throbber
Boronic Acid Compounds as
`Potential Pharmaceutical Agents
`
`WenqianYang,XingmingGao,BingheWang
`
`Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695-8204
`
`Published online 00 Month 2003 in Wiley InterScience (www.interscience.wiley.com).
`DOI 10.1002/med.10043
`
`!
`
`Abstract: Boronic acid compounds have been used, because of their unique structural features, for
`the development of potent enzyme inhibitors, boron neutron capture agents for cancer therapy, and
`as antibody mimics that recognize biologically important saccharides. Consequently, there has
`been a surge of interests in boronic acid compounds. This study reviews the recent development in
`this area during the last six years. ß 2003 Wiley Periodicals, Inc. Med Res Rev, 23 No. 3, 346–368, 2003
`
`Key words: boronic acid; enzyme inhibitor; boron neutron; capture agent; antibody mimics; drug
`delivery
`
`1. INTRODUCTION
`
`Recently, there is an increasing interest in boronic acid compounds. Such an interest stems from
`the tremendous importance of boronic acids in the synthesis of biologically active compounds and
`the use of boronic acid themselves as pharmaceutical agents. In the area of synthetic medicinal
`chemistry, boronic acids are important intermediates that have been widely used in Suzuki cross-
`coupling reactions,1 protection of diols,2 Diels-Alder reactions,3 asymmetric synthesis of amino
`acids,4 selective reduction of aldehydes,5 carboxylic acid activation,6,7 and as a template in organic
`synthesis.8 As potential pharmaceutical agents, boronic acids have been used for the development
`of enzyme inhibitors;9 boron neutron capture therapy (BNCT) agents,10 feedback controlled drug
`delivery polymers,11 saccharide sensors,12–14 and the antibody mimics for cell-surface polysacchar-
`ides.15,16 This review will focus on the recent development in the last six years in the development of
`enzyme inhibitors, BNCT agents, and polymers used for feedback controlled delivery insulin. There
`have been several reviews published in the area of saccharide sensors.13,14 Therefore, this part will not
`be duplicated here.
`
`Correspondence to: Professor BingheWang,Ph.D., Department of Chemistry, North Carolina State University, Raleigh,NC 27695-
`8204. E-mail: binghe _ wang@ncsu.edu
`Contract grant sponsor: National Institutes of Health; Contract Grant numbers: NO1-CO-27184, CA88343,
`DK55062.Contract grant sponsor: North Carolina Biotechnology Center; Contract Grant number: 2001ARG0016.
`
`Medicinal Research Reviews, Vol. 23, No. 3, 346 ^368, 2003
`ß 2003 Wiley Periodicals, Inc.
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 1 of 23
`
`

`
`BORONIC ACID COMPOUNDS
`
`*
`
`347
`
`2. GENERAL PROPERTIES OF BORONIC ACID COMPOUNDS AND
`THEIR IMPLICATIONS IN BIOLOGICAL APPLICATIONS
`
`The utility of boronic acid compounds as pharmaceutical agents is directly related to their unique
`electronic and physicochemical properties. Boron occupies a special place in the periodic table. It is in
`the same period as carbon, but has one less electron. Therefore, it has many similarities with carbon in
`terms of structural features, which make it very useful in the world of carbon in organic and medicinal
`chemistry. The fact that there are many boron-based reagents in organic synthesis reflects this
`structural similarity.17 In medicinal chemistry, the use of boronic acids as enzyme inhibitors to a large
`degree reflects the usefulness of boron as a carbon analog in the binding process, but not in terms
`of reactions, which is the essence of a good enzyme inhibitor. Boronic acids have been used for
`the development of enzyme inhibitors of peptidases/proteases, proteasomes, arginase, nitric oxide
`synthase (NOS), as well as transpeptidases.
