throbber
TUTORIAL REVIEW
`
`www.rsc.org/csr | Chemical Society Reviews
`
`Molecular recognition of oxoanions based on guanidinium receptors
`
`Pascal Blondeau,a Margarita Segura,a Ruth Pe´rez-Ferna´ndez{a and Javier de Mendoza*ab
`
`Received 1st September 2006
`First published as an Advance Article on the web 9th November 2006
`DOI: 10.1039/b603089k
`
`Guanidinium is a versatile functional group with unique properties. In biological systems,
`hydrogen-bonding and electrostatic interactions involving the arginine side chains of proteins are
`critical to stabilise complexes between proteins and nucleic acids, carbohydrates or other proteins.
`Leading examples of artificial receptors for carboxylates, phosphates and other oxoanions, such
`as sulfate or nitrate are highlighted in this tutorial review, addressed to readers interested in
`biology, chemistry and supramolecular chemistry.
`
`1. Introduction
`
`Nature frequently uses guanidinium moieties to coordinate
`different anion groups. Present in the side chain of the amino
`acid arginine, the guanidinium group forms strong ion-pairs
`with oxoanions such as carboxylates or phosphates in enzymes
`and antibodies, and it also contributes to the stabilisation of
`protein tertiary structures via internal salt bridges, mainly with
`carboxylates.1 Not
`surprisingly, guanidinium-based com-
`pounds are found in many drugs and have been extensively
`used in molecular recognition studies, leading to the design
`and synthesis of various receptors for anions.2
`The capacity of the guanidinium group to bind oxoanions is
`due to its geometrical Y-shaped, planar orientation, which
`directs the hydrogen bonding, and to its high pKa value
`(around 12–13),3 which ensures protonation over a wide pH
`
`aInstitute of Chemical Research of Catalonia (ICIQ), 43007 Tarragona,
`Spain
`bDepartment of Organic Chemistry, Universidad Auto´noma de Madrid,
`28049 Madrid, Spain
`{ Current address: Department of Chemistry, University of
`Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.
`
`range. The positive charge is delocalized over the three
`nitrogen atoms, and four out of the five hydrogen bond
`donors present in the guanidinium group of arginine can
`complement bidentate oxoanion acceptors, along the two
`edges available (Fig. 1). This accounts for the geometrical
`versatility of the binding modes. From the energy point of
`view, binding to oxoanions results from both ion-pairing
`and hydrogen bonding, and this turns out to be a difficult
`challenge in highly polar solvents or in water. In fact, the
`binding energy arises from the difference of the energy released
`by the host–guest
`interactions and the energy penalty
`necessary to remove the solvation shell around the host, which
`is quite high in water.
`In proteins, the guanidinium–oxoanion interaction usually
`occurs inside hydrophobic pockets or in areas of low dielectric
`constant. On the contrary,
`in artificial synthetic systems
`designed to work in water or polar solvents, complexation
`takes place in an environment more exposed to solvation
`effects which compete with the donor and acceptor sites,
`causing a substantial decrease of the binding. This is usually
`overcome by increasing the number of charges or hydrogen
`bond donors or by the design of more sophisticated receptors
`
`Pascal Blondeau was born in Le
`Mans, France in 1978. He
`completed his MSc degree at
`University of Montpellier,
`France,
`in 2002 working on
`hybrid self-organized materials
`applied to transport bio-
`mimetism. He is currently doing
`his PhD under the supervision
`of Prof. Javier de Mendoza
`at ICIQ in Tarragona. His
`research interests involve the
`design and synthesis of chiral
`guanidinium receptors for mole-
`cular recognition of anions,
`enantioselective recognition of
`carboxylic acid derivatives as well as transport of amino acids.
`
`Pascal Blondeau
`
`Margarita Segura was born in Leiden, The Netherlands in 1969.
