throbber
Biochemical Pharmacology, Vol. 55, pp. 1541–1549, 1998.
`© 1998 Elsevier Science Inc. All rights reserved.
`
`ISSN 0006-2952/98/$19.00 1 0.00
`PII S0006-2952(97)00684-9
`
`COMMENTARY
`Mechanism of Action of Diazaborines
`Clair Baldock,* Gert-Jan de Boer,† John B. Rafferty,* Antoine R. Stuitje†
`and David W. Rice*‡
`*KREBS INSTITUTE FOR BIOMOLECULAR RESEARCH, DEPARTMENT OF MOLECULAR BIOLOGY AND BIOTECHNOLOGY,
`THE UNIVERSITY OF SHEFFIELD, SHEFFIELD S10 2TN, U.K.; AND †DEPARTMENT OF GENETICS, INSTITUTE OF
`MOLECULAR BIOLOGICAL STUDIES (IMBW), VRIJE UNIVERSITEIT, BIOCENTER AMSTERDAM, DE BOELELAAN 1087,
`1081 HV AMSTERDAM, THE NETHERLANDS
`
`ABSTRACT. The diazaborine family of compounds have antibacterial properties against a range of
`Gram-negative bacteria. Initially, this was thought to be due to the prevention of lipopolysaccharide synthesis.
`More recently, the molecular target of diazaborines has been identified as the NAD(P)H-dependent enoyl acyl
`carrier protein reductase (ENR), which catalyses the last reductive step of fatty acid synthase. ENR from
`Mycobacterium tuberculosis is the target for the front-line antituberculosis drug isoniazid. The emergence of
`isoniazid resistance strains of M. tuberculosis, a chronic infectious disease that already kills more people than any
`other infection, is currently causing great concern over the prospects for its future treatment, and it has
`reawakened interest in the mechanism of diazaborine action. Diazaborines only inhibit ENR in the presence of
`the nucleotide cofactor, and this has been explained through the analysis of the x-ray crystallographic structures
`of a number of Escherichia coli ENR–NAD1– diazaborine complexes that showed the formation of a covalent
`bond between the boron atom in the diazaborines and the 29-hydroxyl of the nicotinamide ribose moiety that
`generates a noncovalently bound bisubstrate analogue. The similarities in catalytic chemistry and in the
`conformation of the nucleotide cofactor across the wider family of NAD(P)-dependent oxidoreductases suggest
`that there are generic opportunities to mimic the interactions seen here in the rational design of bisubstrate
`analogue inhibitors for other NAD(P)H-dependent oxidoreductases.
`BIOCHEM PHARMACOL 55;10:1541–1549,
`1998. © 1998 Elsevier Science Inc.
`
`KEY WORDS. enoyl reductase; diazaborine; isoniazid; lipid biosynthesis; bisubstrate analogues
`
`Diazaborines represent a group of antibacterial drugs of
`which the important structural element is a heterocyclic
`1,2-diazine ring containing a boron as a third hetero atom
`(Fig. 1). Although the antimicrobial activity of these
`compounds was first described in the late 1960s, their
`biological target remained obscure until the 1980s. The first
`clue that diazaborines may interfere with membrane bio-
`synthesis was obtained by Hogenauer and Woisetschlager
`[1]. Using specific mutant strains of Escherichia coli and
`Salmonella typhimurium, they showed that diazaborine in-
`hibits the incorporation of radioactive galactose into the
`LPS§ of these bacteria. This result fitted nicely with earlier
`observations that the antibacterial activity of diazaborines
`is confined almost exclusively to Gram-negative bacteria,
`indicating that they specifically inhibit LPS synthesis,
`which is an integral part of the outer membrane of this
`group of bacteria. This notion triggered the search for new
`diazaborine derivatives and analogues in the treatment of
`bacterial infections, despite the inherent toxicity of boron-
`containing compounds [2].
`
`‡ Corresponding author. Tel. 44-114-222-4242; FAX 44-114-272-8697.
`§ Abbreviations: LPS, lipopolysaccharide; ACP, acyl carrier protein;
`ENR, enoyl ACP reductase; and FAS, fatty acid synthetase.
`
`STRUCTURE–ACTIVITY RELATIONSHIPS
`The schematic structure for compounds that are generally
`referred to as diazaborines is given in Fig. 1A. The more
`systematic name for these compounds is 1,2-dihydro-1-
`hydroxy-2-(organosulfonyl)-areno[d][1,2,3]diazaborines
`(arene 5 benzene, naphthalene, thiophene, furan, pyrrole).