`One unique property of boronic acid is that it is a strong Lewis acid because of the boron open
`shell. Most phenylboronic acids have a pKa in the range of 4.5–8.818 depending upon the phenyl
`substitution.19,20 This means that with the appropriate substitution, boronic acids would have the right
`property for ready conversion from a neutral and trigonal planar sp2 boron to an anionic tetrahedral sp3
`boron (Scheme 1) under physiological conditions. Realizing that the process of cleaving an amide
`bond also requires the conversion of an sp2 carbonyl carbon to a tetrahedral sp3 carbon, it is easy to
`understand that boronic acid compounds would make good transition state analogs for the inhibition
`of hydrolytic enzymes. This is indeed the case. Some 20 years ago, simple alkyl or arylboronic acids
`were recognized as serine protease inhibitors.21 –23 Since then, many boronic acid compounds with an
`appropriate peptide sequences have been designed and synthesized for the development of more
`potent and selective inhibitors.24 When compared with aldehyde-based inhibitors of hydrolytic
`enzymes, the ready conversion of boronic acids to their anionic sp3 form seems to make them better
`transition state analogs.25 Although not the emphasis of this review, it needs to be noted that Matteson
`et al. has established a general synthetic route to chiral a-aminoalkylboronic acid derivatives by
`stereoselective homologation of pinanediol boronic esters.25,26 This enabled the synthesis of many
`potent boronic acid-based enzyme inhibitors. Thereafter, several variations of the general route have
`been developed and used for the synthesis of different kinds of enzyme inhibitors.27 –31
`In addition to being developed as enzyme inhibitors, boron-based compounds (not limited to
`boronic acid compounds although that is the focus of this review) are also being studied for their
`utility as BNCT agents.10,32,33 Such applications are based on the unique property of boron-10,
`which emit a particles upon irradiation with neutron. Since a particles do not travel a long distance
`(a few millimeter), they are ideal for localized radiation therapy. Therefore, targeted delivery of high
`concentrations of boron agents can be used for BNCT of certain tumor.
`The third potential application of boronic acid compounds to be discussed is the development of
`feedback controlled delivery systems for insulin. For such an application, the ideal controlling signal
`is glucose concentration. Boronic acids have the unique properties of forming reversible complexes
`with diol-containing compounds such as sugars.19,34 Therefore, efforts have been made to prepare
`polymers that can respond to glucose concentration variation with permeability changes. Such perme-
`ability changes can in turn be used for controlling the release of insulin from the polymer encapsulation.
`Again, this review will focus on using boronic acid compounds for the development of enzyme
`inhibitors, BNCT agents, and feedback controlled delivery systems for insulin.
`
`Scheme1. Conversion from a neutral and trigonal planar sp2 boronto an anionic tetrahedral sp3 boron.
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 2 of 23
`
`

`
`348
`
`*
`
`YANG, GAO, AND WANG
`
`3. BORONIC ACID COMPOUNDS AS ENZYME INHIBITORS
`
`As discussed in the Introduction, the use of boronic acid compounds as enzyme inhibitors is mostly
`based on their easy conversion between the trigonal and tetrahedral forms (Scheme 1), which make
`them ideal transition state analogs in hydrolytic processes. Therefore, various boronic acid com-
`pounds have been widely studied for their inhibition of hydrolytic enzymes such as proteases.
`The following discussion is divided based on the target enzymes.
