPP)

Isopentenyl diphosphate O- O- O- O-
(IPP) O P O P O- O P O P O- Dimethylallyl diphosphate
Isopentenyl diphosphate
O O O O (DMAPP)
isomerase

Fig. (2). Biosynthesis of IPP via the MVA and the MEP pathway.
1164 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

Table 1. Diseases and their Causative Organisms in Plants and in Humans that may be Alleviated by Inhibition of DXR.a

Causative Organism Infection/Disease Type of Pathogen

Human Pathogen

Helicobacter pylori Gastritis, peptic ulcer, gastric adenocarcinoma, gastric Bacteria


mucosa-associated lymphoid tissue lymphoma

Plasmodium falciparum Malaria Protozoa

Mycobacterim tuberculosis Tuberculosis Bacteria

Bacillus anthracis Anthrax Bacteria

Bordetella purtussis Whooping cough Bacteria

Neisseria meningitidis Meningitis Bacteria

Fusobacterium nucleatum Periodontitis Bacteria

Porphyromonas gingivalis Chronic adult periodontitis and progressive periodontitis Bacteria

Chlamydia trachomatis Chlamydia, Ttachoma Bacteria

Neisseria gonorrhoeae Gonorrhea Bacteria

Treponema pallidum Syphilis Bacteria

Campylobacter jejuni Gastroenteritis Bacteria

Clostridium botulinum Food poisoning Bacteria

Clostridium tetani Tetanus Bacteria

Corynebacterium diphtheriae Diptheria Bacteria

Escherichia coli Enteric bacteria Bacteria

Francisella tularensis Tularemia Bacteria

Haemophilus influenzae Bacteremia and acute bacterial meningitis Bacteria

Pseudomonas aeruginosa Bacteremia Bacteria

Klebsiella pneumoniae Pneumonia Bacteria

Mycobacterium leprae Leprosy Bacteria

Vibrio cholerae Cholera Bacteria

Yersinia pestis Plague Bacteria

Bacillus anthracis Anthrax Bacteria

Toxoplasma gondii Toxoplasmosis Protozoa

Plasmodium vivax Malaria Protozoa

Shigella dysenteriae Shigellosis Bacteria

Salmonella enteritidis Gastroenteritis Bacteria

Chlamydia psittaci Psittacosis Bacteria

Clostridium difficile Antibiotic induced diarrhea, pseudomembranous colitis Bacteria

Enterococcus faecalis Urinary tract infection Bacteria

Salmonella typhi Typhoid fever Bacteria

Salmonella typhimurium Gastroenteritis Bacteria

Propionibacterium acnes Acne vulgaris Bacteria


MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1165

(Table 1) contd….

Causative Organism Infection/Disease Type of Pathogen

Plant Pathogen

Agrobacterium tumefaciens Crown gall disease Bacteria

Ralstonia solanacearum Southern wilt Bacteria


a
This information is obtained from the SWISSPROT protein sequence data bank (http://us.expasy.org/sprot/; DXR was used as the keyword).

cance to plants providing them with better adaptability to vivo, Mg2+ has been suggested to be the relevant divalent
environmental changes [66]. cation in vivo [73, 74, 76]. The substrate for the reaction
catalyzed by the enzyme is DXP. The first evidence for DXP
Interestingly, it has been found that isoprenoids are syn-
as an isoprenoid precursor was provided by the studies of
thesized exclusively via this pathway in various pathogenic
Arigoni et al. and Broers [45]. Since 1-deoxy-D-xylulose
bacteria like Escherichia coli, Mycobacterium tuberculosis,
(DX) was reported to be used by a wild-type Escherichia
Mycobacterium leprae, Helicobacter pylori, Vibrio cholerae
and Bacillus anthracis and in the apicoplast of the apicom- coli strain in place of DXP, [46] the possibility of using DX
to produce MEP via a DXR catalyzed reaction was also in-
plexan protozoan, P. falciparum; refer to Table 1 [34, 53,
vestigated. No oxidation of NADPH was observed under
67]. In addition, knockouts of DXR in E. coli [34, 41] and
such conditions that led to the conclusion that DXP and not
Bacillus subtilis [68] are lethal. However, it has not yet been
proven, if within the infected host, the DXP/MEP pathway is
the sole provider of de novo synthesized isoprenoids in the
pathogen or that the products and/or the intermediates may
be utilized from the alternative pathway operating in the
host. Sole dependence on the DXP/MEP pathway within the
diseased host for IPP/DMAP will increase the significance of
the target since the theoretical potential of this target to cure
disease hinges on the importance of the target in the infected
host cell. Since the amino acid sequences of the constituent
enzymes of the DXP/MEP pathway are highly conserved
(except HMBPP synthase that contains a large plant-specific
internal domain of unknown function) [69] and have no
known human counterparts, [57] the constituent enzymes of
the DXP/ MEP pathway offer the potential for the develop-
ment of novel antibacterial, antimalarial and herbicidal com-
pounds perhaps, without toxicity to humans.

DXR ENZYME: MECHANISTIC STUDIES


The DXR is the second enzyme in the cascade of the
MEP pathway that has been characterized at the enzymatic
and molecular level in Escherichia coli [34, 69] and its
orthologues from the cyanobacteria, Synechocystis, [70] the
plants Arabidiopsis [71] and Mentha piperata [72] and the
malarial parasite, P. falciparum [67] The structural and bio-
chemical studies conducted on this enzyme show that the
DXRs exist as homodimers (Fig. 3) with molecular weights
between 42-45 kDa. The enzyme catalyzes the rearrange-
ment and NADPH-dependent reduction of DXP to MEP.
Studies on Escherichia coli and Mycobacterium tuberculosis
enzymes [73, 74, 89] have afforded direct evidence for the
reversibility of the reaction catalyzed by both enzymes, with
Fig. (3). The ribbon representation of the physiological homodimer
a reaction equilibrium that is essentially in favor of MEP
of DXR. The upper monomer is colored blue (N-terminal domain;
production.
residues 1–150), pink (connective domain; residues 160-270), and
DXR requires a divalent metal ion for catalysis; Co2+, green (C-terminal domain; residues 312-398). The catalytic loop
Mn and Mg2+ have been shown to activate the enzyme in
2+
(residues 186–216) surrounding the inhibitor-binding site is indi-
an optimum concentration of 1mM, [73] albeit with varying cated in cyan. The lower monomer is colored grey with NADPH (in
kinetic parameters [34]. With other divalent metal ions such stick representation and colored yellow) and fosmidomycin (repre-
as Ca2+, Ni2+, Zn2+, minimal or no activity was observed [34, sented as ball and stick and colored by atom type) cocrystallized
73, 75, 76]. However, due to the abundance of its levels in [82].
1166 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

DX is the substrate for the reaction [69] and that DX needs chemical course of the reduction step in Liriodendro tulipif-
to be converted to DXP before it can be used as a substrate era confirmed these findings [80]. The analysis of the crystal
[34]. Also, the conversion of DXP into MEP by DXR re- structures of DXR from E. coli [81, 82] provides additional
quires the presence of NADPH as a cofactor, in preference to support to the above statement.
NADH [34, 77]. The results from recent studies on the My- A similar reaction has already been detailed for acetohy-
cobacterium tuberculosis DXR suggest that although NADH
droxy acid isomeroreductase [83-87]. Although the reaction
can be utilized as a cofactor in vitro, NADPH is the relevant
catalyzed by DXR and acetohydroxy acid isomeroreductase
cofactor in vivo because of its higher kcat/Km values [73].
are related, their overall amino acid sequences are different.
The reaction catalyzed by the enzyme includes both the Moreover, even though both enzymes have a comparable
rearrangement and the reduction of DXP. The presence of a induced fit mechanism, their X-ray crystal structures differ
rearrangement in the MEP pathway was demonstrated by extensively [86, 88]. Like acetohydroxy acid isomeroreduc-
studies on the incorporation of [4,5-13C2] glucose into ho- tase, the catalysis by the E. coli enzyme has been shown to
panoids and ubiquinones from Methylbacterium fujisa- obey an ordered mechanism in which NADPH and a divalent
waense [43] which was in agreement with another similar cation binds first resulting in a minor conformational change
study in Zymomonas mobilis [23]. The experiments on the before substrate binding that causes more pronounced con-
biosynthesis of diterpenoids of Ginkgo biloba embryos led to formational change prior to the enzymatic reaction [76, 89].
similar conclusions [63]. In the first step, the C3-C4 bond in Notably, the results of product inhibition studies and isotope
DXP is broken with concomitant formation of a new bond effect experiments on the Mycobacterium tuberculosis DXR
between C2 and C4 of the substrate to produce a putative suggested a steady-state random mechanism [73]. However,
aldehyde intermediate, 2-C-methyl-D-erythrose 4-phosphate, a preferred order of substrate addition, where NADPH pre-
which in the next step is reduced by NADPH (Fig. 4). cedes DXP, could not be ruled out.
The stereochemistry of the reaction catalyzed by DXR Nevertheless, due to the similarities in the overall reac-
with the recombinant enzymes from Synechocystis [73], E. tions catalyzed by DXR and the acetohydroxy acid iso-
coli [70], and Mycobacterium tuberculosis, [78] shows that meroreductase, it was generally accepted that their mecha-
the C-1 proS hydrogen of MEP is derived from H-3 of DXP nism was also analogues. However, several other studies
and the hydride transferred from C-4 of NADPH is delivered suggested that the aldehyde intermediate could be formed by
to the Re face of the intermediate aldehyde (Fig. 5) [79]. a retro-aldol/aldol mechanism [90]. The proposed mecha-
Based on these results, the enzyme has been classified as a nism for both the reactions is shown in Fig. (6).
class B dehydrogenase [70, 78]. Investigation of the stereo-

OH 1 OH
OH
5 Mg2+ 5 NADPH
2 4 O 4 HO
1 OPO32-
3 OPO32- 2 OPO32-
3
O OH OH OH

DXP 2-C-Methyl-D-erythrose-4-phosphate MEP

Fig. (4). The reaction catalyzed by DXR.