`Systematic syntheses of these compounds, by a reaction of
`(organosulfonyl)hydrazones of arene aldehydes or ketones
`with tribromoborane in the presence of ferric chloride, were
`first described by Grassberger et al. [2]. In this study, the
`activities of approximately 80 different diazaborine deriva-
`tives against bacteria in vitro and in vivo (E. coli septicae-
`mia) were determined. Although, in general, thieno-diaza-
`borines were found to be the most potent inhibitors,
`followed by benzo-diazaborines and furo-diazaborines,
`whereas pyrrolo-diazaborines were totally inactive (Fig.
`1B), this classification oversimplifies the extensive data of
`Grassberger et al. [2].
`To facilitate a more comprehensive understanding of
`the significance of the organosulfonyl side chain, the
`arene group, and various substitutions on this group in
`relation to antibacterial activity of diazaborines, we have
`summarized the most relevant data, in this respect, in
`Table 1.
`
`CFAD v. Anacor, IPR2015-01776
`ANACOR EX. 2009 - 1/10
`
`

`
`1542
`
`C. Baldock et al.
`
`FIG. 1. (A) Structural formulae of a number of different classes of diazaborine; and (B) the MIC values for the best inhibitors in each
`class of diazaborine 1A, 1B, 2, 3 and 4 in an in vivo antibacterial assay against E. coli [2]. MIC 5 minimum inhibitory concentration.
`
`Importance of the Organosulfonyl Side Chain (SO2R2)
`As depicted in Table 1, irrespective of the nature of the
`arene group, diazaborines with propylsulfonyl side chains
`show the highest antibacterial activity. Decreasing the
`chain length of the alkyl group greatly reduces activity
`(Table 1, No. 6 vs No. 7 or No. 11 vs No. 13). Derivatives
`with benzylsulfonyl side chains show considerable activity,
`whereas substitutions on the benzene ring are generally not
`favourable (No. 9 vs No. 8 and 10).
`
`Importance of the Arene Group (X)
`Irrespective of the nature of the organosulfonyl group,
`thieno-diazaborines are more potent than benzo-diazabo-
`rines (No. 1 vs No. 11, No. 12 vs No. 3 and 10). In the
`thieno-diazaborine series, however, the thieno[2,3-d]diaza-
`borines (No. 1– 4) are generally slightly more active than
`their thieno[3,2-d] counterparts (No. 5–10).
`Substitution of a methyl for a hydrogen in position 6
`(Fig. 1) of thieno-diazaborine significantly increases biolog-
`ical activity (e.g. No. 6 vs No. 5), whereas replacement of
`a methyl by bromine has little effect, with the exception of
`the 6-bromo derivative of thieno[2,3-d]diazaborine, which
`is totally inactive (e.g. No. 4 vs No. 9 and 10). Substitution
`of hydrogen in position 7 is probably not advisable, since
`replacement with bromine resulted in a complete loss of
`biological activity (not shown).
`Benzo-diazaborines are generally less active than thieno-
`diazaborines, and substitutions by methyl or halogen (F, Cl,
`Br) on the benzene ring have no marked influence on the
`bacterial activities in vitro. In general, benzo-diazaborine
`derivatives with methyl or halogen in position 5 or 7 (Fig.
`
`1) are less active than the unsubstituted parent compound
`(No. 14; not shown). Substitution (methyl, halogen) in
`position 6 slightly increases antibacterial activity (No. 14 vs
`No. 12). Substitution with polar groups (OH, NH2, NR2,
`NHCOCH3, COOH) in position 6 or 7 generally leads to
`a complete loss of activity (not shown).
`Only a few furo- and pyrrolo-diazaborines (Fig. 1) were
`analysed in the work of Grassberger et al.
`[2]. While
`pyrrolo-diazaborines were found to be completely inactive,
`2 out of the 3 furo-diazaborines tested showed substantial
`antibacterial activity. Both biologically active furo-diazabo-
`rines have a tosyl group attached to the sulfonyl moiety
`(Table 1, No. 15), and a methyl or bromine substitution in
`position 6. Although alkylsulfonyl derivatives were not
`included in the study, it seems very likely that substitution
`of a propyl moiety instead of a tosyl moiety will further
`enhance the biological activity of furo-diazaborines.