`
`A. Serine Protease Inhibitors
`
`Thrombin, as the final serine protease in the blood coagulation cascade, is a promising target for the
`development of an anticoagulant agent. Therefore, there is a great deal of interest in the development
`of thrombin inhibitors. Boronic acid compounds were found to exhibit potent inhibition activities.35
`Recently, through the examination of the X-ray crystal structure of boropeptide (1) bound to
`thrombin, it was found that the 3-phenylpropionyl chain attached to the proline residue forms a
`favorable edge-to-face interaction with the Trp-215 side chain located at the base of the S3 specificity
`pocket of thrombin36 (Figs. 1 and 2). To maximize this edge-to-face interaction, rigidified analogs of 1
`and 2 were designed. In such a design, a cyclohexane ring (3) or a pyrrolidine ring (4) was used to hold
`the phenylpropionyl moiety in an orientation favorable for the interaction with the Trp-215 residue as
`predicted by computer modeling studies based on the X-ray crystal structure. Both constrained
`analogs 3 and 4 showed a twofold increase in potency relative to their unconstrained counterparts
`1 and 2, respectively. In a related effort to maximize the edge-to-face interaction with the Trp-215 side
`chain, the P3 residue of 5, a previously discovered inhibitor, was replaced by benzoic acid-derived
`residues. This afforded an extremely potent thrombin inhibitor, compound 6, which is approximately
`threefold more potent than the lead compound (5).37
`
`Figure1
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 3 of 23
`
`

`
`BORONIC ACID COMPOUNDS
`
`*
`
`349
`
`Figure2. Thebindingofcompound1tothrombin.Three expectedinteractions are shown: (a) interactionoftheaminoside chainwith
`Asp-189 in the S1specificity pocket; (b) tetrahedral complex between the hydroxyl of Ser-195 and the boron of the inhibitor; (c) the
`edge-to-face interaction of the 3-phenylpropionyl (P3) residue withTrp-215 located at the base of the S3 specificity pocket.
`
`One concern with the use of thrombin inhibitors as anti-coagulants is the non-specific inhibition
`of other related enzymes. Earlier studies from DuPont Pharmaceuticals identified DuP-714 as a very
`potent thrombin inhibitor with a Ki of 0.07 nM. However, animal studies indicated that DuP-714
`caused side effect that appears to be related to the undesirable inhibition of complement factor I.
`To design inhibitors with minimal interaction with factor I, it was important to analyze the difference
`in the binding requirements between factor I and thrombin. However, the crystal structure of factor I
`was not available. Therefore, the crystal structure of factor Xa (fXa) was used, working on the
`assumption that the overall conformation of factor I is similar to that of fXa. Crystal structural
`analyses of the inhibitor–enzyme complexes with different inhibitors showed that there were very
`noticeable differences in the P2 pocket. Therefore, a series of b,b-dialkylphenethylglycine P2 analogs
`of DuP-714 were designed and synthesized. These compounds, such as 7 and 8, have greater
`selectivity for thrombin over factor I and improved safety profile.38
`There have also been efforts in designing selective thrombin inhibitors by varying the P1 position.
`For example, incorporation of m-cyano-substituted phenylalanine boronic acid analogues into R-
`(D)Phe-Pro-OH dipeptides produced several highly effective thrombin inhibitors such as H-(D)Phe-
`Pro-boroPhe(m-CN)-OH.39 The cyano group enhances binding by several orders of magnitude.
`Because of its structural and functional similarities with thrombin, trypsin was used as a surrogate
`in the crystal structural studies. The trypsin-H-(D)Phe-Pro-boroPhe(m-CN)-OH (Ki ¼ 0.48 nM)
`complex showed that the aromatic side chain was bound in the P1 binding site and that the cynao group
`acted as a H-bond acceptor for the amide proton of the Gly-219.
`
`Figure3
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 4 of 23
`
`

`
`350
`
`*
`
`YANG, GAO, AND WANG
`
`Figure4
`
`In a separate study, based on the known potent inhibition effect of hirudin on thrombin, a novel peptide
`boronate as thrombin inhibitor was designed and synthesized using solid phase chemistry and suitably
`protected aminoboronates.40 By conjugating a boronic acid moiety with a hirudin-based recognition
`moiety, [-D-PheProBoroBpgOPin]-CO(CH2)3COCly2Hir was synthesized and shown to have a very
`high affinity for the target enzyme (Ki ¼ 0.6 nM). It has a 10-fold higher potency relative to the
`corresponding non-hirudin-containing portion Z-D-PheProBoroBpgOPin (9) or the mixture of non-
`covalently linked units.
