Review pubs.acs.org/CR

Glycopolymer Nanobiotechnology Yoshiko Miura,* Yu Hoshino, and Hirokazu Seto Department of Chemical Engineering, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan systems. To accumulate these weak interactions, the biological ligands are usually able to bind in a multivalent way. In particular, it is well-known that saccharide interactions are greatly affected by the multivalent effect.4 Though saccharide− protein interactions with monomeric sugars are weak, with a dissociation constant (Kd) in the order of millimolar, interactions that involve multivalent binding are much stronger with Kd values on the order of micromolar.5,6 For glycolipids, Kd values for the saccharide−protein interactions become smaller with increasing glycolipid ratios, which suggests the contribution of a multivalent effect.7 Dense microdomains of CONTENTS lipids or “rafts” mediates multivalent saccharide−protein 1. Introduction to Glycopolymers 1673 interactions.8 These lipid raft structures are important in 2. Structure and Function of Glycopolymers 1674 various saccharide-mediated cellular interactions.4 The den2.1. Early Work on Glycopolymers 1674 dritic saccharide structure of glycoproteins enables a strong 2.2. Structure of Glycopolymers and the Multiinteraction with lectins via multivalency. Polysaccharides, such valent Effect 1674 as glycan and glycosaminoglycans, also present saccharides in a 2.3. Controlled Polymerization of Glycopolymers 1676 multivalent way. The natural saccharides have multivalent 2.4. Branched Glycopolymers and Glycodenstructures that enable multivalent binding and, hence, strong drimers 1677 interactions.9 2.5. Physical Properties of Glycopolymers 1677 Saccharide−protein interactions are found throughout living 3. Polymer Nanomedicine using Glycopolymers 1678 systems. Cell surfaces are covered by various saccharides. 3.1. Nanomedicine Using Glycopolymer MultiSaccharide interactions are involved in various biological events, valency 1678 including cell−cell adhesion, cell recognition, and cell differ3.2. Glycopolymers that Control Cell Functions 1680 entiation.4 In addition, pathogens, such as viruses and toxin 3.3. Glycopolymers for Biosensing 1680 proteins, infect cells though saccharide−protein interactions. 3.4. Glycopolymers for Drug Delivery and Gene Notable examples are the interactions between influenza viruses Delivery 1681 and sialyl oligosaccharides, the HIV and HIV receptors, cholera 3.5. Glycopolymer Conjugation to Biomacromotoxins and glycolipids, and Shiga toxins and glycolipids. lecules 1683 4. Conclusion and Outlook 1683 Saccharide interactions are also involved in the folding, Author Information 1684 transportation, and quality control of proteins (Table 1). Corresponding Author 1684 Saccharide recognition proteins, known as lectins, usually Notes 1684 have multimeric structures. The lectins from both plant and Biographies 1684 mammalian cells have been studied.10−12 The plant lectin Acknowledgments 1684 concanavalin A (ConA) has been studied thoroughly since the Abbreviations 1684 1970s.10 The detailed ConA structure was first analyzed by XReferences 1685 ray crystallography,13−19 and this triggered the intense investigation of saccharide−protein interactions.20−24 Studies on ConA have provided extensive information on saccharide− protein interactions. ConA has a homo tetramer structure with 1. INTRODUCTION TO GLYCOPOLYMERS four sugar binding sites. Previous studies have revealed that the Molecular recognition plays an important role in living density of mannose (Man) residues and cross-linking via systems.1,2 Representative molecular recognitions in living multiple binding play important roles in the amplification of organisms are saccharide−protein, protein−protein, and saccharide−protein interactions. antigen−antibody interactions. These biological interactions are possible through molecular interactions, including hydroSpecial Issue: Frontiers in Macromolecular and Supramolecular phobic and electrostatic interactions and hydrogen bonding.3 Science Though each individual molecular interaction is weak, the accumulation of several interactions makes the overall effect Received: April 27, 2015 strong, which results in the ability to control complex biological Published: October 28, 2015 © 2015 American Chemical Society

1673

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Table 1. Examples of Saccharide Recognition by Proteins, Cells, and Pathogens lectin

cell pathogen

target

saccharides

concanavalin A (ConA) Ricinus communis agglutinin(RCA)120, RCA60 peanut agglutinin Sambucus nigra lectin selectins (E-, L-, P-) galectins (−1 ∼ −15) hepatocyte, HepG2 macrophage Shiga toxins (E. coli O-157, H-7, etc.) Vibrio cholera toxin hemagglutinin from influenza virus type A for human hemagglutinin from influenza virus type A for avian E. coli type 1 E. coli P-adhesin HIV-gp120−DC-SIGN

α-Man/α-Glc β-Gal/β-GalNac β-Gal Neu5Ac(α2−6)Gal/GalNAc Sialyl LewisX, Sialyl LewisA β-Gal β-Gal, Gal(β1−4)GlcNAc Man Gb3: Gal(α1−4) Gal(β1−4)GlcCer GM1: Gal(β1−3) GalNAc(β1−4) (Neu5Acα3) Gal(β1−4) GlcCer Neu5Ac(α2−6)Gal Neu5Ac(α2−3)Gal α-Man Gal(α1−4)Galβ Man

1990s.42−47 In early work, the glycopolymers were mainly synthesized by radical polymerization, because the radical reaction is orthogonal to the hydroxyl group on the saccharide side chain. Other glycopolymers have been prepared by a polymer reaction using polymers with functional groups, such as poly(L-glutamic acid),48−51 alginic acid,52,53 and poly(Nacryloxysuccinimide).42 The polymers prepared by free radical polymerization show polydispersity but exhibit large enough multivalent effects to function as biomaterials. The Akaike and Kobayashi groups have cultured hepatocytes with lactose substituted polystyrene poly(N-p-vinylbenzyl-Dlactonamide) (PVLA) in vitro.54−61 Hepatocytes are known to have an affinity for β-Gal and LacNAc (Gal(β1−4)GlcNAc) through the ASGP-R, and Gal is considered one of the key factors in the culture of hepatocytes. However, as we described above, saccharide−protein interactions are generally weak, and it has been difficult to use saccharides for cell-culture materials. The strong interactions of glycopolymers have enabled specific cell cultures via the Gal-ASGP-R specific interaction. The Akaike group has also reported hepatocyte specific drug delivery with PVLA in vivo and in vitro.62−64 This group showed that the saccharide structure and density induces a specific response to hepatocytes, which revealed the curious aspect of saccharide−protein interactions.65−67 The function of the saccharides on lectins had been unclear until the 1990s, because it was unknown what role the weak interactions could play. However, knowledge of the effect of multivalency and the use of synthetic glycopolymers has played an important part in clarifying the function of the saccharides. The Whitesides group designed glycopolymers with sialic acid groups that inhibited the hemagglutinin of influenza viruses in vitro.42−47 These glycopolymers inhibited the aggregation of erythrocytes. Glycopolymers were prepared with various sugar densities, and the polymers showed a much stronger inhibitory effect than monomeric sialic acid. The glycopolymers with more modest sugar densities showed the strongest inhibitory effect, and the polymer structure also affected the inhibition. The universality of the polyvalent interaction has been demonstrated using other polyvalent ligands, including the interactions between a polyvalent peptide and a toxin68 and polyvalent vancomycin and bacteria.69

To date, mammalian lectins have not been studied as thoroughly as plant lectins. Molecular recognition using mammalian lectins was first investigated in studies of the galectins.25−28 Galectins have a specific multimeric structure, which recognizes galactose (Gal) and controls the cell functions of apoptosis, adhesion, and migration. In addition, the functions of a mammalian lectin, the asialoglycoprotein receptor (ASGPR), in hepatocytes have been studied in detail.29,30 It has been reported that the ASGP-R interacts with a Gal cluster and is involved in the clearance of red blood cells. The saccharide interaction with the ASGP-R can be applied to hepatocyte cultures to mediate cell adhesion. Previous studies have clearly shown the importance of the multivalent effect in saccharide−protein interactions. To investigate the multivalent effect, the use of multivalent compounds or “glycoclusters” is indispensable, and many groups have reported syntheses of glycocluster compounds. Examples of glycoclusters include liposomes with glycolipids,31 glycocalixarenes,32 glycocyclodextrins,33 glycopeptides,34 and glycopolymers. Among the various synthetic glycoclusters, glycopolymers have been the subject of much attention (Figure 1).35−40 In this review, we define glycopolymers as polymers

Figure 1. Schematic structures of glycopolymers with linear, dendrimer, and star polymer structures.

carrying pendant saccharides. Since glycopolymers have larger valencies than other multivalent compounds, they show the largest amplification effects in molecular recognition. Glycopolymers are able to be prepared as nanomaterials by controlled polymerization. In this section of the review, we discuss glycopolymers and their application for biotechnology.

2. STRUCTURE AND FUNCTION OF GLYCOPOLYMERS 2.1. Early Work on Glycopolymers

2.2. Structure of Glycopolymers and the Multivalent Effect

A glycopolymer carrying lactose was first synthesized in the 1980s by the Kobayashi group,41 and the Whitesides group investigated glycopolymers and multivalent interactions in the

Early studies demonstrated the multivalent effect of saccharides and its practical usefulness. The use of glycopolymers 1674

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Figure 2. Illustration of the concept of the multivalent effect of glycopolymers: (a) comparison of monovalent and multivalent binding, including the definition of cooperativity; (b) multivalent binding of glycopolymers with different DP to tetrameric lectin; and (c) the multiple binding mode of a polyvalent saccharide to a lectin.

Figure 3. Representative monomer structures for the synthesis of glycopolymers for (a) radical polymerization, including living radical polymerization, (b) ring-opening metathesis polymerization, and (c) polymerization with subsequent saccharide addition.

polymerization (DPs) (based on molecular weight). The Kiessling group studied the affinity to ConA of glycopolymers carrying Man with different DPs, and the polymers with higher DPs showed larger binding affinities.70 The Miura group have studied the binding constants of lactose glycopolymers with different DPs to RCA120.71 Glycopolymers with a high DP

contributes to both enthalpy gain and cooperativity increases on binding. Glycopolymers have polyvalent saccharides and show amplification effects (Figure 2).35−40 The multiple binding of glycopolymers can result in a gain in the binding enthalpy. The multiple binding of a polymer to a lectin has been studied using glycopolymers with different degrees of 1675

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

(RAFT) polymerization,91 and single-electron transfer living radical polymerization (SET-LRP)93 have been successfully applied in the synthesis of glycopolymers. In spite of the bulky structure of the monomers, saccharide derivatives of styrene, methacrylate, and acrylate have been polymerized by living radical polymerization. The first reported living radical polymerization of a glycopolymer was performed by Ohno et al.94 PVLA was polymerized via nitroxide-mediated living radical polymerization using di-tert-butyl nitroxide in DMF, with an acetylated monomer (AcVLA). Other groups have also reported the polymerization of glycopolymers with styrene by nitroxide-mediated living radical polymerization.95−101 Chen et al. reported the synthesis of various poly(styryl sugar)s using 2,2,6,6,-tetramethylpiperidine-1-oxyl (TEMPO).97,98 Kakuchi et al. reported the living radical polymerization of glycopolymers with polystyrene, preparing homopolymer, block copolymer, and star polymers with TEMPO.99−101 ATRP is a representative method of living radical polymerization. Ohno et al. also first reported the ATRP of a glycopolymer using a glucose (Glc) methacrylate derivative (3-O-methacryloxyl-1,2,5,6-di-O-isoproylidine-D-glucoside).102 They reported the preparation of the glycopolymer and block copolymer in high yield. Narain and Armes synthesized glycopolymers, using CuBr with a macroinitiator, containing poly(ethylene oxide) (PEO), poly(propylene oxide), and poly(ε-caprolactone).103−105 The conversion was dependent on the solvent system and the macroinitiator, and they were able to polymerize glycopolymers of 2-lactobionamidoethyl methacrylate (LAMA) and 2-gluconamidoethyl methacrylate (GAMA) in aqueous solutions without any protection of the hydroxyl groups. The advantage of ATRP is the versatility, with the choice of different initiators enabling the synthesis of various types of polymers and use of different solvents. The Maynard group reported the glycopolymers with a functional initiator such as a biotin and pyridyl group.36 Much attention has also been paid to the use of surfaceinitiated ATRP (SI-ATRP) to fabricate biointerfaces. SI-ATRP can be used to produce glyco-polymer brushes, which are useful biointerfaces for glyco-chips and cell cultivation.106−109 The densities of the polymer brushes are able to be controlled by the amount of immobilized initiator.106,107 Recently, a novel method of ATRP, SET-LRP, has been developed by the Percec group.110−112 Living radical polymerization using SET-LRP enables a high yield and fast polymerization. The Haddleton group has reported the controlled polymerization of various polymers using SET-LRP, including glycopolymers.113−115 The Haddleton group is investigating glycopolymers synthesized with precise sequence control of the block-glycopolymers, which is an important concept for polymer nanomedicine. RAFT is another important method of living radical polymerization, which is performed with thiocarbonyl additives.91 RAFT polymerization is suitable to the polymerization of bulky monomers, and there have been various report of polymerization of glycopolymers. McCormick et al. first synthesized a glycopolymer of 2-methacryloxyethyl glucoside with a RAFT reagent.116 The Stenzel group reported living radical polymerization of various glycopolymers with the RAFT method. Glycopolymers have been prepared using polyacrylamide,117−122 polymethacrylate,123−126 polyvinylester,127 polyacrylate,128 and polyvinyl triazole129 with Glc, Man, and Nacetylglucosamine (GlcNAc), resulting in a variety of glycopolymer structures, including simple linear structures,

showed larger binding constants than those with a low DP because of the multiple binding to one lectin (Figure 2b). Glycopolymers with a high DP can cross-link lectin and showed a larger binding affinity because of the enthalpy gain with multiple binding. Another important attribute of glycopolymers is the ability to present multiple binding modes to lectins and cells (Figure 2c). Glycopolymers with a shorter DP show larger affinities than monomeric saccharides, even though the polymers only bind to one sugar recognition site. The polyvalent ligand can provide various binding modes compared with monovalent ligands, which is favorable in terms of entropy. The cooperativity entropy effect is affected, not only by the multiple binding mode but also by the ligand spacer, molecular dynamics,72 and polymer backbone flexibility.1,2 Frequently, flexible linkers are connected to the saccharide to increase the affinity.70 Use of a hydrophobic linker, such as benzene, has been reported to increase the binding affinity because of the hydrophobicity.44 In glycopolymers with phenyl isocyanide, the glycopolymers showed low cooperativity because of the rigid polymer structure.73 The Whitesides group defined the concept of the cooperativity of multiple binding as shown in Figure 2a. The Kiessling group investigated the affinity of various glycopolymers70 and highlighted the complexity of multivalent effects. The binding amplification effect of a glycopolymer is not in simple proportion to the number of saccharides and saccharide recognition units. Generally, the observed Gibbs energy change values are smaller than those obtained from a simple calculation based on the saccharide number (Figure 2a). In glycopolymers, the sugar binding efficiencies are not high because of the limited number of sugar binding sites and the sugar motility, which are possible disadvantages in the use of glycopolymers as polyvalent ligands. Despite these known weak points, most of the glycopolymers studied have been reported to amplify saccharide−protein interactions.6,35−40 Though the basic mechanism of the multivalent effect is the same for all of the lectins, the distance between the saccharide recognition sites is unique for each protein. While the distance between the saccharide recognition sites of ConA is 6.5 nm,13−16,74 those of the AB5 toxins in B subunits,75 such as in Shiga toxins76−78 and the cholera toxin79−81 are much shorter ca. 1−3 nm. The number of sugar recognition sites is more than 5 in AB5 toxins. Therefore, glycopolymers easily provide for several binding sites with AB5 toxins, which results in stronger binding.82−85 Similarly, the hemagglutinin of influenza viruses has densely packed trisaccharide recognition sites.86−88 Many lectins exist on cell membranes. The addition of glycopolymers to the cell induces cross-linking and selfassembly of lectins. The assembly of lectins makes a great contribution in terms of a multivalent effect to the control of cell functions and signal transduction.1,54 2.3. Controlled Polymerization of Glycopolymers

Controlled polymerizations have been reported using various methods;89−93 these methods have also been applied to the preparation of glycopolymers. The examples of monomer structures for controlled polymerization reactions are listed in Figure 3. Even though the monomers possess a bulky side chain that causes steric hindrance, recent advances in polymer chemistry have enabled living polymerization. Various living radical polymerization techniques, including nitroxide-mediated polymerization, 92 atom-transfer radical polymerization (ATRP),89,90 reversible addition-fragment chain transfer 1676

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

block polymers125 star polymers,126 nanoparticles,129−131 and graft-polymers.119 More diverse monomers can be used with RAFT than with ATPR and nitroxide-mediated living radical polymerization. Another advantage of RAFT polymerization is the facile modification of the polymer terminal. The polymer terminal thiocarbonyl group of the RAFT reagent is easily converted to thiol, which is available for many useful modifications, including Au−S bond formation, Michael addition with maleimide, the thiol−ene reaction, and disulfide bond formation.132 These modification methods provide for versatile hybrid materials with glycopolymers (Figure 4).