`
`BORON-FREE ANALOGUES
`A major problem with diazaborines is their inherent toxic
`potential, which is probably due to the arenoboronic acid
`amide moiety [2]. To help design boron-free analogues,
`Grassberger et al. [2] tried to address the question as to
`whether the bicyclic areno-diazaborines themselves are the
`active species or whether hydrolytic cleavage at the BN
`bond to give the corresponding (dihydroxy)arenes is essen-
`tial
`for biological activity of the compounds. For this
`purpose they synthesized the carbacyclic analogue of benzo-
`diazaborine (No. 14). This isoquinoline (Fig. 2A), how-
`ever, was inactive in all test systems. On the other hand,
`the stable N-methyl derivative (Fig. 2B), which served as a
`
`ANACOR EX. 2009 - 2/10
`
`

`
`Mechanism of Action of Diazaborines
`
`1543
`
`TABLE 1. Antibacterial activity of selected diazaborines*
`
`Structure
`
`No.
`
`ED50
`(mg/kg)
`
`MIC† values (mg/mL)
`E. coli E. aerogenes S. typhimurium K. pneumoniae P. mirabilis N. gonorrhoeae
`
`2
`
`4
`
`9
`
`1.25
`
`1.56
`
`2.5
`
`5
`
`6.25
`
`25
`
`0.19
`
`0.31
`
`1.56
`
`0.31
`
`0.31
`
`1.25
`
`1.56
`
`1.25
`
`0.5
`
`0.5
`
`1.25
`
`0.25
`
`.300
`
`.50
`
`.50
`
`.50
`
`.50
`
`.50
`
`.8
`
`4.5
`
`1.56
`
`3.12
`
`28
`
`6.25
`
`25
`
`0.78
`
`3.12
`
`0.39
`
`0.78
`
`1.56
`
`25
`
`1
`
`8
`
`113
`
`.50
`
`.50
`
`.50
`
`.50
`
`.50
`
`.8
`
`1
`
`2
`
`3
`
`4
`
`5
`
`6
`
`7
`
`8
`
`49
`
`3.12
`
`12.5
`
`9
`
`;20
`
`3.12
`
`12.5
`
`10
`
`;15
`
`6.25
`
`11
`
`73
`
`6.25
`
`12
`
`;15
`
`12.5
`
`13
`
`42
`
`14
`
`;25
`
`25
`
`25
`
`25
`
`10
`
`50
`
`.50
`
`.50
`
`15
`
`;10
`
`12.5
`
`25
`
`0.78
`
`0.78
`
`3.12
`
`2.5
`
`3.12
`
`0.31
`
`0.78
`
`1.56
`
`1.25
`
`1.56
`
`0.39
`
`1.56
`
`1.56
`
`1.56
`
`3.12
`
`0.5
`
`1
`
`0.5
`
`2
`
`1
`
`12.5
`
`6.25
`
`25
`
`.8
`
`6.25
`
`3.12
`
`3.12
`
`12.5
`
`1.56
`
`3.12
`
`2
`
`1
`
`*Data excerpted from Ref. 2. Reprinted with permission from J Med Chem 27: 947–953, 1984. © 1984 American Chemical Society.
`†MIC 5 minimum inhibitory concentration that inhibited visible growth of bacteria in trypticase soy broth (in mg/mL). Genus and species not specified in text: E. aerogenes,
`Enterobacter aerogenes; K. pneumoniae, Klebsiella pneumoniae; and P. mirabilis, Proteus mirabilis.
`
`ANACOR EX. 2009 - 3/10
`
`

`
`1544
`
`C. Baldock et al.
`
`FIG. 2. Structural formulae of: (A) the car-
`bacyclic analogue of benzo-diazaborine; and
`(B) the ring-opened derivative of benzo-
`diazaborine.
`
`stable ring-opened analogue of benzo-diazaborine (No. 14),
`showed some biological activity on E. coli, but was at least
`ten times less active than the corresponding diazaborine.
`Although these results seem to support to some extent the
`hypothesis that ring opening is essential
`for biological
`activity, no firm conclusions could be made.
`
`CELLULAR TARGET OF DIAZABORINES
`The antibacterial activity of diazaborines is confined almost
`exclusively to Gram-negative bacteria. A rationale behind
`this observation was first presented by Hogenauer and
`Woisetschlager [1], who demonstrated that a specific thi-
`eno-diazaborine (Table 1, No. 5) inhibited LPS biosynthe-
`sis in both E. coli and Salmonella. In a later paper of
`Turnowsky et al. [3], it was shown that the primary target of
`inhibition was not LPS biosynthesis itself, but rather an
`earlier step in lipid A precursor biosynthesis, such as de novo
`fatty acid biosynthesis or the acyl transfer to the UDP-N-
`acetylglucosamine moiety. In addition, these authors car-
`ried out a thorough molecular genetic analysis of diazabo-
`rine resistance in both E. coli and Salmonella and demon-
`strated that resistance results from a point mutation in the
`envM genes of these bacteria. In an earlier study, the envM
`gene of E. coli was identified as an essential gene, since an
`allelic mutant form (envM392) results in a temperature-
`sensitive growth phenotype [4]. In fact, the similarity of the
`effects on E. coli cells seen after treatment with diazaborine
`or by incubating a temperature-sensitive envM mutant at
`
`42° strongly suggested that the envM protein was the actual
`target of the drug.