HA HA
N N N HA
HB HB
O O O
H2N H2N H2N

O CH3 HO HO
CH3 CH3
O
OH HO OH
HO H3 H
H3
OH H3
OPO32- OPO32-
Re
OPO32-

DXP MEP

Fig. (5). Outline of the stereochemical features of the DXR reaction. (Copyright obtained from Ref. [79]).
MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1167

A B

H H
O O O O
O P O- O P O-
O O
O- O-
OH O
DXP DXP H
-ketol
Retro-aldolization
rearrangement

H
O O O O
HO P O- - O P O-
O O
O- Aldolization O-
OH O

H H
O O NADPH O O
O P O- HO P O-
O O
O- O-
OH OH
MEP

Fig. (6). The stereochemistry of the proposed mechanism for the rearrangement of DXP to MEP; (A) -ketol rearrangement, (B) retro-
aldol/aldol reaction. The shaded circle represents a Mg2+ ion.

As of today, no conclusive evidence that rules out one of quently, the catalytic loop region was only partially ordered
these pathways has been found, but it is certain that the in- in this structure. No structural rearrangement within the en-
termediate(s) remain tightly bound to DXR prior to the zyme was observed assuming that complete ordering of the
NADPH mediated reduction. After reduction, it is suggested loop region requires substrate as well as cofactor binding
that NADP+ is released before the release of MEP [91]. within the binding pocket. The active site consists of phos-
phate and divalent metal binding motifs separated by a hy-
STRUCTURAL STUDIES drophobic region. The phosphonate moiety of fosmidomycin
binds to the site defined by Ser186, Gly187, Ser222,
To date, several crystal structures of DXR from E. coli
Met225, Asn227, Lys228 and Ile250 (Fig. 7). The N-formyl
[81, 82, 92-94] and one from Zymomonas mobilis [95] have
hydroxylamine moiety in fosmidomycin engages the metal
been reported by different research groups. The first crystal
by a bidentate chelation. This structure is believed to mimic
structure of the E. coli DXR published, was an apo-enzyme,
substrate binding of the C2 carbonyl oxygen and the C3 hy-
which revealed the arrangement of the three domains: an N-
terminal domain that binds NADPH; a “connective” domain droxyl group in DXP. These two functionalities along with
several acidic residues (Asp150, Glu152 and Glu231) hold
that contains the active site; and a C-terminal domain also
the metal ion in place forming an octahedral coordination
involved in cofactor binding, to form an overall V-shaped
sphere [92].
molecule with an apparent flexibility observed in the loop
region. The unoccupied site is filled with water molecules The structure of Zymomonas mobilis DXR was solved in
[81]. The second enzyme structure, in complex with NADPH the apo-form and as a binary complex with NADPH. Com-
and a sulfate ion, was also obtained from E. coli. This struc- parison with the E. coli DXR crystal structures revealed that
ture provided insight into the cofactor binding that stabilized except for differences in the residues interacting with the
the protein molecule and exhibited a more ordered flexible adenine ring of NADPH, the two enzymes shared significant
loop region. The sulfate ion was found positioned within the structural homology and highly conserved active site archi-
putative phosphate-binding pocket in the DXR protein [82]. tecture [95]. Crystallographic structures of two biphospho-
The first DXR-ligand complex structure with Mn2+ and fos- nate inhibitors complexed to DXR were published but nei-
midomycin bound was reported along with two other struc- ther NADPH nor a metal ion is present in either structures.
tures of DXR: the apo-enzyme and with Mn2+. Unfortu- Instead, a sulfate ion is observed which fits to the substrate
nately, the NADPH binding site was blocked by the C- phosphate pocket in a similar fashion as seen in one of the
terminus of the neighboring monomer in the crystal. Conse- previous crystal structures [93]. However, phosphonate
1168 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

Fig. (7). A view of the binding mode of fosmidomycin within the active site of E. coli DXR: Fosmidomycin is represented as a ball-and-stick
model colored by atom-type while NADPH as a stick model is indicated in yellow [94].

moieties of these compounds bind to the metal ion instead of nificantly reduced kcat but with minor changes in Km for both
the phosphate-binding site that is utilized by fosmidomycin. DXP and NADPH. Mutations of three conserved His resi-
Besides, the loop region forms a different conformation dues to Gln, recognized by comparing DXRs from E. coli
when it is located over the bisphosphonate inhibitors rather with those from other bacteria and plants, to identify the
than that when it is over fosmidomycin [91]. Recently, the residue crucial for catalytic activity of the enzyme were also
ternary structures of the DXR-NADPH-fosmidomycin, the made. The His153Gln mutation resulted in a 10-fold de-
DXR-NADPH-substrate and the selenomethionine-labeled crease in kcat while demonstrating a 3.5-fold increase in
DXR-NADPH-fosmidomycin complex in a tight binding KmDXP, with no effect on the KmNADPH. Although surrounded
closed enzyme conformation were solved (Fig. 8) [94]. The by conserved residues, this residue is not in close proximity
DXR-NADPH-fosmidomycin complex revealed a large con- of the substrate. However, it has been implicated to have a
formational change in the “connective” domain to give a structural role [82]. The mutation of either His257 or His209
relatively hydrophobic binding pocket that tightly binds the to a Gln, resulted in increased Km for both DXP and
inhibitor. The narrow size of the active site in the DXR- NADPH but drastically reduced kcat, suggesting that these
fosmidomycin-NADPH complex explains why larger ana- residues have no enzymatic role in vivo. Recently, site di-
logues of DXP or fosmidomycin have little or no inhibitory rected mutagenesis of highly conserved Trp204 in the
activity against the enzyme [94]. The absence of an expected Synechocystis DXR (Trp212 in E. coli DXR) revealed the
metal ion from the structure was attributed to the acidic crys- important role of this residue in substrate/inhibitor binding.
tallization conditions (pH = 5) used at which the affinity of The authors have suggested that it may also have a role in
DXR for metal is low. Furthermore, this structure had a fully discriminating substrates for DXR [96]. These findings have
ordered loop conformation that closed upon the active site been well supported by those available from published DXR
making interactions with the ligand. crystal structures.
Site-directed mutagenesis studies of the recombinant E.
INHIBITORS OF DXR: THE STATE-OF-THE-ART
coli protein revealed the importance of conserved histidine
and glutamate residues for activity [69]. Glu231 was found Fosmidomycin (also known as FR-31564 or 3-(N-formyl-
to be the most important residue for catalysis, with a sug- N-hydroxyamino) propylphosphonic acid; Fig. 9) [28, 50]
gested role in formation of an octahedral complex with the was identified as a natural antibiotic from Streptomyces lav-
metal ion along with other strictly conserved residues. Muta- endulae in the late 1970s [97]. This compound was also
tion of this residue to a Lys resulted in an enzyme with sig- found to exert herbicidal activity [98, 99]. Studies on the
MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1169

Fig. (8). A representation of the E. coli DXR in a tight binding conformation with the inhibitor (PDB ID: 1Q0L). The NADPH molecule
(stick representation) and fosmidomycin (depicted in ball and stick) has been shown. Helices and sheets are represented as red cylinders and
yellow arrows, respectively [94].

inhibition of the biosynthesis of menaquinone and caro- [92]. Recent studies have defined fosmidomycin as a mixed-
tenoids in Micrococcus luteus attributed this effect to the type (both competitive and non-competitive) inhibitor of the
inhibition of isoprenoid biosynthesis [100]. Kuzuyama et al. recombinant enzyme of E. coli (Ki = 38 nM) [38]. It has also
performed a database search to identify specific inhibitors of been reported to be a slow, tight-binding competitive inhibi-
the non-mevalonate pathway [28]. This was accomplished by tor of DXR [94]. Its activity as a competitive inhibitor of the
identifying compounds that were active against E. coli and Zymomonas mobilis DXR was tested to a Ki of 0.6 μM [74].
Bacillus subtilis, which possess the non-mevalonate path- The antibiotic also inhibits the enzyme from higher plants
way, but was inactive against Staphylococcus aureus, which [50, 103]. Fosmidomycin and its acetyl derivative, FR-
has the classical acetate/mevalonate pathway [101]. This 900098 (2, Fig. 9) inhibit the recombinant DXR from P. fal-
search led to the identification of fosmidomycin as a putative ciparum in a dose-dependent manner [67]. Both compounds
pathway-specific inhibitor that was active against most are effective antimalarial agents, since mice infected with
Gram-negative and some Gram-positive bacteria. Plasmodium vinckei became free of parasites upon treatment
[50, 103]. Fosmidomycin also cured uncomplicated P. falci-
Further studies confirmed that fosmidomycin was a re-
parum malaria in humans [104, 105]. However, a high rate
markably non-toxic, potent, specific inhibitor of DXR [50,
of recrudescence was observed with treatment regimens of
94]. In an in vitro assay with the purified recombinant E. coli
less than 4 days precluding the use of fosmidomycin as a
DXR, it was shown that fosmidomycin inhibited the DXR
activity in a dose-dependent manner with an IC50 value of monotherapeutic agent. This led to a combination of fosmi-
domycin with clindamycin that has been shown to be well
8.2 nM. Also, in an E. coli growth assay, the inhibitory effect
tolerated and highly efficacious in the treatment of uncom-
of fosmidomycin was overcome by the addition of 0.025%
plicated malaria and such therapies would seem to be more
of 2-C-methylerythritol, the free alcohol of MEP, to the me-
promising [106]. Clinical studies to assess the efficacy and
dium. Moreover, in the presence of the above supplementing
agent, fosmidomycin had no effect on the growth of an E.
coli DXR knockout mutant. These results led to the conclu- OH O OH O
sion that fosmidomycin inhibited DXR activity [38, 50]. A H N P O- N P O-
similar conclusion was reached from studies of isoprenoid O- O-
biosynthesis in barley (Hordeum vulgare L.), tomato (Ly- O O
2
copersicon esculentum L.), and duckweed (Lemna sp.) [102] 1

