You are on page 1of 9

Advanced Drug Delivery Reviews 57 (2005) 1733 – 1741

www.elsevier.com/locate/addr

Commentary

Molecularly imprinted polymers: A bridge to


advanced drug delivery B
Börje Sellergren a,*, Chris J. Allender b
a
INFU, Universität Dortmund, D-44221 Dortmund, Germany
b
Welsh School of Pharmacy Cardiff University, Cardiff XCF10 3XF, UK
Received 20 March 2005; accepted 29 July 2005
Available online 25 October 2005

Abstract

Specific molecular recognition is a fundamental requirement of living systems on which processes as diverse as neural
transmittance, respiration, immune defence, cellular differentiation and nutrition rely. It is therefore not surprising that scientists
have invested huge amounts of time and effort into harnessing, and more recently mimicking, biological function. A number of
synthetic approaches have been developed and one of the most promising of these is molecular imprinting. Molecular imprinted
polymers have routinely been used, as robust and effective synthetic molecular receptors, in a diverse range of technologies. But
it is perhaps in the area of drug delivery, in particular dintelligent drug releaseT and dmagic bulletT drug targeting, that significant
future opportunities lie.
D 2005 Elsevier B.V. All rights reserved.

Keywords: Molecularly imprinted polymers; Controlled drug delivery; Targeted drug delivery; Magic bullet; Synthetic receptors

1. Introduction cesses of life, information transfer and reaction cata-


lysis, rely on the specific interaction of low molecular
Specific molecular recognition is a fundamental weight molecules with macromolecular dhosts.T In the
requirement of living systems and, through millions majority of such events the macromolecule is a pro-
of years and countless rounds of evolutionary optimi- tein. Processes as diverse as neural transmittance,
zation, dbiologyT has become a master of the art. At the respiration, immune defence, cellular differentiation
cellular and sub-cellular level the fundamental pro- and nutrition all rely on the basic principle of specific
molecular recognition. It is therefore not surprising
that scientists have invested huge amounts of time
B
This commentary is part of the Advanced Drug Delivery and effort, initially into harnessing the potential of
Reviews theme issue on bMolecularly imprinted polymers: Technol- biological molecular recognition, antibodies and en-
ogy and applicationsQ, Vol. 57/12, 2005.
* Corresponding author. Tel.: +49 231 755 4082; fax: +49 231
zymes, and more recently in trying to mimic these
7554084. properties in synthetic materials. A number of syn-
E-mail address: borje@infu.uni-dortmund.de (B. Sellergren). thetic approaches have been developed that aim to
0169-409X/$ - see front matter D 2005 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2005.07.010
1734 B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741

Fig. 1. Schematic generalisation of the molecular imprinting process.

mimic the dynamic non-covalent molecular recogni- solving in the areas of preparative chemical separa-
tion approach favored by biology. Inherently, all of tions [3], solid phase extraction [4,5] and sensing [6].
these approaches aim to solve the problem in the same In addition, proof of concept of their use as scaven-
way, by creating a binding cavity or binding frame- gers to remove undesirable compounds from foods or
work within which functional chemical groups can be biological fluids [7], and as screening tools in drug
positioned so that reciprocal non-covalent interactions discovery [8] has also been demonstrated (Fig. 2).
between the binding site and a specific target analyte
are favored. One of the most promising of these
approaches is molecular imprinting. 2. A brief history of imprinting
Molecularly imprinted polymers (MIPs) are poly-
mers prepared in presence of a template (Fig. 1) [1,2] Molecular imprinting is not a new science. The
that serves as a mould for the formation of template earliest reports of imprinting go back to the early
complementary binding sites. Thus, MIPs can be 1930s when a Soviet chemist M.V. Polyakov [9] pre-
programmed to recognize a large variety of target pared a number of silica gels and observed that when
structures with antibody-like affinities and selectiv- prepared in the presence of a solvent additive the
ities. These properties, in addition to the robustness resulting silica demonstrated preferential binding
and ease of preparation of these artificial receptors, capacity for that solvent. Although Polyakov contin-
have made them extremely attractive for problem ued with this work until the1950s his findings were

Fig. 2. Scheme outlining the main applications envisaged for MIPs.