`
`BIOCHEMICAL TARGET OF DIAZABORINES
`Despite the demonstration that both inhibition of wild-
`type E. coli cells by diazaborine and shifting the envM392
`mutant to the nonpermissive temperature result in imme-
`diate cessation of fatty acid biosynthesis, the actual func-
`tion of the envM protein in fatty acid biosynthesis re-
`mained obscure until more recently. The notion that the
`envM protein might actually be the ENR component of the
`bacterial fatty acid synthetase (FAS II) was only recognized
`a few years ago on the basis of amino acid sequence
`homology with the purified ENR protein and the corre-
`sponding cDNA sequence from oilseed rape (Brassica na-
`pus) [5, 6]. ENR catalyses the last reductive step in the
`cyclic process of fatty acid elongation, as depicted in Fig. 3.
`Direct evidence that the E. coli envM gene encodes a
`diazaborine-sensitive ENR was provided independently by
`two laboratories in 1994 [7, 8]. In addition, it was estab-
`lished that the envM392 (ts) allele encoded an extremely
`temperature-sensitive ENR and that diazaborine is a spe-
`cific inhibitor of this E. coli enzyme [8]. The enzyme studies
`also showed that NAD1 is required as a cofactor for both
`the inhibition and the binding of diazaborine to the ENR
`enzyme [8]. Based on the fact that the envM gene encodes
`the ENR component of the E. coli fatty acid synthetase, this
`gene was recently renamed fabI [7].
`
`FIG. 3. Schematic representation of the fatty
`acid synthetase cycle.
`
`ANACOR EX. 2009 - 4/10
`
`

`
`Mechanism of Action of Diazaborines
`
`1545
`
`FIG. 4. Protein sequence alignment of E. coli, B. napus (rape
`seed) and M. tuberculosis ENRs in the region of the Gly93Ser
`and Ser94Ala mutations, associated with diazaborine and isoni-
`azid resistance in E. coli and M. tuberculosis ENR, respectively.
`
`MOLECULAR GENETICS OF
`DIAZABORINE RESISTANCE
`The demonstration that ENR is the target of diazaborine
`and the observation that all diazaborine-resistant mutants
`isolated thus far have the same Gly93Ser (see Fig. 4) amino
`acid substitution in this gene justify the conclusion that the
`primary effect of diazaborine is on core fatty acid biosyn-
`thesis, whereas the effects on,
`for example, membrane
`integrity and LPS biosynthesis are of a pleiotropic nature
`[8]. It was also shown that both the rape seed ENR and the
`equivalent E. coli enzyme encoded by the diazaborine-
`resistant allele are insensitive to the drug [8]. In this
`respect,
`it is important to realize that both rape seed
`plantlets and E. coli strains with a resistant fabI allele are
`still sensitive to diazaborine at concentrations above 20 and
`200 mg/mL, respectively (Stuitje AR, unpublished observa-
`tion). This observation suggests that other targets for
`diazaborine may exist in these organisms.
`The insensitivity of the plant ENR towards diazaborine
`has facilitated gene replacement experiments, demonstrat-
`ing that the coding sequence of the essential E. coli fabI
`gene can be replaced by a cDNA sequence encoding rape
`seed ENR. Although the resulting E. coli strain shows
`slightly different growth characteristics and membrane fatty
`
`acid composition, it is viable under laboratory conditions,
`demonstrating that plant ENR can functionally replace its
`counterpart in the bacterial multi-enzyme FAS system [8].
`
`SENSITIVITY OF ENR TO OTHER DRUGS
`Recently, it has become evident that besides diazaborines,
`ENR is a potential target for at least two other drugs (see
`Fig. 5), the bleaching herbicide diflufenican [N-(2,4-
`difluorophenyl)-2-[3-(trifluoromethyl)phenoxy]-3-pyridin-
`ecarboxyamide] and the antituberculosis drug isoniazid
`[INH; isonicotinic acid hydrazide].