The crystal structure of E. coli DXR-metal-fosmidomy- Fig. (9). Inhibitors of the DXR enzyme, Fosmidomycin (1) and
cin complex revealed a substrate-like binding of the inhibitor FR900098 (2).
1170 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

safety of the combination of fosmidomycin with other anti- none (8) ring have also been synthesized although their bio-
malarial drugs have already been initiated in humans [107- logical evaluation was reported in a model plant cell, Ca-
109]. Consequently, both fosmidomycin and FR900098 af- tharanthus roseus, in terms of its cell growth and the ability
forded valuable leads for further development towards clini- to accumulate a monoterpenoid indole alkaloid, ajmalicine,
cal use. Fig. (9) [117]. It was found that fosmidomycin, as a control,
resulted in no effect on Catharanthus roseus cell growth but
Additionally, these compounds have low toxicity and
inhibited production of ajmalicine, which has been proven to
their half-life in serum is very short [110]. Moreover, both
originate from the MEP pathway [118], in a dose-dependent
the compounds have poor oral bioavailability, with a resorp-
manner with an IC50 of 10 M [119]. Of the twelve phos-
tion rate in the range of 20-40% [111] which may be attrib-
phonic esters and acids studied, none affected Catharanthus
uted to low lipophilicity resulting from ionization of polar
phosphonate group at physiological pH values that restricts roseus cell growth, but four of them (including 6, 7 and 8)
showed a significant inhibition (45-85% at 125 M) of ajma-
their usage as single therapeutic agent. Since FR900098 pos-
licine accumulation.
sessed an approximately 2-fold greater antimalarial activity,
attempts were made to improve its efficacy upon oral ad- Recently, the synthesis of substituted 3-[2-(diethoxy-
ministration by synthesizing prodrugs [112-114]. These pro- phosphoryl) propyl] oxazolo [4, 5 - b] pyridine - 2(3H) - ones by
drugs were obtained by transforming the charged phospho- functionalization at 6-position with various substituents like
nate moiety to yield phosphodiaryl (3), [112] acyloxyalkyl aryl, vinyl, carbonyl chains as fosmidomycin analogues have
esters (4) [113] and alkoxycarbonyloxyethyl (5) [114], with been reported by the same group [120]. Some of the reported
increased oral antimalarial efficacy in mice infected with the derivatives (including 9; Fig. 10) were found to be more
rodent malaria parasite, Plasmodium vinckei (Fig. 10). powerful inhibitors of ajmalicine production than fosmido-
mycin. The inhibitory effect of these analogues and those
Structural modifications directed towards the synthesis of
hydroxyurea [115] and carboxylic acid [116] analogues of synthesized previously, against DXR has not been investi-
gated which is necessary to confirm that these compounds
fosmidomycin and FR900098 have also been reported, how-
are, in fact, DXR inhibitors. It has been suggested that inhib-
ever no enzyme inhibition studies of these compounds have
iting the MEP pathway of plants could be useful in the
been reported to date. Fosmidomycin analogues containing a
search of novel antibiotics, antimalarials and herbicides
benzoxazolone (6), benzoxazolothione (7) or oxazolopyridi-
[121]. If positive results are obtained after further testing of

OH O 3 R= O

N P OR
O
OR
O 4 R=
O
O
5 R=
O O

Cl Cl Br

N
O O O
O N P OC2H5 O N P OC2H5 O N P OC2H5
OC2H5 OC2H5 OC2H5
O S O
6 7 8

N OH O OH O
O N P OH N P O-
O O
O N P OC2H5 OH O-
O O
OC2H5
O
9 10 11

Fig. (10). Structural analogues of fosmidomycin as DXR inhibitors.


MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1171

these compounds for their specific activity against DXR, it P. falciparum strains. Of the several active analogues that
will only corroborate the above statement and may provide were reported, compound 12 (Fig. 11) was found to be 3
interesting starting points for the development of antibacte- times more potent than FR900098 in inhibiting the growth of
rial as well as antimalarial agents. P. falciparum. Moreover, the authors have suggested that the
In a study by Woo et al., several analogues of fosmido- presence of the aromatic ring would improve the ability of
these compounds to cross the parasitic cell membrane.
mycin were synthesized and evaluated against recombinant
DXR from Synechocystis sp. PCC6803 [122]. In this study The synthesis of conformationally restricted fosmidomy-
the polar phosphonate moiety was modified to a phosphate, cin analogues has also been reported by incorporating the C-
carboxylate and sulfamate while the hydroxamic moiety was 1 and C-2 atoms in a three-membered ring [126]. The (1R,
substituted with N-formyl, N-methyl and N-acetyl, N-methyl 2S)-trans-analogue 13 showed an important inhibitory activ-
or converted to the reversed hydroxamate. It was found that ity toward E. coli DXR, whereas 14 appeared to be as effec-
although the phosphate analogue of FR900098 (10) was de- tive as fosmidomycin in growth inhibition of Dd2 and 3D7
termined to be more potent mixed inhibitor of Synechocystis strains of P. falciparum. Conversely, synthesis of its -
DXR (Ki = 2nM) than fosmidomycin, due to the phosphate phenyl-substituted analogue led only to the cis-isomer 15,
moiety in its structure, it was prone to cleavage by phopha- which appeared to have no significant activity toward E. coli
tases that inactivated this compound. The other two changes DXR.
led to large increase in the Ki values. Similarly, the hydrox-
In addition to the aforementioned inhibitors, several
amic group was suggested to be highly sensitive to changes
structural analogues of DXP have shown inhibitory activity
with even a minor modification leading to decreased inhibi-
against DXR. Two DXP analogues, each lacking a hydroxyl
tion.
group at C-3 (16) and C-4 (17) respectively, were designed
In another study by Mercklé et al., a fragment-based ap- and synthesized as substrates for DXR. However, they were
proach was employed in an attempt to understand the inhibi- found to be reversible, mixed-type inhibitors of the E. coli
tion of DXR by fosmidomycin using a kinetic assay based on DXR with Ki values of 800 M and 120 M respectively
consumption of NADPH and fosmidomycin [125]. A series (Fig. 12) [127]. This study established the importance of
of fosmidomycin fragments and cyclic analogues (11) were hydroxyl groups in enzyme catalysis. A similar conclusion
synthesized and examined. While the fragments showed was reached in another study, in which the corresponding 3-
weak inhibition mostly in the mM range the cyclic analogues and 4-fluoro analogues (18, 19) of DXP acted as noncom-
showed high-M binding at best. petitive inhibitors with Ki values of 444 M and 733M
respectively [128].
Modification of the propylene spacer between the hy-
droxamic moiety and the phophonate moiety has been Phaosiri and Proteau reported the synthesis of six DXP
scarcely reported in the literature. A previous attempt to analogues and their evaluation against recombinant Synecho-
shorten the three-carbon spacer by Hemmi et al. only com- cystis PCC6803 DXP reductoisomerase [129]. Five of these
promised antibacterial activity, which shows that the length acted as relatively weak competitive inhibitors when com-
of this carbon chain is critical for optimal activity [123]. pared to fosmidomycin. In accordance with the previous
More recently, Haemers et al. reported the synthesis of - report, the absence of the C3 hydroxyl (16, Ki = 150 M)
aryl-substituted fosmidomycin analogues [124]. In this and C4 hydroxyl (17, Ki = 30 M), the Km DXP for both of
study, the influence of aromatic substituents in the -position which was found to be approximately six-fold lower than Ki,
of the phosphonate moiety was investigated. The activity of supports the idea that the hydroxyl at these positions are not
these analogues was evaluated against E. coli DXR and two critical for binding but essential for catalysis. Moreover, the

OH O OH H O OH H
H N P OH R N P OH N
OH OH
O O H O HO P O
OH
12 Cl 13 R=H
15
14 R=CH3
Cl

Compound 12 13 14 15

E. coli 0.059 0.313 0.050 > 30

IC50 (μ M) P. falciparum Dd2 0.028 2.0 0.48 --

P. falciparum 3D7 0.090 2.1 0.32 --

Fig. (11). Conformationally restricted fosmidomycin analogues.