`She received her MSc degree in chemistry (1993) from the
`
`University of Granada, Spain
`and her PhD degree (1998)
`f r o m t h e U n i v e r s i d a d
`Auto´ noma (Madrid), super-
`vised by Prof. Javier de
`Mendoza, working on mole-
`cular recognition of oxoanions
`from phosphodiesters, uronic
`acids and dipeptides. After
`postdoctoral research at the
`University of Parma, Italy
`(1998–2000) worki ng on
`carbohydrate recognition in
`water in the group of Prof.
`Rocco Ungaro, she again joined
`de Mendoza’s group in Madrid
`as a postdoctoral researcher (2000–2003) leading a project on
`supramolecular donor–acceptor electroactive systems linked by
`multiple hydrogen-bonding. Since 2004 she has been the Group
`Coordinator at Prof. de Mendoza’s group at ICIQ in Tarragona.
`
`Margarita Segura
`
`198 | Chem.Soc.Rev., 2007, 36, 198–210
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
` / Journal Homepage
`
` / Table of Contents for this issue
`
`Eye Therapies Exhibit 2040, 1 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`to a-helical stabilization and sometimes destabilization of
`peptides and proteins.6
`Guanidinium salt bridges play also important roles in
`enzyme active sites. Typical examples are carboxypeptidase
`A,7a creatine kinase,7b
`fumarate reductase,7c and malate
`dehydrogenase.7d
`Protein–protein hetero-dimerization processes are often
`mediated by salt bridges involving arginine on one molecule
`and phosphorylated amino acids on the other. For example,
`phosphorylation of the OH group of a serine residue in a
`receptor enables
`the simultaneous
`interaction with two
`adjacent arginine residues of another receptor. On the other
`hand, phosphorylation of serines (or threonines) adjacent to
`the arginines of the same molecule slows down the attraction
`between the receptors.8
`A related case is the involvement of the guanidinium group
`in cell–cell and cell–matrix adhesion motifs such as the tri-
`peptide sequence RGD (arginine-glycine-aspartate). Adhesive
`proteins like fibronectin, osteopontin, vitronectin and col-
`lagens display the RGD sequence at their cell recognition site
`in extracellular matrices,9 which is recognized by at least one
`member of the structural related integrins, a family of a,b
`hetero-dimeric transmembrane cellular receptors (Fig. 2).10 On
`the cytoplasmatic side of the plasma membrane, the receptors
`connect the extracellular matrix to the cytoskeleton.
`Thus, osteopontin (OPN), a multifunctional phosphorylated
`glycoprotein recognized as a key molecule in a multitude of
`biological processes such as bone mineralization or cancer
`metastasis, contains an integrin-binding RGD sequence. A
`significant regulation of OPN function is mediated through
`post-translational phosphorylation and glycosylation, a pro-
`cess that is essential for osteoclast attachment.11 Osteoclasts
`are cells that actively reabsorb old bones so that a new bone
`may be replaced. Osteoporosis (bone loss) occurs when
`osteoclasts reabsorb bone faster than the osteoblasts cells are
`producing it.
`
`Fig. 1 The guanidinium group of arginine and its two possible
`binding modes with carboxylates.
`
`where the access to the solvent is restricted. In this review,
`several examples on how this has been achieved in natural
`systems and in artificial guanidinium receptors are provided.
`
`2. Guanidinium–oxoanion ion pairs in proteins and
`nucleic acids
`
`2.1. Proteins
`
`the forefront of research
`Protein structure has been at
`studies with the goal of better understanding the function of
`these biomolecules in the chemistry, physiology and pathology
`of the cell. Proteins are remarkably flexible and susceptible
`to the influence of the environment. Both intramolecular
`and intermolecular interactions involving the protein and
`the solvent define the native conformation.4 To perform its
`function, a protein has to fold properly, a task where the
`various intra-protein or inter-protein interactions, as well
`as the interactions of
`the protein with metals or other
`molecules (such as co-factors, lipids or carbohydrates), are
`essential elements of control. An illustrative example of
`misfolding is the prion protein, which results in aggregated
`copies of the protein causing the ‘‘Mad Cow Disease’’ deadly
`condition.5 Hydrogen-bonded salt bridges, such as those
`involving guanidinium–carboxylate, are relevant contributors
`
`Ruth Pe´ rez was born in
`Madrid, Spain in 1975. She
`obtained her degree in chemis-
`try (organic chemistry) at
`the Universidad Auto´ noma
`(Madrid). She was awarded a
`European PhD at
`the same
`university in October 2005
`under the supervision of Prof.