`Although the primary mode of action of diflufenican is
`on carotenoid biosynthesis, with phytoene synthase being
`the main target, it was also reported to inhibit plant fatty
`acid synthetase in vitro [9]. In fact, Ashton et al. [10]
`demonstrated that both plant and E. coli ENR are also a
`target for diflufenican. Although it has structural similari-
`ties, in part, with pyridine nucleotides, the mode of action
`of diflufenican is still obscure, since it is clearly not a
`general
`inhibitor of pyridine nucleotide-dependent en-
`zymes. The NADPH-dependent b-keto reductase compo-
`nent of FAS, for example, is not inhibited by diflufenican
`[10].
`Isoniazid has been used since 1952 as one of the most
`effective drugs for the treatment and prophylaxis of tuber-
`culosis. A single missense mutation in the inhA gene of
`Mycobacterium tuberculosis can confer resistance to this
`drug. Based on the similarities of the corresponding inhA
`protein to E. coli (40%) and rape seed ENR (37%), it was
`demonstrated recently that this gene also encodes an
`NADH-specific ENR [11]. Despite the fact that the crystal
`structure of the inhA protein is solved and that the
`mutation that leads to resistance to isoniazid, Ser94Ala
`(Fig. 4), maps to a region close to the nucleotide binding
`site [11], little is known about the mode of action of
`isoniazid at the molecular level. Further mechanistic studies
`on enzyme inhibition by isoniazid have been seriously
`hampered, because it is now recognized that isoniazid is a
`prodrug that is activated by mycobacteria to an as yet
`
`FIG. 5. Structural formulae of diflufenican and
`isoniazid.
`
`ANACOR EX. 2009 - 5/10
`
`

`
`1546
`
`C. Baldock et al.
`
`FIG. 6. (A) Schematic diagram of the ENR tetramer with the cofactor bound. The ribbon trace of E. coli ENR is shown coloured in
`red, white, green, and cyan for each subunit, and the NAD1 is shown in an all-atom representation, coloured by atom (produced using
`MIDAS [14]). (B) Schematic diagram of a single subunit of the ENR–NAD1 complex. The ribbon trace of E. coli ENR is shown
`coloured in rainbow colours from red at the N-terminus to blue at the C-terminus, and the NAD1 is shown in an all-atom
`representation, coloured by atom (produced using MIDAS [14]). (C) Photograph of an ENR–NAD1– diazaborine crystal. (D)
`Schematic diagram of a single subunit of the ENR–NAD1–thieno-diazaborine complex. The ribbon trace of E. coli ENR is shown in
`green; the NAD1 and diazaborine are shown in an all-atom representation and coloured cyan and blue, respectively. The loop that
`orders on diazaborine binding is highlighted in red (produced using MIDAS [14]). (E) Active site of the ENR–NAD1–thieno-
`diazaborine complex. The Ca backbone trace is shown in cyan, with the NAD1 and diazaborine shown in an all-atom representation
`and coloured by atom (produced using MIDAS [14]). (F) Fourier map of the NAD1–thieno-diazaborine complex at 2.2 Å resolution
`with the final refined structure superimposed (produced using BOBSCRIPT [15]).
`
`unknown species. This activation process is thought to
`involve the catalase-peroxidase KatG [12].
`
`molecular basis by which diazaborine inhibits the E. coli
`enzyme.
`
`MOLECULAR BASIS OF
`DIAZABORINE ACTION
`Given the importance of ENR as a drug target, recent
`crystallographic studies have sought to understand the
`
`Structure and Mechanism of ENR
`E. coli ENR is a homo-tetramer of subunit Mr of approxi-
`mately 28,000 and, following crystallization [13], its struc-
`ture was solved to 2.1 Å resolution (Fig. 6A), by a
`
`ANACOR EX. 2009 - 6/10
`
`

`
`Mechanism of Action of Diazaborines
`
`1547
`
`Structural Studies on the E. coli
`ENR–NAD1–Diazaborine Complexes
`To understand the mode of diazaborine action, crystals of
`two E. coli ENR–NAD1– diazaborine complexes were pre-
`pared (Fig. 6C) [in the presence of a thieno (No. 5) and a
`benzo-diazaborine (No. 14)], and the structures were solved
`independently by molecular replacement using the struc-
`ture of E. coli ENR–NAD1 as a search model to 2.2 and 2.6
`Å, respectively [17]. Unlike the situation for the ENR–
`NAD1 complex, in both the diazaborine complexes the
`loop between Leu195 and Met206 is well defined (Fig. 6D)
`and provides two residues, Ile200 and Phe203, whose side
`chains are in van der Waals’ contact with the non-boron-
`containing 5- and 6-membered rings of the thieno- and
`benzo-diazaborines, respectively.