Synthetic nanoparticles of branched glycopolymers are one of the interesting types of polymer for biological applications. Glycodendrimers have been intensively studied as models of glycoproteins. One of the features of dendrimers is a uniform molecular structure and a molecular weight similar to proteins. Aoi et al. first reported the synthesis of glycodendrimers by the modification of polyamidoamine (PAMAM) dendrimers.153 The Roy group has reported the synthesis of various types of glycodendrimers.154 For example, this group has prepared dendritic sialosides with PAMAM dendrimer backbones and investigated the affinity of these dendrimers to a sialic acidrecognizing lectin. The affinity was analyzed by the inhibition of glycopolymer−lectin binding, which was ca. 100 times higher than with the monomeric saccharide in view of IC50.155,156 Since PAMAM dendrimers are commercially available, the synthesis of various glycodendrimers based on this scaffold have been reported. Wolfenden et al. reported the syntheses of αMan modified glycodendrimers from the isothiocyanate reaction with amines and found the affinity of the glycodendrimers to ConA was related to the multivalency of the generated glycodendrimers.157 Modified PAMAM dendrimers, as well as other dendrimers, have been investigated for use in nanomedicine. The various attributes of glycodendrimers, such as size, sugar multivalency, electrostatic properties, self-assembling properties, and protein resistance, have an effect on biological funcions.157,158 These properties of glycodendrimers are a function of the structure of the dendrons. Glycodendrimers with poly(propyleneimine) have been investigated as drug carriers.159,160 The Percec group has reported the synthesis of Janus glycodendrimers and analyzed the resulting selfassembling structures to provide information on specific molecular recognition.161−163 Similar to the preparation of glycopolymers, click reactions have been investigated for glycodendrimer synthesis, which enables facile and precise syntheses without the use of protecting groups.164,165 Hyperbranched and star polymers can also provide spherical multivalent structures. The synthetic techniques to prepare these polymers are based on the synthesis of the linear glycopolymers as discussed in section 2.3. The Stenzel group has reported the synthesis of star glycopolymers.128 The Müller group has reported hyperbranched glycopolymers prepared via ATRP, including nanoparticle and nanocarbon hybrid-glycopolymers.166,167 The Perrier group reported the hyper branched glycopolymers by RAFT living radical polymerization.168 The hyperbranched polymers were easily synthesized by living radical polymerization, using the addition of a branching reagent. Hyperbranched polymers spontaneously form nanoparticles, which are useful in nanomedicine and as drug carriers. The application of these branched polymers to nanomedicine will be described in section 3.

Figure 4. Terminal functionalization of glycopolymer via RAFT living radical polymerization.

Recently, the use of a two-step reaction has been reported, which enables facile and precise glycopolymer preparation. Since the bulky side group of a saccharide hinders the polymerization reaction, it is difficult to use living radical polymerization of saccharide derivatives to provide polymers with a particular design. Hence, monomers with small reactive functional groups are polymerized using living radical polymerization, and the subsequent polymers are then modified by further chemical reactions.133−136 The Haddleton group has reported the living radical polymerization of acetylenesubstituted methacrylate, to which azide-terminated saccharides were added by click chemistry.137−139 The Bertozzi group prepared and polymerized a methyl vinyl ketone, and prepared a glycopolymer by an addition reaction with aminooxyterminated saccharides.140−142 Furukawa et al. reported glycopolymer preparation by the addition reaction of sugars to an aminooxy side chain,143 which is the reverse of the Bertozzi method. Boyer et al. prepared glycopolymers via living radical polymerization of an activated ester [3-(benzylsulfanylthiocarbonylsulfanyl)-propionic acid] and amide formation with an amine-terminated saccharide.144 You et al. also reported glycopolymer synthesis using a thiol−ene reaction.145,146 The combination of living radical polymerization and the addition of saccharides via an orthogonal reaction is a facile method to prepare glycopolymers with well-defined structures. Ring-opening metathesis polymerization (ROMP) is another important method to fabricate well-defined glycopolymers because it is tolerant of bulky side-chains and harsh solvents. The Kiessling group has reported the preparation of glycopolymers via ROMP.147−151 This group prepared various glycopolymers with different molecular weights and densities, which were investigated in terms of the molecular recognition of proteins, cells, and bacteria, including details of the molecular recognition and the control of cell functions. Saccharide addition to polymers via ROMP with succinimide norbornene has also been reported in the synthesis of well-defined glycopolymers.152

2.5. Physical Properties of Glycopolymers

Since glycopolymers have hydrophilicity from the saccharides and hydrophobicity from the polymer backbone, the polymers are amphiphilic. This amphiphilicity induces the self-assembly of glycopolymers in aqueous solutions. Polystyrene-substituted glycopolymers form helical rodlike structures because of the amphiphilicity and chiral properties of the glycopolymers. Wataoka et al. have reported rodlike structures formed by maltopentaose and lactose carrying polystyrene polymers using small-angle X-ray scattering analysis.169−171 Polystyrene carrying lactose (PVLA) glycopol-

2.4. Branched Glycopolymers and Glycodendrimers

Spherical polymer structures of glycopolymers are important for the use of polymers in medicine and drug delivery. 1677

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Figure 5. Self-assembling structures of glycopolymers. (a) Block glycopolymers formed various kinds of self-assembling structures based on the molecular packing parameters and (b) block glycopolymers with amphiphilicity formed nanoparticles.

methacrylate. The resulting self-assembling structures formed nanospheres, wormlike micelles, and vesicles, and have potential applications as drug carriers. The formation of the structures was controlled by the monomer ratio and degree of polymerization. The Stenzel group has reported the synthesis of various block-glycopolymers, including stimuli-responsive glycopolymer micelles.180,181 Other groups have also reported the formation of polymer micelles of block glycopolymers, based on the amphiphilicity and self-assembling properties (Figure 5b).104−108,145,146 Although uniform self-assembled structures of glycopolymers have been seldom reported, many groups have reported the formation of nanoparticles. Dendrimers are also useful as a component of supramolecules. The Percec group has investigated self-assembling dendrimers under various conditions.182 This group has reported the preparation of Janus type glycodendrimers, which formed vesicles in aqueous solution.161−163 The vesicles (glycodendrimersomes) are molecular assemblies with diameters of several hundred nanometers, which were observed to regulate biological signaling with galectins. The molecular interactions could be controlled by altering the structure of the glycodendrimersomes.

ymers formed isotactic structures and showed strong circular dichroism based on the amphiphilicity and chirality of the polymers.172 PVLA polymers formed helical rodlike structures with a hydrophobic cavity. The Miura group reported the nanowire formation with the self-assembling ability of PVLA glycopolymers for nanowire formation.173 The hydrophobic cavity of the glycopolymer incorporated π-conjugated polymers of polythiophene to form the polymer nanowire, which is similar to the complex formation of natural polysaccharides.174 Though glycopolymers with polystyrene form rodlike structure in aqueous solution, the glycopolymers can be adsorbed onto hydrophobic substrates. Akaike et al. reported the coating of PVLA polymers onto hydrophobic polystyrene, which was used to prepare hepatocyte cultures.55 The Miura group investigated the polymer adsorption on the hydrophobic interface.175,176 A glass substrate was made hydrophobic using a silane coupling reagent, and the hydrophobic−hydrophilic micropattern was prepared by photolithography. The glycopolymers adsorbed only on the hydrophobic part. The saccharide micropattern was easily prepared by the simple immersion of the patterned substrate in the glycopolymer solution. The glycopolymer micropattern was applied to the cell cultivation of two kinds of cells. Block copolymers of glycopolymers with hydrophobic polymers have been used for the preparation of polymer micelles.125 It is well-known that lipids form various supramolecules, including micelles, rodlike micelles, vesicles, and hexagonal reverse micelles.177 The Armes group has reported various self-assembled block glycopolymers (Figure 5a).178 The self-assembling structure of lipids is determined by the packing paramters.179 The Armes group synthesized well-defined amphiphilic glycopolymers and investigated in detail the lipidlike self-assembling properties of the glycopolymers, using block polymers with galactose methacrylate and hydrophobic

3. POLYMER NANOMEDICINE USING GLYCOPOLYMERS 3.1. Nanomedicine Using Glycopolymer Multivalency

As mentioned in the Introduction to section 2.2, the multivalency of glycopolymers amplifies the molecular interactions with lectins, bacteria, viruses, and cells. Glycopolymers strongly interact with pathogens and are useful in polymer medicines. In some cases, the affinities of glycopolymers are comparable to those of antibodies and antigens. 1678

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Figure 6. Concepts of glycopolymer nanomedicines and applications.

The inhibitory effect of amyloid β aggregation was dependent on the saccharide density and the molecular weight of the glycopolymer. The inhibition of viruses by glycopolymers has been reported. As mentioned in section 1, the saccharide−protein interaction is related to the infection of viruses.4 The influenza viruses infect cells by the binding of hemagglutinin and neuraminidase to sialyl oligosaccharides on the cell surface. The Whitesides43−47 and Kobayashi48,196−198 groups have reported glycopolymers that interact with influenza viruses. Strong interactions between the hemagglutinin molecules of various types (A, B, C, and avian) of influenza viruses and glycopolymers having polyacrlyamide,43 polystyrene,196 and poly(glutamate)197 backbones have been reported. The Roy group has synthesized glycodendrimers and glycopolymers with sialic acid that bound to the hemagglutinin moieties of various types of influenza viruses,199 where the strongest inhibitory effect was more than 1000 times higher than with the monomeric sialic acid. The glycodendrimer efficiently inhibited infection of the influenza virus in mammalian cells. Another example of saccharide-mediated viral infection is the HIV. Multivalent α-Man on the envelope glycoprotein of the HIV interacts with dendritic cell-specific ICAM-grabbing nonintegrin (DC-SIGN). Becer et al. reported that a glycopolymer carrying α-Man was able to interact with DC-SIGN.200−203 Glycopolymers carrying Man were synthesized by living radical polymerization and click chemistry, and the resulting polymer inhibited the binding of DC-SIGN to gp120. The greatest inhibitory effect was exhibited by the glycopolymer with the highest Man density. Saccharides are able to influence the inflammatory response through binding to selectins. Glycopolymers have been used as selectin inhibitors. Selectins are classified as L-, P-, and Eselectins.4 Miyauchi et al. prepared polyacrylamide polymers carrying sialyl Lewis X, which interacted with E-selectins and inhibited the cell adhesion of HL-60.204,205 Sasaki et al. synthesized polyacrylamide derivatives carrying sialyl Lewis X mimics that bound to L-selectin.206,207 The Chaikof group reported PEO dendrimer-like glycopolymers, which interacted with L-selectin to inhibit U937 cell adhesion.208 The Kiessling group has synthesized glycopolymers with sulfated groups, and the sulfo-Lewis X moieties were shown to interact with selectins.209 Oligosaccharides are needed for the synthesis of glycopolymers that may be useful in medicine. However, the syntheses of oligosaccharides are usually tedious and proceed in low yield. To overcome these synthetic difficulties, multivalent oligosaccharide mimics have been investigated. Sasaki et al. have developed the glycomodule method, where a few different types of saccharides are displayed along one polymer to mimic

Saccharide interactions have already been used in medicines, including in the influenza virus inhibitors, oseltamivir and zanamivir,183 where interactions, such as hydrophobicity and charge, are induced to amplify the affinity. The overall interaction of a glycopolymer with a target is the accumulation of several weak interactions, which is similar to antibody binding. The strong interaction between glycopolymers and target pathogens may be useful in the development of novel polymer-based therapeutics (Figure 6). The molecular recognition of glycopolymers with lectins has been reported by many groups. Interestingly, many lectins are toxic and often pathogenic but are able to be inhibited by glycopolymers, glycodendrimers, glycopolymer micelles, and glycopolymer brushes. For example, β-Gal or lactosesubstituted polymers are able to bind to the deadly biological toxin ricin.184 The Stenzel group has reported a glycopolymer that was able to bind to ricin, where the density and linker structure of the glycopolymer was critical to the ricin binding. Nagatsuka et al. prepared a glycopolymer-conjugate carrying lactose with magnetic nanoparticles and gold nanoparticles, which bound to ricin.185−187 The glycopolymer captured ricin for detection and removed ricin from a solution. AB5 toxins are also toxic and pathogenic. Glycopolymers carrying saccharide ligands efficiently inhibit AB5 toxic proteins, such as Shiga toxins and the cholera toxin.188 Dohi et al. synthesized a glycopolymer carrying a trisaccharide segment of Gb3, which effectively neutralized Shiga toxin-1 and E. coli O157 by multivalent effect.84 Gargano also reported that a glycopolymer with a trisaccharide was able to inhibit Shiga toxin.83 Glycodendrimers with Gb3 trisaccharides have also been reported to inhibit Shiga toxins.189 The inhibition of the Shiga toxins was dependent on the distance between sugars and the linker structure of the glycopolymers. Shiga toxin-1 showed a strong interaction with glycopolymers, independent of the linker structure, but the affinity of the glycopolymers to Shiga toxin-2 was strongly dependent on the linker structure. Glycopolymers carrying Gal have been shown to inhibit cholera toxin,190−192 though the natural ligand is not Gal but GM1. The Kiick group has investigated the cholera toxin inhibition in detail and found that appropriate sugar density (ca. 3.5 nm) is important for strong binding. Richards et al. also found that the interaction between a glycopolymer and the cholera toxin is strongly related to the linker structure of the glycopolymer.193 Inhibition of the cholera toxin was also found to be dependent on the density of the sugar groups. Another toxic protein, the Alzheimer’s amyloid β protein, has been observed to be neutralized by glycopolymers. We have synthesized glycopolymers carrying 6-sulfo-β-D-GlcNAc.194,195 This sulfonated glycopolymer mimics glycosaminoglycan, and inhibited the aggregation of amyloid β, neutralizing the toxicity. 1679

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

oligosaccharide functions.206 Jones et al. have synthesized a glycopolymer with secondary binding motifs that mimics the branched structure of oligosaccharides.210 Most of the studies of polymer medicine were carried out in vitro. Since the glycopolymers have strong activities to the target due to the multivalent effect, glycopolymers have a great potential in vivo for series of novel medicines though they needs clinical experiment. 3.2. Glycopolymers that Control Cell Functions

Figure 8. Glycopolymers with lipid terminals were displayed on the cell surfaces, and saccharide−cell functions were analyzed. The inset shows the representative structure of glycopolymers for glycocalyx engineering. The polymers were synthesized by RAFT living radical polymerization of polyMVK with saccharide addition. Various saccharides with aminooxy groups were conjugated with the polymers.