`J. de Mendoza and Prof. P.
`Prados working in the field of
`protein–ligand interactions,
`self-assembly and dynamic
`evolution based on guanidi-
`nium–oxoanion interactions.
`In November 2005 she joined
`Prof. Jeremy Sanders’ group at the University of Cambridge as a
`postdoctoral researcher and her current work concerns molecular
`recognition using a dynamic combinatorial approach.
`
`Ruth Pe´rez-Ferna´ndez
`
`Javier de Mendoza (Barcelona, Spain 1944) is Professor of
`Organic Chemistry at Universidad Auto´noma (Madrid) and
`
`the ICIQ
`Group Leader at
`(Tarragona). He obtained
`MSc and PhD degrees in
`Pharmacy (Barcelona, 1967
`and 1971). He has been
`Assistant Professor (1971–
`1975), Associate Professor
`( 1 9 7 5 – 1 9 8 0 ) a n d F u l l
`Professor at the Universities
`of Barcelona, Bilbao, Alcala´
`de Henares and Auto´ noma
`(Madrid). He pursued a post-
`doctoral stay (1971–1972) in
`Montpellier (France) under the
`supervision of Prof. Robert
`Jacquier and Dr Jose´ Elguero.
`He is Chevalier de l’Ordre du Me´rite of France since 1994 and he
`was awarded in 1999 with the Research National Prize and Medal
`of the Spanish Royal Chemical Society. Javier de Mendoza
`pioneered the introduction of Supramolecular Chemistry in Spain
`and his current research interests range from molecular recogni-
`tion to calixarene chemistry, self-assembly and catalysts design.
`
`Javier de Mendoza
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Chem.Soc.Rev., 2007, 36, 198–210 | 199
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 2 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`(Fig. 3).12 Due to the diversity of binding modes in this
`system, the 39 arginine residues present in the four histone
`proteins forming the nucleosome core may be divided into
`three groups: a first group of 20 arginine residues involved
`in histone–histone interactions not contacting DNA, followed
`by 7 arginines which enter the minor groove of DNA and are
`essential for histone–DNA binding; and a final group of
`12 arginines which show direct guanidinium–phosphate salt
`bridge interaction. Methylation of arginine residues in the
`histone core leads to a conformational change allowing DNA
`transcription.13 In this way, the transcription of genes can be
`regulated.
`
`3. Guanidinium-based artificial receptors for
`oxoanions
`
`reported in the late 1970’s
`first
`Lehn and co-workers
`guanidinium-containing macrocycles for the recognition of
`32 in water.14 The weak association constants
`phosphate PO4
`(Ka 5 50 (1), 158 (2) and 251 (3) M21, pH titrations) can
`be explained in terms of the more delocalised charge of
`guanidinium over ammonium and accounts for the electro-
`static prevailing interaction.
`The guanidinium can be incorporated into a bicyclic
`framework (Fig. 4a) in order to improve its solubility in
`apolar solvents, where the hydrogen bonds are stronger, and to
`avoid the anti conformation, not suitable for hydrogen-
`bonding to oxoanions (Fig. 4b). As a result, the hydration
`of the cation is reduced and the conformational freedom
`restricted. Inserted into a decaline framework, the guanidi-
`nium cation becomes therefore an almost ideal complement
`for oxoanions, since both NH protons are docking sites for
`the two syn lone pairs of the oxoanion. The resulting ionic
`
`Fig. 2 Scheme of the binding of the RGD sequence to integrins in
`cell–cell and cell–matrix adhesion processes.
`
`2.2. Nucleic acids
`
`Proteins that interact with nucleic acids have a key role in
`biological processes. They are necessary for the control of the
`genetic information, replication, packaging and protection.