`
`Binding of Diazaborine
`Analysis of the diazaborine binding sites showed that both
`the diazaborine compounds bind in a closely related man-
`ner, adjacent to the nicotinamide ring of the cofactor, in a
`pocket formed by the side chains of Tyr146, Tyr156,
`Met159, Ile200, Phe203, Leu100, Lys163, and the main
`chain peptide between Gly93 and Ala95. The bicyclic rings
`of the diazaborines form a face-to-face interaction with the
`nicotinamide ring, allowing the formation of extensive
`p–p stacking interactions with additional van der Waals’
`interactions between the rings and the side chains of
`Tyr156, Tyr146, Phe203, and Ile200. The major difference
`between the binding of the two diazaborines is that their
`respective tosyl and propyl groups occupy subtly modified
`positions. The tosyl moiety lies perpendicular to the bicy-
`clic ring and interacts with the main chain peptide between
`Gly93 and Ala95 and the side chain of Leu100, whereas the
`propyl moiety folds back onto the planar bicyclic ring
`system in a manner reminiscent of a scorpion’s tail and
`forms interactions with the side chain of Met159 and
`Ile200 and the main chain peptide of both Gly93 and
`Phe94 (Fig. 6E). Additional interactions made by both
`drugs include hydrogen bonds between the boron hydroxyl
`and the phenolic hydroxyl of Tyr156 and between a
`nitrogen in the boron-containing ring and an ordered
`solvent molecule.
`
`MECHANISM OF DIAZABORINE RESISTANCE
`Previous studies have shown that a Gly93Ser mutation in
`E. coli ENR leads to diazaborine resistance [3, 8] and
`analysis of the structure shows that the alpha carbon of
`Gly93 lies close to the sulphonyl group of the diazaborine.
`Modelling studies show that in the absence of changes to
`the main-chain torsion angles in the Gly93Ser mutant, the
`Cb atom of the serine side-chain would be unacceptably
`close to the two oxygens of the sulphonyl group of the
`diazaborine (2.1 and 2.6 Å, respectively). Therefore, resis-
`tance to diazaborine can be explained by the serine side-
`
`FIG. 7. Proposed catalytic mechanism of reduction of the double
`bond in an enoyl substrate by ENR.
`
`combination of isomorphous replacement and molecular
`replacement using the B. napus ENR structure (Protein
`Data Bank entry 1ENO; [16]) as a search model [17]. A
`notable feature in the structure of this enzyme is a region of
`10 amino acid residues from Leu195 to Met206 that forms
`a disordered loop between strand b6 and helix a6, which
`borders the nucleotide binding site (Fig. 6B). In the B.
`napus structure, residues in the equivalent region (residues
`Ala240 to Thr249) have high temperature factors and have
`been implicated in substrate binding [16]. Therefore, this
`disorder may reflect the fact that the acyl substrate is not
`present in the crystals of the E. coli enzyme.
`The ENR subunit comprises a single domain of dimen-
`sions 45 3 48 3 50 Å and is formed from seven b-strands
`(b1–b7), creating a parallel b-sheet with seven flanking
`a-helices (a1–a7). The b-sheet is flanked on one side by
`helices a1, a2, and a7 and on the other by helices a3, a4,
`and a5 with a6 sitting along the “top edge” of the b-sheet
`above the COOH-terminal ends of strands b6 and b7. This
`fold is highly reminiscent of the Rossmann fold commonly
`found in dinucleotide binding enzymes [18]. The cofactor is
`bound in a similar and extended conformation to that
`observed in other dehydrogenases at the COOH-terminal
`end of the b sheet with the nicotinamide ring lying deep in
`a pocket on the enzyme surface.
`A plausible mechanism for the catalytic activity of ENR
`has been described previously [16]. This involves the
`hydride transfer from the C-4 position of the NADH to the
`C-3 position at the double bond in the enoyl substrate. This
`leads to the formation of an enolate anion intermediate
`that can then be protonated on the oxygen to form an enol;
`subsequent tautomerization of the enol would then lead to
`the production of the reduced acyl product. Residues
`implicated in this mechanism include Lys163 whose role is
`thought to be to stabilize the positive charge of the
`transition state and Tyr156, thought to be the proton donor
`to the enolate anion (Fig. 7). Both of these residues are
`conserved in the sequences of the ENRs from E. coli, M.