1172 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

Ki for compound 20 and the Km for DXP were found to be by analogue 27 demonstrates that a carbonyl group is not
similar, indicating the importance of the stereochemistry of required for inhibition. Interestingly, amine 28 (Fig. 13) did
both hydroxyl moieties for catalysis. The inhibitory activity not inhibit the enzyme. Since Mg2+ forms complexes with
of compound 21 was explained to be due to either the incor- amines, the lack of inhibition may reflect that the amino
rect alignment of the compound within the active site or due group in 28 is protonated. Also, the hydroxylamine analogue
to a steric interaction upon rearrangement. In fact, a recently 29 (Fig. 13) did not possess inhibitory activity that was at-
reported mutation study, using this compound, confirmed tributed to the extremely confined volume of the active site,
that it was steric interaction with an amino acid side chain which is observed after the enzyme binds its substrates. The
that prevents DXR from using the compound as a substrate chain of analogue 29 was one atom longer than that of the
[96]. It was suggested that the lack of turnover of compound potent hydroxamic acid isostere of fosmidomycin, stressing
22 (Fig. 12) by the enzyme was perhaps, due to lower elec- the importance of the three-carbon chain length separating
trophilicity at the carbonyl moiety when compared to the the hydroxamic and the phophonate group.
substrate DXP.
The synthesis of two novel related inhibitors of DXR, the
Synthesis and evaluation of fluoro analogues of DXP 4-(Hydroxyamino)-4-oxobutylphosphonic acid, 30 (Fig. 14)
[130] (23, 24) as modest inhibitors with IC50 values of 2.0 and its methylated analogue, 31 (Fig. 14) has been reported
and 3.4 mM respectively, and 6.2 mM for compound 21, also by Kuntz et al. [132]. The two inhibitors were characterized
revealed the importance of steric interactions within the ac- by the same features, a negatively charged phosphonate moi-
tive site substantiating the findings of Phaosiri et al. ety and a chelating hydroxamate group linked by a chain of
the same length as that found between the functional groups
More recently, the same group reported the synthesis of
of fosmidomycin, but in a different arrangement. Like fos-
six DXP analogues of which four compounds (22; Fig. 12
midomycin, the hydroxamic acids 30 and 31 were also com-
and 25-27; Fig. 12) inhibited the E. coli DXR with IC50 val-
ues ranging from 0.25 to 1.0 mM. Their activity was attrib- petitive slow-binding inhibitors with the Ki values of both
compounds against DXP as 169 nM for 30 and 54 nM for 31.
uted to their size that approximated that of the substrate,
DXP, and their ability to coordinate with the active site diva- In addition to targeting the enzyme active site, ap-
lent metal ion [131]. Although none of the DXP analogues proaches focused on developing inhibitors to block the
were as potent as fosmidomycin, inhibition by 22 and 25-27 NADPH binding site of the enzyme have also been investi-
suggested that negatively charged or neutral donor atoms are gated. Several novel adenosine derivatives that displayed
accepted as chelators of the active site Mg2+ ion. Inhibition moderate but significant activity against the P. falciparum

O OH O F O
P O- P O- P O-
O O O
O- O- O-
O OH O O OH

16 17 18

OH O OH O OH O
P O- P O- P O-
O O O
O- O- O-
O F O OH O OH

19 20 21

OH O OH O OH O
H2N P O- F3C P O- HF2C P O-
O O
O- O- O-
O OH O OH O OH

22 23 24

Compound 16 17 18 19 20 21 22 23 24

E. coli 800 120 -- -- -- 6200 253 2000 3400


IC50 (μ M)
Synechocystis PCC6803 150 30 444 733 180 630 90 -- --

Fig. (12). Analogues of substrate, DXP as DXR inhibitors.


MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1173

OH O OH O OH OH O
HO P O- O P O- P O-
O O O
O- O- O-
O OH O OH O OH

25 26 27

NH2 OH O OH OH O
P O- HN P O-
O O
O- O-
O OH O OH

28 29

Compound 25 26 27 28 29

IC50 (μM) - E. coli 551 1024 310 > 5000 > 5000

Fig. (13). Substrate analogues as DXR inhibitors.

strain Dd2 in the low-micromolar range were reported for compounds while the test set consisted of 16 compounds. A
DXR inhibition by Herforth et al. However, the binding to statistically significant model with the regression coefficient,
the NADPH binding site was not confirmed in the study r2 = 0.86 and predictive power, q2 = 0.46 was obtained.
[133]. Also, the fact that the E. coli DXR was utilized for
Recently, a 3D model of DXR enzyme from P. falcipa-
DXR inhibition studies when the in vivo target would be the
rum has also been determined by means of comparative
P. falciparum DXR further complicates the issue. Addition- modeling by Singh et al. [136]. In this study, a structure of
ally, application of a high-throughput screen to DXR, which
the enzyme-inhibitor complex was also modeled by docking
is based on the ability of a compound to compete with a pep-
fosmidomycin into the putative active site. The proposed
tide “surrogate ligand”, at either or both DXP and NADPH
model helped to identify several critical ligand-receptor con-
binding sites of the E. coli DXR enzyme, has also been ex-
tact points in the binding site that might serve as the key mo-
ploited. A library of 32,000 compounds was screened from
lecular determinants of ligand recognition by the DXR en-
which 89 potent and specific inhibitors were registered as zyme. The authors suggest that the model may be used as a
hits. However, the structure-activity data for these com-
significant tool to enhance the understanding of interactions
pounds have not been reported [134].
of DXR inhibitors with the protein at the atomic level. Such
modeling studies may prove useful in the early design and
OH O OH O development of inhibitors by either de novo drug design or
HN P O- N P O- virtual screening of large chemical databases leading to de-
O O velopment of new agents against the enzyme.
O- O-
O O
30
CONCLUSION
31
Despite substantial progress, the quest for therapeutically
Fig. (14). Inhibitors of DXR. significant inhibitors of DXR has not yet led to the develop-
ment of drugs that are clinically useful. Several inhibitors of
Molecular modeling techniques have been employed to DXR have been identified, with fosmidomycin and its acetyl
establish a detailed and extensive structure-activity relation- analogue being the most potent to date. Although these com-
ship for forty-three fosmidomycin analogues, tested as in- pounds exhibit poor pharmacological properties and proba-
hibitors of E. coli DXR in order to predict affinity data for bly do not have the potential to become drugs, they provide a
new inhibitors [135]. The adaptations of fields for molecular reference for the evaluation of DXR inhibition in cell lines.
comparison (AFMoC) approach, a 3D-QSAR technique that Studies on analogues of fosmidomycin suggest that it will be
incorporated information about the interaction properties of difficult to improve upon the inhibition qualities of fosmi-
the surrounding protein available from several published domycin. Although several X-ray crystal structures of DXR
DXR X-ray structures, was implemented. This technique have been published, these structures still leave the active
was shown to overcome the drawbacks of field-based meth- fosmidomycin conformation and the detailed reaction
ods that relies solely on ligand-based information and makes mechanism undetermined. The evaluation of DXR inhibition
no use of available information about the interaction proper- by the fosmidomycin analogues should aid in defining the
ties of the surrounding protein from the existing crystal structural requirements for the design of potent DXR inhibi-
structure. The binding affinities of the inhibitors spanned tors in conjunction with the X-ray structural data and avail-
over 4 logarithmic units. The training set consisted of 27 able SAR studies. Efforts towards understanding and eluci-
1174 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