`Arginine is again essential for the interaction of proteins with
`DNA. In the nucleosome, in which the DNA winds around the
`arginine-rich histone, the amino acid side chains clearly show a
`direct
`interaction with the DNA phosphodiester chains
`
`Fig. 3 Stereoview of a double stranded DNA interacting with arginines of H2A histones along the major groove. (Reprinted with permission from
`Subirana et al.12 Biopolymers, 2003, 69, 432–439. Copyright (2003) Wiley Periodicals, Inc.)
`
`200 | Chem.Soc.Rev., 2007, 36, 198–210
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 3 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`gave Ka 5 7 6 106 M21.17 The crystal structure of an acetate
`salt confirmed the formation of
`two strong symmetric
`(N…O
`hydrogen bonds between the host and the guest
`2.850 A˚ ). This first binding study confirmed the good match
`of oxoanions by guanidinium receptors through ion pair and a
`linear array of hydrogen bonds in apolar solvents.
`for
`We developed receptor 5 (chloride as counterion)
`aromatic carboxylates, but the stability constant with TBA
`p-nitrobenzoate was much lower (Ka 5 1.6 6 103 M21, 1H
`NMR titrations in CDCl3).18 This example illustrates the
`competition with the initial counterion and the importance of
`the counterion in binding strength:
`in this case the tetra-
`phenylborate counterion results in significantly weaker bind-
`ing than chloride. Thus, poorly coordinating counterions such
`as hexafluorophosphate or tetraphenylborate are necessary if
`strong binding constants are desired.
`The strong deshielding of the NH signals in the 1H NMR
`spectrum of 5?p-nitrobenzoate indicates
`the presence of
`hydrogen bonds. Moreover, stacking interactions between
`the naphthoyl side arms and the p-nitrophenyl moiety are
`evidenced by the shifting of the aromatic signals. Despite their
`ionic character, hosts 4 and 5 are insoluble in water but soluble
`in chlorinated solvents. Thus,
`liquid–liquid extractions of
`water solutions of carboxylate salts give quantitatively the ion
`pair in the organic solvent, free from any competing ion.
`
`DD-AA (donor-donor–acceptor-acceptor) hydrogen-bonded
`complex is particularly stable and geometrically well defined.
`Due to the large pKa difference between guanidinium and
`carboxylic acids (ca. 9 pKa units in water) a trans-protonation
`that would destroy the salt bridge and give a less robust AD-
`DA hydrogen bond interaction15 is unlikely, although it could
`occur in non-polar solvents, where the differences in pKa are
`substantially reduced. Finally, C2 symmetry can be introduced
`into the molecule by two stereogenic centres at the vicinal
`atoms, allowing chiral recognition of the oxoanion guest. Such
`a chiral bicyclic guanidinium binding subunit can be con-
`veniently prepared in multigram quantities in nine steps from
`chiral amino acids (asparagine and methionine).16
`The association constant between bicyclic guanidinium
`derivatives and carboxylates are quite high in chloroform or
`apolar solvents. Thus, UV titrations between 4 (tetraphenyl-
`borate salt) and tetrabutylammonium (TBA) p-nitrobenzoate
`
`Hamilton and co-workers synthesised bis-acylguanidinium
`salt 6 as a receptor for phosphodiesters. The binding constant
`with TBA diphenylphosphate (Ka 5 4.6 6 104 M21, measured
`by UV in CH3CN), was one order of magnitude higher than
`with a simpler benzoylguanidinium tetraphenylborate.19 The
`carbonyl groups contribute to the binding in two ways: they
`increase the acidity of the guanidinium NHs (but not to such an
`extent that trans-protonation can occur) and they pre-organise
`the host by intramolecular hydrogen bonds (chelation effect).
`The combination of these two factors and the additional
`hydrogen bonding from the guanidinium groups allows strong
`complexation in more polar solvents, such as acetonitrile.
`Schmidtchen studied guanidinium–carboxylate interactions
`by isothermal titration calorimetry (ITC).20 The isotherm
`
`a) Chiral bicyclic guanidinium receptor. b) Anti and syn
`Fig. 4
`conformations of guanidinium group.