`tuberculosis, B. napus, Anabaena spheroides, and Haemophilus
`influenzae.
`
`ANACOR EX. 2009 - 7/10
`
`

`
`1548
`
`C. Baldock et al.
`
`chain of the mutant encroaching into the drug-binding site
`and causing severe steric hindrance. In B. napus ENR, the
`equivalent residue to Gly93 is an alanine, and the B. napus
`enzyme is insensitive to diazaborine, presumably because a
`steric clash with the methyl group of the alanine prevents
`diazaborine from binding. In M. tuberculosis ENR, the
`Ser94Ala mutation, which leads to resistance to isoniazid
`[19], maps to a similar region of the structure as Gly93 in E.
`coli ENR. However, in contrast to the evidence supporting
`the steric mechanism responsible for diazaborine resistance
`in E. coli ENR, Dessen and coworkers [11] proposed that
`the serine to alanine mutation in M. tuberculosis ENR
`causes the disruption of a hydrogen-bonding network asso-
`ciated with the cofactor binding, which leads to a lower
`affinity for the cofactor and a consequent reduction in the
`binding of the isoniazid-derived inhibitor.
`
`MECHANISM OF DIAZABORINE ACTION
`Analysis of the drug complex showed that the distance
`between the boron atom of the diazaborine and the 29OH
`of the nicotinamide ribose was approximately 1.7 Å, com-
`parable with a B—O covalent bond length of 1.6 Å. The
`quality of the electron density map (particularly for the
`thieno-diazaborine complex at 2.2 Å) implies that the
`errors in coordinates are very small, and thus the interac-
`tion between these two atoms is covalent. This is further
`supported by continuous electron density between the
`29OH of the nicotinamide ribose and the boron and the
`unambiguous identification of the position of the hydroxyl
`oxygen to which the boron is linked, which can be seen to
`form part of a tetrahedral, rather than a trigonal, arrange-
`ment as required if the boron forms four covalent bonds
`(Fig. 6F). This finding provides a clear explanation for the
`strong inhibitory properties of the diazaborines and for the
`requirement of NAD1 for diazaborine binding. The forma-
`tion of a covalent bond by the boron atom in diazaborines
`is similar to the mechanism of the boronic acid inhibitors of
`serine proteases, which chemically modify the active site
`serine to give a tetrahedral adduct [20].
`The position of the aromatic bicyclic ring of the diaza-
`borine above the nicotinamide ring strongly resembles the
`proposed model for the binding of the enoyl substrate
`suggested from studies on B. napus ENR [16] with the
`proposed position for the negatively charged oxygen of the
`enolate anion of the substrate close to that of the boron
`atom in the drug. The amino group of the putative catalytic
`lysine (Lys163) of ENR is only 4.1 Å from the boron atom,
`and, therefore, this residue may afford partial stabilization
`of the negatively charged boron, in a manner similar to its
`proposed role in the stabilization of the enolate anion
`during catalysis. Thus, it is clear that the inhibitor action of
`the diazaborines derives,
`in part,
`from their structural
`resemblance to the substrate of the enzyme.
`The formation of a covalent bond between the enzyme-
`bound NAD1 and diazaborine generates a tight, non-
`covalently bound bisubstrate analogue. In this respect,
`
`diazaborines are similar to inhibitors of pyridoxal phos-
`phate-containing enzymes (e.g. gabaculine), which co-
`valently modify the cofactor [21]. The best analogy is
`perhaps with 5-fluoro-2-deoxyuridylic acid, which acts as a
`potent inhibitor of thymidylate synthase [22]. This inhibi-
`tor also modifies one of the substrates (the deoxynucleo-
`side) to covalently modify the other substrate (methylene
`tetrahydrofolate) to form a bisubstrate analogue. Potent
`bisubstrate inhibitors of other enzymes with nucleotide
`substrates have also been described (for example, the
`polyoxin inhibitors of chitin synthase [23] and various
`synthetic inhibitors of protein kinase C [24]). Hitherto, no
`such good bisubstrate inhibitors have been described for
`NAD(P)-dependent oxidoreductases. Examples of this type
`designed to inhibit 3-hydroxy-3-methyl glutaryl CoA re-
`ductase were only very weak inhibitors of cholesterol
`biosynthesis, possibly not only because of the lack of a
`moiety to mimic the adenosine diphosphoribose but also,
`quite probably, because of steric problems in the active site
`associated with the nature of the linkage that utilized the
`C4 atom of the nicotinamide ring [25]. Therefore, an
`important feature of the diazaborine study is that it indi-
`cates the type of linkage that might be used to create a
`bisubstrate analogue with the necessary geometry to occupy
`the active site cleft within a pyridine nucleotide-dependent
`enzyme. This may prove crucial in the design of a new
`generation of antibacterial agents against a range of drug-
`resistant organisms including M. tuberculosis. Furthermore,
`the broad spectrum antibacterial activity of diazaborines
`against organisms such as Enterobacter, Neisseria gonor-
`rhoeae, Proteus, and Salmonella [2] suggests that bisubstrate
`analogues designed from the structure/activity profiles of
`the diazaborines may be effective as broad spectrum anti-
`biotics and may even be targeted against organisms such as
`the multi-drug-resistant strains of staphylococcus that are
`proving to be a problem for the current range of antibiotics.