Most saccharides are present on the cell surface to enable interaction with other cells. Therefore, glycopolymers can be used in cell cultivation and as scaffolds for tissue engineering. In particular, glycopolymers have been investigated for use in hepatocyte cultivation.55,56 The interaction of glycopolymers with hepatocytes induces a round shape because of a strong interaction with the ASGP-R. The Kiessling group has reported precise control of the synthesis of glycopolymers using ROMP,71,150−152 to analyze molecular recognition with lectins and cells. Glycopolymers carrying β-Gal, with different degrees of polymerization, were synthesized and signaling with E. coli was investigated (Figure 7).211−213 The Gal receptor exists on the cell surface, and the

conjugation with DPPE.218,219 Galectin multimerization using glycopolymers has been investigated, and it was found that lactose-substituted glycopolymers in the cell membrane play important roles in galectin-3 multimerization.214 In other research, glycopolymers with GalNAc have been prepared, and it was found that the glycopolymers regulated integrin assembly on the cell surface, which affected cancer cell growth and survival.216 The role of the saccharide has also been investigated using sialic acid modified polymers. PolyMVK was treated with various saccharides, including Sialyl-LacNAc, GD3, and sialyl Lewis X.215 Activation of human natural killer cells (NK) cells was observed to be controlled by the Siglec interaction. A glycopolymer carrying sialic acid was inserted in the cell membrane, and it was found that the interaction of the glycopolymer with Siglec affected the NK cell cytotoxicity. These glycopolymer investigations suggest that multiple sialic acid residues reduce the cytotoxicity of NK cells for cancer cell survival. These results show that glycopolymer technology provides a novel method and an interesting tool to investigate cell functions. There is clear evidence that the addition of certain glycopolymers can control cell viability and functions. This technology has potential in the treatment of cancer and other diseases. These studies are investigated at the level of basic scientific research in vitro, but it is useful to clarify the biological role of saccharides.

Figure 7. Glycopolymer carrying Gal synthesized via ROMP induced clustering of the Gal receptor and chemoreceptor. Clustering of the chemoreceptors suggests intercellular protein interactions.

glycopolymers showed a strong affinity with E. coli because of the multivalent effect with the Gal receptor (lectin) on the cell surfaces. Interestingly, not only lectins on the cell but also other chemoreceptors were affected by the glycopolymers. Lectins on the cell surface interact with other chemoreceptors by inter− receptor communications. The addition of a glycopolymer induced clustering of the lectins and subsequently other chemoreceptors. The migration of the cell was dependent on the DP of the glycopolymer. A single glycopolymer was able to cross-link the lectin and chemoreceptors and induced a motility response and chemotaxis. A fluorescent-labeled glycopolymer to monitor cell behavior has also been prepared. The Bertozzi group synthesized glycopolymers via RAFT living radical polymerization.140−142 The RAFT reagent was conjugated with a phospholipid of 1,2-bis(diphenylphosphino)ethane (DPPE), and poly(methylvinyl ketone) (polyMVK) was prepared by living radical polymerization. Then, saccharides having an aminooxy group were added to polyMVP by a condensation reaction. DPPE-terminated glycopolymers were easily anchored to the cell membrane, which was termed “Glycocalyx engineering” (Figure 8). Glycopolymers with various bioactive saccharides were prepared, and the saccharide function was investigated as an artificial extracellular matrix.214−217 The glycopolymers were inserted in the cell membrane in an extended conformation, because of the

3.3. Glycopolymers for Biosensing

The molecular recognition ability of saccharides can be used as a molecular recognition tool. If the saccharide−protein interaction is strong enough, the interactions can be used to produce bio devices, which function similarly to antibodies. One of the important applications of antibodies is for biosensing and diagnosis; hence, the use of the saccharide− protein interaction in glycopolymers is a promising alternative to antibodies for use in biosensing and diagnosis.220 In addition, saccharides are generally hydrophilic, which inhibits the nonspecific adsorption of protein.221 Biosensors are divided into a sensing part and a molecular recognition unit. The sensing parts are often composed of devices that make use of techniques, such as surface plasmon resonance (SPR), quartz crystal microbalance, field-effect transistor, and other electrochemical techniques. The sensing parts are usually made of substrates, such as gold, semiconductors, and metal nanoparticles. The immobilization of glycopolymers enables biosensing via saccharide−protein interactions. Gold is frequently used as a substrate in biosensing devices. Gold substrates are easily modified by Au−S bond formation. 1680

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Figure 9. Glycopolymer modified gold nanoparticles and application for biosensors: (a) preparation of glycopolymer modified gold nanoparticles via RAFT living radical polymerization and (b) biosensors with glycopolymer modified gold nanoparticles.

polythiophenes with sialic acid had a UV peak at approximately 400 nm, and the addition of influenza viruses (influenza virus A and B) induced a red shift of approximately 10 nm, despite there being no response to the sialic acid recognition lectin. The Bunz group reported accurate lectin and E. coli detection using saccharide-modified poly(p-phenylene ethynylene).234,235 Recently, imaging techniques, such as magnetic resonance and fluorescence have been used for biosensing and diagnosis. The Whittaker group has reported imaging using glycopolymers (Figure 10).236,237 Hyperbranched polymers were synthesized with RAFT living radical polymerization and modified with click chemistry. A 19F label was incorporated with trifluoroethyl acrylate.236 The obtained polymer was used for 19F magnetic resonance imaging. A glycopolymer has also been prepared with magnetic nanoparticles coated with blockglycopolymers carrying Man and Glc.237 The interaction between the glycopolymer and ConA and the cellular uptake of the nanoparticles were monitored by the relaxation times of the magnetic resonance. Pfaff et al. has investigated fluorescent and magnetic imaging with glycopolymer-modified nanoparticles.238 Lu et al. has used fluorescent imaging with BODIPY-labeled glycopolymers carrying Gal.239 Glycopolymer imaging is still in its infancy; however, the magnetic resonance imaging of saccharides has been already applied to biochemical analyses such as brain imaging,240 and glycopolymers are likely to be used as diagnosis tools in the near future. Biosensing is mainly utilized in vitro. Since the saccharide− protein interaction is basically specific, biosensing with saccharides is practical if the interaction is amplified by glycopolymer multivalency. Bioimagings are carried out mainly in vitro and still a little far from practical use in vivo and needs clinical experiments.

As mentioned in section 2.3, thiol-terminated glycopolymers are obtained by RAFT polymerization and degradation of the polymer terminal. We have modified gold substrates and gold nanoparticles with glycopolymers carrying Man, Gal, and GlcNAc and could detect lectins with high affinities by SPR and observed color changes (Figure 9).222,223 The glycopolymers carrying Man (poly(Man-co-acrylamide)) were synthesized by RAFT living radical polymerization, and the polymer terminal was converted to a thiol. The gold nanoparticles were modified by incubation with a polymer solution. Glycopolymermodified gold nanoparticles are optically and electrically active, which is useful for biosensing. The glycopolymer-modified gold nanoparticles showed a pink color with a plasmon peak at approximately 530 nm. The addition of ConA changed the color of the gold nanoparticle solution, and the plasmon peak was red-shifted from 530 to 590 nm because of the crosslinking of the gold nanoparticles. Not only the proteins but also the sugar recognition bacterium (E. coli ORN 178) were able to be detected using glycopolymer-gold nanoparticles.223 On the basis of the color of the gold nanoparticles, glycopolymermodified gold nanoparticles have been used as the biosensor of a lateral flow assay.222,224 The gold nanoparticle has also been used for detection, based on the redox active label from Au(0) to Au3+ using differential pulse voltammetry. We have detected ConA using glycopolymer-modified gold nanoparticles and an electrochemical assay.225 Kitano et al. reported the detection of lectins (ConA and RCA120) using glycopolymers on gold nanoparticles using local SPR.226,227 The Maynard group reported the immobilization of glycopolymer on a gold substrate to analyze the interaction with SPR.228,229 The Narain groups immobilized biotinylated glycopolymer-modified gold nanoparticles on the substrate for detecting interaction of lectins with SPR and diffractive optics technology.230,231 In this review, we mainly focus on glycopolymers synthesized by addition polymerization. However, glycopolymers of πconjugated polymers have been reported for use in biosensing. The Charych group reported various π-conjugated glycopolymers used for biosensing.232,233 Saccharide-modified polydiacetylene (PDA) lipid micelles were prepared by UV irradiation. The UV spectra of sialic acid modified PDA was changed by the addition of the influenza virus, resulting in a red shift and decrease in absorbance. Saccharide-modified polythiophenes have also been investigated by this group. Glyco-

3.4. Glycopolymers for Drug Delivery and Gene Delivery

The interaction of saccharides with specific cells is known, interactions include Gal−hepatocyte, Mal−macrophage, and Glc−cancer cell interactions.4 Glycopolymers have shown early promise as cell specific drug carriers. The Akaike group has demonstrated the hepatocyte-specific uptake of glycopolymers carrying lactose.59 Wang et al. reported drug delivery system based on the interaction with glycopolymer and ASGP-R.241 They reported the doxorubicin delivery encapsulation in the glycopolymer micelles and HepG2 specific delivery via ASGP-R interaction. Chen et al. synthesized the biodegradable 1681

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Figure 11. Cell specific drug delivery with glycopolymer micelles. The block polymer carrying galactose was synthesized by ring opening polymerization, and doxorubicin was adsorbed by hydrophobic interactions. The polymer showed HepG2 specific uptake and drug release.

ymer−nanoparticle also showed pH responsive by degradation at low pH. Glycopolymer having amino group or carboxylic acid group are able to be response to pH. The ionic property of those groups was changed due to the change between amine and ammonium ion or carboxylic acid and carboxylate. Liu et al. reported the block copolymer of poly(2-(diethylamino)ethyl methacrylate)-co-(3-O-methacryloy-glucopyranose)) via RAFT polymerization and pH responsive micelle formation in the acidic condition. 245 Kim et al. reported pH sensitive glycopolymer gel for drug delivery, where the swelling ratio of the polymer was changed by pH.246 Glucose responsive drug carriers have been also reported for insulin delivery for the diabetes patient. The glycopolymer with phenylboronic acid form particles247 or gel248 by the bond formation between hydroxyl group of glycopolymer and phenylboronic acid. The self-assembled particle or gel was decomposed with addition of glucose because of the sequential bond dissociation. Those materials are useful for glucose sensitive drug delivery for the diabetes patient. The Stenzel group also reported the glycopolymer nanocapusules, which are responsive in reductive condition with glutathione.249 They prepared glycopolymer nanocapsules by postpolymerization reaction and investigated the glutathione-specific hydrophilic drug (gemcitabine) release to pancreatic cancer cells. Recently, gene delivery using glycopolymers has also been investigated. Not only the drug with small molecules but also the biomacromolecules of DNA and RNA are important for delivery systems based on the development of gene therapy. Since genes are macromolecules, the design of delivery materials are different from drug delivery. The conjugation of a cationic segment to the glycopolymer is important for complexation with DNA and RNA for gene delivery. The Narain group has studied gene delivery with various glycopolymers in detail, including diblock and random copolymers, and hyperbranched polymers of the glycopolymer

Figure 10. Hyperbranched glycopolymer with a fluorinated segment for MRI-imaging. The polymer was synthesized by RAFT living radical polymerization and click chemistry.

glycopolymer via ring opening reaction and prepared polymer micelles, which attained the hepatoma cell specific delivery of doxorubicin.242 Recent advances in polymer chemistry have enabled the design of drug carriers using glycopolymers. As we described in the previous section, block copolymer of glycopolymers have self-assembling properties to form micelles and vesicles. The size and properties of the polymer carrier affect the drug delivery behavior. The hydrophobic segment can provide a container for the hydrophobic medicines, such as doxorubicin. Hence, block-glycopolymer micelles are useful for cell specific drug delivery (Figure 11).181,243,244 The Armes group has prepared various kinds of self-assembled glycopolymers with hydrophobic segments.178 Several glyco-block copolymers were synthesized, with variations in the ratios of the galactosylated segments and hydrophobic segments and the molecular weights. The self-assembled structures varied from wormlike and spherical micelles to vesicles. Affinity of the wormlike micelles and vesicles with galectin was demonstrated. The glycopolymer vesicles with Gal were biocompatible and enabled intracellular delivery to Human Dermal Fibroblasts (HDF) cells. The environmental responsive materials are required for sophisticated drug delivery systems. The well-known method is the utilization of thermoresponsive polymer of poly(Nisopropylacrylamide). The Stenzel group investigated glucosamine substituted glycopolymer−nanoparticle containing poly(N-isopropylacrylamide), which have shown temperatureresponsive properties for drug delivery.120 The same glycopol1682

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

(3-gluconamidopropyl methacrylamide) with a cationic segment (2-amino ethyl methacrylamide) (PAEMA).250,251 DNA formed a complex with the cationic segment of PAEMA by electrostatic interaction. The transfection efficiency was affected by the type of polymer, molecular weight, and monomer ratio. The random copolymer and hyperbranched polymers showed better transfection efficiency because of better uptake to the target cells. The Narain group also showed enhancement DNA transfection using glycopolymer conjugates of gold nanoparticles and carbon nanotubes.252,253 The same group also reported siRNA delivery with cationic glycopolymer, which efficiently exhibited knockdown of cell growth factor EGFR.254 The Obata group reported gene delivery systems with glycopolymer having Man.255,256 They prepared poly(2-(α-Dmannnopyranosyloxy)ethyl-co-(2-dimethylaminoethyl methacylates)), which showed the affinity to ConA and the DNA condensed ability. The same group also reported the overall transfection efficiency of block and statistical copolymer and found the statistical copolymer showed the better transfection efficiency. The polymer micelles are one of the prominent materials for drug delivery and gene delivery. Though most of the experiments were still at the basic level and need more research, target specific and stimuli responsive delivery systems are expected to be of practical use in the near future.

Figure 12. Glycopolymer conjugation to proteins: (a) polyvalent trehalose-substituted protein for resistance to heat shock and (b) polyvalent antigenic carbohydrate for amplification of the immune response.

3.5. Glycopolymer Conjugation to Biomacromolecules

New types of biomolecules are able to be fabricated by conjugation of glycopolymers to other biomacromolecules. In section 3.2, we described glycopolymer conjugation to the cell membrane as an artificial extracellular matrix. Glycopolymers synthesized via living polymerization are easily modified at the polymer terminal, and the glycopolymer terminal is able to be conjugated to proteins. Biotin-terminated RAFT and ATRP initiator reagents have been used to synthesize glycopolymers conjugated with streptavidin.228−231 The glycopolymers with biotin also bound to proteins, substrates, and nanoparticles.257,258 Gupta et al. conjugated glycopolymers to virus capsid proteins,259 where glycopolymers were prepared via ATRP and the polymer conjugated with click chemistry and the virus-glycopolymer conjugates exhibited a new method to prepare uniform nanocapsules.238 The Maynard group has conjugated a trehalose glycopolymer to a protein (Figure 12a).260,261 The glycopolymers were synthesized by RAFT polymerization, and the polymer was conjugated to lysozyme by disulfide bond formation. Due to the trehalose activity and multivalency, the lysozyme with trehalose showed stability to heat shock. Conjugation of saccharides to a protein has advantages in terms of the immune response. Since saccharides are involved in diseases and cancer, immunization using saccharides is important in view of production of antibodies. However, the immunogenicity of saccharides is generally weak, and amplification of the immune response is indispensable. Bundle et al. conjugated glycopolymers with albumin, which amplified the immune response (Figure 12b).262 Glycopolymers carrying β-mannan trisaccharides with polyacrylamide were prepared, and the polymer was conjugated with chicken albumin, which was then used for immunization in mice. The glycopolymer conjugate showed a robust and strong immune response, which was advantageous for antibody production. A glycopolymer with tetanus toxin has also been prepared, which also induced a strong immune response. The Haddleton group has prepared a

glycopolymer conjugate with bovine serum albumin using a maleimide-terminated glycopolymer, which induced an immune response.263 A glycopolymer with gold nanoparticles has also been reported to show a strong immune response because of the multivalency of the polymer.264 The amplification of immunogenicity using glycopolymers is a new target for vaccine development. DNA can be used in nanotechnology for the creation of DNA origami.265 Matsuura et al. conjugated welldefined glycopolymers with DNA and controlled the affinity using DNA technologies.266,267 Glycopolymer conjugates with DNA were prepared having half-zip structures, which were incubated with a DNA platform to construct well-defined glycopolymer brushes, which showed strong interactions with lectins.