dating the mechanism of the transformation of DXP to MEP [6] De Rosa M, Gambacorta A, Gliozzi A. Structure, biosynthesis, and
catalyzed by the enzyme will no doubt improve efforts to- physicochemical properties of archaebacterial lipids. Microbiol
Rev 1986; 50: 70-80.
wards this goal. However, rational design of inhibitors based [7] Mills JT, Furlong ST, Dawidowicz EA. Plasma membrane bio-
upon the X-ray structure of target enzyme has the prospects genesis in eukaryotic cells: Translocation of newly synthesized
of yielding drugs against the enzyme. Biochemical ap- lipid. Proc Natl Acad Sci USA 1984; 81: 1385-8.
proaches, together with computational approaches, should [8] Hind G, Olson JM. Electron transport pathways in photosynthesis.
Ann Rev of Plant Physiol 1968; 19: 249-82.
provide the required thrust to identify many compounds, [9] Rademacher W. Growth retardants: Effects on gibberellin biosyn-
which may become potent drugs in the future. While much thesis and other metabolic pathways. Ann Rev of Plant Physiol and
remains to be learned, considerable progress has been made Plant Mol Biol 2000; 51: 501-31.
in recent years and the prospects of obtaining clinically ef- [10] Fenton JW, Jeske WP, Catalfamo JL, Brezniak DV, Moon DG,
fective DXR inhibitors seems, for the first time, tangible. Shen GX. Statin drugs and dietary isoprenoids downregulate pro-
tein prenylation in signal transduction and are antithrombotic and
prothrombolytic agents. Biochem (Moscow) 2002; 67: 85-91.
ACKNOWLEDGEMENTS Translated from Biokhimiya 2002; 67: 99-106.
[11] Bramley PM. Isoprenoid metabolism. In: Dey PM, Harborne JB
The authors thank Dr. Marcelo J. Nieto for critical read- Eds, Plant Biochemistry. London, Academic Press 1997; 417-37.
ing of the manuscript and his insightful suggestions. [12] Duke O, Dayan FE, Romagni JG, Rimando AM. Natural products
as sources of herbicides: current status and future trends. Weed Res
ABBREVIATIONS 2000; 40: 99-111.
[13] Sacchettini JC, Poulter CD. Creating isoprenoid diversity. Science
MEP = 2-C-methyl-D-erythritol 4-phosphate 1997; 277: 1788-9.
[14] Banthorpe DV, Charlwood BV, Francis MJ. The biosynthesis of
DXR = 1-deoxy-D-xylulose-5-phosphate monoterpenes. Chem Rev 1972; 72: 115-55.
reductoisomerase [15] Beytia ED, Porter JW. Biochemistry of polyisoprenoid biosynthe-
sis. Annu Rev Biochem 1976; 45: 113-42.
DX = 1-deoxy-D-xylulose-5-phosphate [16] Amdur BH, Rilling H, Bloch K. The enzymatic conversion of
mevalonic acid to squalene. J Am Chem Soc 1957; 79: 2646-7.
NADPH = Nicotinamide adenine dinucleotide [17] Rodwell VW, Beach MJ, Bischoff KM, Bochar DA, Darnay BG,
phosphate Friesen JA, et al. 3-Hydroxy-3-methylglutaryl-CoA reductase.
Methods Enzymol 2000; 324: 259–80.
IPP = Isopentenyl diphosphate [18] Zhou D, White RH. Early steps of isoprenoid synthesis. Biochem J
DMAPP = Dimethylallyl diphosphate 1991; 273: 627b-34.
[19] (a) Cane DE, Rossi T. The biosynthsis of pentalenolactone. Tetra-
MVA = Mevalonate hedron Lett 1979; 20: 3639-42. (b) Cane DE, Rossi T, Tillman A.
Stereochemical studies of isoprenoid biosynthesis. J Am Chem Soc
HMG-CoA = 3-hydroxy-3-methylglutaryl Coenzyme A 1981; 103: 1838-43.
[20] (a) Bach TJ, Lichtenthaler HK. Mevinolin, a highly specific inhibi-
ATP = Adenosine triphosphate tor of microsoma 3-hydroxy-3-methyl-glutarylcoenzyme A reduc-
tase of radish plants. Z Naturforsch 1982; 37C: 46-50 (b) Bach TJ,
DXP = 1-deoxy-D-xylulose-5-phosphate Lichtenthaler HK. Inhibition of mevalonate biosynthesis and of
CTP = Cytidine triphosphate plant growth by the fungal metabolite mevinolin. In: Wintermanns
JFGM, Kuiper PJC Eds, Biochemistry and metabolism of plant lip-
CDP-ME = 4-diphosphocytidyl-2C-methyl-D- ids. Amsterdam: Elsevier Biochemical Press 1982; 515-21. (c)
erythritol Bach TJ, Lichtenthaler HK. Inhibition by mevinolin of plant
growth, sterol formation and pigment accumulation. Physiol. Plant
CDP-ME2P = 4-diphosphocytidyl-2-C-methyl-D- 1983; 59: 50-60. (d) Schindler S, Bach TJ, Lichtenthaler HK. Dif-
erythritol-2-phosphate ferential inhibition by mevinolin of prenyllipid accumulation in
radish seedlings. Z Naturforsch 1985; 40C: 208-14.
ADP = Adenosine diphosphate [21] Rohmer M, Knani M, Simonin P, Sutter B, Sahm H. Isoprenoid
biosynthesis in bacteria: a novel pathway for the early steps leading
MECP = 2C-methyl-D-erythritol-2,4- to isopentenyl diphosphate. Biochem J 1993; 295: 517-24.
cyclodiphosphate [22] Flesch G, Rohmer M. Prokaryotic hopanoids: the biosynthesis of
the bacteriohopane skeleton. Formation of isoprenic units from two
CMP = Cytidine monophosphate distinct acetate pools and a novel type of carbon/carbon linkage be-
tween a triterpene and D-ribose. Eur J Biochem 1988; 175: 405-11.
REFERENCES [23] Rohmer M, Seemann M, Horbach S, Bringer-Meyer S, Sahm H.
Glyceraldehyde 3-phosphate and pyruvate as precursors of iso-
References 137-139 are related articles recently published in prenic units in an alternative non-mevalonate pathway for terpenoid
Current Pharmaceutical Design. biosynthesis. J Am Chem Soc 1996; 118: 2564-6.
[24] Eisenreich W, Menhard B, Hylands PJ, Zenk MH, Bacher A. Stud-
[1] Connolly JD, Hill RA. In: Dictionary of terpenoids. New York, ies on the biosynthesis of taxol: the taxane carbon skeleton is not of
Chapman and Hall 2002; 2156. mevalonoid origin. Proc Natl Acad Sci USA 1996; 93: 6431-6.
[2] Rohdich F, Bacher A, Eisenreich W. Perspectives in anti-infective [25] Schwender J, Seemann M, Lichtenthaler HK, Rohmer M. Biosyn-
drug design. The late steps in the biosynthesis of the universal ter- thesis of isoprenoids (carotenoids, sterols, prenyl side-chains of
penoid precursors, isopentenyl diphosphate and dimethylallyl chlorophylls and plastoquinone) via a novel pyruvate/gly-
diphosphate. Bioorg Chem 2004; 32: 292-308. ceraldehyde 3-phosphate non-mevalonate pathway in the green
[3] Chappell J. Biochemistry and molecular biology of the isoprenoid alga Scenedesmus obliquus. Biochem J 1996; 316: 73-80.
biosynthetic pathway in plants. Ann Rev Plant Physiol Plant Mol [26] Lichtenthaler HK, Schwender J, Disch A, Rohmer M. Biosynthesis
Biol 1995; 46: 521-47. of isoprenoids in higher plant chloroplasts proceeds via a me-
[4] McGarvey DJ, Croteau R. Terpenoid metabolism. Plant Cell 1995; valonate-independent pathway. FEBS Lett 1997; 400: 271-4.
7: 1015-26. [27] Rodríguez-Concepción M, Boronat A. Elucidation of the meth-
[5] Croteau R, Kutchan TM, Lewis NG. In: Buchanan B, Gruissem W, ylerythritol phosphate pathway for isoprenoid biosynthesis in bac-
Jones R Eds, Biochemistry and molecular biology of plants. teria and plastids. A metabolic milestone achieved through genom-
Rockville, MD, USA, ASPB 2000; 1250-68. ics. Plant Physiol 2002; 130: 1079-89.
MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1175