B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741 1735

seldom cited outside of Eastern Europe. A later study, covalent interactions during rebinding. Importantly, in
also using silica, was to have more of an impact. In order to improve subsequent non-covalent binding
1949 a senior student of Linus Pauling, Frank Dickey, geometry, Whitcombe’s approach incorporated a sacri-
published the results of experiments where silica gels ficial spacer group that was designed to be lost during
had been prepared in the presence of dyes [10]. Dickey template removal.
observed that after removal of the dpatterningT dye the The noncovalent approach however is still by far
silica would rebind the same dye in preference to the the most widely used approach in MIP synthesis.
others. Silica imprinting continued during the 1950s Several of its drawbacks can be overcome by the use
and 1960s but the number of publications in the area of stoichiometrically associating monomer-template
remained low. However, in 1972 a step change in systems [33–35,49]. This has resulted in a range of
molecular imprinting occurred when the group of receptors exhibiting high capacity and effective recog-
Guenter Wulff reported that they had successfully nition properties in aqueous media.
prepared a molecularly imprinting organic polymer
[11]. Wulff used what is now termed a dcovalent
approachT to prepare an organic molecularly imprinted 3. Molecular imprinting in drug delivery
polymer capable of discriminating between the enan-
tiomers of glyceric acid. Subsequently, throughout the Synthetic molecularly selective receptors such as
1970s and 1980s, Wulff’s group published extensively MIPs have broad application in many areas of science
using this approach. The second major break through but perhaps the area of greatest potential, and probably
in organic polymer imprinting occurred in 1981 when an area of greatest challenge, is that of therapeutics and
Mosbach and Arshady reported that they had prepared medical therapy. Examples of robust near real-time
an organic MIP using non-covalent interactions only diagnostic sensors, as discussed within this issue in
[12]. This approach was termed the dnon-covalent an article by Hillberg et al., and MIPs with ddrug-
approach,T as opposed to the covalent approach likeT effects, such as a cholesterol selective MIPs devel-
favored by Wulff, and it was this approach, with its oped for use as oral adsorbers [14] and imprinted bile
simple, seemingly trivial methodology, that triggered acid sequestrants [15,16], have triggered widespread
the explosion in molecular imprinting that was to occur interest in MIPs as fully synthetic molecularly selective
during the 1990s. To this day the non-covalent versus materials. In addition MIPs incorporated into mem-
covalent debate continues with both sides being cham- branes are being increasingly investigated in the con-
pioned. However, it is generally accepted that there text of bioseparations and biopurification [17]. For
are pros and cons to both approaches and in 1995 instance, biocompatible membranes imprinted with
Whitcombe et al. reported an intermediate approach uric acid have been developed for use in selective
that appeared to combine the advantages of both blood purification [18], whilst the area of drug separa-
approaches [13]. This approach relies on covalent tion and analysis is fully reviewed in an article by
interaction during the polymerization stage but non- Richard Ansell contained in this issue. But it is in the

Fig. 3. Intelligent dallostericT drug release from MIP carrier.


1736 B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741

Fig. 4. Targeted drug delivery using a molecularly imprinted carrier.

area of drug delivery that some of the most significant technologies that can bring about intelligent drug
opportunities lie, and what is particularly exciting is release or can target a therapeutic pay-load to a parti-
that to date, researchers have barely scratched the sur- cular site of action. Intelligent drug release refers to
face as to the potential of molecular imprinting. the release, in a predictable way, of a therapeutic agent
But MIPs are not entirely new in the area of drug in response to specific stimuli such as the presence of
delivery. They have been applied usefully as excipi- another molecule, whilst drug targeting is best exem-
ents in controlled release delivery systems and this is plified by the dmagic bulletT approach where a drug
discussed in detail by Cunliffe at al. later on in this conjugated to a targeting vector, such as an antibody
issue. A number of publications report that MIPs have or a peptide, interacts with specific sites of interac-
been used to modify drug release from solid dosage tions. A good example of this might be a cell surface
forms and that by modifying MIP composition release epitope. In both of these areas molecular imprinting
can be tuned. Although this work is of obvious value, has very real potential. For instance, intelligent con-
it does not exploit the real benefit of molecular trolled release could be achieved by the competitive
imprinting and that is selectivity. The MIP drug displacement of a drug by a structurally related cross-
release profiles reported in the literature are not excit- reactant. This type of direct displacement is probably
ing, similar profiles could have been obtained by the simplest way of generating a release profile in
simple modification of a non-imprinted polymer. response to a second analyte but other dallosteric-likeT
However, there is huge potential in drug delivery for phenomenon are also plausible (Fig. 3).