4. CONCLUSION AND OUTLOOK This review has outlined the technology for the use of glycopolymers nanotechnology for biotechnology application. Much attention to the application of saccharides in medicines and biomaterials has been paid, as there is the potential for the development of novel medicines, drug-delivery systems, antibodies, and biomaterials. The bottleneck in the application of saccharides is the weak interaction of saccharides in the monomeric state, but this can be amplified by the presence of multivalency. However, the process of molecular recognition by saccharides is not totally clear, and the precise preparation of multivalent saccharides is important for future application of saccharides in nanomedicine. The living polymerization of glycopolymers is important to control the preparation of multivalent glycoclusters. Glycopolymers synthesized via living radical polymerization enable the design of specific polymers with a multivalent effect and conjugation to proteins and other materials, which is indispensable for the development of glycopolymer nanomedicine. The physical properties of glycopolymers are also important to control the self-assembling structure of polymer micelles and particles. Advances in 1683

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

glycopolymer are the key for the practical application of saccharide science.

was appointed Associate Professor. His research is focused on the development of general procedures to create synthetic polymer nanomaterials that function as antibody and enzymes.

AUTHOR INFORMATION Corresponding Author

*E-mail: [email protected]. Tel: +81-92-8022749. Notes

The authors declare no competing financial interest. Biographies

Dr. Hirokazu Seto received his Doctor degree of Engineering in 2011 from Saga University. In the same year, he joined Prof. Miura’s group in Kyushu University. In 2013, he became a Researcher of Japan Society for Promotion of Science (JSPS) Research Fellowships. His research interest is chemical engineering with glycopolymer technologies and polymeric catalysis.

ACKNOWLEDGMENTS We are grateful for the financial support provided by the Grantin-Aid for Scientific Research B (15H03818), and Challenging Exploratory Research (26620106).

Yoshiko Miura is currently Professor at Kyusyu University. Yoshiko Miura received her B Eng. Degree in 1995 and M Eng. Degree in 1997 from the Kyoto University under the supervision of Professor Yukio Imanishi. She received Dr Eng. Degree in 2000 from the Kyoto University under the supervision of Professor Shiro Koabayashi. She spent her postdoctoral period from 2000 to 2001 at the University of Pennsylvania in Professor Virgil Percec’s group. In 2001, she joined the Department of Molecular Design and Engineering, Nagoya University, as Assistant Professor and started the research of glycomaterials with Professor Kazukiyo Kobayashi. In 2005, she was appointed Associate Professor at the Schools of Materials Science in Japan Advanced Institute of Science and Technology. In 2010, she was appointed Professor at the Department of Chemical Engineering, at Kyushu University. Her research interests are biomaterial fabrication with glycopolymers and glyco-nanoparticles.

ABBREVIATIONS ASGP-R asialoglycoprotein receptor ATRP atom transfer radical polymerization Cer ceramide ConA concanavalin A DC-SIGN dendritic cell-specific ICAM-grabbing non-integrin E. coli Escherichia coli FET field-effect transistor Gal galactose GAMA 2-gluconamidoethyl methacrylate GalNAc N-acetyl galactosamine Glc glucose GlcNAc N-acetyl glucosamine HDF human dermal fibroblasts LacNAc Gal(β1−4) GlcNAc LAMA 2-lactobionamidoethyl methacrylate LSPR local surface plasmon resonance Man mannose MVK methylvinyl ketone Neu5Ac N-acetylneuraminic acid, sialic acid NK human killer cells PAMAM polyamidoamine PDA polydiacetylene PNA peanut agglutinin PVLA poly(N-p-vinylbenzyl-D-lactonamide) QCM quartz crystal microbalance RAFT reversible addition fragment chain transfer RCA Ricinus communis agglutinin SET-LRP single-electron transfer living radical polymerization SI-ATRP surface-initiated atom transfer radical polymerization SNA sumbucus nigra lectin

Dr. Yu Hoshino was born in Kanagawa Prefecture, Japan, in 1978. He received his Doctor degree in 2006 from Tokyo Institute of Technology under the supervision of Professor Yoshio Okahata. He spent his postdoctoral period from 2006 to 2010 at the University of California, Irvine, in Professor Kenneth J. Shea’s group, and began the study of nanoparticle−peptide interaction. In 2010, he joined the Department of Chemical Engineering, Kyushu University. In 2013, he 1684

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews SPR TEMPO

Review

(23) Mori, T.; Toyoda, M.; Ohtsuka, T.; Okahata, Y. Kinetic Analyses for Bindings of Concanavalin A to Dispersed and Condensed Mannose Surfaces on a Quartz Crystal Microbalance. Anal. Biochem. 2009, 395, 211−216. (24) Okahata, Y.; Niikura, K.; Sugiura, Y.; Sawade, M.; Morii, T. Kinetic Studies of Sequence-Specific Binding of GCN4-bZIP Peptides to DNA Strands Immobilized on a 27-MHz Quartz-Crystal Microbalance. Biochemistry 1998, 37, 5666−5672. (25) Bourne, Y.; Bolgiano, B.; Liao, D.-I.; Strecker, G.; Cantau, P.; Herzberg, O.; Feizi, T.; Cambillau, C. Crosslinking of Mammalian Lectin (Galectin-1) by Complex Biantennary Saccharides. Nat. Struct. Biol. 1994, 1, 863−870. (26) Xu, X.-C.; Ei-Naggar, A. K.; Lotan, R. Differential Expression of Galectin-1 and Galectin-3 in Thyroid Tumors. Potential Diagnostic Implications. Am. J. Pathol. 1995, 147, 815−822. (27) Vlassara, H.; Li, Y. M.; Imani, F.; Wojciechowicz, D.; Yang, Z.; Liu, F.-T.; Cerami, A. Identification of Galectin-3 as a High-Affinity Binding Protein for Advanced Glycation End Products (AGE): A New Member of the AGE-Receptor Complex. Mol. Med. 1995, 1, 634−646. (28) Perillo, N. L.; Pace, K. E.; Seilhamer, J. J.; Baum, L. G. Apoptosis of T Cells Mediated by Galectin-1. Nature 1995, 378, 736−739. (29) Ashwell, G.; Harford, J. Carbohydrate-Specific Receptors of the Liver. Annu. Rev. Biochem. 1982, 51, 531−554. (30) Sharon, N.; Lis, H. History of Lectins: from Hemagglutinins to Biological Recognition Molecules. Glycobiology 2004, 14, 52R−62R. (31) Surolia, A.; Bachhawat, B. K.; Podder, S. K. Interaction between Lectin from Ricinus communis and Liposomes Containing Gangliosides. Nature 1975, 257, 802−804. (32) Sansone, F.; Casnati, A. Multivalent Glycocalixarenes for Recognition of Biological Macromolecules: Glycocalyx Mimics Capable of Multitasking. Chem. Soc. Rev. 2013, 42, 4623−4639. (33) Andre, S.; Kaltner, H.; Furuike, T.; Nishimura, S.-I.; Gabius, H.I. Persubstituted Cyclodextrin-Based Glycoclusters as Inhibitors of Protein−Carbohydrate Recognition Using Purified Plant and Mammalian Lectins and Wild-Type and Lectin-Gene-Transfected Tumor Cells as Targets. Bioconjugate Chem. 2004, 15, 87−98. (34) Lundquist, J. J.; Debenham, S. D.; Toone, E. J. Multivalency Effects in Protein-Carbohydrate Interaction: the Binding of the ShigaLike Toxin 1 Binding Subunit to Multivalent C-Linked Glycopeptides. J. Org. Chem. 2000, 65, 8245−8250. (35) Zhang, Q.; Haddleton, D. M. Synthetic Glycopolymers: Some Recent Developments. Adv. Polym. Sci. 2013, 262, 39−60. (36) Vázquez-Dorbatt, V.; Lee, J.; Lin, E. W.; Maynard, H. D. Synthesis of Glycopolymers by Controlled Radical Polymerization Techniques and Their Applications. ChemBioChem 2012, 13, 2478− 2487. (37) Ghadban, A.; Albertin, L. Synthesis of Glycopolymer Architectures by Reversible-Deactivation Radical Polymerization. Polymers 2013, 5, 431−526. (38) Kiessling, L. L.; Grim, J. C. Glycopolymer Probes of Signal Transduction. Chem. Soc. Rev. 2013, 42, 4476−4491. (39) Sunasee, R.; Adokoh, C. K.; Darkwa, J.; Narain, R. Therapeutic Potential of Carbohydrate-based Polymeric and Nanoparticle Systems. Expert Opin. Drug Delivery 2014, 11, 867−884. (40) Ahmed, M.; Wattanaarsakit, P.; Narain, R. Recent Advances in the Preparation of Glycopolymer Bioconjugates. Eur. Polym. J. 2013, 49, 3010−3033. (41) Kobayashi, K.; Sumitomo, H.; Ina, Y. Synthesis and Functions of Polystyrene Derivatives Having Pendant Oligosaccharides. Polym. J. 1985, 17, 567−575. (42) Mammen, M.; Dahmann, G.; Whitesides, G. M. Effective Inhibitors of Hemagglutination by Influenza Virus Synthesized from Polymers Having Active Ester Groups. Insight into Mechanism of Inhibition. J. Med. Chem. 1995, 38, 4179−4190. (43) Spaltenstein, A.; Whitesides, G. M. Polyacrylamides Bearing Pendant α-Sialoside Groups Strongly Inhibit Agglutination of Erythrocytes by Influenza Virus. J. Am. Chem. Soc. 1991, 113, 686− 687.

surface plasmon resonance 2,2,6,6,-tetramethylpieperidine 1-oxy

REFERENCES (1) Mammen, M.; Choi, S.-K.; Whitesides, G. M. Polyvalent Interactions in Biological Systems: Implications for Design and Use of Multivalent Ligands and Inhibitors. Angew. Chem., Int. Ed. 1998, 37, 2754−2794. (2) Choi, S.- K. Synthetic Multivalent Molecules: Concepts and Biomedical Applications; John Wiley & Sons, Inc.: Hoboken, 2004. (3) Reed, A. E.; Curtiss, L. A.; Weinhold, F. Intermolecular Interactions from a Natural Bond Orbital, Donor-Acceptor Viewpoint. Chem. Rev. 1988, 88, 899−926. (4) Taylor, M. E.; Drickamaer, K. Introduction to Glycobiology; Oxford Press: London, 2002. (5) Dam, T. K.; Brewer, C. F. Thermodynamic Studies of LectinCarbohydrate InteractIons by Isothermal Titration Calorimetry. Chem. Rev. 2002, 102, 387−430. (6) Lundquist, J. J.; Toone, E. J. The Cluster Glycoside Effect. Chem. Rev. 2002, 102, 555−578. (7) Simons, K.; Ikonen, E. Functional Rafts in Cell Membranes. Nature 1997, 387, 569−572. (8) Noble, G. T.; Flitsch, S. L.; Liem, K. P.; Webb, S. J. Assessing the Cluster Glycoside Effect during the Binding of concanavalin A to Mannosylated Artificial Lipid Rafts. Org. Biomol. Chem. 2009, 7, 5245− 5254. (9) Brewer, C. F.; Miceli, M. C.; Baum, L. G. Clusters, Bundles, Arrays and Lattices: Novel Mechanisms for Lectin−SaccharideMediated Cellular Interactions. Curr. Opin. Struct. Biol. 2002, 12, 616−623. (10) Loris, R. Principles of Structures of Animal and Plant Lectins. Biochim. Biophys. Acta, Gen. Subj. 2002, 1572, 198−208. (11) Gabius, H.-J.; Andre, S.; Jimenez-Barbero, J.; Romero, A.; Solis, D. From Lectin Structure to Functional Glycomics: Principles of the Sugar Code. Trends Biochem. Sci. 2011, 36, 298−313. (12) Nilsso, C. L. Lectins: Analytical Technologies; Elsevier Science: New York, 2011. (13) Jack, A.; Weinzierl, J.; Kalb, A. J. An X-ray Crystallographic Study of Demetallized Concanavalin A. J. Mol. Biol. 1971, 58, 389− 390. (14) Greer, J.; Kaufman, H. W.; Kalb, A. J. The Structure of the Saccharide-Binding Site of Concanavalin A. J. Mol. Biol. 1970, 48, 365−366. (15) Hardman, K. D.; Ainsworth, C. F. Structure of concanavalin A at 2.4-Ang Resolution. Biochemistry 1972, 11, 4910−4919. (16) Rini, J. M.; Hardman, K. D.; Einspahr, H.; Suddath, F. L.; Carver, J. P. X-ray Crystal Structure of a Pea Lectin-Trimannoside Complex at 2.6 Å Resolution. J. Biol. Chem. 1993, 268, 10126−10132. (17) Becker, J. W.; Reeke, G. N., Jr.; Cunningham, B. A.; Edelman, G. M. Structure of Concanavalin A at 2.4 Å Resolution. Nature 1976, 259, 406−409. (18) Becker, J. W.; Reeke, G. N., Jr.; Wang, J. L.; Cunningham, B. A.; Edelman, G. M. Favin Versus Concanavalin A: Circularly Permuted Amino acid Sequences. J. Biol. Chem. 1975, 250, 1513−1524. (19) Naismith, J. H.; Field, R. A. Structural basis of Trimannoside Recognition by Concanavalin A. J. Biol. Chem. 1996, 271, 972−976. (20) Dam, T. K.; Roy, R.; Das, S. K.; Oscarson, S.; Brewer, C. F. Binding of Multivalent Carbohydrates to Concanavalin A and Dioclea Grandiflora Lectin, Thermodynamic Analysis Of The “Multivalency Effect. J. Biol. Chem. 2000, 275, 14223−14230. (21) Dam, T. K.; Roy, R.; Page, D.; Brewer, C. F. Epitopes of Multivalent Carbohydrates to Concanavalin A As Determined by “Reverse” Isothermal Titration Microcalorimetry. Biochemistry 2002, 41, 1359−1363. (22) Yonzon, C. R.; Jeoung, E.; Zou, S.; Schatz, G. C.; Mrksich, M.; Van Duyne, R. P. A Comparative Analysis of Localized and Propagating Surface Plasmon Resonance Sensors: The Binding of Concanavalin A to a Monosaccharide Functionalized Self-Assembled Monolayer. J. Am. Chem. Soc. 2004, 126, 12669−12676. 1685