[28] Kuzuyama T, Shimizu T, Takahashi S, Seto H. Fosmidomycin, a [46] Schwender J, Zeidler J, Groner R, Muller C, Focke M, Braun S, et
specific inhibitor of 1-deoxy-D-xylulose 5-phosphate reducto- al. Incorporation of 1-deoxy-D-xylulose into isoprene and phytol
isomerase in the non-mevalonate pathway for terpenoid biosynthe- by higher plants and algae. FEBS Lett 1997; 414: 129-34.
sis. Tetrahedron Lett 1998; 39: 7913-6. [47] Cvejic JH, Rohmer M. CO2 as main carbon source for isoprenoid
[29] Lois LM, Campos N, Putra SR, Danielsen K, Rohmer M, Boronat biosynthesis via the mevalonate-independent methylerythritol 4-
A. Cloning and characterization of a gene from E. coli encoding a phosphate route in the marine diatoms Phaeodactylum tricornutum
transketolase-like enzyme that catalyzes the synthesis of D-1- and Nitzschia ovalis. Phytochemistry 2000; 53: 21-8.
deoxyxylulose 5-phosphate, a common precursor for isoprenoid, [48] Eberl M, Hintz M, Reihenberg Aistiy, Kollas AK, Weisner J,
thiamin, and pyridoxol biosynthesis. Proc Natl Acad Sci USA Jomaa H. Microbial isoprenoid biosynthesis and human gamma
1998; 95: 2105-10. delta T cell activation. FEBS Lett 2003; 544: 4-10.
[30] Wungsintaweekul J, Herz S, Hecht S, Eisenreich W, Feicht R, [49] Zeidler JG, Lichtenthaler HK, May HU, Lichtenthaler FW. Is iso-
Rohdich F, et al. Phosphorylation of 1-deoxy-D-xylulose by D- prene emitted by plants synthesized via the novel isopentenylpyro-
xylulokinase of Escherichia coli. Eur J Biochem 2001; 268: 310-6. phosphate pathway? Z Naturforsch 1997; 52C: 15-23
[31] White RH. Stable isotope studies on the biosynthesis of the thiazole [50] Zeidler JG, Schwender J, Muller C, Wiesner J, Weidemeyer C,
moiety of thiamin in Escherichia coli. Biochemistry 1978; 17: Beck E, et al. Inhibition of the non-mevalonate 1-deoxy-D-
3833-40. xylulose-5-phosphate pathway of plant isoprenoid biosynthesis by
[32] David S, Estramareix B, Fischer JC, Therisod M. 1-deoxy-d-threo- fosmidomycin. Z Naturforsch 1998; 53C: 980-6.
2-pentulose: The precursor of the five-carbon chain of the thiazole [51] Lichtenthaler HK, Rohmer M, Schwender J. Two independent
of thiamine. J Am Chem Soc 1981; 103: 7341-2. biochemical pathways for isopentenyl diphosphate and isoprenoid
[33] Hill RE, Sayer BG, Spenser JD. Biosynthesis of vitamin B6: incor- biosynthesis in higher plants. Physiol Plant 1997; 101: 643-52.
poration of d-1-deoxyxylulose. J Am Chem Soc 1989; 111: 1916-7. [52] Eisenreich W, Schwarz M, Cartayrade A, Arigoni D, Zenk MH,
[34] Takahashi S, Kuzuyama T, Watanabe H, Seto H. A 1-deoxy-D- Bacher A. The deoxyxylulose phosphate pathway of terpenoid bio-
xylulose-5-phosphate reductoisomerase catalyzing the formation of synthesis in plants and microorganisms. Chem Biol 1998; 5: R221-
2-C-methyl-D-erythritol-4-phosphate in an alternative non- 33.
mevalonate pathway for terpenoid biosynthesis. Proc Natl Acad Sci [53] Lichtenthaler HK. The 1-deoxy-D-xylulose 5-phosphate pathway
USA 1998; 18: 9879-84. of isoprenoid biosynthesis in plants. Ann Rev Plant Physiol Plant
[35] Rohdich F, Wungsintaweekul J, Fellermeier M, Sagner S, Herz S, Mol Biol 1999; 50: 47-65.
Kis K, et al. Cytidine 5'-triphosphate-dependent biosynthesis of [54] Rohmer M. The discovery of a mevalonate-independent pathway
isoprenoids: YgbP protein of Escherichia coli catalyzes the forma- for isoprenoid biosynthesis in bacteria, algae and higher plants. Nat
tion of 4-diphosphocytidyl-2-C-methylerythritol. Proc Natl Acad Prod Rep 1999; 16: 565-74.
Sci USA 1999; 96: 11758-63. [55] Eisenreich W, Rohdich F, Bacher A. Deoxyxylulose phosphate
[36] Luttgen H, Rohdich F, Herz S, Wungsintaweekul J, Hecht S, pathway to terpenoids. Trends Plant Sci 2001; 6: 78-84.
Schuhr CA, et al. Biosynthesis of terpenoids: YchB protein of Es- [56] Rodriguez-Concepcion M, Boronat A. Elucidation of the meth-
cherichia coli phosphorylates the 2-hydroxy group of 4- ylerythritol phosphate pathway for isoprenoid biosynthesis in bac-
diphosphocytidyl-2C-methyl-D-erythritol. Proc Natl Acad Sci USA teria and plastids. A metabolic milestone achieved through genom-
2000; 97: 1062-7. ics. Plant Physiol 2002; 130: 1079-89.
[37] Herz S, Wungsintaweekul J, Schuhr CA, Hecht S, Luttgen H, [57] Lange BM, Rujan T, Martin W, Croteau R. Isoprenoid biosynthe-
Sagner S, et al. Biosynthesis of terpenoids: YgbB protein converts sis: the evolution of two ancient and distinct pathways across ge-
4-diphosphocytidyl-2C-methyl-D-erythritol-2-phosphate to 2C- nomes. Proc Natl Acad Sci USA 2000; 97: 13172-7.
methyl-D-erythritol 2,4-cyclodiphosphate. Proc Natl Acad Sci USA [58] Hoeffler JF, Hemmerlin A, Grosdemange-Billiard C, Bach TJ,
2000; 97: 2486-90. Rohmer M. Isoprenoid biosynthesis in higher plants and in Es-
[38] Hintz M, Reichenberg A, Altincicek B, Bahr U, Gschwind RM, cherichia coli: on the branching in the methylerythritol phosphate
Kollas AK, et al. Identification of (E)-4-hydroxy-3-methyl-but-2- pathway and the independent biosynthesis of isopentenyl diphos-
enyl pyrophosphate as a major activator for human gamma-delta T phate and dimethylallyl diphosphate. Biochem J 2002; 366: 573-83.
cells in Escherichia coli. FEBS Lett 2001; 509: 317-22. [59] Disch A, Hemmerlin A, Bach TJ, Rohmer M. Mevalonate-derived
[39] Hecht S, Eisenreich W, Adam P, Amslinger S, Kis K, Bacher A, et isopentenyl diphosphate is the biosynthetic precursor of ubiquinone
al. Studies on the nonmevalonate pathway to terpenes: the role of prenyl side in tobacco BY-2 cells. Biochem J 1998; 331: 615-21.
the GcpE (IspG) protein. Proc Natl Acad Sci USA 2001; 98: [60] Goodwin TW. Incorporation of 14CO2 , [2-14C] acetate and [2-14C]
14837-42. mevalonic acid into -carotene in etiolated maize seedlings. Bio-
[40] Rohdich F, Hecht S, Gartner K, Adam P, Krieger C, Amslinger S, chem J 1958; 68: 26-7.
et al. Studies on the non-mevalonate terpene biosynthetic pathway: [61] Hemmerlin A, Hoeffler JF, Meyer O, Tritsch D, Kagan IA, Gros-
metabolic role of IspH (LytB) protein. Proc Natl Acad Sci USA demange-Billiard C, et al. Cross-talk between the cytosolic me-
2002; 99: 1158-63. valonate and the plastidial methylerythritol phosphate pathways in
[41] Rodriguez-Conception M, Campos N, Lois LM, Maldonado C, tobacco bright yellow-2 cells. J Biol Chem 2003; 278: 26666-76.
Hoeffler JF, Grosdemange-Billiard C, et al. Genetic evidence of [62] Nabeta K, Ishikawa T, Okuyama H. Sesqui- and di-terpene biosyn-
branching in the isoprenoid pathway for the production of isopen- thesis from 13C labelled acetate and mevalonate in cultured cells of
tenyl diphosphate in Escherichia coli. FEBS Lett 2000; 473: 328- Heteroscyphus planus. J Chem Soc Perkin Trans I 1995; 3111-5.
32. [63] Schwarz MK. Terpen-Biosynthese in Ginkgo biloba: Eine überra-
[42] Rohdich F, Zepeck F, Adam P, Hecht S, Kaiser J, Laupitz R, et al. schende Geschichte. Dissertation, Eidgenössische Hochschule Zü-
The deoxyxylulose phosphate pathway of isoprenoid biosynthesis: rich 1994.
studies on the mechanisms of the reactions catalyzed by IspG and [64] Schwender J, Gemünden C, Lichtenthaler HK. Chlorophyta exclu-
IspH protein. Proc Natl Acad Sci USA 2003, 100: 1586-91. sively use the 1-deoxyxylulose 5-phosphate/ 2-C-methylerythritol-
[43] Rohmer M, Knani M, Simonin P, Sutter B, Sahm H. Isoprenoid 4-phosphate pathway for the biosynthesis of isoprenoids. Planta
biosynthesis in bacteria: a novel pathway for the early steps leading 2001; 212: 416-23.
to isopentenyl diphosphate. Biochem J 1993; 295: 517-24. [65] Lichtenthaler HK. Evolution of carotenoid and isoprenoid biosyn-
[44] Disch A, Schwender J, Muller C, Lichtenthaler HK, Rohmer M. thesis in photosynthetic and non photosynthetic organisms. 16th In-
Distribution of the mevalonate and glyceraldehyde phos- ternational Plant Lipid Symposium. Budapest, Hungary. June 1-4,
phate/pyruvate pathways for isoprenoid biosynthesis in unicellular 2004.
algae and the cyanobacterium Synechocystis PCC 6714. Biochem J [66] Rodriguez-Concepcion M. The MEP pathway: A new target for the
1998; 333: 381-8. development of Herbicides, antibiotics and antimalarial drugs. Curr
[45] (a) Arigoni D, Sagner S, Latzel C, Eisenreich W, Bacher A, Zenk Pharm Des 2004; 10: 2391-400.
MH. Terpenoid biosynthesis from 1-deoxy-D-xylulose in higher [67] Jomaa H, Wiesner J, Sanderbrand S, Altincicek B, Weidemeyer C,
plants by intramolecular skeletal rearrangement. Proc Natl Acad Hintz M, et al. Inhibitors of the non-mevalonate pathway of isopre-
Sci USA 1997; 94: 10600-5. (b) Broers STJ. Dissertation (Eidge- noid biosynthesis as antimalarial drugs. Science 1999; 285: 1573-6.
nossische Technische Hochschule, Zurich) 1994.
1176 Current Pharmaceutical Design, 2007, Vol. 13, No. 11 Singh et al.