`Factor Xa (fXa) is another important protease in the coagulation cascade, which occupies
`the juncture of the intrinsic and extrinsic clotting pathways. The physiological role of fXa is the
`proteolytic cleavage of prothrombin to thrombin. Therefore, development of inhibitors against
`fXa should be an attractive method of thrombosis prevention. During the screening of a series of
`conformationally restricted boropeptide thrombin inhibitors, a borolysine compound (10) containing
`a 2-(2-cyanophenylthio)benzoyl in the P3 position was found to be a potent fXa inhibitor.41 It has a
`16-fold higher potency relative to the corresponding compound 11 without the nitrile moiety.
`The serine proteases subtilisin Carlsberg and a-chymotrypsin are commercially available
`enzymes for which high-resolution X-ray crystal structures are known. Therefore, they are good
`targets for probing the factors responsible for determining the structural and stereospecificity of
`enzymes toward unnatural substrates and inhibitors. Enantiomeric 1-acetomido boronic acid analogs
`of the L- and D-forms of alanine, phenylalanine, p-fluorophenyalanine, p-chlorophenylalanine, and
`1-naphthyanaline were synthesized and evaluated as inhibitors of the serine proteases subtilisin
`Carlsberg and a-chymotrypsin.42 All of the boronic acids examined are powerful competitive inhibi-
`tors of both enzymes. The L-enantiomers are generally more potent than the D-enantiomers. However,
`[1-acetamido-2-(1-naphthyl)ethyl]boronic acid showed a dramatic reversal of the normal stereo-
`selectivity preference with the D-enantiomer being 25-fold more potent than the L-enantiomer.
`Molecular modeling analyses of the possible binding modes of the inhibitors indicate that the
`stereoselectivity reversal is because of S1-pocket orientation differences between the naphthyl group
`and the aromatic chains of the phenylalanine analogs.
`To explore the possibility of forming a peptide boronate adduct in the serine protease active site
`that mimics the first tetrahedral intermediate in the peptide hydrolysis mechanism, peptidyl boronic
`acids (12), (13), and (14) were designed and synthesized.43 This design intended to take advantage of
`an intramolecular process hoping to overcome the inherent disadvantage of ternary adduct formation
`(Fig. 5a) by tethering P0 components to the peptidyl boronic acid (Fig. 5b). The complex boronates
`thus prepared are potent inhibitors of a-chymotrypsin. However, the affinity of 12 is neither time- nor
`pH-dependent, which would be expected for a covalent inhibitor, and it only shows a moderately
`increase in affinity compared to compounds 15, 16, and 17 that can not form a diester adduct. These
`results do not follow the predictions. The authors suggested that either boronate ester formation
`was not occurring, or that the energy derived from binding of the S0 binding fragment and boronate
`ester formation was not sufficient to offset the flexibility or binding characteristics of the linking
`group. They thus believed that a more detailed structural investigation was needed to confirm this
`interpretation.
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 5 of 23
`
`

`
`BORONIC ACID COMPOUNDS
`
`*
`
`351
`
`Figure5. Proposedbindingofserineproteasein (a) aternarycomplex withaboronicacidand (b) withanintramolecular tetrahedral
`boronic acid adduct.
`
`The bacterial b-lactamases catalyze the degradation of b-lactam antibiotics through an incredibly
`efficient hydrolysis of the lactam bond, which lead to antibiotic resistance to the b-lactam family of
`antibiotics. With bacterial resistance to b-lactam antibiotics continuing to increase, identification
`of new structural classes of b-lactamase inhibitors is of great clinical importance. TEM-1 is a
`representative member of the group 2b or class A b-lactamases that has achieved particular clinical
`notoriety.44 Based on the crystallographic structure of acyl-enzyme intermediate of TEM-1 bound
`to a substrate, penicillin G,45 (1R)-1-acetamido-2-(3-carboxyphenyl)ethane boronic acid (18) was
`designed to mimic the critical interactions observed in the penicillin G/TEM-1 complex and was
`found to be a potent TME-1 inhibitor.46 The structure of the b-lactamase TEM-1 has been solved in a
`complex with boronic acid (18) at 1.7 A˚ resolution, which suggested a novel transition state of the
`deacylation step in the b-lactamase-catalyzed reaction pathway. Using the same strategy, further
`structure-guided incorporation of additional side chain functionalities (20) or hydrogen bonding
`
`Figure6
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 6 of 23
`
`

`
`352
`
`*
`
`YANG, GAO, AND WANG
`
`groups (19) to maximize energetically favorable interactions of the boronate inhibitors and TEM-1
`b-lactamase was investigated.47 As designed, compounds 19 and 20 are highly effective deacylation
`transition state analogue inhibitors. The high-resolution crystallographic structures of these two
`inhibitors covalently bound to TEM-1 showed interesting and unanticipated changes in the active site
`area, including strong hydrogen bond formation, water displacement, and rearrangement of side
`chains, which provided new insights for the further design of this class of inhibitors.