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

(44) Sigal, G. B.; Mammen, M.; Dahmann, G.; Whitesides, G. M. Polyacrylamides Bearing Pendant α-Sialoside Groups Strongly Inhibit Agglutination of Erythrocytes by Influenza Virus: The Strong Inhibition Reflects Enhanced Binding through Cooperative Polyvalent Interactions. J. Am. Chem. Soc. 1996, 118, 3789−3800. (45) Kingery-Wood, J. E.; Williams, K. W.; Sigal, G. B.; Whitesides, G. M. The Agglutination of Erythrocytes by Influenza Virus is Strongly Inhibited by Liposomes Incorporating an Analog of Sialyl Gangliosides. J. Am. Chem. Soc. 1992, 114, 7303−7305. (46) Choi, S.-K.; Mammen, M.; Whitesides, G. M. Monomeric Inhibitors of Influenza Neuraminidase Enhance the Hemagglutination Inhibition Activities of Polyacrylamides Presenting Multiple CSialoside Groups. Chem. Biol. 1996, 3, 97−104. (47) Lees, W. J.; Spaltenstein, A.; Kingery-Wood, J. E.; Whitesides, G. M. Polyacrylamides Bearing Pendant alpha.-Sialoside Groups Strongly Inhibit Agglutination of Erythrocytes by Influenza A Virus: Multivalency and Steric Stabilization of Particulate Biological Systems. J. Med. Chem. 1994, 37, 3419−3433. (48) Kobayashi, K.; Tawada, E.; Akaike, T.; Usui, T. Artificial Glycopolypeptide Conjugates: Simple Synthesis of Lactose- and N,NDiacetylchitobiose-Substituted Poly(L-glutamic acid)s through N-βGlycoside Linkages and Their Interaction with Lectins. Biochim. Biophys. Acta, Gen. Subj. 1997, 1336, 117−122. (49) Zeng, X.; Murata, T.; Kawagishi, H.; Usui, T.; Kobayashi, K. Synthesis of Artificial N-Glycopolypeptides Carrying N-Acetyllactosamine and Related Compounds and Their Specific Interactions with Lectins. Biosci., Biotechnol., Biochem. 1998, 62, 1171−1178. (50) Zeng, X.; Murata, T.; Kawagishi, H.; Usui, T.; Kobayashi, K. Analysis of Specific Interactions of Synthetic Glycopolypeptides Carrying N-Acetyllactosamine and Related Compounds with Lectins. Carbohydr. Res. 1998, 312, 209−217. (51) Kobasa, D.; Takada, A.; Shinya, K.; Hatta, M.; Halfmann, P.; Theriault, S.; Suzuki, H.; Nishimura, H.; Mitamura, K.; Sugaya, N.; Usui, T.; Murata, T.; Maeda, Y.; Watanabe, S.; Suresh, M.; Suzuki, T.; Suzuki, Y.; Feldmann, H.; Kawaoka, Y. Enhanced Virulence of Influenza A Viruses with the Hemagglutinin of the 1918 Pandemic Virus. Nature 2004, 431, 703−707. (52) Chung, T. W.; Yang, J.; Akaike, T.; Cho, K. Y.; Nah, J. W.; Kim, S. I.; Cho, C. S. Preparation of Alginate/Galactosylated Chitosan Scaffold for Hepatocyte Attachment. Biomaterials 2002, 23, 2827− 2834. (53) Yang, J.; Goto, M.; Ise, H.; Cho, C. S.; Akaike, T. Galactosylated Alginate as a Scaffold for Hepatocytes Entrapment. Biomaterials 2002, 23, 471−479. (54) Kobayashi, A.; Akaike, T.; Kobayashi, K.; Sumitomo, H. Enhanced Adhesion and Survival Efficiency of Liver Cells in Culture Dishes Coated with a Lactose-Carrying Styrene Homopolymer. Makromol. Chem., Rapid Commun. 1986, 7, 645−650. (55) Kobayashi, A.; Goto, M.; Kobayashi, K.; Akaike, T. ReceptorMediated Regulation of Differentiation and Proliferation of Hepatocytes by Synthetic Polymer Model of Asialoglycoprotein. J. Biomat. Sci., Polym. Ed. 1995, 6, 325−342. (56) Kobayashi, A.; Kobayashi, K.; Akaike, T. Control of Adhesion and Detachment of Parenchymal Liver Cells using Lactose-Carrying Polystyrene as Substratum. J. Biomater. Sci., Polym. Ed. 1992, 3, 499− 508. (57) Kobayashi, K.; Sumitomo, H.; Kobayashi, A.; Akaike, T. Oligosaccharide-Carrying Styrene-Type Macromers. Polymerization and Specific Interactions Between the Polymers and Liver Cells. J. Macromol. Sci., Chem. 1988, 25, 655−667. (58) Mitsuaki, G.; Hirohumi, Y.; Chia-Wun, C.; Akira, K.; Tatsuki, S.; Atsushi, M.; Seiki, K.; Kazukiyo, K.; Toshihiro, A. Lactose-Carrying Polystyrene as a Drug Carrier: Investigation of Body Distributions to Parenchymal Liver Cells using 125I-Labelled Lactose-Carrying Polystyrene. J. Controlled Release 1994, 28, 223−233. (59) Matsushita, N.; Kobayashi, K.; Akaike, T.; Oda, H.; Yoshida, A. Induction of Cytochrome P-450s and Expression of Liver-specific Genes in Rat Primary Hepatocytes Cultured on Different Extracellular Matrices. Biosci., Biotechnol., Biochem. 1994, 58, 1514−1516.

(60) Tobe, S.; Takei, Y.; Kobayashi, K.; Akaike, T. ReceptorMediated Formation of Multilayer Aggregates of Primary Cultured Adult Rat Hepatocytes on Lactose-Substituted Polystyrene. Biochem. Biophys. Res. Commun. 1992, 184, 225−230. (61) Kobayashi, K.; Goto, M.; Sekine, T.; Masumoto, A.; Yamamoto, N.; Yamamoto, N.; Akaike, T. Regulation of Differentiation and Proliferation of Rat Hepatocytes by Lactose-Carrying Polystyrene. Artif. Organs 1992, 16, 564−567. (62) Cho, C.-S.; Kobayashi, A.; Takei, R.; Ishihara, T.; Maruyama, A.; Akaike, T. Receptor-Mediated Cell Modulator Delivery to Hepatocyte using Nanoparticles Coated with Carbohydrate-Carrying Polymers. Biomaterials 2000, 22, 45−51. (63) Cho, C.; Cho, K.; Park, I.; Kim, S.; Sasagawa, T.; Uchiyama, M.; Akaike, T. Receptor-Mediated Delivery of All Trans-Retinoic Acid to Hepatocyte using Poly(L-lactic acid) Nanoparticles Coated with Galactose-Carrying Polystyrene. J. Controlled Release 2001, 77, 7−15. (64) Watanabe, Y.; Liu, X.; Shibuya, I.; Akaike, T. Functional Evaluation of Poly-(N-p-vinylbenzyl-O-β-Dgalactopyranosyl-[1−4]-Dgluconamide) (PVLA) as a Liver Specific Carrier. J. Biomater. Sci., Polym. Ed. 2000, 11, 833−848. (65) Cho, C.; Seo, S.; Park, I.; Kim, S.; Kim, T.; Hoshiba, T.; Harada, I.; Akaike, T. Galactose-Carrying Polymers as Extracellular Matrices for Liver Tissue Engineering. Biomaterials 2006, 27, 576−585. (66) Lee, J. S.; Kim, S. H.; Kim, Y. J.; Akaike, T.; Kim, S. C. Hepatocyte Adhesion on a Poly[N-p-vinylbenzyl-4-O-β-D-galactopyranosyl-D-glucoamide]-Coated Poly(l-lactic acid) Surface. Biomacromolecules 2005, 6, 1906−1911. (67) Hoshiba, T.; Nagahara, H.; Cho, C.-S.; Tagawa, Y.-i.; Akaike, T. Primary Hepatocyte Survival on Non-Integrin-Recognizable Matrices Without the Activation of Akt Signaling. Biomaterials 2007, 28, 1093− 1104. (68) Mourez, M.; Kane, R. S.; Mogridge, J.; Metallo, S.; Deschatelets, P.; Sellman, B. R.; Whitesides, G. M.; Collier, R. J. Designing a Polyvalent Inhibitor of Anthrax Toxin. Nat. Biotechnol. 2001, 19, 958− 961. (69) Rao, J.; Lahiri, J.; Isaacs, L.; Weis, R. M.; Whitesides, G. M. A Trivalent System from Vancomycin·D-Ala-D-Ala with Higher Affinity Than Avidin·Biotin. Science 1998, 280, 708−711. (70) Gestwicki, J. E.; Cairo, C. W.; Strong, L. E.; Oetjen, K. A.; Kiessling, L. L. Influencing Receptor−Ligand Binding Mechanisms with Multivalent Ligand Architecture. J. Am. Chem. Soc. 2002, 124, 14922−14933. (71) Miura, Y.; Koketsu, D.; Kobayashi, K. Synthesis and Properties of a Well-Defined Glycopolymer via Living Radical Polymerization. Polym. Adv. Technol. 2007, 18, 647−651. (72) Hoshino, Y.; Nakamoto, M.; Miura, Y. Control of ProteinBinding Kinetics on Synthetic Polymer Nanoparticles by Tuning Flexibility and Inducing Conformation Changes of Polymer Chains. J. Am. Chem. Soc. 2012, 134, 15209−15212. (73) Hasegawa, T.; Kondoh, S.; Matsuura, K.; Kobayashi, K. Rigid Helical Poly(glycosyl phenyl isocyanide)s: Synthesis, Conformational Analysis, and Recognition by Lectins. Macromolecules 1999, 32, 6595− 6603. (74) Bittiger, H.; Schnebi, H. P. Concanavalin A as a Tool; John Wiley & Sons Ltd: Hoboken, 1976. (75) Merritt, E. A.; Hol, W. G. J. AB 5 Toxins. Curr. Opin. Struct. Biol. 1995, 5, 165−171. (76) St. Hilaire, P. M.; Boyd, M. K.; Toone, E. J. Interaction of the Shiga-like Toxin Type 1 B-Subunit with Its Carbohydrate Receptor. Biochemistry 1994, 33, 14452−14463. (77) Fraser, M. E.; Fujinaga, M.; Cherney, M.; Melton-Celsa, A. R.; Twiddy, E. M.; O'Brien, A. D.; James, M. N. G. Structure of Shiga Toxin Type 2 (Stx2) from Escherichia coli O157:H7. J. Biol. Chem. 2004, 279, 27511−27517. (78) Ling, H.; Boodhoo, A.; Hazes, B.; Cummings, M. D.; Armstrong, G. D.; Brunton, J. L.; Read, R. J. Structure of the Shigalike Toxin I B-Pentamer Complexed with an Analogue of Its Receptor Gb3. Biochemistry 1998, 37, 1777−1788. 1686

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

(79) Merritt, E. A.; Sarfaty, S.; Akker, F. V. D.; L'Hoir, C.; Martial, J. A.; Hol, W. G. J. Crystal Structure of Cholera Toxin B-Pentamer Bound to Receptor GM1 Pentasaccharide. Protein Sci. 1994, 3, 166− 175. (80) Zhang, R. G.; Scott, D. L.; Westbrook, M. L.; Nance, S.; Spangler, B. D.; Shipley, G. G.; Westbrook, E. M. The Threedimensional Crystal Structure of Cholera Toxin. J. Mol. Biol. 1995, 251, 563−573. (81) Zhang, R. G.; Westbrook, M. L.; Westbrook, E. M.; Scott, D. L.; Otwinowski, Z.; Maulik, P. R.; Reed, R. A.; Shipley, G. G. The 2.4 Å Crystal Structure of Cholera Toxin B Subunit Pentamer: Choleragenoid. J. Mol. Biol. 1995, 251, 550−562. (82) Gargano, J. M.; Ngo, T.; Kim, J. Y.; Acheson, D. W. K.; Lees, W. J. Multivalent Inhibition of AB5 Toxins. J. Am. Chem. Soc. 2001, 123, 12909−12910. (83) Dohi, H.; Nishida, Y.; Mizuno, M.; Shinkai, M.; Kobayashi, T.; Takeda, T.; Uzawa, H.; Kobayashi, K. Synthesis of an Artificial Glycoconjugate Polymer Carrying Pk-Antigenic Trisaccharide and its Potent Neutralization Activity Against Shiga-Like Toxin. Bioorg. Med. Chem. 1999, 7, 2053−2062. (84) Miyagawa, A.; Kurosawa, H.; Watanabe, T.; Koyama, T.; Terunuma, D.; Matsuoka, K. Synthesis of Glycoconjugate Polymer Carrying Globotriaose as Artificial Multivalent Ligand for Shiga ToxinProducing Escherichia coli O157: H7. Carbohydr. Polym. 2004, 57, 441−450. (85) Miyagawa, A.; Watanabe, M.; Igai, K.; Kasuya, M. C. Z.; Natori, Y.; Nishikawa, K.; Hatanaka, K. Development of Dialyzer with Immobilized Glycoconjugate Polymers for Removal of Shiga-toxin. Biomaterials 2006, 27, 3304−3311. (86) Ha, Y.; Stevens, D. J.; Skehel, J. J.; Wiley, D. C. X-ray Structures of H5 Avian and H9 Swine Influenza Virus Hemagglutinins Bound to Avian and Human Receptor Analogs. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 11181−11186. (87) Stevens, J.; Blixt, O.; Tumpey, T. M.; Taubenberger, J. K. Structure and Receptor Specificity of the Hemagglutinin from an H5N1 Influenza Virus. Science 2006, 312, 404−410. (88) Sui, J.; Hwang, W. C.; Perez, S.; Wei, G.; Aird, D. Structural and Functional Bases for Broad-Spectrum Neutralization of Avian and Human Influenza A Viruses. Nat. Struct. Mol. Biol. 2009, 16, 265−273. (89) Kamigaito, M.; Ando, T.; Sawamoto, M. Metal-Catalyzed Living Radical Polymerization. Chem. Rev. 2001, 101, 3689−3746. (90) Braunecker, W. A.; Matyjaszewski, K. Controlled/Living Radical Polymerization: Features, Developments, and Perspectives. Prog. Polym. Sci. 2007, 32, 93−146. (91) Chong, Y.; Le, T. P.; Moad, G.; Rizzardo, E.; Thang, S. H. A More Versatile Route to Block Copolymers and Other Polymers of Complex Architecture by Living Radical Polymerization: The RAFT Process. Macromolecules 1999, 32, 2071−2074. (92) Hawker, C. J.; Bosman, A. W.; Harth, E. New Polymer Synthesis by Nitroxide Mediated Living Radical Polymerizations. Chem. Rev. 2001, 101, 3661−3688. (93) Lligadas, G.; Rosen, B. M.; Monteiro, M. J.; Percec, V. Solvent Choice Differentiates SET-LRP and Cu-Mediated Radical Polymerization with Non-First-Order Kinetics. Macromolecules 2008, 41, 8360−8364. (94) Ohno, K.; Tsujii, Y.; Miyamoto, T.; Fukuda, T.; Goto, M.; Kobayashi, K.; Akaike, T. Synthesis of a Well-Defined Glycopolymer by Nitroxide-Controlled Free Radical Polymerization. Macromolecules 1998, 31, 1064−1069. (95) Götz, H.; Harth, E.; Schiller, S. M.; Frank, C. W.; Knoll, W.; Hawker, C. J. Synthesis of Lipo-Glycopolymer Amphiphiles by Nitroxide-Mediated Living Free-Radical Polymerization. J. Polym. Sci., Part A: Polym. Chem. 2002, 40, 3379−3391. (96) Ting, S. R. S.; Min, E. H.; Escalé, P.; Save, M.; Billon, L.; Stenzel, M. H. Lectin Recognizable Biomaterials Synthesized via Nitroxide-Mediated Polymerization of a Methacryloyl Galactose Monomer. Macromolecules 2009, 42, 9422−9434.