Fig. 5. Targeted drug delivery and facilitated internalisation using a MIP.


B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741 1737

Using MIPs to target drug delivery is also an origin of some of these issues it is important to under-
exciting concept (Fig. 4). The drug, coupled either stand some background.
covalently or non-covalently to the MIP, would be The majority of MIPs reported in the literature are
released when the MIP binds to its target on the highly reticulated network polymers consisting of a
surface of a cell. This concept could be extended so common matrix structure and binding sites formed
that the binding of the MIP to the cell surface would by a template present during the polymer synthesis
bring about internalisation of the MIP–drug complex (Fig. 1). The three dimensional arrangement of the
and subsequent drug release (Fig. 5). binding functional groups in MIPs is obtained by
Although these targeting applications are certainly linking the functional monomers covalently or non-
someway-off in terms of in vivo evaluation, molecular covalently to the template during polymerization
imprinting technology is close to the point where an in (Fig. 6). Removal of the template from the formed
vitro demonstration would be plausible. Currently, a polymer then generates a structure complementary to
wide range of polymers are being studied for con- the template structure. These sites can be reoccupied
trolled drug delivery, including a number of biocom- by the template or an analogous structure by refor-
patible acrylates and vinyl polymers, so the step mation of the binding interactions present during
change from current best practice is not great. In synthesis or, alternatively, weaker kinetically more
addition, biodegradable polymers, and the potential favorable interactions.
for biodegradable MIPs, would provide further flex-
ibility in polymer morphology and the resulting 4.1. Imprinting approaches
dosage form. It is therefore feasible that a working
system might comprise a surface imprinted (refer to Essentially, three approaches currently exist to gen-
the article by Mayes and Whitcombe in this issue) erate high fidelity imprinted sites, these are distin-
nanoparticulate MIP, selective for some cell surface guishable by the nature of the linkage during
epitope, conjugated to the active drug. synthesis and during rebinding.
Already chemically responsive gels have been The first example of molecular imprinting of
reported that release drug loads only in response to organic network polymers introduced by Wulff et al.
specific chemical stimuli [19,20], whilst chemically later followed by Shea et al. was based on a covalent
responsive materials [21,22] could also be envisaged attachment strategy, i.e., covalent monomer–template,
to act as smart valves in microfluidic systems, clos- covalent polymer–template [25,26]. This approach has
ing or opening channels in response to chemical the advantage of a known stoichiometry between the
signals [23]. Although MIPs are good at binding functional monomer and the template. Provided that
specific target molecules they have been less suc- the template can be recovered in high yields, a high
cessful in catalysing chemical reactions. However, density of well defined sites can be expected. One
using stable transition state analogues as templates, problem with this approach is the limited number of
recent advances indicate that there is potential in the covalent linkages that satisfy these criteria. Further-
future for MIPs to compete with their biological more, considerable synthetic effort may be required to
counterparts [24]. prepare the template and slow kinetics is often
observed for rebinding by reformation of the covalent
bond. This approach is therefore difficult to combine
4. Molecular imprinting: the challenges with applications where fast on-off kinetics is required.
In this respect, the use of sacrificial spacers has found
However, before some of these ideas can be further more widespread use [13]. Here the functional mono-
developed a number of technology issues need to be mer is bound to the template through a disposable
more fully addressed and these include the perfor- spacer that is removed after polymerization is com-
mance of MIP in fully aqueous environments, pleted. This results in a disposition of the functional
imprinting of very polar molecules and bio-macromo- groups allowing rebinding to occur through hydrogen
lecules, mass transfer, binding site population, capa- bonding interactions. Therefore, this approach can be
city and template bleed. But in order to appreciate the more amenable to chromatographic applications and
1738 B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741

Fig. 6. Strategies used to place binding or catalytic functional groups at defined positions in imprinted sites of network polymers.