`AmpC b-lactamase is a representative member of the class C b-lactamases that is among
`the most clinically important b-lactamase enzymes. Using the crystallographic structure of the
`m-aminophenylboronic acid–Escherichia coli AmpC b-lactamase complex, several types of boronic
`acid compounds were modeled into the AmpC binding site, and a total of 37 boronic acids were
`evaluated for b-lactamase inhibition. Among these inhibitors, benzo[b]thiophene-2-boronic acid is
`the most potent compound with Ki of 27 nM for AmpC.48 These boronic acid inhibitors were also
`found to potentiate the activity of b-lactam antibiotics. The X-ray crystallographic structure of
`benzo[b]thiophene-2-boronic acid in complex with AmpC was determined for probing the specific
`interactions between the enzyme and the inhibitor.49 The complex structure revealed several
`previously unknown interactions. The inhibitor was found to complement the conserved, R1-amide
`binding region of AmpC. Concerted interactions between one of the boronic acid oxygen atoms,
`Tyr-150, and an ordered water molecule implicate a mechanism for acid/base catalysis and a direction
`for hydrolytic attack in the enzyme catalyzed reaction. Further antimicrobial assays showed that
`benzo[b]thiophene-2-boronic acid significantly potentiated the activity of cephalosporin against
`AmpC-producing resistant bacteria.
`Hepatitis C virus (HCV) is the major cause of transfusion-associated hepatitis and community-
`acquired hepatitis worldwide.50 NS3 serine protease is one of the most intensively studied and best
`understood targets for antiviral therapy against HCV. Derived from previously identified potent
`hexapeptide aldehyde inhibitors of the HCV proteinase,51 a set of analogous boronic acid inhibitors
`were designed and synthesized. A high throughput method of synthesizing such boronic acid
`compounds was developed by using a resin bound diol as a boronic acid protecting group and
`immobilization linking point. Therefore, a hexapeptide boronic acid library was established by
`parallel synthesis using an Advanced Chemtech 496 synthesizer, from which compounds 21
`(Ki ¼ 80 nm) and 22 (Ki ¼ 80 nm) were found to be highly potent inhibitors of the HCV NS3
`protease.52
`
`Figure7
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 7 of 23
`
`

`
`BORONIC ACID COMPOUNDS
`
`*
`
`353
`
`Dipeptidyl peptidase IV (DPPIV) is a membrane-bound serine protease found on the surface
`of a variety of mammalian cells. Its inhibition could cause the suppression of the T-cell-mediated
`immune response both in vitro and in vivo. This enzyme cleaves a dipeptide from the amino
`terminus of polypeptides where the penultimate residue is proline. Based on dipeptides that possess a
`proline or proline mimic in the P1 position, different kinds of inhibitors of this enzyme have been
`developed.53 –55 Among them, several boronic acid dipeptides have been found to be exceptionally
`potent inhibitors. To reveal the structure-activity relationships associated with variations of the P2
`position of the dipeptide inhibitors, a series of proline boronic acid containing dipeptides (Fig. 8) were
`designed, synthesized, and assayed for their ability to inhibit DPPIV.56 It was found that inhibitory
`activity requires the (R)-stereoisomer of boroproline in the P1 position. A number of substituents
`tested, both polar and nonpolar amino acids, are tolerated in the P2 position except the unnatural
`amino acids, a,a-disubstituted amino acids and glycine.