(97) Chen, Y. M.; Wulff, G. Synthesis of Poly(styryl sugar)s by TEMPO Mediated Free Radical Polymerization. Macromol. Chem. Phys. 2001, 202, 3426−3431. (98) Chen, Y. M.; Wulff, G. Amphiphilic Block Copolymers with Pendent Sugar as Hydrophilic Segments and Their Surface Properties. Macromol. Chem. Phys. 2001, 202, 3273−3278. (99) Narumi, A.; Matsuda, T.; Kaga, H.; Satoh, T.; Kakuchi, T. Glycoconjugated Polymer II. Synthesis of Polystyrene-block-poly(4vinylbenzyl glucoside) and Polystyrene-block-poly(4-vinylbenzyl maltohexaoside) via 2,2,6,6-Tetramethylpiperidine-1-oxyl-Mediated Living Radical Polymerization. Polym. J. 2001, 33, 939−945. (100) Narumi, A.; Satoh, T.; Kaga, H.; Kakuchi, T. Glycoconjugated Polymer. 3. Synthesis and Amphiphilic Property of Core-Glycoconjugated Star-Shaped Polystyrene. Macromolecules 2002, 35, 699−705. (101) Narumi, A.; Matsuda, T.; Kaga, H.; Satoh, T.; Kakuchi, T. Synthesis of Amphiphilic Triblock Copolymer of Polystyrene and Poly(4-vinylbenzyl glucoside) via TEMPO-Mediated Living Radical Polymerization. Polymer 2002, 43, 4835−4840. (102) Ohno, K.; Goto, A.; Fukuda, T.; Xia, J.; Matyjaszewski, K. Kinetic Study on the Activation Process in an Atom Transfer Radical Polymerization. Macromolecules 1998, 31, 2699−2701. (103) Narain, R.; Armes, S. P. Synthesis of Low Polydispersity, Controlled-Structure Sugar Methacrylate Polymers under Mild Conditions without Protecting Group Chemistry. Chem. Commun. 2002, 2776−2777. (104) Narain, R.; Armes, S. P. Synthesis and Aqueous Solution Properties of Novel Sugar Methacrylate-Based Homopolymers and Block Copolymers. Biomacromolecules 2003, 4, 1746−1758. (105) Narain, R.; Armes, S. P. Direct Synthesis and Aqueous Solution Properties of Well-Defined Cyclic Sugar Methacrylate Polymers. Macromolecules 2003, 36, 4675−4678. (106) Yang, Q.; Tian, J.; Hu, M.-X.; Xu, Z.-K. Construction of a Comb-like Glycosylated Membrane Surface by a Combination of UVInduced Graft Polymerization and Surface-Initiated ATRP. Langmuir 2007, 23, 6684−6690. (107) Meng, X.-L.; Fang, Y.; Wan, L.-S.; Huang, X.-J.; Xu, Z.-K. Glycopolymer Brushes for the Affinity Adsorption of RCA120: Effects of Thickness, Grafting Density, and Epitope Density. Langmuir 2012, 28, 13616−13623. (108) Raynor, J. E.; Petrie, T. A.; Fears, K. P.; Latour, R. A.; García, A. J.; Collard, D. M. Saccharide Polymer Brushes To Control Protein and Cell Adhesion to Titanium. Biomacromolecules 2009, 10, 748−755. (109) Chernyy, S.; Jensen, B. E.; Shimizu, K.; Ceccato, M.; Pedersen, S. U.; Zelikin, A. N.; Daasbjerg, K.; Iruthayaraj, J. Surface Grafted Glycopolymer Brushes to Enhance Selective Adhesion of HepG2 Cells. J. Colloid Interface Sci. 2013, 404, 207−214. (110) Rosen, B. M.; Lligadas, G.; Hahn, C.; Percec, V. Synthesis of Dendritic Macromolecules through Divergent Iterative Thio-Bromo “Click” Chemistry and SET-LRP. J. Polym. Sci., Part A: Polym. Chem. 2009, 47, 3940−3948. (111) Lligadas, G.; Percec, V. Ultrafast SET-LRP of Methyl Acrylate at 25 °C in Alcohols. J. Polym. Sci., Part A: Polym. Chem. 2008, 46, 2745−2754. (112) Lligadas, G.; Rosen, B. M.; Bell, C. A.; Monteiro, M. J.; Percec, V. Effect of Cu(0) Particle Size on the Kinetics of SET-LRP in DMSO and Cu-Mediated Radical Polymerization in MeCN at 25 °C. Macromolecules 2008, 41, 8365−8371. (113) Geng, J.; Lindqvist, J.; Mantovani, G.; Haddleton, D. M. Simultaneous Copper (I)-Catalyzed Azide-Alkyne Cycloaddition (CuAAC) and Living Radical Polymerization. Angew. Chem., Int. Ed. 2008, 47, 4180−4183. (114) Zhang, Q.; Anastasaki, A.; Li, G.-Z.; Haddleton, A. J.; Wilson, P.; Haddleton, D. M. Multiblock Sequence-Controlled Glycopolymers via Cu(0)-LRP Following Efficient Thiol−Halogen, Thiol−Epoxy and CuAAC Reactions. Polym. Chem. 2014, 5, 3876−3883. (115) Zhang, Q.; Wilson, P.; Anastasaki, A.; McHale, R.; Haddleton, D. M. Synthesis and Aggregation of Double Hydrophilic Diblock Glycopolymers via Aqueous SET-LRP. ACS Macro Lett. 2014, 3, 491− 495. 1687

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Implementing Modern Macromolecular Design. Macromol. Rapid Commun. 2011, 32, 1123−1143. (133) Becer, C. R. The Glycopolymer Code: Synthesis of Glycopolymers and Multivalent Carbohydrate−Lectin Interactions. Macromol. Rapid Commun. 2012, 33, 742−752. (134) Becer, C. R.; Babiuch, K.; Pilz, D.; Hornig, S.; Heinze, T.; Gottschaldt, M.; Schubert, U. S. Clicking Pentafluorostyrene Copolymers: Synthesis, Nanoprecipitation, and Glycosylation. Macromolecules 2009, 42, 2387−2394. (135) Babiuch, K.; Wyrwa, R.; Wagner, K.; Seemann, T.; Hoeppener, S.; Becer, C. R.; Linke, R.; Gottschaldt, M.; Weisser, J. r.; Schnabelrauch, M.; et al. Functionalized, Biocompatible Coating for Superparamagnetic Nanoparticles by Controlled Polymerization of a Thioglycosidic Monomer. Biomacromolecules 2011, 12, 681−691. (136) Babiuch, K.; Becer, C. R.; Gottschaldt, M.; Delaney, J. T.; Weisser, J.; Beer, B.; Wyrwa, R.; Schnabelrauch, M.; Schubert, U. S. Adhesion of Preosteoblasts and Fibroblasts onto Poly (pentafluorostyrene)-Based Glycopolymeric Films and Their Biocompatibility. Macromol. Biosci. 2011, 11, 535−548. (137) Ladmiral, V.; Mantovani, G.; Clarkson, G. J.; Cauet, S.; Irwin, J. L.; Haddleton, D. M. Synthesis of Neoglycopolymers by a Combination of “Click Chemistry” and Living Radical Polymerization. J. Am. Chem. Soc. 2006, 128, 4823−4830. (138) Slavin, S.; Burns, J.; Haddleton, D. M.; Becer, C. R. Synthesis of Glycopolymers via Click Reactions. Eur. Polym. J. 2011, 47, 435− 446. (139) Nurmi, L.; Lindqvist, J.; Randev, R.; Syrett, J.; Haddleton, D. M. Glycopolymers via Catalytic Chain Transfer Polymerisation (CCTP), Huisgens Cycloaddition and Thiol−ene Double Click Reactions. Chem. Commun. 2009, 2727−2729. (140) Godula, K.; Rabuka, D.; Nam, K. T.; Bertozzi, C. R. Synthesis and Microcontact Printing of Dual End-Functionalized Mucin-like Glycopolymers for Microarray Applications. Angew. Chem., Int. Ed. 2009, 48, 4973−4976. (141) Rabuka, D.; Forstner, M. B.; Groves, J. T.; Bertozzi, C. R. Noncovalent Cell Surface Engineering: Incorporation of Bioactive Synthetic Glycopolymers into Cellular Membranes. J. Am. Chem. Soc. 2008, 130, 5947−5953. (142) Godula, K.; Bertozzi, C. R. Synthesis of Glycopolymers for Microarray Applications via Ligation of Reducing Sugars to a Poly(acryloyl hydrazide) Scaffold. J. Am. Chem. Soc. 2010, 132, 9963−9965. (143) Furukawa, J.-I.; Shinohara, Y.; Kuramoto, H.; Miura, Y.; Shimaoka, H.; Kurogochi, M.; Nakano, M.; Nishimura, S.-I. Comprehensive Approach to Structural and Functional Glycomics Based on Chemoselective Glycoblotting and Sequential Tag Conversion. Anal. Chem. 2008, 80, 1094−1101. (144) Boyer, C.; Davis, T. P. One- pot Synthesis and Biofunctionalization of Glycopolymers via RAFT Polymerization and Thiol−ene Reactions. Chem. Commun. 2009, 6029−6031. (145) You, L.; Schlaad, H. An Easy Way to Sugar-Containing Polymer Vesicles or Glycosomes. J. Am. Chem. Soc. 2006, 128, 13336− 13337. (146) Schlaad, H.; You, L.; Sigel, R.; Smarsly, B.; Heydenreich, M.; Mantion, A.; Mašić, A. Glycopolymer Vesicles with an Asymmetric Membrane. Chem. Commun. 2009, 1478−1480. (147) Kanai, M.; Mortell, K. H.; Kiessling, L. L. Varying the Size of Multivalent Ligands: The Dependence of Concanavalin A Binding on Neoglycopolymer Length. J. Am. Chem. Soc. 1997, 119, 9931−9932. (148) Manning, D. D.; Hu, X.; Beck, P.; Kiessling, L. L. Synthesis of Sulfated Neoglycopolymers: Selective P-Selectin Inhibitors. J. Am. Chem. Soc. 1997, 119, 3161−3162. (149) Mortell, K. H.; Gingras, M.; Kiessling, L. L. Synthesis of Cell Agglutination Inhibitors by Aqueous Ring-Opening Metathesis Polymerization. J. Am. Chem. Soc. 1994, 116, 12053−12054. (150) Mortell, K. H.; Weatherman, R. V.; Kiessling, L. L. Recognition Specificity of Neoglycopolymers Prepared by Ring-Opening Metathesis Polymerization. J. Am. Chem. Soc. 1996, 118, 2297−2298.

(116) Lowe, A. B.; Sumerlin, B. S.; McCormick, C. L. The Direct Polymerization of 2-Methacryloxyethyl Glucoside via Aqueous Reversible Addition-Fragmentation Chain Transfer (RAFT) Polymerization. Polymer 2003, 44, 6761−6765. (117) Bernard, J.; Hao, X.; Davis, T. P.; Barner-Kowollik, C.; Stenzel, M. H. Synthesis of Various Glycopolymer Architectures via RAFT Polymerization: From Block Copolymers to Starsolymerization. Biomacromolecules 2006, 7, 232−238. (118) Zhang, L.; Bernard, J.; Davis, T. P.; Barner-Kowollik, C.; Stenzel, M. H. Acid-Degradable Core-Crosslinked Micelles Prepared from Thermosensitive Glycopolymers Synthesized via RAFT Polymerization. Macromol. Rapid Commun. 2008, 29, 123−129. (119) Stenzel, M. H.; Zhang, L.; Huck, W. T. S. TemperatureResponsive Glycopolymer Brushes Synthesized via RAFT Polymerization Using the Z-group Approach. Macromol. Rapid Commun. 2006, 27, 1121−1126. (120) Pearson, S.; Allen, N.; Stenzel, M. H. Core-shell Particles with Glycopolymer Shell and Polynucleoside Core via RAFT: From Micelles to Rods. J. Polym. Sci., Part A: Polym. Chem. 2009, 47, 1706−1723. (121) Min, E. H.; Ting, S. R.; Billon, L.; Stenzel, M. H. ThermoResponsive Glycopolymer Chains Grafted onto Honeycomb Structured Porous Films via RAFT Polymerization as a Thermo-dependent switcher for Lectin Concanavalin a Conjugation. J. Polym. Sci., Part A: Polym. Chem. 2010, 48, 3440−3455. (122) Ting, S. R. S.; Min, E. H.; Zetterlund, P. B.; Stenzel, M. H. Controlled/Living ab Initio Emulsion Polymerization via a Glucose RAFTstab: Degradable Cross-Linked Glyco-Particles for Concanavalin A/FimH Conjugations to Cluster E. coli Bacteria. Macromolecules 2010, 43, 5211−5221. (123) Albertin, L.; Stenzel, M. H.; Barner-Kowollik, C.; Foster, L. J. R.; Davis, T. P. Well-Defined Diblock Glycopolymers from RAFT Polymerization in Homogeneous Aqueous Medium. Macromolecules 2005, 38, 9075−9084. (124) Albertin, L.; Stenzel, M. H.; Barner-Kowollik, C.; Davis, T. P. Effect of an Added base on (4-Cyanopentanoic acid)-4-dithiobenzoate Mediated RAFT Polymerization in Water. Polymer 2006, 47, 1011− 1019. (125) Albertin, L.; Stenzel, M.; Barner-Kowollik, C.; Foster, L. J. R.; Davis, T. P. Well-Defined Glycopolymers from RAFT Polymerization: Poly(methyl 6-O-methacryloyl-α-d-glucoside) and Its Block Copolymer with 2-Hydroxyethyl Methacrylate. Macromolecules 2004, 37, 7530−7537. (126) Granville, A. M.; Quemener, D.; Davis, T. P.; Barner-Kowollik, C.; Stenzel, M. H. Chemo-enzymatic Synthesis and RAFT Polymerization of 6-O-Methacryloyl Mannose: A Suitable Glycopolymer for Binding to the Tetrameric Lectin Concanavalin A. Macromol. Symp. 2007, 255, 81−89. (127) Bernard, J.; Favier, A.; Zhang, L.; Nilasaroya, A.; Davis, T. P.; Barner-Kowollik, C.; Stenzel, M. H. Poly(vinyl ester) Star Polymers via Xanthate-Mediated Living Radical Polymerization: From Poly(vinyl alcohol) to Glycopolymer Stars. Macromolecules 2005, 38, 5475−5484. (128) Escalé, P.; Ting, S. S.; Khoukh, A.; Rubatat, L.; Save, M.; Stenzel, M. H.; Billon, L. Synthetic Route Effect on Macromolecular Architecture: from Block to Gradient Copolymers based on Acryloyl Galactose Monomer using RAFT Polymerization. Macromolecules 2011, 44, 5911−5919. (129) Hetzer, M.; Chen, G.; Barner-Kowollik, C.; Stenzel, M. H. Neoglycopolymers Based on 4-Vinyl-1, 2, 3-Triazole Monomers Prepared by Click Chemistry. Macromol. Biosci. 2010, 10, 119−126. (130) Ting, S. R. S.; Gregory, A. M.; Stenzel, M. H. Polygalactose Containing Nanocages: The RAFT Process for the Synthesis of Hollow Sugar Balls. Biomacromolecules 2009, 10, 342−352. (131) Chen, Y.; Espeel, P.; Reinicke, S.; Du Prez, F. E.; Stenzel, M. H. Control of Glycopolymer Nanoparticle Morphology by a One-Pot, Double Modification Procedure Using Thiolactones. Macromol. Rapid Commun. 2014, 35, 1128−1134. (132) Roth, P. J.; Boyer, C.; Lowe, A. B.; Davis, T. P. RAFT Polymerization and Thiol Chemistry: A Complementary Pairing for 1688

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

(151) Mann, D. A.; Kanai, M.; Maly, D. J.; Kiessling, L. L. Probing Low Affinity and Multivalent Interactions with Surface Plasmon Resonance: Ligands for Concanavalin A. J. Am. Chem. Soc. 1998, 120, 10575−10582. (152) Strong, L. E.; Kiessling, L. L. A General Synthetic Route to Defined, Biologically Active Multivalent Arrays. J. Am. Chem. Soc. 1999, 121, 6193−6196. (153) Aoi, K.; Itoh, K.; Okada, M. Globular Carbohydrate Macromolecules ″Sugar Balls″. 1. Synthesis of Novel SugarPersubstituted Poly(amido amine) Dendrimers. Macromolecules 1995, 28, 5391−5393. (154) Roy, R. Syntheses and Some Applications of Chemically Defined Multivalent Glycoconjugates. Curr. Opin. Struct. Biol. 1996, 6, 692−702. (155) Roy, R.; Zanini, D.; Meunier, S. J.; Romanowska, A. SolidPhase Synthesis of Dendritic Sialoside Inhibitors of Influenza A Virus Haemagglutinin. J. Chem. Soc., Chem. Commun. 1993, 1869−1872. (156) Zanini, D.; Roy, R. Synthesis of New α-Thiosialodendrimers and Their Binding Properties to the Sialic Acid Specific Lectin from Limax f lavus. J. Am. Chem. Soc. 1997, 119, 2088−2095. (157) Wolfenden, M. L.; Cloninger, M. J. Mannose/GlucoseFunctionalized Dendrimers To Investigate the Predictable Tunability of Multivalent Interactions. J. Am. Chem. Soc. 2005, 127, 12168− 12169. (158) Turnbull, W. B.; Stoddart, J. F. Design and Synthesis of Glycodendrimers. Rev. Mol. Biotechnol. 2002, 90, 231−255. (159) Ashton, P. R.; Boyd, S. E.; Brown, C. L.; Nepogodiev, S. A.; Meijer, E.; Peerlings, H. W. I.; Stoddart, J. F. Synthesis of Glycodendrimers by Modification of Poly(propylene imine) Dendrimers. Chem. - Eur. J. 1997, 3, 974−984. (160) Dutta, T.; Jain, N. K. Targeting Potential and anti-HIV Activity of Lamivudine Loaded Mannosylated Poly(propyleneimine) Dendrimer. Biochim. Biophys. Acta, Gen. Subj. 2007, 1770, 681−686. (161) Percec, V.; Leowanawat, P.; Sun, H.-J.; Kulikov, O.; Nusbaum, C. D.; Tran, T. M.; Bertin, A.; Wilson, D. A.; Peterca, M.; Zhang, S.; et al. Modular Synthesis of Amphiphilic Janus Glycodendrimers and Their Self-Assembly into Glycodendrimersomes and Other Complex Architectures with Bioactivity to Biomedically Relevant Lectins. J. Am. Chem. Soc. 2013, 135, 9055−9077. (162) Zhang, S.; Moussodia, R. O.; Sun, H. J.; Leowanawat, P.; Muncan, A.; Nusbaum, C. D.; Chelling, K. M.; Heiney, P. A.; Klein, M. L.; André, S.; Roy, R.; Gabius, H.-J.; Percec, V. Mimicking Biological Membranes with Programmable Glycan Ligands Self-Assembled from Amphiphilic Janus Glycodendrimers. Angew. Chem. 2014, 126, 11079− 11083. (163) Zhang, S.; Xiao, Q.; Sherman, S. E.; Muncan, A.; RamosVicente, A. D. M.; Wang, Z.; Hammer, D. A.; Williams, D.; Chen, Y.; Pochan, D. J.; Vértesy, S.; André, S.; Klein, M. L.; Gabius, H.-J.; Percec, V. Glycodendrimersomes from Sequence-Defined Janus Glycodendrimers Reveal High Activity and Sensor Capacity for the Agglutination by Natural Variants of Human Lectin. J. Am. Chem. Soc. 2015, 137, 13334−13344. (164) Killops, K. L.; Campos, L. M.; Hawker, C. J. Robust, Efficient, and Orthogonal Synthesis of Dendrimers via Thiol-ene “Click” Chemistry. J. Am. Chem. Soc. 2008, 130, 5062−5064. (165) Fernandez-Megia, E.; Correa, J.; Rodríguez-Meizoso, I.; Riguera, R. A Click Approach to Unprotected Glycodendrimers. Macromolecules 2006, 39, 2113−2120. (166) Pfaff, A.; Müller, A. H. Hyperbranched Glycopolymer Grafted Microspheres. Macromolecules 2011, 44, 1266−1272. (167) Gao, C.; Muthukrishnan, S.; Li, W.; Yuan, J.; Xu, Y.; Müller, A. H. Linear and Hyperbranched Glycopolymer-Functionalized Carbon Nanotubes: Synthesis, Kinetics, and Characterization. Macromolecules 2007, 40, 1803−1815. (168) Semsarilar, M.; Ladmiral, V.; Perrier, S. Highly Branched and Hyperbranched Glycopolymers via Reversible Addition−Fragmentation Chain Transfer Polymerization and Click Chemistry. Macromolecules 2010, 43, 1438−1443.