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Chem.Soc.Rev., 2007, 36, 198–210 | 201
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 4 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`binding curve between 7 (bromide) and tetraethylammonium
`acetate in acetonitrile (Ka 5 2.0 6 105 M21) revealed that the
`process was both entropically and enthalpically favourable
`for a 1 : 1 complex. Although thermodynamic parameters
`could be determined in both CH3CN and DMSO, the reaction
`in MeOH produced too little heat to allow quantification of the
`association constant. This result shows that the stabilization of
`the guanidinium–carboxylate is not only due to the strong
`electrostatic interactions (DHu) but also to a favourable release
`of solvent molecules (DSu), which strongly emphasises the
`importance of solvation in host–guest interactions, a factor
`often neglected in receptor design.
`The thermodynamic aspects of dicarboxylate recognition
`by artificial
`receptors with increasingly acidic hydrogen
`bond donor groups such as two ureas (8), thioureas (9), or
`guanidiniums (10 and 11) in polar solvents (from DMSO to
`water) were studied by Hamilton (Fig. 5).21
`As expected, association constants with carboxylate groups
`(12 and 13) increase with hydrogen acidity but are decreased in
`more polar solvents. While guanidinium–carboxylate associa-
`tion in DMSO is enthalpically driven, in more polar solvents
`such as methanol or water the association becomes an
`entropically driven process due to the liberation of solvent
`molecules upon binding.
`Anslyn and co-workers developed receptor 14, with three
`guanidinium moieties into a 1,3,5-triethyl-2,4,6-trimethylben-
`zene preorganized tripod platform,22 showing selective binding
`towards citrate 15 in pure water (Ka 5 6.9 6 103 M21, 1H
`
`NMR titrations). The host was able to complex citrate even
`from a crude extract of orange juice, which highlights its
`selectivity relative to other carboxylates. This receptor shows
`how the solvent competition can be overcome by accumulation
`of hydrogen bond donors (three guanidinium subunits) in a
`suitable fashion.
`
`The same principles inspired Schmuck’s 2-(guanidinio-
`carbonyl)-1H-pyrroles (Fig. 6), designed to complex carboxyl-
`ate groups in highly competitive media, such as water.23
`Whereas the simple guanidinium cation 16 (pKa 5 13) does not
`show any sign of complexation with carboxylates in aqueous
`DMSO,
`the increased acidity of
`the acylguanidinium 17
`(pKa 5 7–8), rises the binding affinity (Ka 5 50 M21). An
`additional hydrogen bond from the pyrrole NH (as in 18)
`increases the association significantly (Ka 5 130 M21) and the
`additional amide group (19) adds a further hydrogen donor
`well oriented to reach the anti oxygen lone pair (Ka 5 770 M21).
`The predicted geometries have been confirmed by X-ray
`
`Fig. 5 Urea- (8), thiourea- (9) and guanidinium-based (10–11) receptors and association data for dicarboxylates 12 and 13 by isothermal titration
`calorimetry.
`
`202 | Chem.Soc.Rev., 2007, 36, 198–210
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 5 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`Fig. 6 Guanidinium cations 16–20 and their carboxylate complexes.
`
`crystal structures. Even dipeptides are bound efficiently in
`water by a receptor such as 20 (Ka 5 54300 M21 for
`Val-Val).24 A similar scaffold has been used in a combinatorial
`approach showing the importance of additional interactions
`caused by the side arm to improve selectivity.
`
`4. Chiral guanidines for the enantioselective
`recognition of carboxylates
`
`Chiral discrimination of anions based on abiotic receptors is
`still an underdeveloped area of supramolecular chemistry.
`Enantiomerically pure compounds are usually obtained by
`asymmetric synthesis, crystallisation of diastereomeric salts,
`kinetic resolution of racemic mixtures or chiral chromato-
`graphy. An interesting alternative to these methods is the
`separation of enantiomers based on the complementarity of a
`receptor. Those processes based on the translocation of a guest
`between immiscible phases
`(chromatography,
`extraction,
`membrane transport) are particularly attractive. If the receptor
`is chiral, one of the enantiomers can be complexed preferen-
`tially and a kinetic resolution could be achieved. Moreover, the
`process needs only a catalytic amount of receptor since it can
`transfer several substrate molecules across the phases, without
`being removed from its own (stationary or liquid) phase.