[68] Kobayashi K, Ehrlich SD, Albertini A, Amati G, Andersen KK, [89] Hoeffler JF, Tritsch D, Grosdemange-Billiard C, Rohmer M. Iso-
Arnaud M, et al. Essential Bacillus subtilis genes. Proc Natl Acad prenoid biosynthesis via the methylerythritol phosphate pathway.
Sci USA 2003; 100: 4678-83. Mechanistic investigations of the 1-deoxy-D-xylulose 5-phosphate
[69] Querol J, Campos N, Imperial S, Boronat A, Rodríguez- reductoisomerase. Eur J Biochem 2002; 269: 4446-57.
Concepción M. Functional analysis of the Arabidopsis thaliana [90] (a) Johnson AE, Tanner ME. Epimerization via carbon-carbon
GCPE protein involved in plastid isoprenoid biosynthesis. FEBS bond cleavage. L-ribulose-5-phosphate 4-epimerase as a masked
Lett 2002; 514: 343-6. class II aldolase. Biochemistry 1998; 37: 5746-54. (b) Lee LV, Vu
[70] Proteau PJ, Woo YH, Williamson RT, Phaosiri C. Stereochemistry MV, Cleland WW. 13C and deuterium isotope effects suggest an
of the reduction step mediated by recombinant 1-deoxy-D-xylulose aldol cleavage mechanism for L-ribulose-5-phosphate 4-epimerase.
5-phosphate isomeroreductase. Org Lett 1999; 1: 921-3. Biochemistry 2000; 39: 4808-20. (c) Hoeffler JF, Tritsch D, Gros-
[71] Schwender J, Muller C, Zeidler J, Lichtenthaler HK. Cloning and demange-Billiard C, Rohmer M. Isoprenoid biosynthesis via the
heterologous expression of a cDNA encoding 1-deoxy-D-xylulose- methylerythritol phosphate pathway. Mechanistic investigations of
5-phosphate reductoisomerase of Arabidopsis thaliana. FEBS Lett the 1-deoxy-D-xylulose 5-phosphate reductoisomerase. Eur J Bio-
1999; 455: 140-44. chem 2000, 69: 4446-57. (d) Fox DT, Poulter CD. Mechanistic
[72] Lange BM, Croteau R. Isoprenoid biosynthesis via a mevalonate- studies with 2-C-methyl-D-erythritol-4-phosphate synthase from
independent pathway in plants: cloning and heterologous expres- Escherichia coli. Biochemistry 2005; 44: 8360-8.
sion of 1-deoxy-D-xylulose-5-phosphate reductoisomerase from [91] Segel IH. In: Enzyme Kinetics. Behavior and analysis of rapid
peppermint. Arch Biochem Biophys 1999; 365: 170-4. equilibrium and steady-state enzyme systems. New York. John
[73] Argyrou A, Blanchard JS. Kinetic and chemical mechanism of Wiley & Sons, Inc., 1975; 653-5.
Mycobacterium tuberculosis 1-deoxy-D-xylulose-5-phosphate iso- [92] Steinbacher S, Kaiser J, Eisenreich W, Huber R, Bacher A,
meroreductase. Biochemistry 2004; 43: 4375-84. Rohdich F. Structural basis of fosmidomycin action revealed by the
[74] Koppisch AT, Fox DT, Blagg BS, Poulter CD. E. coli MEP syn- complex with 2-C-methyl-D-erythritol-4-phosphate synthase
thase: steady-state kinetic analysis and substrate binding. Biochem- (IspC). Implications for the catalytic mechanism and antimalarial
istry 2002; 41: 236-43. drug development. J Biol Chem 2003; 278: 18401-7.
[75] Grolle S, Bringer-Meyer S, Sahm H. Isolation of the dxr gene of [93] Yajima S, Hara K, Sanders JM, Yin F, Ohsawa K, Wiesner J, et al.
Zymomonas mobilis and characterization of the 1-deoxy-D- Crystallographic structures of two bisphosphonate: 1-
xylulose 5-phosphate reductoisomerase. FEMS Microbiol Lett deoxyxylulose-5-phosphate reductoisomerase complexes. J Am
2000; 191: 131-7. Chem Soc 2004; 126: 10824-5.
[76] Yin X, Proteau PJ. Characterization of native and histidine-tagged [94] Mac Sweeney A, Lange R, Fernandes RP, Schulz H, Dale GE,
deoxyxylulose 5-phosphate reductoisomerase from the cyanobacte- Douangamath A, et al. The crystal structure of E. coli 1-deoxy-D-
rium Synechocystis sp. PCC6803. Biochim Biophys Acta 2003; xylulose-5-phosphate reductoisomerase in a ternary complex with
1652: 75-81. the antimalarial compound fosmidomycin and NADPH reveals a
[77] Kuzuyama T, Takahashi S, Watanabe H, Seto H. Direct formation tight-binding closed enzyme conformation. J Mol Biol 2005; 345:
of 2-C-methyl-D-erythritol-4-phosphate from 1-deoxy-D-xylulose 115-27.
5-phosphate by 1-deoxy-D-xylulose 5-phosphate reductoisomerase, [95] Ricagno S, Grolle S, Bringer-Meyer S, Sahm H, Lindqvist Y,
a new enzyme in the non-mevalonate pathway to isopentenyl Schneider G. Crystal structure of 1-deoxy-d-xylulose-5-phosphate
diphosphate. Tetrahedron Lett 1998; 39: 4509-12. reductoisomerase from Zymomonas mobilis at 1.9 Å resolution.
[78] Radykewicz T, Rohdich F, Wungsintaweekul J, Herz S, Kis K, Biochim Biophys Acta 2004; 1698: 37-44.
Eisenreich W, et al. Biosynthesis of terpenoids: 1-deoxy-D- [96] Fernandes RPM, Phaosiri C, Proteau PJ. Mutation in the flexible
xylulose-5-phosphate reductoisomerase from Escherichia coli is a loop of 1-deoxy-D-xylulose 5-phosphate reductoisomerase broad-
class B dehydrogenase. FEBS Lett 2000; 465: 157-60. ens substrate utilization. Arch Biochem Biophys 2005; 444: 159-
[79] Proteau PJ. 1-Deoxy-D-xylulose 5-phosphate reductoisomerase: An 64.
overview. Bioorg Chem 2004; 32: 483-93. [97] Okuhara M, Kuroda Y, Goto T, Okamoto M, Terano H, Kohsaka
[80] Arigoni D, Giner JL, Sagner S, Wungsintaweekul J, Zenk MH, Kis M, et al. Studies on new phosphonic acid antibiotics. III. Isolation
K, et al. Stereochemical course of the reduction step in the forma- and characterization of FR-31564, FR-32863 and FR-33289. J An-
tion of 2-C-methyl erythritol from the terpene precursor 1- tibiot (Tokyo) 1980; 33: 24-8.
deoxyxylulose in higher plants. Chem Commun 1999; 1127-8. [98] Kamuro Y, Kawai T, Kakiuchi T. Fujisawa Pharmaceutical Co.
[81] Reuter K, Sanderbrand S, Jomaa H, Wiesner J, Steinbrecher I, Ltd., European Patent 0256785B1. 1988.
Beck E, et al. Crystal structure of 1-deoxy-D-xylulose-5-phosphate [99] Patterson DR. Rohm and Haas Co. US Patent 4693742. 1987.
reductoisomerase, a crucial enzyme in the non-mevalonate pathway [100] Shigi Y. Inhibition of bacterial isoprenoid synthesis by fosmido-
of isoprenoid biosynthesis. J Biol Chem 2002; 277: 5378-84. mycin, a phosphonic acid-containing antibiotic. J Antimicrob
[82] Yajima S, Nonaka T, Kuzuyama T, Seto H, Ohsawa K. Crystal Chemother 1989; 24: 131-45.
structure of 1-deoxy-D-xylulose-5-phosphate reductoisomerase [101] Horbach S, Sahm H, Welle R. Isoprenoid biosynthesis in bacteria:
complexed with cofactors: implications of a flexible loop move- two different pathways? FEMS Microbiol Lett 1993; 111: 135-40.
ment upon substrate binding. J Biochem (Tokyo) 2002; 131: 313-7. [102] Zerdler J, Schwender J, Muller C, Wiesner J, Weidemeyer C, Beck
[83] Arfin SM, Umbarger HE. Purification and properties of the aceto- E, et al. Inhibition of the non-mevalonate 1-deoxy-D-xylulose-5-
hydroxy acid isomeroreductase of Salmonella typhimurium. J Biol phosphate pathway of plant isoprenoid biosynthesis by fosmido-
Chem 1969; 244: 1118-27. mycin. Z Naturforsch 1998; 53c: 980-6.
[84] Shematek EM, Diven WF, Arfin SM. Subunit structure of - [103] Fellermeier M, Kis K, Sagner S, Maier U, Bacher A, Zenk MH.
acetohydroxy acid isomeroreductase from Salmonella typhimurium. Cell-free conversion of 1-deoxy-D-xylulose-5-phosphate and 2-C-
Arch Biochem Biophys 1973; 158: 126-31. methyl-D-erythritol-4-phosphate into -carotene in higher plants
[85] Hofler JG, Decedue CJ, Luginbuhl GH, Reynolds JA, Burns RO. and its inhibition by fosmidomycin. Tetrahedron Lett 1999; 40:
The subunit structure of alpha-acetohydroxyacid isomeroreductase 2743-6.
from Salmonella typhimurium. J Biol Chem 1975; 250: 877-82. [104] Wiesner J, Borrmann S, Jomaa H. Fosmidomycin for the treatment
[86] (a) Chunduru SK, Mrachko GT, Calvo KC. Mechanism of the ketol of malaria. Parasitol Res 2003; 90: S71-6.
acid reductoisomerase. Steady state analysis and metal ion re- [105] Lell B, Ruangweerayut R, Wiesner J, Missinou MA, Schindler A,
quirement. Biochemistry 1989; 28: 486-93. (b) Dumas R, Job D, Baranek T, et al. Fosmidomycin, a novel chemotherapeutic agent
Ortholand JY, Emeric G, Greiner A, Douce R. Isolation and kinetic for malaria. Antimicrob Agents Chemother 2003; 47: 735-8.
properties of acetohydroxy acid isomeroreductase from spinach [106] Borrmann S, Adegnika AA, Matsiegui P-B, Issifou S, Schindler A,
(Spinacia oleracea) chloroplasts overexpressed in Escherichia coli. Mawili-Mboumba DP, et al. Fosmidomycin-clindamycin for Plas-
Biochem J 1992; 288: 865-74. modium falciparum infections in African children. J Infect Dis
[87] Aulabaugh A, Schloss JV. Oxalyl hydroxamates as reaction- 2004; 189: 901-8.
intermediate analogues for ketol-acid reductoisomerase. Biochem- [107] Borrmann S, Lundgren I, Oyakhirome S, Impouma B, Matsiegui P-
istry 1990; 29: 2824-30. B, Adegnika AA, et al. Fosmidomycin plus clindamycin for treat-
[88] Koshland DE Jr, Neet KE. The catalytic and regulatory properties ment of pediatric patients aged 1 to 14 years with Plasmodium fal-
of enzymes. Annu Rev Biochem 1968; 37: 359-410. ciparum malaria. Antimicrob Agents Chemother 2006; 50: 2713-8.
MEP Pathway for Novel Antimalarial, Antibacterial and Herbicidal Drug Discovery Current Pharmaceutical Design, 2007, Vol. 13, No. 11 1177