(169) Wataoka, I.; Urakawa, H.; Kobayashi, K.; Akaike, T.; Schmidt, M.; Kajiwara, K. Structural Characterization of Glycoconjugate Polystyrene in Aqueous Solution. Macromolecules 1999, 32, 1816− 1821. (170) Wataoka, I.; Urakawa, H.; Kobayashi, K.; Ohno, K.; Fukuda, T.; Akaike, T.; Kajiwara, K. Molecular Specification of Homopolymer of Vinylbenzyl-Lactose-Amide in Aqueous Solution. Polym. J. 1999, 31, 590−594. (171) Wataoka, I.; Kobayashi, K.; Kajiwara, K. Effect of the Carbohydrate Side-Chain on the Conformation of a Glycoconjugate Polystyrene in Aqueous Solution. Carbohydr. Res. 2005, 340, 989−995. (172) Shimojo, S.; Cho, C.-S.; Park, I.-K.; Kunou, M.; Goto, M.; Akaike, T. Helical Structure of Sugar-Carrying Polystyrene in Aqueous Solution by Circular Dichroism. Carbohydr. Res. 2003, 338, 2129− 2133. (173) Fukuda, T.; Inoue, Y.; Koga, T.; Matsuoka, M.; Miura, Y. Encapsulation of Polythiophene by Glycopolymer for Water-soluble Nanowire. Chem. Lett. 2011, 40, 864−866. (174) Star, A.; Steuerman, D. W.; Heath, J. R.; Stoddart, J. F. Starched Carbon Nanotubes. Angew. Chem., Int. Ed. 2002, 41, 2508− 2512. (175) Miura, Y.; Sato, H.; Ikeda, T.; Sugimura, H.; Takai, O.; Kobayashi, K. Micropatterned Carbohydrate Displays by SelfAssembly of Glycoconjugate Polymers on Hydrophobic Templates on Silicon. Biomacromolecules 2004, 5, 1708−1713. (176) Sato, H.; Miura, Y.; Saito, N.; Kobayashi, K.; Takai, O. A Micropatterned Multifunctional Carbohydrate Display by an Orthogonal Self-Assembling Strategy. Biomacromolecules 2007, 8, 753−756. (177) Antonietti, M.; Förster, S. Vesicles and Liposomes: A SelfAssembly Principle Beyond Lipids. Adv. Mater. 2003, 15, 1323−1333. (178) Ladmiral, V.; Semsarilar, M.; Canton, I.; Armes, S. P. Polymerization-Induced Self-Assembly of Galactose-Functionalized Biocompatible Diblock Copolymers for Intracellular Delivery. J. Am. Chem. Soc. 2013, 135, 13574−13581. (179) Israelachvili, J. N.; Mitchell, D. J.; Ninham, B. W. Theory of self-assembly of hydrocarbon amphiphiles into mi-celles and bilayers. J. Chem. Soc., Faraday Trans. 2 1976, 72, 1525−1568. (180) Pearson, S.; Scarano, W.; Stenzel, M. H. Micelles Based on Gold-Glycopolymer Complexes as New Chemotherapy Drug Delivery Agents. Chem. Commun. 2012, 48, 4695−4697. (181) Gregory, A.; Stenzel, M. H. Synthesis of Hollow Polymeric Nanoparticles for Protein Delivery via Inverse Miniemulsion Periphery RAFT polymerization. Prog. Polym. Sci. 2012, 37, 38−105. (182) Percec, V.; Wilson, D. A.; Leowanawat, P.; Wilson, C. J.; Hughes, A. D.; Kaucher, M. S.; Hammer, D. A.; Levine, D. H.; Kim, A. J.; Bates, F. S. Self-Assembly of Janus Dendrimers into Uniform Dendrimersomes and Other Complex Architectures. Science 2010, 328, 1009−1014. (183) Kim, C. U.; Lew, W.; Williams, M. A.; Liu, H.; Zhang, L.; Swaminathan, S.; Bischofberger, N.; Chen, M. S.; Mendel, D. B.; Tai, C. Y.; Laver, W. G.; Stevens, R. C. Influenza Neuraminidase Inhibitors Possessing a Novel Hydrophobic Interaction in the Enzyme Active Site: Design, Synthesis, and Structural Analysis of Carbocyclic Sialic Acid Analogues with Potent Anti-Influenza Activity. J. Am. Chem. Soc. 1997, 119, 681−690. (184) Kumar, J.; McDowall, L.; Chen, G.; Stenzel, M. H. Synthesis of Thermo-Responsive Glycopolymers via copper Catalysed Azide− Alkyne ‘Click’ Chemistry for Inhibition of Ricin: the Effect of Spacer between Polymer Backbone and Galactose. Polym. Chem. 2011, 2, 1879−1886. (185) Nagatsuka, T.; Uzawa, H.; Ohsawa, I.; Seto, Y.; Nishida, Y. Use of Lactose against the Deadly Biological Toxin Ricin. ACS Appl. Mater. Interfaces 2010, 2, 1081−1085. (186) Nagatsuka, T.; Uzawa, H.; Sato, K.; Ohsawa, I.; Seto, Y.; Nishida, Y. Glycotechnology for Decontamination of Biological Agents: A Model Study Using Ricin and Biotin-Tagged Synthetic Glycopolymers. ACS Appl. Mater. Interfaces 2012, 4, 832−837. (187) Nagatsuka, T.; Uzawa, H.; Sato, K.; Kondo, S.; Izumi, M.; Yokoyama, K.; Ohsawa, I.; Seto, Y.; Neri, P.; Mori, H.; et al. Localized 1689

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

Homopolymer at Preventing Cell Adhesion. Bioorg. Med. Chem. Lett. 1997, 7, 985−988. (205) Miyauchi, H.; Yuri, M.; Tanaka, M.; Kawamura, N.; Hayashi, M. Synthesis and Inhibitory Effects of Bivalent Sialyl Lewis X Analogs at Preventing Cell Adhesion. Bioorg. Med. Chem. Lett. 1997, 7, 989− 992. (206) Sasaki, K.; Nishida, Y.; Tsurumi, T.; Uzawa, H.; Kondo, H.; Kobayashi, K. Facile Assembly of Cell Surface Oligosaccharide Mimics by Copolymerization of Carbohydrate Modules. Angew. Chem., Int. Ed. 2002, 41, 4463−4467. (207) Nishida, Y.; Tsurumi, T.; Sasaki, K.; Watanabe, K.; Dohi, H.; Kobayashi, K. Design and Synthesis of C3-Symmetric LewisX Antigen. Org. Lett. 2003, 5, 3775−3778. (208) Rele, S. M.; Cui, W.; Wang, L.; Hou, S.; Barr-Zarse, G.; Tatton, D.; Gnanou, Y.; Esko, J. D.; Chaikof, E. L. Dendrimer-like PEO Glycopolymers Exhibit Anti-Inflammatory Properties. J. Am. Chem. Soc. 2005, 127, 10132−10133. (209) Gordon, E. J.; Strong, L. E.; Kiessling, L. L. GlycoproteinInspired Materials Promote the Proteolytic Release of Cell Surface LSelectin. Bioorg. Med. Chem. 1998, 6, 1293−1299. (210) Jones, M. W.; Otten, L.; Richards, S. J.; Lowery, R.; Phillips, D. J.; Haddleton, D. M.; Gibson, M. I. Glycopolymers with Secondary Binding Motifs Mimic Glycan Branching and Display Bacterial Lectin Selectivity in Addition to Affinity. Chem. Sci. 2014, 5, 1611−1616. (211) Gestwicki, J. E.; Kiessling, L. L. Inter-Receptor Communication through Arrays of Bacterial Chemoreceptors. Nature 2002, 415, 81−84. (212) Kiessling, L. L.; Gestwicki, J. E.; Strong, L. E. Synthetic Multivalent Ligands as Probes of Signal Transduction. Angew. Chem., Int. Ed. 2006, 45, 2348−2368. (213) Gestwicki, J. E.; Strong, L. E.; Kiessling, L. L. Tuning Chemotactic Responses with Synthetic Multivalent Ligands. Chem. Biol. 2000, 7, 583−591. (214) Mauris, J.; Mantelli, F.; Woodward, A. M.; Cao, Z.; Bertozzi, C. R.; Panjwani, N.; Godula, K.; Argüeso, P. Modulation of Ocular Surface Glycocalyx Barrier Function by a Galectin-3 N-terminal Deletion Mutant and Membrane Anchored Synthetic Glycopolymers. PLoS One 2013, 8, e72304. (215) Hudak, J. E.; Canham, S. M.; Bertozzi, C. R. Glycocalyx Engineering Reveals a Siglec-based Mechanism for NK Cell Immunoevasion. Nat. Chem. Biol. 2013, 10, 69−75. (216) Paszek, M. J.; DuFort, C. C.; Rossier, O.; Bainer, R.; Mouw, J. K.; Godula, K.; Hudak, J. E.; Lakins, J. N.; Wijekoon, A. C.; Cassereau, L.; et al. The Cancer Glycocalyx Mechanically Primes IntegrinMediated Growth and Aurvival. Nature 2014, 511, 319−325. (217) Rabuka, D.; Parthasarathy, R.; Lee, G. S.; Chen, X.; Groves, J. T.; Bertozzi, C. R. Hierarchical Assembly of Model Cell Surfaces: Synthesis of Mucin Mimetic Polymers and Their Display on Supported Bilayers. J. Am. Chem. Soc. 2007, 129, 5462−5471. (218) Grogan, M. J.; Kaizuka, Y.; Conrad, R. M.; Groves, J. T.; Bertozzi, C. R. Synthesis of Lipidated Green Fluorescent Protein and Its Incorporation in Supported Lipid Bilayers. J. Am. Chem. Soc. 2005, 127, 14383−14387. (219) Godula, K.; Umbel, M. L.; Rabuka, D.; Botyanszki, Z.; Bertozzi, C. R.; Parthasarathy, R. Control of the Molecular Orientation of Membrane-Anchored Biomimetic Glycopolymers. J. Am. Chem. Soc. 2009, 131, 10263−10268. (220) Jelinek, R.; Kolusheva, S. Carbohydrate Biosensors. Chem. Rev. 2004, 104, 5987−6016. (221) Singhvi, R.; Kumar, A.; Lopez, G. P.; Stephanopoulos, G. N.; Wang, D. I.; Whitesides, G. M.; Ingber, D. E. Engineering Cell Shape and Function. Science 1994, 264, 696−698. (222) Takara, M.; Toyoshima, M.; Seto, H.; Hoshino, Y.; Miura, Y. Polymer-Modified Gold Nanoparticles via RAFT Polymerization: a Detailed Study for a Biosensing Application. Polym. Chem. 2014, 5, 931−939. (223) Toyoshima, M.; Miura, Y. Preparation of GlycopolymerSubstituted Gold Nanoparticles and Their Molecular Recognition. J. Polym. Sci., Part A: Polym. Chem. 2009, 47, 1412−1421.

Surface Plasmon Resonance Detection of Biological Toxins Using Cell Surface Oligosaccharides on Glyco Chips. ACS Appl. Mater. Interfaces 2013, 5, 4173−4180. (188) Branson, T. R.; Turnbull, W. B. Bacterial Toxin Inhibitors based on Multivalent Scaffolds. Chem. Soc. Rev. 2013, 42, 4613−4622. (189) Nishikawa, K.; Matsuoka, K.; Watanabe, M.; Igai, K.; Hino, K.; Hatano, K.; Yamada, A.; Abe, N.; Terunuma, D.; Kuzuhara, H. Identification of the Optimal Structure Required for a Shiga Toxin Neutralizer with Oriented Carbohydrates to Function in the Circulation. J. Infect. Dis. 2005, 191, 2097−2105. (190) Polizzotti, B. D.; Kiick, K. L. Effects of Polymer Structure on the Inhibition of Cholera Toxin by Linear Polypeptide-Based Glycopolymers. Biomacromolecules 2006, 7, 483−490. (191) Liu, S.; Kiick, K. L. Architecture Effects on the Binding of Cholera Toxin by Helical Glycopeptides. Macromolecules 2008, 41, 764−772. (192) Polizzotti, B. D.; Maheshwari, R.; Vinkenborg, J.; Kiick, K. L. Effects of Saccharide Spacing and Chain Extension on Toxin Inhibition by Glycopolypeptides of Well-Defined Architecture. Macromolecules 2007, 40, 7103−7110. (193) Richards, S.-J.; Jones, M. W.; Hunaban, M.; Haddleton, D. M.; Gibson, M. I. Probing Bacterial-Toxin Inhibition with Synthetic Glycopolymers Prepared by Tandem Post-Polymerization Modification: Role of Linker Length and Carbohydrate Density. Angew. Chem., Int. Ed. 2012, 51, 7812−7816. (194) Miura, Y.; Yasuda, K.; Yamamoto, K.; Koike, M.; Nishida, Y.; Kobayashi, K. Inhibition of Alzheimer Amyloid Aggregation with Sulfated Glycopolymers. Biomacromolecules 2007, 8, 2129−2134. (195) Fukuda, T.; Matsumoto, E.; Onogi, S.; Miura, Y. Aggregation of Alzheimer Amyloid β Peptide (1−42) on the Multivalent Sulfonated Sugar Interface. Bioconjugate Chem. 2010, 21, 1079−1086. (196) Tsuchida, A.; Kobayashi, K.; Matsubara, N.; Muramatsu, T.; Suzuki, T.; Suzuki, Y. Simple Synthesis of Sialyllactose-Carrying Polystyrene and its Binding with Influenza Virus. Glycoconjugate J. 1998, 15, 1047−1054. (197) Totani, K.; Kubota, T.; Kuroda, T.; Murata, T.; Kazuya, I.-P.; Suzuki, T.; Suzuki, Y.; Kobayashi, K.; Ashida, H.; Yamamoto, K. Chemoenzymatic Synthesis and Application of Glycopolymers Containing Multivalent Sialyloligosaccharides with a Poly(L-glutamic acid) Backbone for Inhibition of Infection by Influenza Viruses. Glycobiology 2003, 13, 315−326. (198) Sasaki, K.; Nishida, Y.; Kambara, M.; Uzawa, H.; Takahashi, T.; Suzuki, T.; Suzuki, Y.; Kobayashi, K. Design of N-Acetyl-6-Sulfo-β-DGlucosaminide-Based Inhibitors of Influenza Virus Sialidase. Bioorg. Med. Chem. 2004, 12, 1367−1375. (199) Reuter, J. D.; Myc, A.; Hayes, M. M.; Gan, Z.; Roy, R.; Qin, D.; Yin, R.; Piehler, L. T.; Esfand, R.; Tomalia, D. A.; et al. Inhibition of Viral Adhesion and Infection by Sialic-Acid-Conjugated Dendritic Polymers. Bioconjugate Chem. 1999, 10, 271−278. (200) Becer, C. R.; Gibson, M. I.; Geng, J.; Ilyas, R.; Wallis, R.; Mitchell, D. A.; Haddleton, D. M. High-Affinity Glycopolymer Binding to Human DC-SIGN and Disruption of DC-SIGN Interactions with HIV Envelope Glycoprotein. J. Am. Chem. Soc. 2010, 132, 15130− 15132. (201) Zhang, Q.; Collins, J.; Anastasaki, A.; Wallis, R.; Mitchell, D. A.; Becer, C. R.; Haddleton, D. M. Sequence-Controlled Multi-Block Glycopolymers to Inhibit DC-SIGN-gp120 Binding. Angew. Chem. 2013, 125, 4531−4535. (202) Zhang, Q.; Su, L.; Collins, J.; Chen, G.; Wallis, R.; Mitchell, D. A.; Haddleton, D. M.; Becer, C. R. Dendritic cell lectin-targeting sentinel-like unimolecular glycoconjugates to release an anti-HIV drug. J. Am. Chem. Soc. 2014, 136, 4325−4332. (203) Kurtulus, I.; Yilmaz, G.; Ucuncu, M.; Emrullahoglu, M.; Becer, C. R.; Bulmus, V. A New Proton Sponge Polymer Synthesized by RAFT Polymerization for Intracellular Delivery of Biotherapeutics. Polym. Chem. 2014, 5, 1593−1604. (204) Miyauchi, H.; Tanaka, M.; Koike, H.; Kawamura, N.; Hayashi, M. Synthesis and Inhibitory Effect of a Sialyl Lewis X-Acrylamide 1690