`
`B. ProteasomeInhibitors
`
`The proteasome is a eukaryotic cytoplasmic protease complex that has several distinct catalytic sites.
`It plays a major role in cellular pathways for the breakdown and processing of proteins to peptides and
`amino acids.57 It is not surprising that defects of various components of this enzyme result in a range
`of human diseases including Angelman’s syndrome,58 cervical cancer,58 and Alzheimer’s disease59
`among others. As a result, these components provide attractive targets for therapeutic intervention.9
`The proteasome showing chymotrypsin-like activity was reported to be the first member of a newly
`identified class of threonine proteases. Recently, some selective and novel dipeptide aldehyde
`inhibitors of the chymotrypsin-like activity of the proteasome complex have been reported.60 Based
`on the dipeptide aldehyde inhibitors, a boropeptide compound 25 was designed and synthesized as an
`inhibitor of the chymotrypsin-like activity of proteasome with IC50 value of 8 nM.61
`Using the same strategy, a series of tri- and di-peptidyl boronic acid analogues were designed62
`based on the replacement of the corresponding aldehyde function of previously reported proteasome
`inhibitors.63 Bioassay of these compounds revealed that the incorporation of a boronic acid moiety
`in this series resulted in dramatically enhanced potency compared to the corresponding peptidyl
`aldehyde compounds. The enhancement in potency is presumed to be because of the formation of a
`stable tetrahedral boronic acid intermediate with the N-terminal threonine residue of the catalytically
`active proteasome b-subunits. In addition, compounds such as 2664 (PS-341) offer the advantages of
`low molecular weight and easy to synthesize. Furthermore, these compounds exhibited extremely
`high selectivity for the proteasome over common serine proteases. As shown in Table I, PS-341
`represents at least 500-fold selectivity for the 20S proteasome over 4 other proteases. This selectivity
`is because of the structural feature of PS-341. As a dipeptide, PS-341 does not fulfill the requirements
`of enzymes such as chymotrypsin and elastase for S3 and S4 subsite binding for optimal activity.
`The P1 position of this dipeptide boronic acid with a leucine residue does not match the preference of
`thrombin for basic residues at that position. All these results make compounds such as PS-341 very
`promising as new therapeutics for the treatment of cancer and inflammatory diseases. Many studies
`have been conducted aimed at developing PS-341 as a new agent in cancer therapy.65 –71 This
`compound is currently under Phase I clinical evaluation in advanced cancer patients.
`
`Figure8
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 8 of 23
`
`

`
`354
`
`*
`
`YANG, GAO, AND WANG
`
`Figure9
`
`To characterize the inhibition of peptidyl boronic acids against different forms of proteasomes
`and to explore their structure-activity relationships, a series of di- and tri-peptidyl boronic acids
`(such as (Bz)-Phe-boroLeu and (Cbz)-Leu-Leu-boroLeu pinacol ester) were designed. The peptidyl
`boronic acids were tested on the chymotrypsin-like activity of purified mammalian 20S and 26S
`proteasomes assayed with succinyl-Leu-Leu-Val-Tyr-amidomethylcoumarin as the substrate. The
`inhibition of 20S proteasomes is competitive but only slowly reversible. The Ki values for the best
`inhibitors are in the range 10–100 nM,72 but the compounds tested are much less effective on other
`proteasome activities measured with other substrates.