`In this context, a useful concept, developed for chiral
`chromatography, is the three-point binding rule, which states
`that a minimum of three simultaneous interactions between the
`chiral stationary phase and for instance one of the enantiomers
`are necessary to achieve enantioselection, with at least one of
`these interactions being stereochemically dependent.25 For
`anions, receptors based on ammonium groups, amides, ureas,
`thioureas and guanidinium moieties, as well as porphyrins,
`saphyrins, or metal-containing ligands have been employed.
`Only chiral guanidines aimed at the discrimination of the
`enantiomers of amino acids will be reviewed here.
`The first example of chiral recognition of a carboxylate by a
`guanidinium-based receptor was reported by de Mendoza in
`1989.18 Indeed, compound 5 was shown to extract enantio-
`meric salts of N-protected amino acids, such as tryptophan,
`
`with modest selectivities (up to 17% excess of N-Ac-L-Trp or
`N-Boc-L-Trp were extracted by (S,S)-5 from water to chloro-
`form). 1H NMR titrations of the triethylammonium salts of
`N-acetyltryptophan in CDCl3 gave Ka 5 1000 and 500 M21
`for the L- and D-enantiomers, respectively. For the non
`protected, strongly solvated zwitterionic amino acids, receptor
`(S,S)-21 was designed.26 The compound features non self-
`complementary binding sites for carboxylate (the guanidinium
`function) and ammonium (a crown ether moiety), preventing
`the receptor from internal collapse, and an aromatic planar
`surface (a naphthalene ring) as a third point for additional
`stacking interactions
`(Fig. 7a). Up to 40% of
`racemic
`tryptophan or phenylalanine were extracted by (S,S)-21 from
`saturated aqueous neutral solutions into dichloromethane,
`with a ca. 80% content of the L-enantiomer. Reciprocally,
`chiral host (R, R)-21 extracts mainly D-Trp.
`
`Further guanidinium receptors were then synthesised in
`order to optimise the binding and extraction properties, and
`were tested as membrane carriers (U-tube tests with dichloro-
`methane between two water phases).27 Interestingly, both
`single liquid–liquid extractions and U-tube transport experi-
`ments revealed that 22 and 23 transported Trp with degrees of
`selectivity comparable with 21. This suggests that the aromatic
`naphthoyl group does not play a significant role in the
`discrimination process. Even compound 24,
`lacking the
`potential p–p interaction, was enantioselective, although to
`a lesser extent. Another binding mode was then proposed,
`without participation of the naphthoyl arm, as the outcome of
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Chem.Soc.Rev., 2007, 36, 198–210 | 203
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 6 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`Fig. 7 a) Three-point binding mode for receptor (S,S)-21 and L-Trp.26b b) Two-point binding mode for L-Trp.27 c) Two-point binding mode
`for D-Trp.27
`
`the apolar solvent, causing the overall energy to increase
`(Fig. 7b,c).27
`A series of receptors for N-protected amino acids, bearing
`guanidinium and carbamate moieties anchored to the curved
`and lipophilic surface of cholic acid (compounds 25–27) have
`been reported by Davis and co-workers.28 The chirality is
`provided by the steroidal framework, the guanidinium as well
`as the carbamate groups establishing the ion pair and
`hydrogen bonds with the substrate. All these hosts efficiently
`extract (52–87%) N-acyl a-amino acids from an aqueous phos-
`phate buffer solution (pH 7.4) into chloroform. Compound 25
`showed high enantioselectivity (up to 7 : 1, 1H NMR
`measurements) for several N-acyl a-amino acids although this
`selectivity decreased dramatically for the more hindered N-Boc
`derivatives. On the contrary, chiral discrimination increased
`(9 : 1) with derivatives 26 and 27, carrying the more acidic
`carbamoyl groups.