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

(242) Chen, W.; Meng, F.; Cheng, R.; Deng, C.; Feijen, J.; Zhong, Z. Biodegradable Glycopolymer-b-poly (ε-caprolactone) Block Copolymer Micelles: Versatile Construction, Tailored Lactose Functionality, and Hepatoma-targeted Drug Delivery. J. Mater. Chem. B 2015, 3, 2308−2317. (243) Suriano, F.; Pratt, R.; Tan, J. P.; Wiradharma, N.; Nelson, A.; Yang, Y.-Y.; Dubois, P.; Hedrick, J. L. Synthesis of a Family of Amphiphilic Glycopolymers via Controlled Ring-Opening Polymerization of Functionalized Cyclic Carbonates and Their Application in Drug Delivery. Biomaterials 2010, 31, 2637−2645. (244) Dai, X. H.; Dong, C. M. Synthesis, Self-Assembly and Recognition Properties of Biomimetic Star-Shaped Poly(ε-caprolactone)-b-Glycopolymer Block Copolymers. J. Polym. Sci., Part A: Polym. Chem. 2008, 46, 817−829. (245) Liu, L.; Zhang, J.; Lv, W.; Luo, Y.; Wang, X. Well-Defined pHSensitive Block Glycopolymers via Reversible Addition-Fragmentation Chain Transfer Radical Polymerization: Synthesis, Characterization, and Recognition with Lectin. J. Polym. Sci., Part A: Polym. Chem. 2010, 48, 3350−3361. (246) Kim, B.; Peppas, N. A. Synthesis and Characterization of pHsensitive Glycopolymers for Oral Drug Delivery Systems. J. Biomater. Sci., Polym. Ed. 2002, 13, 1271−1281. (247) Jin, X.; Zhang, X.; Wu, Z.; Teng, D.; Zhang, X.; Wang, Y.; Wang, Z.; Li, C. Amphiphilic Random Glycopolymer based on Phenylboronic acid: Synthesis, Characterization, and Potential as Glucose-sensitive Matrix. Biomacromolecules 2009, 10, 1337−1345. (248) Kotsuchibashi, Y.; Agustin, R. V. C.; Lu, J.-Y.; Hall, D. G.; Narain, R. Temperature, pH, and Glucose Responsive Gels via Simple Mixing of Boroxole-and Glyco-based Polymers. ACS Macro Lett. 2013, 2, 260−264. (249) Utama, R. H.; Jiang, Y.; Zetterlund, P. B.; Stenzel, M. H. Biocompatible Glycopolymer Nanocapsules via Inverse Miniemulsion Periphery RAFT Polymerization for the Delivery of Gemcitabine. Biomacromolecules 2015, 16, 2144−2156. (250) Ahmed, M.; Narain, R. The Effect of Polymer Architecture, Composition, and Molecular Weight on the Properties of Glycopolymer-Based Non-Viral Gene Delivery Systems. Biomaterials 2011, 32, 5279−5290. (251) Ahmed, M.; Narain, R. The Effect of Molecular Weight, Compositions and Lectin Type on the Properties of Hyperbranched Glycopolymers as Non-viral Gene Delivery Systems. Biomaterials 2012, 33, 3990−4001. (252) Ahmed, M.; Deng, Z.; Liu, S.; Lafrenie, R.; Kumar, A.; Narain, R. Cationic Glyconanoparticles: Their Complexation with DNA, Cellular Uptake, and Transfection Efficiencies. Bioconjugate Chem. 2009, 20, 2169−2176. (253) Ahmed, M.; Jiang, X.; Deng, Z.; Narain, R. Cationic GlycoFunctionalized Single-Walled Carbon Nanotubes as Efficient Gene Delivery Vehicles. Bioconjugate Chem. 2009, 20, 2017−2022. (254) Ahmed, M.; Wattanaarsakit, P.; Narain, R. Cationic Glyconanogels for Epidermal Growth Factor Receptor (EGFR) Specific siRNA Delivery in Ovarian Cancer Cells. Polym. Chem. 2013, 4, 3829− 3836. (255) Obata, M.; Kobori, T.; Hirohara, S.; Tanihara, M. Synthesis of Poly [2-(α-d-mannopyranosyloxy) ethyl-co-2-dimethylaminoethyl methacrylates] and its Lectin-binding and DNA-Condensing Properties. Polymer 2012, 53, 4672−4677. (256) Obata, M.; Kobori, T.; Hirohara, S.; Tanihara, M. Aqueous RAFT Synthesis of Block and Statistical Copolymers of 2-(α-dmannopyranosyloxy) ethyl methacrylate with 2-(N, N-dimethylamino) ethyl methacrylate and Their Application for Nonviral Gene Delivery. Polym. Chem. 2015, 6, 1793−1804. (257) Lee, S.-G.; Brown, J. M.; Rogers, C. J.; Matson, J. B.; Krishnamurthy, C.; Rawat, M.; Hsieh-Wilson, L. C. End-functionalized Glycopolymers as Mimetics of Chondroitin Sulfate Proteoglycans. Chem. Sci. 2010, 1, 322−325. (258) Sun, X.-L.; Faucher, K. M.; Houston, M.; Grande, D.; Chaikof, E. L. Design and Synthesis of Biotin Chain-Terminated Glycopol-

(224) Ishii, J.; Toyoshima, M.; Chikae, M.; Takamura, Y.; Miura, Y. Preparation of Glycopolymer-Mediated Gold Nanoparticles and a New Approach for a Lateral Flow Assay. Bull. Chem. Soc. Jpn. 2011, 84, 466−470. (225) Ishii, J.; Chikae, M.; Toyoshima, M.; Ukita, Y.; Miura, Y.; Takamura, Y. Electrochemical Assay for Saccharide−Protein Interactions using Glycopolymer-Modified Gold Nanoparticles. Electrochem. Commun. 2011, 13, 830−833. (226) Morokoshi, S.; Ohhori, K.; Mizukami, K.; Kitano, H. Sensing Capabilities of Colloidal Gold Modified with a Self-Assembled Monolayer of a Glucose-Carrying Polymer Chain on a Glass Substrate. Langmuir 2004, 20, 8897−8902. (227) Kitano, H.; Nakada, H.; Mizukami, K. Interaction of Wheat Germ Agglutinin with an N-Acetylglucosamine-Carrying Telomer Brush Accumulated on a Colloidal Gold Monolayer. Colloids Surf., B 2008, 61, 17−24. (228) Vázquez-Dorbatt, V.; Maynard, H. D. Biotinylated Glycopolymers Synthesized by Atom Transfer Radical Polymerization. Biomacromolecules 2006, 7, 2297−2302. (229) Bontempo, D.; Maynard, H. D. Streptavidin as a Macroinitiator for Polymerization: In Situ Protein−Polymer Conjugate Formation. J. Am. Chem. Soc. 2005, 127, 6508−6509. (230) Narain, R.; Housni, A.; Gody, G.; Boullanger, P.; Charreyre, M.-T.; Delair, T. Preparation of Biotinylated Glyconanoparticles via a Photochemical Process and Study of Their Bioconjugation to Streptavidin. Langmuir 2007, 23, 12835−12841. (231) Jiang, X.; Housni, A.; Gody, G.; Boullanger, P.; Charreyre, M.T. R. S.; Delair, T.; Narain, R. Synthesis of Biotinylated α-dMannoside or N-Acetyl β-D-Glucosaminoside Decorated Gold Nanoparticles: Study of Their Biomolecular Recognition with Con A and WGA Lectins. Bioconjugate Chem. 2010, 21, 521−530. (232) Okada, S.; Peng, S.; Spevak, W.; Charych, D. Color and Chromism of Polydiacetylene Vesicles. Acc. Chem. Res. 1998, 31, 229− 239. (233) Baek, M.-G.; Stevens, R. C.; Charych, D. H. Design and Synthesis of Novel Glycopolythiophene Assemblies for Colorimetric Detection of Influenza Virus and E. coli. Bioconjugate Chem. 2000, 11, 777−788. (234) Phillips, R. L.; Kim, I.-B.; Tolbert, L. M.; Bunz, U. H. Fluorescence Self-Quenching of a Mannosylated Poly(p-phenyleneethynylene) Induced by Concanavalin A. J. Am. Chem. Soc. 2008, 130, 6952−6954. (235) Phillips, R. L.; Kim, I.-B.; Carson, B. E.; Tidbeck, B. r.; Bai, Y.; Lowary, T. L.; Tolbert, L. M.; Bunz, U. H. Design and Synthesis of Novel Glycopolythiophene Assemblies for Colorimetric Detection of Influenza Virus and E. coli. Macromolecules 2008, 41, 7316−7320. (236) Thurecht, K. J.; Blakey, I.; Peng, H.; Squires, O.; Hsu, S.; Alexander, C.; Whittaker, A. K. Functional Hyperbranched Polymers: Toward Targeted in Vivo 19F Magnetic Resonance Imaging Using Designed Macromolecules. J. Am. Chem. Soc. 2010, 132, 5336−5337. (237) Basuki, J. S.; Esser, L.; Duong, H. T.; Zhang, Q.; Wilson, P.; Whittaker, M. R.; Haddleton, D. M.; Boyer, C.; Davis, T. P. Magnetic Nanoparticles with Diblock Glycopolymer Shells Give Lectin Concentration-Dependent MRI Signals and Selective Cell Uptake. Chem. Sci. 2014, 5, 715−726. (238) Pfaff, A.; Schallon, A.; Ruhland, T. M.; Majewski, A. P.; Schmalz, H.; Freitag, R.; Müller, A. H. Magnetic and Fluorescent Glycopolymer Hybrid Nanoparticles for Intranuclear Optical Imaging. Biomacromolecules 2011, 12, 3805−3811. (239) Lu, Z.; Mei, L.; Zhang, X.; Wang, Y.; Zhao, Y.; Li, C. WaterSoluble BODIPY-Conjugated Glycopolymers as Fluorescent Probes for Live Cell Imaging. Polym. Chem. 2013, 4, 5743−5750. (240) van Kasteren, S. I.; Campbell, S. J.; Serres, S.; Anthony, D. C.; Sibson, N. R.; Davis, B. G. Glyconanoparticles Allow Pre-Symptomatic in vivo Imaging of Brain Disease. Proc. Natl. Acad. Sci. U. S. A., 2009, 106, 18−2310.1073/pnas.0806787106. (241) Wang, Y.; Zhang, X.; Yu, P.; Li, C. Glycopolymer Micelles with Reducible Ionic Cores for Hepatocytes-targeting Delivery of DOX. Int. J. Pharm. 2013, 441, 170−180. 1691

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Chemical Reviews

Review

ymers for Surface Glycoengineering. J. Am. Chem. Soc. 2002, 124, 7258−7259. (259) Gupta, S. S.; Raja, K. S.; Kaltgrad, E.; Strable, E.; Finn, M. Virus−Glycopolymer Conjugates by Copper(I) Catalysis of Atom Transfer Radical Polymerization and Azide−Alkyne Cycloaddition. Chem. Commun. 2005, 4315−4317. (260) Mancini, R. J.; Lee, J.; Maynard, H. D. Trehalose Glycopolymers for Stabilization of Protein Conjugates to Environmental Stressors. J. Am. Chem. Soc. 2012, 134, 8474−8479. (261) Lee, J.; Lin, E.-W.; Lau, U. Y.; Hedrick, J. L.; Bat, E.; Maynard, H. D. Trehalose Glycopolymers as Excipients for Protein Stabilization. Biomacromolecules 2013, 14, 2561−2569. (262) Lipinski, T.; Kitov, P. I.; Szpacenko, A.; Paszkiewicz, E.; Bundle, D. R. Synthesis and Immunogenicity of a Glycopolymer Conjugate. Bioconjugate Chem. 2011, 22, 274−281. (263) Geng, J.; Mantovani, G.; Tao, L.; Nicolas, J.; Chen, G.; Wallis, R.; Mitchell, D. A.; Johnson, B. R.; Evans, S. D.; Haddleton, D. M. Site-Directed Conjugation of “Clicked” Glycopolymers To Form Glycoprotein Mimics: Binding to Mammalian Lectin and Induction of Immunological Function. J. Am. Chem. Soc. 2007, 129, 15156−15163. (264) Parry, A. L.; Clemson, N. A.; Ellis, J.; Bernhard, S. S.; Davis, B. G.; Cameron, N. R. ‘Multicopy Multivalent’ Glycopolymer-Stabilized Gold Nanoparticles as Potential Synthetic Cancer Vaccines. J. Am. Chem. Soc. 2013, 135, 9362−9365. (265) Han, D.; Pal, S.; Nangreave, J.; Deng, Z.; Liu, Y.; Yan, H. DNA Origami with Complex Curvatures in Three-Dimensional Space. Science 2011, 332, 342−346. (266) Kim, K.; Matsuura, K.; Kimizuka, N. Binding of Lectins to DNA Micro-Assemblies: Modification of Nucleo-Cages with LactoseConjugated Psoralen. Bioorg. Med. Chem. 2007, 15, 4311−4317. (267) Matsuura, K.; Hibino, M.; Yamada, Y.; Kobayashi, K. Construction of Glyco-Clusters by Self-Organization of SiteSpecifically Glycosylated Oligonucleotides and Their Cooperative Amplification of Lectin-Recognition. J. Am. Chem. Soc. 2001, 123, 357−358.

1692

DOI: 10.1021/acs.chemrev.5b00247 Chem. Rev. 2016, 116, 1673−1692

Glycopolymer Nanobiotechnology.

Glycopolymer Nanobiotechnology. - PDF Download Free
4MB Sizes 1 Downloads 13 Views