`
`C. Arginase Inhibitors
`
`Arginase plays a crucial role in the regulation of diverse metabolic pathways such as ureagenesis
`and nitric oxide (NO) biosynthesis. Recently, the synthesis and evaluation of nonreactive arginine
`analogues as possible enzyme inhibitors or receptor antagonists have attracted much attention. The
`X-ray crystal structure of rat liver arginase shows that the trimeric metaloenzyme contains a binuclear
`manganese cluster in the active site of each subunit required for maximal catalytic activity.73
`Using the information of X-ray crystal structure of the ternary arginase–ornithine–borate complex in
`which the manganese-bridging solvent molecule of the native enzyme is displaced by an oxygen of the
`tetrahedral borate anion, the first boronic acid analogue of arginine, 2(S)-amino-6-boronohexanoic
`acid (27), was designed, synthesized, and evaluated as an inhibitor for arginase.74 The inhibitory
`activity of compound 27 against Mn2 þ
`-arginase was evaluated using a radioactive assay to yield an
`IC50 of 0.8 mM. The crystal structure of the complex between arginase and 27 was also determined.75
`It was found that compound 27 binds as the tetrahedral boronate anion that mimics the intermediate of
`a metal-activated hydroxide mechanism. The tight binding and high specificity of compound 27 allow
`for the further study the physiological role of arginase in regulating the NO-dependent biological
`processes. Significant enhancement of nonadrenergic, noncholinergic nerve-mediated relaxation
`of penile corpus cavernosum smooth muscle was observed with compound 27 and these results
`suggested that arginase inhibition sustained L-arginine concentrations for NOS activity. Therefore,
`human penile arginase is a potential target for therapeutic intervention in the treatment of erectile
`dysfunction.
`
`Table I. Enzyme Inhibitory Profile of Compound 26 (PS-341)
`
`Enzyme
`
`20S proteasome
`Human leukocyte elastase
`Human cathepsin G
`Human chymotrypsin
`Thrombin
`
`Ki (nM)
`
`0.62
`2,300
`630
`320
`13,000
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 9 of 23
`
`

`
`BORONIC ACID COMPOUNDS
`
`*
`
`355
`
`Figure10
`
`Another similar boronic acid-based arginine analogue S-(2-boronoethyl)-L-cystein (28), in
`which a sulfur atom was introduced, was also designed and synthesized.76 Biological test showed it as
`a slow-binding competitive inhibitor of arginase with a Ki value of 0.4–0.6 mM. The X-ray crystal
`structure of the arginase-28 complex was also determined, and the structure of the complex revealed
`that the binding mode also mimics the tetrahedral intermediate in the arginine hydrolysis reaction as
`compound 27 does. Similarly, compound 28 also causes significant enhancement of NO-dependent
`smooth muscle relaxation in human penile corpus cavernosum tissue. Further biological studies77
`demonstrated that both compounds 27 and 28 are classical, competitive inhibitors of human type II
`arginase at pH 7.5 with Ki values of 0.25 and 0.31 mM, respectively. However, at pH 9.5, both were
`found to be slow-binding inhibitors of the enzyme with Ki values of 8.5 and 30 nM, respectively.
`It is expected that the pKa of the boronic moiety being8.5.19 Therefore, the compounds are expected
`in the tetrahedral form at pH 9.5 and trigonal form at pH 7.5. This could help to explain the enhanced
`potency at pH 9.5.
`
`D. OtherEnzymeInhibitors
`
`NO displays potent activities in the cardiovascular system as well as in the central and peripheral
`nervous systems. NO and its co-product L-citrulline are produced by the oxidation of L-arginine by
`NOS (Scheme 2). The selective modulation of NO biosynthesis offers the opportunity for therapeutic
`intervention of neurodegenerative diseases, among others. Based on the mechanism proposed for NO
`biosynthesis, two boronic acid analogues (29 and 30) of L-arginine were designed and synthesized as
`potential substrates or inhibitors of NOS.78 The abilities of the boro-L-arginine 29 and 30 to generate
`NO and to inhibit [3H]L-citrulline formation from [3H]L-arg were investigated in the presence of
`purified recombinant neuronal and inducible NOSs. The Na-acetyl derivative 30 did not lead to any
`significant NO formation and poorly inhibited L-citrulline formation (IC50 > 500 mM). However,
`the unprotected boro-L-arginine 29 selectively inhibited L-citrulline formation catalyzed by the
`inducible NOS (IC50 ¼ 50 mM) compared to the neuronal isoform (IC50 ¼ 300 mM). These results
`demonstrated the feasibility of using boronic acid compounds for the inhibition of this enzyme and the
`strict substrate specificity of NOSs.