furthermore allows more freedom in the choice of subtle structural differences in the template, similar
polymerization conditions (vide infra). to the selectivity displayed by antibodies.
However, the most widely used approach in
imprinting involves functional monomers that are 4.2. Solutions to the problems
chosen to associate non-covalently with the template
[27,28]. Here the template is directly mixed with one Advances made in the areas of host–guest chem-
or several functional monomers followed by polymer- istry, polymer and materials chemistry and analytical
ization. It can thereafter be easily extracted from the chemistry, each associated with the different steps
polymer and recycled. Generally, the resulting materi- in the MIP synthesis schemes, have guided the
als can be directly used to perform separations with way to much improved imprinting protocols. Thus
high affinity and selectivity for instance as chromato- approaches to generate MIPs tailored not only for
graphic stationary phases. This approach is very static molecular recognition but also for rapid binding
attractive since it is simple, it delivers MIPs showing and release possibly associated with a secondary func-
high affinity for its target and it is broadly applicable. tion, i.e., a measurable signal such as in sensors or a
Thus, using essentially the same procedure, MIPs mechanical work such as in stimuli-responsive gels,
targeting a large variety of small lipophilic targets are now available.
can be obtained. Conceivably, a simple commodity
monomer such as methacrylic acid (MAA) can be 4.2.1. High throughput synthesis and computational
used to create good binding sites for a large variety techniques
of template structures containing hydrogen bond- or The optimization of the molecular recognition
proton-accepting functional groups [29]. Commonly, properties under equilibrium conditions has been
these binding sites are capable of discriminating aided by computational [30] and high throughput
B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741 1739

synthesis (HTS) and screening techniques [31]. These small, the binding kinetics can also be expected to
techniques have mainly been used to rapidly identify strongly improve. Recently developed precipitation
suitable functional monomers for a given template but polymerization techniques [38] and microemulsion
are equally useful for optimizing ratios of monomers polymerization techniques [39] appear very promising
in co- or higher polymers. Recently, an HTS technique in this regard.
was successfully used to optimize terpolymers for
minimal nonspecific binding of lipophilic targets in 4.2.5. Composites
biological fluids [32]. In order to decouple the binding site formation
from the generation of a particular morphology,
4.2.2. New host monomers grafting techniques may be applied. Thus starting
Commonly, the monomers used in the above pro- from a support (e.g., a porous membrane [40], por-
tocols are commercially available and individually ous particles [41]) thin layers of imprinted polymers
provide typically only weak interactions with the may be grafted on its surface. Combining the graft-
template molecule. This leads to site heterogeneity, ing with controlled radical polymerization techniques
low saturation capacity and nonspecific binding. In offers a mean to perform this grafting in a controlled
order to address this issue, the monomer tool box may manner leading to a tunable polymer film thickness
be extended drawing inspiration from the area of [42]. In contrast to the polymer beads the thin film
host–guest chemistry. Thus designed functional composites have shown strongly improved mass
monomers are capable of stoichiometrically associat- transfer properties.
ing with the template in the imprinting step and sub-
sequently to allow strong binding in the rebinding step 4.2.6. Interfacial imprinting techniques
[33–35]. This minimizes site heterogeneity due to the By confining the binding sites to accessible sur-
functional monomer–template complex distribution. faces, mass transfer may be considerably enhanced
and the sites may be accessed by larger target mole-
4.2.3. The polymer matrix cules or when surface–surface recognition is required
The polarity and functionality of the material sur- as would be the case when using MIP-nanoparticles to
face need to be matched to the matrix environment for target drug delivery (Fig. 4). This is possible by
which the materials are designed. As mentioned above employing interfacial imprinting techniques where
water compatibility can be achieved by tuning the the template is confined at the interface between a
composition of terpolymers including hydrophilic monomer rich and a monomer poor phase [43,44]. If
comonomers or the use hydrophilic crosslinkers the template penetrates into the monomer rich phase a
[32,50]. Alternatively the organic polymer matrix surface imprint is generated which would be directly
may be entirely replaced by an inorganic one. Thus associated with a pore or a void tunable by the choice
sol–gel imprinting is growing in importance and sev- of monomer poor phase. In this regard, the immobi-
eral impressive examples of molecular recognition in lized template approach, making use of template mod-
aqueous systems using these materials have been ified porous silica, appears particularly promising
reported [36,37]. [45–47]. Here, the template is coupled to the silica
surface whereafter the pore system is filled with the
4.2.4. Polymer beads and nanoparticles imprint monomer mixture. Polymerization and subse-
The development of bead polymerization techni- quent removal of the silica by etching leave behind an
ques serves a twofold objective. First it addresses the organic polymer corresponding to the replica of the
technical issues related to the conventional monolith original silica pore system. In a recent extension of the
procedure to produce MIPs, e.g., low yields of useful protocol, peptides were synthesised by solid phase
particles, irregular particle sizes, time-consuming synthesis techniques on the silica surface [47]. The
crushing sieving techniques. Secondly, packed beds resulting material could recognize the peptide used as
of monodisperse spherical particles will exhibit less template but more importantly it also exhibited a high
band broadening effects than corresponding beds of affinity for larger peptides. Again this is an important
irregular particles. If the particles are sufficiently marker for future drug targeting studies.
1740 B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741

5. Conclusions [12] R. Arshady, M. Mosbach, Synthesis of substrate-selective


polymers by host–guest polymerization, Macromol. Chem.
Phys.-Makromol. Chem. 182 (1981) 687 – 692.
With no ambition to be comprehensive, the above [13] M.J. Whitcombe, M.E. Rodriguez, P. Villar, E.N. Vulfson, A
examples and discussion show that molecular imprint- new method for the introduction of recognition site function-
ing is alive and well. Recent enabling techniques are ality into polymers prepared by molecular imprinting—synth-
likely to open the door to a range of exciting applica- esis and characterization of polymeric receptors for
cholesterol, J. Am. Chem. Soc. 117 (1995) 7105 – 7111.
tions in the near future. Particularly important is the
[14] B. Sellergren, J. Wieschemeyer, K.-S. Boos, D. Seidel,
ability to engineer binding sites and the material Imprinted polymers for selective adsorption of cholesterol
morphology in a relatively simple and controlled fash- from gastro-intestinal mimicking fluids, Chem. Mater. 10
ion. The future will tell how far the imprinting engi- (1998) 4037 – 4046.
neers can take MIPs, and whether the fully synthetic [15] C.C. Huval, X. Chen, S.R. Holmes-Farley, W.H. Mandeville,
antibody is destined to become fact or fiction. This S.C. Polomoscanik, R.J. Sacchiero, P.K. Dhal, Molecularly
imprinted bile acid sequestrants: synthesis and biological stu-
current issue complements the recent Advanced Drug dies, in: B. Kofinas, B. Sellergren, M.J. Roberts (Eds.), Mole-
Delivery Review issue edited by Nicholas Peppas on cularly Imprinted Materials, vol 787, MRS, Warrendale, PA,
bIntelligent Therapeutics: Biomimetic systems and 2004, pp. 85 – 90.
nanotechnology in drug deliveryQ [48]. [16] C.C. Huval, M.J. Bailey, W.H. Braunlin, S.R. Holmes-Farley,
W.H. Mandeville, J.S. Petersen, R.J. Sacchiro, S.C. Polomos-
canik, X. Chen, P.K. Dhal, Novel cholesterol lowering poly-
meric drugs obtained by molecular imprinting, Macro
References molecules 34 (2001) 1548 – 1550.
[17] M. Ulbricht, Membrane separations using molecularly
[1] B. Sellergren (Ed.), Molecularly imprinted polymers, man imprinted polymers, J. Chromatogr., B-Anal. Technol.
made mimics of antibodies and their applications in Analytical Biomed. Life Sci. 804 (2004) 113 – 125.
Chemistry, in: Techniques and Instrumentation in Analytical [18] C. Cristallini, G. Ciardelli, N. Barbani, P. Giusti, Acryloni-
Chemistry, vol 23, Elsevier, Amsterdam, 2001. trile–acrylic acid copolymer membrane imprinted with uric
[2] B. Sellergren, Imprinted polymers with memory for small acid for clinical uses, Macromol. Biosci. 4 (2004) 31 – 38.
molecules, proteins, or crystals, Angew. Chem., Int. Ed. 39 [19] E. Oral, N.A. Peppas, Responsive and recognitive hydrogels
(2000) 1031 – 1037. using star polymers, J. Biomed. Mater. Res., Part A 68A
[3] B. Sellergren, Enantiomer separations using designed (2004) 439 – 447.
imprinted chiral phases, in: G. Subramanian (Ed.), Chiral [20] P. Bures, Y. Huang, E. Oral, N.A. Peppas, Surface modifications
Separation Techniques, 2nd edition, Wiley-VCH, Weinheim, and molecular imprinting of polymers in medical and pharma-
2001, pp. 153 – 184. ceutical applications, J. Control. Release 72 (2001) 25 – 33.
[4] F. Lanza, B. Sellergren, Molecularly imprinted extraction [21] M. Watanabe, T. Akahoshi, Y. Tabata, D. Nakayama, Mole-
materials for highly selective sample cleanup and analyte cular specific swelling change of hydrogels in accordance with
enrichment, Adv. Chromatogr. 41 (2001) 137 – 173. the concentration of guest molecules, J. Am. Chem. Soc. 120
[5] L.I Andersson, L. Schweitz, Solid-phase extraction on molecu- (1998) 5577 – 5578.
larly imprinted polymers, Handb. Anal. Sep. 4 (2003) 45 – 71. [22] Y. Kanekiyo, R. Naganawa, H. Tao, pH-responsive molecu-
[6] K. Haupt, K. Mosbach, Molecularly imprinted polymers and larly imprinted polymers, Angew. Chem., Int. Ed. 42 (2003)
their use in biomimetic sensors, Chem. Rev. 100 (2000) 3014 – 3016.
2495 – 2504. [23] T. Miyata, N. Asami, T. Uragami, A reversibly antigen respon-
[7] M.J. Whitcombe, C. Alexander, E.N. Vulfson, Smart polymers sive hydrogel, Nature 399 (1999) 766 – 769.
for the food industry, Trends Food Sci. Technol. 8 (1997) [24] G. Wulff, Imprinted catalysts, Chem. Rev. 102 (2002) 1.
140 – 145. [25] G. Wulff, A. Sarhan, Use of polymers with enzyme-analogous
[8] Y. Yu, L. Ye, K. Haupt, K. Mosbach, Formation of a class of structures for the resolution of racemates, Angew. Chem., Int.
enzyme inhibitors (drugs), including a chiral compound, by Ed. Engl. 11 (1972) 341.
using imprinted polymers or biomolecules as molecular-scale [26] K.J. Shea, T.K. Dougherty, Molecular recognition on synthetic
reaction vessels, Angew. Chem. 114 (2002) 4640 – 4643. amorphous surfaces—the influence of functional-group posi-
[9] M.V. Polyakov, Adsorption properties and structure of silica tioning on the effectiveness of molecular recognition, J. Am.
gel Zhur, Fiz. Khim. 2 (1931) 799 – 805. Chem. Soc. 108 (1986) 1091 – 1093.
[10] F.H. Dickey, Specific adsorbents, Proc. Natl. Acad. Sci. 35 [27] L. Andersson, B. Sellergren, K. Mosbach, Imprinting of amino
(1949) 227 – 229. acid derivatives in macroporous polymers, Tetrahedron Lett.
[11] G. Wulff, A. Sarhan, Über die Anwendung von enzymanalog 25 (1984) 5211 – 5214.
gebauten Polymeren zur Racemattrennung, Angew. Chem. 84 [28] B. Sellergren, B. Ekberg, K. Mosbach, Molecular imprinting
(1972) 364. of amino acid derivatives, J. Chromatogr. 347 (1985) 1 – 10.
B. Sellergren, C.J. Allender / Advanced Drug Delivery Reviews 57 (2005) 1733–1741 1741

[29] B. Sellergren, M. Lepistoe, K. Mosbach, Highly enantioselec- [40] M. Ulbricht, M. Belter, U. Langenhangen, W.W. Schneider,
tive and substrate-selective polymers obtained by interactions. Novel molecularly imprinted polymer (MIP) composite mem-
NMR and chromatographic studies on the nature of recogni- branes via controlled surface and pore functionalizations,
tion, J. Am. Chem. Soc. 110 (1988) 5853 – 5860. Desalination 149 (2002) 293 – 295.
[30] S.A. Piletsky, K. Karim, E.V. Piletska, C.J. Day, D.W. Free- [41] C. Sulitzky, B. Rückert, A.J. Hall, F. Lanza, K. Unger, B.
bairn, C. Legge, A.P.F. Turner, Recognition of ephedrine enan- Sellergren, Grafting of molecularly imprinted polymer films
tiomers by molecularly imprinted polymers designed using a on silica supports containing surface-bound free radical initia-
computational approach, Analyst 126 (2001) 1826 – 1830. tors, Macromolecules 35 (2002) 79 – 91.
[31] F. Lanza, B. Sellergren, Molecularly imprinted polymers via [42] B. Sellergren, B. Ruckert, A.J. Hall, Layer-by-layer grafting of
high-throughput and combinatorial techniques, Macromol. molecularly imprinted polymers via iniferter modified sup-
Rapid Commun. 25 (2004) 59 – 68. ports, Adv. Mater. 14 (2002) 1204 – 1208.
[32] B. Dirion, Z. Cobb, E. Schillinger, L.I. Andersson, B. Sell- [43] S. Nakamura, H. Takeuchi, H. Nakamura, T.M. Maeda, Mole-
ergren, Water-compatible molecularly imprinted polymers cularly imprinted resin for recognition amino acids using self-
obtained via high-throughput synthesis and experimental assembly at o/w interface, Trans. Mater. Res. Soc. Jpn. 24
design, J. Am. Chem. Soc. 125 (2003) 15101 – 15109. (1999) 453 – 456.
[33] G. Wulff, T. Gross, R. Schönfeld, Enzyme models based on [44] M.A. Markowitz, P.R. Kust, G. Deng, P.E. Schön, J.S. Dor-
molecularly imprinted polymers with strong esterase activity, dick, D.S. Clark, B.P. Gaber, Silica solgel catalysis, Langmuir
Angew. Chem., Int. Ed. Engl. 36 (1997) 1962 – 9164. 16 (2000) 1759.
[34] C. Lübke, M. Lübke, M.J. Whitcombe, E.N. Vulfson, [45] E. Yilmaz, K. Haupt, K. Mosbach, The use of immobilized
Imprinted polymers prepared with stoichiometric template– templates—a new approach, Angew. Chem., Int. Ed. 39 (2000)
monomer complexes: efficient binding of ampicillin from 2115 – 2118.
aqueous solutions, Macromolecules 33 (2000) 5098 – 5105. [46] M.M. Titirici, A.H. Hall, B. Sellergren, Hierarchically
[35] P. Manesiotis, A.J. Hall, M. Emgenbroich, M. Quaglia, E. de imprinted stationary phases: mesoporous polymer beads con-
Lorenzi, B. Sellergren, An enantioselective imprinted receptor taining surface-confined binding sites for adenine, Chem.
for Z-glutamate exhibiting a binding induced colour change, Mater. 14 (2002) 21 – 23.
Chem. Commun. (2004) 2278 – 2279. [47] M.M. Titirici, A.H. Hall, B. Sellergren, Hierarchical imprint-
[36] T. Kunitake, S.-W. Lee, Molecular imprinting in ultrathin ing using crude solid phase peptide synthesis products as
titania gel films via surface sol–gel process, Anal. Chim. templates, Chem. Mater. 15 (2003) 822 – 824.
Acta 504 (2004) 1 – 6. [48] N.A. Peppas (Ed.), Intelligent Therapeutics: Biomimetic
[37] S. Marx, A. Zaltsman, I. Turyan, D. Mandler, Parathion sensor Systems and Nanotechnology in Drug Delivery,
based on molecularly imprinted sol–gel films, Anal. Chem. 76 Advanced Drug Delivery Reviews, vol. 56 (issue 11),
(2004) 120 – 126. 2004, pp. 1529 – 1688.
[38] J. Wang, P.A.G. Cormack, D.C. Sherrington, E. Khoshdel, [49] B. Sellergren, Imprinted dispersion polymers. New easily
Monodisperse, molecularly imprinted polymer microspheres accessible affinity stationary phases, J. Chromatogr. 673
prepared by precipitation polymerization for affinity (1994) 133.
separation applications, Angew. Chem., Int. Ed. 42 (2003) [50] P. Manesiotis, A.J. Hall, J. Courtouis, K. Irgum, B. Sellergren,
5336 – 5338. An artificial riboflavine receptor via a template analogue
[39] G.E.M. Tovar, I. Kraeuter, C. Gruber, Molecularly imprinted imprinting strategy, Angew. Chem. Int. Ed. 44 (2005)
polymer nanospheres as fully synthetic affinity receptors, Top. 3902 – 3906.
Curr. Chem. 227 (2003) 125 – 144.

You might also like