`The highly lipophilic receptor 28 was then synthesised in
`gram amounts for transport studies with N-acetylphenylal-
`anine either in U-tube bulk liquid membranes (dichloro-
`methane) or with hollow-fibre membrane contactors (2.5%
`octanol in hexane).29 High enantioselectivity and transport
`rates were observed in the U-tubes (27% of N-Ac-Phe
`transported in 24 h with 56% e.e.), as well as with the large
`scale hollow fibre system (ca. 70 equiv. of substrate trans-
`ported after 48 hours) although in this case the initial
`selectivity (ca. 30%) decreased over time.
`Guanidiniocarbonyl pyrrole systems have also been tested
`for enantioselection. Schmuck reported host 29 which was able
`to bind strongly carboxylates in water.23 Despite its flexible
`structure and the fact that it bears only one chiral centre, this
`
`molecular dynamics calculations with explicit solvent mole-
`cules. In this model, binding of D-Trp exposes a highly polar
`area of the receptor (around the crown ether nitrogen) to
`
`204 | Chem.Soc.Rev., 2007, 36, 198–210
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 7 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`receptor showed enantioselectivity towards N-acetylalanine
`(Ka 5 1610 and 910 M21 for N-Ac-L-Ala and N-Ac-D-Ala,
`respectively), a remarkable result considering the small size
`of alanine’s side chain. Curiously, other amino acids with
`bulkier
`side
`chains
`(such as N-acetylphenylalanine or
`N-acetyltryptophan) showed only slight differences in binding
`for both enantiomers.
`More recently, tris-cationic receptors based on the guanidi-
`niocarbonyl pyrrole scaffold were developed by combinatorial
`chemistry. One compound (30, R1 5 R2 5 Lys; R3 5 Phe)
`showed efficient binding to the sequence D-Glu-L-Lys-D-Ala-
`D-Ala-OH (31) with Ka . 104 M21 in buffered water.30 This
`peptide sequence is related to the bacterial peptidoglycan
`that is recognised by the vancomycin family of antibiotics,
`preventing formation of the cell wall.
`
`5. Phosphate, sulfate and nitrate recognition
`
`In addition to carboxylates or phosphodiesters, other oxo-
`anions such as phosphate, sulfate and nitrate are biologically
`relevant31 and chemically challenging to recognise, due to their
`22) and
`weak basicity. At neutral pH, phosphate (as HPO4
`sulfate present a tetrahedral binding mode with two negative
`charges, although nitrate has a trigonal planar binding motif
`with just one negative charge. Thus, two guanidines are
`required for phosphate and sulfate but only one is needed for
`nitrate, among other hydrogen bond donor atoms. The design
`of suitable linkers between the hydrogen donors with optimal
`orientation and maximum participation of host’s lone pairs
`constitutes the major issue in the field.
`
`5.1. Phosphates
`
`Anslyn and co-workers showed metallo-receptor 32 selective
`22) and
`binding for monoprotonated phosphate
`(HPO4
`22) over other anions (such as AcO2, NO3
`2,
`arsenate (HAsO4
`
`HCO32 or Cl2) at biological pH (Ka 5 104 M21 in 98 : 2
`water/methanol, UV/vis and ITC titrations).32 Hosts bearing
`only the Cu(II) centre were less effective (Ka 5 102 M21),
`highlighting the role of the cavity and the presence of the
`guanidinium groups. Thermodynamic data showed that
`22 with guanidinium derivative 32 was
`association of HPO4
`both enthalpically and entropically driven, whereas complexa-
`tion with an ammonium analogue was mainly governed
`by entropy. The different mode of binding was rationalized
`in terms of the different solvation energies of both binding
`groups.
`Ferrocenyl-based receptor 33 gives moderately strong
`
`complexes with pyrophosphate P2O742 (Ka 5 4600 M22,
`50% methanol–water) showing a 2 : 1 host–guest stoichio-
`metry.33 The presence of a redox active subunit (the ferrocene)
`allows its use as an electrochemical sensor for this biologically
`relevant anion. Another receptor binding pyrophosphate in a
`2 : 1 fashion (Ka 5 1.2 6 108 M22 and 1.0 6 104 M21 for 2 : 1
`and 1 : 1 complexes, respectively) is guanidinium 34, contain-
`ing a fluorescent pyrene subunit, which appeared to be highly
`42 over a variety of anions.34 Moreover, 1H
`selective for P2O7
`NMR suggests that the two hosts in the 2 : 1 complex are self-
`assembled through pyrene–pyrene stacking interactions.