`Cysteine proteases are involved in many disease processes. Developing effective inhibitors for
`these enzymes is of great pharmaceutical interest. The catalysis mechanism of cysteine protease is
`similar in a number of ways to that of serine protease. Both types of enzymes use a histidine-imidazole
`residue as a proton shuttle and both utilize nucleophilic catalysis. In the case of cysteine proteases, the
`cysteine thiol is the nucleophile, and for serine proteases the serine hydroxyl group is the nucleophile.
`
`Scheme2. Illustration of NO biosynthesis.
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 10 of 23
`
`

`
`356
`
`*
`
`YANG, GAO, AND WANG
`
`Figure11
`
`There were fewer examples of inhibition of cysteine proteases by boronic acid compounds, although
`boronic acids are often very potent transition state analogue inhibitors of serine proteases. Taking
`papain as the target protease, two peptidyl boronic acids (31 and 32) were prepared and assayed as
`potential transition state analogue inhibitors.79 The design of these peptidyl boronic acid substrates
`was derived from the substrate specificity and X-ray structures of papain. However, no inhibition
`could be detected at concentrations up to 10 mM. The reasons for the lack of inhibition were
`investigated with molecular modeling. Molecular mechanics and semi-empirical quantum mechanics
`calculations indicated that the absence of inhibition was because of boronic acid–cysteine protease
`tetrahedral complexes being thermodynamically less stable than their preceding non-covalent
`EI-complexes. In contrast, an analogous boronic acid–serine protease tetrahedral complex was
`calculated to be more stable than its precursor EI-complex because of the oxyanion hole stabilization
`of the tetrahedral intermediate. Therefore, such studies suggest that boronic acid compounds may not
`be good inhibitor candidates for cysteine proteases in general.
`g-Glutamyl transpeptidase (g-GT) is a membrane-associated enzyme that displays a crucial role
`in the metabolism of glutathione and is also a marker for neoplasia and cell transformation. This
`enzyme appears to function analogously to a serine protease. Based on the proposed transition state
`of g-GT, which was very similar to the proposed ternary complex of g-GT with serine and borate,
`L-2-amino-4-boronobutanoic acid (33) was designed and evaluated as a structural analog of the
`putative ternary complex. Compound 33 was found to be a potent inhibitor of the enzyme with a Ki
`value of 17 nM.80 The structural similarity of compound 33 to glutamate implicated that it might
`serve as a substrate for some glutamate-dependant enzymes or receptors. 13C-NMR studies demon-
`strated that transamination of pyruvate by compound 33 yielded alanine in the presence of glutamic
`pyruvic transaminase. Effects of compound 33 on the growth of cultured rat live cell line ARL-15C1
`and ARL-16T2 were investigated. At the high concentration of 1 mM of 33, there was a significant
`reduction in cell count for both cell lines. In contrast, growth inhibition of both cell lines by 10 mM
`compound 33 could be observed only in low cysteine media.
`
`4. BORONIC ACID COMPOUNDS AS BNCT AGENTS
`
`In addition to being used as enzyme inhibitors, boron compounds can also be used in BNCT,
`first proposed in 1936.32 BNCT was based on the unique feature of boron-10 that becomes lithium
`and emits a-particles upon irradiation with neutron. Because a-particles are very damaging and
`only travel a very short distance, they are ideal candidates for localized cancer radiation therapy.10
`Successful application of BNCT requires the development of boron compounds that specifically
`deliver substantial quantities of 10B to the target cells.81 –84
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1060 - Page 11 of 23
`
`

`
`BORONIC ACID COMPOUNDS
`
`*
`
`357
`
`Critical to the development of BNCT is the synthesis of boron-containing compounds that
`selectively target tumor cells. Numerous boron-containing compounds have been sy

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