[108] Borrmann S, Ayola A, Adegnika AA, Moussavou F, Oyakhirome [124] Haemers T, Wiesner J, Poecke SV, Goeman J, Henschker D, Beck
S, Esser G, et al. Short-course regimens of artesunate- E, et al. Synthesis of alpha-substituted fosmidomycin analogues as
fosmidomycin in treatment of uncomplicated Plasmodium falcipa- highly potent Plasmodium falciparum growth inhibitors. Bioorg
rum malaria. Antimicrob Agents Chemother 2005; 49: 3749-54. Med Chem Lett 2006; 16: 1888-91.
[109] Wiesner J, Henschker D, Hutchinson DB, Beck E, Jomaa H. In [125] Merckle L, de Andres-Gomez A, Dick B, Cox RJ, Godfrey CR. A
vitro and in vivo synergy of fosmidomycin, a novel antimalarial fragment-based approach to understanding inhibition of 1-deoxy-
drug, with clindamycin. Antimicrob Agents Chemother 2002; 46: D-xylulose-5-phosphate reductoisomerase Chembiochem 2005; 6:
2889-94. 1866-74.
[110] Murakawa T, Sakamoto H, Fukada S, Konishi T, Nishida M. [126] Devreux V, Wiesner J, Goeman JL, Van der Eycken J, Jomaa H,
Pharmacokinetics of fosmidomycin, a new phosphonic acid antibi- Van Calenbergh S. Synthesis and biological evaluation of cyclo-
otic. Antimicrob Agents Chemother 1982; 21: 224-30. propyl analogues of fosmidomycin as potent Plasmodium falcipa-
[111] Kuemmerle HP, Murakawa T, De Santis F. Pharmacokinetic rum growth inhibitors. J Med Chem 2006; 49: 2656-60.
evaluation of fosmidomycin, a new phosphonic acid antibiotic. [127] Hoeffler JF, Tritsch D, Grosdemange-Billiard C, Rohmer M. Iso-
Chemioterapia 1987; 6: 113-9. prenoid biosynthesis via the methylerythritol phosphate pathway.
[112] Reichenberg A, Wiesner J, Weidemeyer C, Dreiseidler E, Sander- Mechanistic investigations of the 1-deoxy-D-xylulose 5-phosphate
brand S, Altincicek B, et al. Diaryl ester prodrugs of FR900098 reductoisomerase. Eur J Biochem 2002; 269: 4446-57.
with improved in vivo antimalarial activity. Bioorg Med Chem Lett [128] Wong A, Munos JW, Devasthali V, Johnson KA, Liu HW. Study
2001; 11: 833-5. of 1-deoxy-D-xylulose-5-phosphate reductoisomerase: synthesis
[113] Ortmann R, Wiesner J, Reichenberg A, Henschker D, Beck E, and evaluation of fluorinated substrate analogues. Org Lett 2004; 6:
Jomaa H, et al. Acyloxyalkyl ester prodrugs of FR900098 with im- 3625-8.
proved in vivo antimalarial activity. Bioorg Med Chem Lett 2003; [129] Phaosiri C, Proteau PJ. Substrate analogs for the investigation of
13: 2163-6. deoxyxylulose 5-phosphate reductoisomerase inhibition: synthesis
[114] Ortmann R, Wiesner J, Reichenberg A, Henschker D, Beck E, and evaluation. Bioorg Med Chem Lett 2004; 14: 5309-12.
Jomaa H, et al. Alkoxycarbonyloxyethyl ester prodrugs of [130] Fox DT, Poulter CD. Synthesis and evaluation of 1-deoxy-D-
FR900098 with improved in vivo antimalarial activity. Arch Pharm xylulose 5-phosphoric acid analogues as alternate substrates for
(Weinheim) 2005; 338: 305-14. methylerythritol phosphate synthase. J Org Chem 2005; 70: 1978-
[115] Kurz T, Geffken D, Wackendorff C. Hydroxyurea analogues of 85.
fosmidomycin. Z Naturforsch 2003; 58B: 106-10. [131] Walker JR, Poulter CD. Synthesis and evaluation of 1-deoxy-D-
[116] Kurz T, Geffken D, Wackendorff C. Carboxylic acid analogues of xylulose 5-phosphate analogues as chelation-based inhibitors of
fosmidomycin. Z Naturforsch 2003; 58B: 457-61. methylerythritol phosphate synthase. J Org Chem 2005; 70: 9955-
[117] Courtois M, Mincheva Z, Andreu F, Rideau M, Viaud-Massuard 9.
MC. Synthesis and biological evaluation with plant cells of new [132] Kuntz L, Tritsch D, Grosdemange-Billiard C, Hemmerlin A,
fosmidomycin analogues containing a benzoxazolone or oxa- Willem A, Bach TJ, et al. Isoprenoid biosynthesis as a target for
zolopyridinone ring. J Enzyme Inhib Med Chem 2004; 19: 559-65. antibacterial and antiparasitic drugs: phosphonohydroxamic acids
[118] Contin A, der Heijden V, Lefeber AWM, Verpoorte R. The iridoid as inhibitors of deoxyxylulose phosphate reductoisomerase. Bio-
glycoside secologanin is derived from the novel triosephos- chem J 2005; 386: 127-35.
phate/pyruvate pathway in a Catharanthus roseus cell culture. [133] Herforth C, Wiesner J, Heidler P, Sanderbrand S, Van Calenbergh
FEBS Lett 1998; 434: 413-6. S, Jomaa H, et al. Antimalarial activity of N (6)-substituted adeno-
[119] (a) Mincheva Z, Curtois M, Creche J, Ridaeu M, Viaud-Massard sine derivatives. Part 3. Bioorg Med Chem 2004; 12: 755-62.
MC. One-pot synthesis of functionalized 4,5-dihydroisoxazole de- [134] Gottlin EB, Benson RE, Conary S, Antonio B, Duke K, Payne ES,
rivatives via nitrile oxides and biological evaluation with plant et al. High-throughput screen for inhibitors of 1-deoxy-d-xylulose
cells. Bioorg Med Chem 2003; 12: 191-7. (b) Hong SB, Hughes 5-phosphate reductoisomerase by surrogate ligand competition. J
EH, Shanks JV, San KY, Gibson SI. Role of the non-mevalonate Biomol Screen 2003; 8: 332-9.
pathway in indole alkaloid production by Catharanthus roseus [135] Silber K, Heidler P, Kurz T, Klebe G. AFMoC enhances predictiv-
hairy roots. Biotechnol Prog 2003; 19: 1105-8. ity of 3D QSAR: a case study with DOXP reductoisomerase. J Med
[120] Mincheva Z, Courtois M, Andreu F, Rideau M, Viaud-Massuard Chem 2005; 48: 3547-63.
MC. Fosmidomycin analogues as inhibitors of monoterpenoid in- [136] Singh N, Cheve G, Avery MA, McCurdy CR. Comparative protein
dole alkaloid production in Catharanthus roseus cells. Phytochem- modeling of 1-deoxy-D-xylulose-5-phosphate reductoisomerase
istry 2005; 66: 1797-803. enzyme from Plasmodium falciparum: A potential target for anti-
[121] Lichtenthaler HK, Zeidler J, Schwender J, Muller C. The non- malarial drug discovery. J Chem Inf Model 2006; 46: 1360-70.
mevalonate isoprenoid biosynthesis of plants as a test system for [137] Rodriguez-Concepcion M. The MEP pathway: a new target for the
new herbicides and drugs against pathogenic bacteria and the ma- development of herbicides, antibiotics and antimalarial drugs. Curr
laria parasite. Z Naturforsch 2000; 55 C: 305-13. Pharm Des 2004; 10(19): 2391-400.
[122] Woo YH, Fernandes RP, Proteau PJ. Evaluation of fosmidomycin [138] Romanelli F, Smith KM, Hoven AD. Chloroquine and hydroxy-
analogs as inhibitors of the Synechocystis sp. PCC6803 1-deoxy-D- chloroquine as inhibitors of human immunodeficiency virus (HIV-
xylulose-5-phosphate reductoisomerase. Bioorg Med Chem 2006; 1) activity. Curr Pharm Des 2004; 10(21): 2643-8.
14: 2375-85. [139] Seeber F, Aliverti A, Zanetti G. The plant-type ferredoxin-NADP+
[123] Hemmi K, Takeno H, Hashimoto M, Kamiya T. Studies on phos- reductase/ferredoxin redox system as a possible drug target against
phonic acid antibiotics. IV. Synthesis and antibacterial activity of apicomplexan human parasites. Curr Pharm Des 2005; 11(24):
analogs of 3-(N-acetyl-N-hydroxyamino)-propylphosphonic acid 3159-72.
(FR-900098). Chem Pharm Bull (Tokyo) 1982; 30: 111-8.

You might also like