`
`Polyanionic messenger inositol-1,4,5-triphosphate (36) was
`recognized by receptor 35 bearing up to six guanidinium
`subunits on top of a pre-organized 2,4,6-triethylbenzene
`platform.35 Steric gearing causes the guanidinium groups to
`converge toward the cavity. As a result, a cleft-like cavity is
`formed. Since 35 has no chromophore, the binding constant
`(Ka 5 2.2 6 104 M21 in a buffered solution, 1.0 6 108 M21 in
`MeOH) was measured by competition with a fluorescent guest
`(5-carboxyfluorescein), which is released in the presence of the
`preferred guest 36.
`
`Schmidtchen designed a urethane-linked bis-guanidinium
`receptor 37 for the binding of ditopic tetrahedral anions.36 A
`binding constant of 106 M21 in water for p-nitrophenyl
`phosphate and cytidine-59-phosphate was determined by 1H
`NMR. Such a high value in a polar solvent was explained by
`the simultaneous complexation of both guanidinium groups to
`the tetrahedral guest.
`Recently, binding studies between 38 and 4 and phosphates
`of different sizes were measured by both 1H NMR and ITC in
`acetonitrile.37 For 38, 1H NMR gave a curve fitting for a 1 : 1
`stoichiometry whereas ITC predicted a 1 : 2 host–guest binding
`model and revealed that the binding was not caused by a large
`enthalpic contribution but to a strong entropic factor instead.
`
`This journal is ß The Royal Society of Chemistry 2007
`
`Chem.Soc.Rev., 2007, 36, 198–210 | 205
`
`Published on 09 November 2006. Downloaded by University of Oxford on 8/23/2022 2:32:36 PM.
`
`View Article Online
`
`Eye Therapies Exhibit 2040, 8 of 13
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`Calorimetry indeed prevents misleading conclusions from
`NMR in cases where rapid interconverting species are in
`equilibrium. Thus,
`introduction of several hydrogen bond
`donors in the receptor scaffold counteracts rather than
`enhances the enthalpic stabilization of the host–guest complex.
`
`Therefore, no detailed studies comparing binding constants
`with sulfates and other anions are available. However, despite
`both phosphate and sulfate being tetrahedral, the latter is less
`basic, thus the affinity for guanidinium receptors is shifted
`towards phosphate.
`In 1996 de Mendoza and co-workers reported on chiral
`bicyclic bis-guanidinium (40 and 41) and tetrakis-guanidinium
`(42) salts whose sulfate counterions, unlike the corresponding
`chloride salts, required hydrogen donors from two different
`molecules to balance the charges and to fully wrap around the
`anion, since the spacer CH2SCH2 is simply too short to use
`guanidines from the same chain (Fig. 8).39 Therefore, two
`subunits are forced to self-assemble orthogonally around the
`tetrahedral anion in a double-helical
`structure
`(sulfate
`helicates). 1H NMR spectra showed large downfield shifts of
`guanidinium NH’s as dimers or tetramers complexed sulfate
`anion. Moreover, ROESY spectra confirmed intermolecular
`contacts due to the folded conformation.
`A recent computational study concluded that for simple
`sulfate–guanidinium interactions several minima of similar
`
`Macrocycle 39, based on the chiral bicyclic guanidinium
`subunit, has been designed by de Mendoza and co-workers to
`afford six strong hydrogen bonds oriented towards its cavity to
`facilitate wrapping around tetrahedral oxoanions.38 Although
`diphenylphosphate was readily extracted from water,
`the
`binding constant could not be measured from the tetra-
`phenylborate salt (Ka . 105 M21) by NMR in CDCl3.
`H

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