`However, given the inherent toxic potential of boron [2], it
`is obviously important to consider the possibility of design-
`ing a pre-formed bisubstrate analogue substituting the
`boron for another atom in order to develop a range of
`inhibitors with minimal side-effects.
`
`GENERIC APPLICATIONS
`The similarities in chemistry catalysed by the family of
`NAD(P)-dependent oxidoreductases give rise to generic
`opportunities for the creation of a series of novel enzyme
`inhibitors based on a related chemistry. Across the family of
`enzymes that belong to this class, the catalytic cycle of
`oxidation/reduction necessarily leads to a situation where
`the p electron system of a substrate approaches the face of
`the nicotinamide ring. Furthermore, the glycosidic bond
`between the nicotinamide ring and its associated ribose
`moiety generally adopts only one of two conformations that
`differ by a rotation of approximately 180° and that lead to
`the presentation of either the pro-4R or pro-4S hydrogen of
`the NADH to the active site. These conformations merely
`
`ANACOR EX. 2009 - 8/10
`
`

`
`Mechanism of Action of Diazaborines
`
`1549
`
`result in a shift in the position of the carboxyamide moiety
`of the nicotinamide ring on the enzyme surface and do not
`affect the relative positions of the nicotinamide ring to its
`ribose group. Moreover, the structural analysis of members
`of this family has shown that the active site in these
`enzymes is consistently positioned in the same relative
`orientation to the nicotinamide ribose. Thus, for the subset
`of dehydrogenases where there is sufficient space in the
`structure around the 29OH group of the nicotinamide
`ribose, there is an excellent opportunity to mimic the
`chemistry seen in the diazaborine–NAD1 complex in the
`synthesis of new enzyme inhibitors. A number of NAD(P)-
`dependent oxidoreductases are known to be drug targets,
`including dihydrofolate reductase, the target for the anti-
`cancer agent methotrexate [26, 27], steroid 5a-reductase,
`the target for finasteride, used to treat benign prostatic
`hyperplasia [28], and inosine monophosphate dehydroge-
`nase (IMPDH), the target for the immunosuppressant,
`mycophenolic acid (MPA) [28]. For example, there is
`considerable similarity between the ENR–NAD1– diazabo-
`rine complex and the orientation of the inosine-59-mono-
`phosphate thioimidate intermediate and the inhibitor
`MPA in IMPDH [29], suggesting that the 29-hydroxyl of
`the inosine may be linked to the inhibitor in a manner
`similar to that seen in the diazaborine–NAD1 complex.
`This suggests that the utilization of the ribose hydroxyl to
`create a bisubstrate analogue might find important applica-
`tions in other areas of medicinal chemistry.
`
`This work is supported by grants from the Biotechnology and Biological
`Sciences Research Council (BBSRC). C. B. is funded by a Zeneca
`Agrochemicals supported CASE award. J. B. R. is a BBSRC David
`Phillips Research Fellow. The Krebs Institute is a designated BBSRC
`Biomolecular Science Centre.
`
`References
`1. Hogenauer G and Woisetschlager M, A diazaborine deriva-
`tive inhibits lipopolysaccharide biosynthesis. Nature 293:
`662– 664, 1981.
`2. Grassberger MA, Turnowsky F and Hildebrandt J, Preparation
`and antibacterial activities of new 1,2,3-diazaborine deriva-
`tives and analogues. J Med Chem 27: 947–953, 1984.
`3. Turnowsky F, Fuchs K, Jeschek C and Hogenauer G, envM
`genes of Salmonella typhimurium and Escherichia coli. J Bacteriol
`171: 6555– 6565, 1989.
`4. Egan AF and Russell RRB, Condition

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket