Sei sulla pagina 1di 10

Sensors and Actuators B 147 (2010) 765–774

Contents lists available at ScienceDirect

Sensors and Actuators B: Chemical


journal homepage: www.elsevier.com/locate/snb

Review

Hydrogel-based devices for biomedical applications


Kosmas Deligkaris ∗ , Tadele Shiferaw Tadele, Wouter Olthuis, Albert van den Berg
University of Twente, Department of Electrical Engineering, P.O. Box 217, 7500 AE Enschede, The Netherlands

a r t i c l e i n f o a b s t r a c t

Article history: This review paper presents hydrogel-based devices for biomedical applications. The first part of the
Received 18 January 2010 paper gives a comprehensive, qualitative, theoretical overview of hydrogels’ synthesis and operation.
Received in revised form 25 March 2010 Crosslinking methods, operation principles and transduction mechanisms are discussed in this part. The
Accepted 26 March 2010
second part includes applications of hydrogel devices in specific fields of interest. Sensing, fluid control,
Available online 2 April 2010
drug delivery, nerve regeneration and other biomedical applications constitute the main focus of this
part. The aim of this paper is to briefly present recent advances of the field, without neglecting older
Keywords:
ones, and to discuss important or novel concepts of each.
Stimuli-responsive hydrogel
Chemical sensor © 2010 Elsevier B.V. All rights reserved.
Fluid control
Drug delivery
Nerve regeneration

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
2. Synthesis of hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 766
2.1. Physical crosslinking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 766
2.2. Chemical crosslinking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 766
3. Theoretical studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 766
4. Operation principles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 767
5. Transduction mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 767
5.1. Optical methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
5.2. Conductometric and amperometric methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
5.3. Mechanical methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
5.3.1. Microcantilevers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
5.3.2. Bending plate transducers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
6. Hydrogel applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
6.1. Sensing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
6.1.1. pH sensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
6.1.2. Additional chemical sensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
6.2. Array networks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
6.3. Fluid control and drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 770
6.4. Artificial muscles and nerve regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 770
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 774

1. Introduction

∗ Corresponding author. Tel.: +31 611179743. Hydrogels are three-dimensional hydrophilic polymer net-
E-mail addresses: k.deligkaris@student.utwente.nl, kraig33@gmail.com works made up of water-soluble polymers, crosslinked to form a
(K. Deligkaris). water-insoluble hydrogel. They are able to swell and retain a sig-

0925-4005/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.snb.2010.03.083
766 K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774

nificant portion of water when placed in an aqueous solution [1]. Besides, polymers can also interact by charge interaction
The amount of water in the polymer matrix is at least 20% [2] (Fig. 1b) or by forming hydrogen bonds between them (Fig. 1c),
and can reach values of 99% by weight [3]. Hydrogels containing acting as a physical crosslink between the polymers. Charge inter-
more than 95% water are termed superabsorbent and have high action can occur between a polymer and a small molecule or
biocompatibility due to their large degree of water retention and between two oppositely charged polymers [4]. Hydrogen and other
their physiochemical similarity with the native extracellular matrix non-covalent bonds are much weaker than covalent ones.
both compositionally and mechanically [4]. In addition, they can be
made to be biodegradable. Hydrogels can be classified into different 2.2. Chemical crosslinking
groups based on their:
When the bonds between the polymers are of covalent nature,
• physical structure: amorphous, semicrystalline, hydrogen- the crosslinking is of chemical character. Covalent interactions are
bonded or supramolecular; much stronger than non-covalent, providing excellent mechanical
• electric charge: ionic (charged) or neutral; stability. Chemical crosslinking methods include radical polymer-
• crosslink: physically or chemically crosslinked; ization, chemical reaction of complementary groups, high energy
• responses to external effects: stimulus-sensitive and -insensitive irradiation and enzyme usage [25]. For chemical crosslinking, con-
ones; trary to physical crosslinking, crosslinking agents are needed which
• origin: synthetic and natural. may react with other substances [23].

Research on hydrogels started as early as in 1960s with a novel 3. Theoretical studies


paper on poly(2-hydroxyethyl methacrylate) by Wichterle and
Lim [5]. In 1968, it was successfully predicted that the net repul- Theoretical study of hydrogel networks has the purpose of
sion between a polymer network and a poor solvent can cause a revealing the structure and configuration of their chains and ana-
phase transition and a change in the swelling degree (and thus to lyzing hydrogels’ kinetics by means of modeling. One of the most
its volume) [6]. Other groups also reported a phase transition in well-known theoretical studies is the equilibrium swelling theory
their experiments induced by changes in the environment [7,8]. [26]. Flory and Rehner developed the theory of the equilibrium
Up to date hydrogels’ use has increased dramatically. Many dif- swelling of crosslinked polymer gels by modeling them as a Gaus-
ferent synthesis techniques have been presented [9–13]. Review sian distribution of polymer chains. In their analysis the swelling
papers have been also published in this field [14–19] presenting of the polymer network was dependent on the elastic forces of the
the theoretical background and applications in specific areas such polymer chains and the thermodynamic compatibility of the poly-
as molecular imprinting [15], micro-total analysis systems (␮TAS) mer and water molecules. Thus, the free energy change of a neutral
[17] and microsensors [19]. However, to our knowledge, there has hydrogel can be expressed as:
not been a recent review paper presenting a complete overview of
hydrogel biomedical applications. The purpose of this paper is to G = Gel + Gmix
give a thorough, qualitative overview of the field, while presenting
where Gel represents the elastic retractive forces and Gmix the
recent developments of hydrogel devices for biomedical applica-
thermodynamic compatibility of the polymer and the swelling
tions and practical issues arising during synthesis, modeling or
agent [27]. In case of ionic hydrogels, several additional factors have
use.
to be taken in account such as degree of ionization, nature of the
counter ions and ionization equilibrium. As the ionic content of the
2. Synthesis of hydrogels hydrogel increases, repulsive forces among the charges develop and
the hydrogel swells. When the charge density decreases the hydro-
Hydrogels are formed by physical or chemical crosslinks of gel shrinks. In addition to the thermodynamic equilibrium used
homopolymers or copolymers, being appropriately used to give in neutral networks, chemical potential of the ions should also be
the three-dimensional structures specific mechanical and chemical in equilibrium. Detailed information can be found in the original
characteristics. The crosslink can be formed by covalent [20,21] or paper and in various reference books [1,2]. When the assumption
non-covalent interactions [22–24]. Covalently crosslinked hydro- of a Gaussian distribution is not adequate non-Gaussian models can
gels are also called chemical gels while non-covalent gels are be used [28,29].
termed physical. The crosslink can take place after or at the same The equilibrium weight swelling ratio can be used to describe
time as the copolymerization [1]. Chemical hydrogels provide good the degree of swelling and can be measured by gravimetric tech-
mechanical strength but suffer from side-effects [23]. Detailed niques (e.g. [30]). The hydrogel is immersed in distilled water until
information about various physical and crosslinking methods has it reaches its equilibrium state. It is then taken out and after wip-
been published by Hennink and van Nostrum [25]. ing out excess water it is being weighted. The equilibrium swelling
ratio of the hydrogel, SReq , can be calculated from measured values
2.1. Physical crosslinking as follows:
Ws
Physical gels are three-dimensional networks where the poly- SReq =
Wd
mer chains bond through non-covalent interactions. Junction zones
form when the separate polymer chains interact over a certain where Ws is the weight of swollen hydrogel in equilibrium and Wd
length and not point to point (point crosslinks). One of the ways is the initial dry weight of the hydrogel.
to form physical crosslinking is hydrophobic interaction (Fig. 1a) When a hydrogel swells according to a change in a stimulus, the
in which hydrophobic blocks are coupled to hydrophilic blocks rate at which the hydrogel’s volume change takes place could be
creating a polymer amphiphile. When temperature is increased more important than the equilibrium swelling ratio.
the hydrophobic blocks aggregate. The polymer concentration, the A widely used parameter describing hydrogel swelling is the
hydrophobic’s block length and the chemical structure of the poly- hydrogel’s characteristic time of swelling (), which is proportional
mer all affect the temperature in which the phase change takes to the square of the characteristic length of the gel (L) and inversely
place [4]. proportional to the diffusion coefficient of the gel network in the
K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774 767

Fig. 1. (a) Hydrophobic interactions drive in situ physical gelation, (b) in situ physical gelation driven by charge interactions, (c) physical gelation driven by hydrogen bonding
interactions, which can be disrupted by shear. Reprinted from [4] with permission from Elsevier.

solvent (D) as follows [31]: Thermo-responsive gels go through a phase transition in


response to external temperature stimulus. The temperature
L2
= dependence can be positive [70] or negative [71,72]. With positive
D temperature dependence the swelling increases with increasing
The characteristic length is the radius for spherical hydrogels temperature, while for negative it decreases. Polymers contain-
and the thickness for hydrogel sheets. The diffusion coefficient of ing mostly hydrophilic groups are water-soluble, while polymers
hydrogel networks is in the order of 10−7 cm2 /s [31,32]. A 1-mm- containing mostly hydrophobic groups are water-insoluble. For
thick gel slab with a diffusion coefficient of 10−7 cm2 /s takes over negative temperature sensitivity the polymers are soluble in water
an hour to reach 50% of the equilibrium swelling and more than 6 h at low temperatures due to their hydrophilic content, but they col-
to reach 90% of equilibrium [33]. This slow swelling property can lapse above a certain temperature due to increased hydrophobic
been found useful in applications where a slow, steady response interactions and exhibit phase separation from the solution. This
is needed but the majority of hydrogels applications are in micro- temperature is termed the Lower Critical Solution Temperature
scaled systems. (LCST).
A common group of monomers, used in the synthesis of
4. Operation principles temperature-sensitive hydrogels, are the N-alkyl acrylamides. A
well-known monomer from this group is N-isopropylacrylamide
The family of stimulus-sensitive hydrogels includes (NIPAAm). This monomer has side chains which have favorable
pH-sensitive [34–66], temperature-sensitive hydrogels interactions with water in the form of hydrogen bonds. The
[10,18,36,38,39,53,58–60,62,67–69] and hydrogels with mul- efficiency of the hydrogen bonding process has negative tem-
tiple sensitivities [36,53,58,62]. Temperature- and pH-sensitive perature dependence. At lower temperatures, hydrogen bonding
hydrogels are very different in their physical behavior and between hydrophilic segments of the polymer chain and the water
swelling mechanism. The following paragraphs discuss the pH- molecules dominates, leading to enhanced dissolution in water.
and temperature-sensitive hydrogels independently. As the temperature increases, hydrophobic interactions among
pH-sensitive polymers are produced by adding acidic or basic hydrophobic segments become strengthened, while hydrogen
functional groups to the polymer backbone; these either accept or bonding becomes weaker. At higher temperatures the hydrogen
release protons in response to appropriate pH and ionic strength bond based crosslinks will break up causing a shrinking in the
changes in aqueous media. hydrogel.
Neutral polymer gels show less swelling than polyelectrolyte van der Linden et al. [73] have presented a new method for the
gels. The more ionized a hydrogel is, the more charges will exist, photolithographical deposition of temperature-sensitive hydro-
increasing the electrostatic repulsion between the polymer chains. gels, while investigating many of the hydrogel’s characteristics. One
The network is more hydrophilic then, and the swelling degree is of them was the effect of the crosslinking density on its swelling
increased. The degree of ionization of these hydrogels depends on behavior. Their results (Fig. 2) indicate that the more the crosslink-
the number of pendant acidic or basic groups in the hydrogel. ing density the less the swelling.
Hydrogels containing acidic groups become more ionized at
higher pH and basic hydrogels show more ionization at low pH. 5. Transduction mechanisms
Thus, acidic hydrogels tend to swell more as the pH of the surround-
ing solution increases while basic hydrogels exhibit the opposite Sensor transducers are components, which convert the non-
behavior. electrical changes of properties of the stimuli-responsive hydrogel
768 K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774

Fig. 3. (a) Microcantilevers, (b) bending plate transducer with a piezo-resistive ele-
ment, and (c) capacitive bending plate sensor. Reproduced from [19].

5.3. Mechanical methods

5.3.1. Microcantilevers
Microcantilevers can transduce changes of mass, temperature,
heat, or stress, into bending (static mode) or a change in resonance
frequency (dynamic mode). Hydrogel swelling leads to alteration
of surface stress, which in turn statically bends the microcantilever.
Fig. 2. Swelling response for different crosslinking densities. Fabricated poly- For dynamic mode, a change in the measurand alters the reso-
N-isopropylacrylamide (PNIPAAm) hydrogels clearly show negative temperature nant frequency which is measured after proper excitation of the
dependence. A polyacrylamide hydrogel used as a reference was not temperature
structure. This form of output is considered digital, since it can be
sensitive. Reproduced from [73] by permission of the Royal Society of Chemistry.
directly connected to digital circuits and is independent from ana-
log levels. This approach has many advantages when compared to
into an electrical signal. Transducers can exploit the mechanical analog output (static mode). It minimizes errors from analog to dig-
work performed by hydrogels or observe alteration in hydrogel ital conversion and the transduction sensitivity of such elements is
properties such as density, volume and stiffness. Various trans- considered to be among the highest. The sensitivity of the resonant
duction methods have been presented up to date for measuring sensor is of crucial importance in this case.
hydrogel properties changes. These include optical, conductomet- Dynamic modeling of a microcantilever can be complicated and
ric, amperometric and mechanical methods and are presented various simplifications such as linearity, absence of damping effects
below. and assumption of rigid supports may be needed [79]. Fig. 3a illus-
trates the operation principle in the static mode. A number of
publications using microcantilevers can be found in the literature
5.1. Optical methods [41,42,47,80,81] while review papers have been published by Car-
rascosa et al. [82] and Lang et al. [83].
The optical methods for detecting the volume change of hydro-
gels have been widely explored with different techniques. Two 5.3.2. Bending plate transducers
similar optical techniques are fluorescence resonance energy trans- These transducers usually include a piezo-resistive
fer (FRET) [50] and non-radiative energy transfer (NRET) [74]. Both Wheatstone-bridge and are commonly used as pressure sen-
techniques involve a donor fluorescent dye emitting a photon sors [48,51,84]. The stimuli-sensitive hydrogel is placed in a fixed
which is consumed by another dye. Distance between the dyes, volume between a stiff/rigid grate, which is permeable for the
and thus of the swelling, can be measured with these techniques. stimulus, and the bending plate (Fig. 3b). When the hydrogel swells,
Another optical method makes use of diffraction of light from a the plate deflects resulting in a change of the resistance of the
hydrogel containing a crystalline colloidal array (CCA) [37,75,76]. piezo-resistive bridge [19]. Device modeling can be accomplished
The particles in the CCA are regularly spaced and cause the gel to by means of finite element modeling (FEM). Trinh et al. presented a
display Bragg diffraction dependent on the lattice spacing. When hydrogel-based silicon piezo-resistive pH sensor [51]. The authors
the hydrogel changes volume, the spacing of the particles in the CCA state the need for modeling and optimization of the devices, which
is changed, causing a shift in the wavelengths of Bragg diffraction. was accomplished with ANSYS software. Membrane deflection,
membrane stress and voltage output results were computed and
measured with a good agreement between them although the
5.2. Conductometric and amperometric methods response time of the real structure was slower than the model
prediction. The authors give possible reasons for this such as
Conductometric methods employ the measurement of resis- that gel extension was assumed to happen instantaneously and
tance values which vary according to the measurand [35]. A thin time constants used for calculation were determined during free
hydrogel layer was deposited on a planar interdigitated conduc- swelling experiments and not inside a silicon chip cavity.
tivity electrode array. The hydrogel changes volume in response to A capacitive bending plate sensor as shown in Fig. 3c has been
pH changes leading to a corresponding increase or decrease in ion presented by Strong et al. [85]. The bending of the plate changes the
mobility inside the hydrogel layer and a change in conductivity. A distance of the capacitor plates resulting in a change of its capaci-
common problem, loss of adhesion is acknowledged by the authors. tance which is inversely proportional to the distance between the
Lesho and Sheppard [77] examined the adhesion of polymer hydro- plates. This change in capacitance leads to a change in the resonance
gel films to oxidized silicon wafers and conductometric sensors. frequency of a passive LC circuit.
Among the results were that adhesion was dependent on pH val-
ues while temperature effects were minimal. Another method of 6. Hydrogel applications
detection is amperometric [78], in which measurement of the elec-
tric current after application of a potential difference between two This part describes hydrogels’ applications from various
electrodes provides information regarding the measurand. research groups and discusses their practical issues. Hydrogels that
K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774 769

6.1.2. Additional chemical sensors


Hydrogels have been used for various sensing purposes either
directly or indirectly [45,49]. In direct methods the measurand
directly affects the output of the system. In indirect methods the
measurand affects an intermediate quantity, for example pH, which
in turn alters the output of the system. Many medically significant
sensors have been created including carbon dioxide sensors [45,49],
lactate sensors [86], sensors for rheochemical characterization of
solutions [87], oxygen sensors [56,88] and ion sensors [80,89].
One of the significant problems a designer of a hydrogel sensor
has to solve is the sensor’s selectivity. This problem is of out-
most importance in biomedical conditions where the environment
(human body) is not completely controlled (although its proper-
ties might be known) and invasive methods are preferred to be
avoided. Complications arise when, for example, an ion sensor pro-
duces non-zero output analogous to the concentration of a second
ion, or when it is affected by temperature.
Fig. 4. Equilibrium cantilever deflection as a function of pH around the polymer.
Reprinted with permission from [41], © 2002, American Institute of Physics. Temperature sensitivity is acknowledged by Herber et al. [49],
where the authors acquire measurements in room temperature and
in the operational temperature during biomedical use. In this case
can sense various external stimuli have been presented. The field the increase of temperature has a positive effect in the response
of sensing seems to attract most attention and therefore it is logical time of the sensor. However, the output of the sensor was reduced
to start with this theme. Other applications include drug deliv- with increasing temperature for a constant concentration of the
ery, fluid control, artificial muscles and nerve regeneration and are measurand. Fortunately, the relationship was found to be linear,
discussed later. making possible a compensation technique. In non-linear cases,
linearization methods are employed increasing the complexity and
6.1. Sensing cost of the final device.
The issue of other elements affecting the operation of the sensor
6.1.1. pH sensors is explored on two typical papers of ion sensors using a micro-
Bashir et al. [41] have presented one of the most sensitive cantilever [80,89]. In both papers the authors have examined the
micromechanical pH sensors, having an ability to sense a change influence of other ions in the sensors. The pH was held constant
of 5 × 10−5 pH by using an atomic force microscope for deflection during these experiments, so its effect on the sensor was minimal.
measurement. It is fabricated by coating a polymer network on The conclusion was that, for the particular sensors, some ions have
a cantilever. A change in pH leads to a change in surface stress minor effects while others had significant effects at large concen-
and expansion of the polymer, which in turn bends the cantilever. trations only.
Various simplifications were made since the actual model is quite
complicated. More specifically, the structure was modeled as a 6.2. Array networks
composite beam with a constant modulus of elasticity and constant
polymer thickness. In reality, the modulus of elasticity decreases Recently presented papers describe exploitation of array tech-
while the polymer thickness increases with a pH increase. These niques for various purposes. One of these is the characterization
simplifications, as the authors acknowledge, may justify the small of liquid analytes [90]. A fluorimetric micro-spot array using a
differences between theoretical and experimental results as illus- non-specific recognition function is used for the analysis of liquid
trated in Fig. 4. samples. The array was composed of binary mixtures of various flu-
A conductimetric pH microsensor is presented by Sheppard et orescence dyes embedded in a hydrogel matrix. By visual extraction
al. [35]. The sensor resistance changes as much as 45% per pH unit of the changes in each spot and in the array as a whole, the method
at physiological pH values. Different conductivity-over-pH graphs seems to be effectively used as a recognition tool. The specific
were produced for different copolymer compositions. The authors array was composed of 32 individual spots (doubly redundant, for
found that the interfacial impedance between the gel and the elec- reproducibility demonstration) and was prepared from single flu-
trode plays a significant role in the total impedance and that the orescent dyes and binary mixtures. Image substraction techniques
loss of adhesion (Section 5.2) between the substrate and the gel were used to visualize the post pattern differences, assigned as the
leads to a decrease in sensitivity. fingerprint of the analyte liquid (Fig. 5).
A pH sensor based on a reversible, mass-changing pH- An application of hydrogel arrays in cell biology research is
responsive hydrogel is presented by Ruan et al. [43]. The authors presented by Jongpaiboonkit et al. [91]. Due to the vast number
achieved increased sensitivity by increasing the fraction of the of parameters, cell behavior in a natural environment is difficult
acrylic acid in the poly(acrylic acid-co-isooctyl) acrylate copolymer. to analyze. An adaptable, automated approach for 3D cell culture
Excellent adhesion was established by pre-coating the sensor with within hydrogel arrays was proposed. As the authors conclude:
a layer of polyurethane, and adding 1-(3-dimethylaminopropyl)-3
ethylcarbodiimide (EDC) into the hydrogel solution. The output of “our results indicate that the adaptable, array-based format
the sensor is encoded in the frequency of the output signal. developed in this study may be useful as an enhanced through-
More recently, Maruyama et al. [57] presented a different put platform for 3-D culture of a variety of cell types”.
approach in local pH sensing. Their patterned hydrogel film is made Another fluorescent microarray sensor [56] is able to sense
of UV photosensitive resin. The output is a change in color (color dissolved oxygen and pH in a cell culture, with a response time
difference for red) of the hydrogel and is obtained by a CCD cam- appropriate for its purpose. The output of the device is optical, while
era. This form of output is one of the most easily recognizable for the whole measurement technique is non-invasive. Other studies
humans, although small details may not be easily distinguished include hydrogel-based protein arrays for analyte sensing [46] and
with the human eye. quantification of epidermal growth factor receptor (EGFR, a trans-
770 K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774

Fig. 6. Operation of a fluid flow controller. The volume response of two different
hydrogels with respect to the pH of the surrounding fluid. Top, the fractional change
in diameter (fD ) of the hydrogels with respect to pH. Bottom, images showing a
device that directs (sorts) a fluid stream on the basis of its pH. The hydrogel gating
the right branch (circles on the top graph) expands in base and contracts in acid. The
hydrogel gating the left branch (squares on the top graph) behaves in the opposite
manner (expands in acid and contracts in base). At a pH of 7.8, the flow is directed
down the left branch. At a pH of 4.7, the flow is directed down the right branch. Both
hydrogels expand to shut off the flow when the pH is changed to 6.7. Scale bars,
300 mm. Reprinted by permission from Macmillan Publishers Ltd.: Nature [94], ©
2000.

Fig. 5. Operation principle of a hydrogel array used for characterization of liquid


term functionality of the enclosed cells and improve the mechanical
analytes. (a) Reference image (dry array) and (b) image under pure methanol. Spot
pattern obtained after image subtraction (a-b) fluorescence diminished spot pattern.
stability of the capsules which can be used in controlled drug deliv-
(b-a) Fluorescence intensified spot pattern. Reprinted from [90] with permission ery. This approach can also be used in other therapeutic methods
from Elsevier. such as central nervous system (CNS) diseases and various patho-
logical problems.
Pachyman is a fungus polysaccharide, well-known in China
membrane protein, which if overexpressed could result in cancer)
for its medical applications, harmless to the human body [101].
activity in cell lysates [92].
A biodegradable pachyman-based hydrogel has been devised for
controlled release of protein drugs with full preservation of the
6.3. Fluid control and drug delivery protein’s stability and enzymatic activity [64]. The authors state
the possibilities of this technique for site-specific protein–drugs
Since the sensing of any measurand is accompanied by a volume delivery.
change of the hydrogel, they can be easily used as actuators or as a A transcutaneous vaccination system has been also studied
combination of sensing–actuation mechanisms. Hydrogel elements [102]. Benefits include the lack of need for specialized personnel,
are soft materials and therefore, able to completely seal the fluid lack of needles, lack of specialized facilities and others. The specific
pathway, minimizing leakage [93]. The most common approach vaccination system using hydrogels, successfully delivered anti-
for controlling the fluid flow is by having the hydrogel swelling genic proteins into the epidermal layer in quantities that exceeded
in response to a stimulus and blocking the fluid’s pathway [94]. the theoretical expectations.
This approach is illustrated in Fig. 6. In Figs. 6a and c one hydrogel Reversing the fluid flow, a sampling mechanism, a micronee-
sphere (with a stimulus-dependent state) is in a shrunken state, dle and a micro-glucose sensor constitute a so-called intelligent
allowing the fluid to pass either to the right or to the left (fluid flow mosquito being able to pump in and sense the glucose concentra-
is represented by an arrow). In Fig. 6b both spheres are in a swelling tion of a solution [103]. The 90% response time was 30–40 s, typical
state, completely blocking the fluid’s pathway (indicated by an X). for such applications. This artificial “mosquito” is disposable and so,
Possible stimuli found in the literature are temperature [93], pH it does not bear any fear of transmitting diseases from patient to
[94,95] and glucose [95,96]. patient. Possible improvements include better geometrical design
The suitability of hydrogels for drug delivery stems from the and faster sampling mechanism.
fact that they are biocompatible and can be made to be biodegrad-
able. They do not pose any risk of rejection by the immune system
and when entering the human body they slowly degrade, releas- 6.4. Artificial muscles and nerve regeneration
ing the drug they carry. However, poorly soluble drugs are difficult
to entrap in the hydrogel matrix and Chen et al. [97] use a drug Even from the 1989 researchers had realized the tremendous
entrapment strategy to increase the efficiency of such methods. capabilities of hydrogels to act as artificial muscles [104]. A more
In cell microencapsulation techniques, cells are immobilized detailed study of artificial muscles, acknowledging that the muscle
within capsules [98]. Immobilization of therapeutically active is both a system and a material has been recently done [105]. The
cells (drug-secreting) within a polymer matrix surrounded by a authors state advantages of polyacrylamide (PAAM) hydrogels for
semi-permeable membrane protects them from immune system such applications such as:
rejection [99] which makes use of immunosuppressants unneces-
sary. Orive et al. [100] were successful in designing biomimetic • Properties and materials similar to living tissue.
cell-hydrogel capsules to promote even further the in vivo long- • Biocompatibility.
K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774 771

Fig. 7. Representative fluorescent images of agarose-treated and agarose/BDNF-treated spinal cords with NF-160 kDa stain. (A) An image of agarose-treated spinal cord
section at 10×. Scale bar = 50 ␮m. (B and C) Enlarged images (20×) of the white boxes in A. This shows that the axons stopped before entering the lesions and that they formed
bulbs; (D) agarose/BDNF-treated spinal cord at 10×; (E and F) enlarged images (20×) of the white boxes in (D). These images show that the axons did not bulb at the end as
the axons did in the agarose-treated spinal cords and the axons infiltrated the scaffold, thus crossing the interface. Reprinted from [115] with permission from Elsevier.

• Not biodegradable. dent or disease. This difference has been attributed to the different
• Chemical manipulation of properties. environment in PNS and CNS lesion sites. Main barriers to regener-
• Adjustable shape. ation are the glial scar, which is composed from reactive astrocytes
• Low-cost. and proteoglycans, degenerating myelin (myelin debris) and oligo-
dendrocytes [106,107]. Chondroitin sulfate proteoglycans are the
The main disadvantage of hydrogels, namely their slow response principal inhibitory component of glial scars but their effect can
time could not pass undetected however. This could mean that be reduced or even eliminated with chondroitinase ABC (ChABC)
hydrogel-based artificial muscles cannot be effectively used when [108,109]. Besides the glial scar formation, CNS injury results in cell
fast response is required, although to our knowledge there have not death and pseudocyst formation further restricting regeneration.
been any detailed studies comparing the kinetics of real muscles By promoting nerve growth factors (substances promoting
against artificial hydrogel muscles. Also, in contrary to some drug nerve growth) and other therapeutic agents (such as ChABC)
delivery methods, when using hydrogels as artificial muscles they researchers are focusing on changing the environmental conditions
should be made able to withstand the physiological environment in CNS lesion sites, and thus restoring nerve regeneration ability.
without degradation. Hydrogels can be used in spinal cord injury repair [110–115] by
Hydrogels can be also used for nervous system repair. The cen- acting as scaffolds bridging the gap between lesions [110,112,115]
tral nervous system (CNS) consists of the brain and the spinal or by delivering neurotrophic factors [113,115], favoring regenera-
cord, while the peripheral nervous system (PNS) connects CNS to tion of neural connections. Syková et al. [111] found good adhesion
the rest of the human body. The PNS can recover after damage of HEMA hydrogels in the host tissue, bridging the whole spinal
while the CNS has limited capacity of replacing damaged or lost cord lesion site. The same group noticed a smaller pseudocyst vol-
neurons, resulting in a permanent loss of function following an acci- ume in hydrogels implanted 1 week after damage when compared
772 K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774

to immediate implantation. However, when a solid hydrogel is [15] M.E. Byrne, K. Park, N.A. Peppas, Molecular imprinting within hydrogels, Adv.
implanted, the surgical procedure itself will cause damage. This Drug Deliv. Rev. 54 (1) (2002) 149–161.
[16] A.S. Hoffman, Hydrogels for biomedical applications, Adv. Drug Deliv. Rev. 54
damage could be spared by using liquid hydrogels solidifying (1) (2002) 3–12.
inside the lesion site [110,114,115]. Jain et al. [115] investigated [17] H.J. van der Linden, S. Herber, W. Olthuis, P. Bergveld, Stimulus-sensitive
an agarose scaffold, geling in situ and conformally filling an irreg- hydrogels and their applications in chemical (micro)analysis, The Analyst 128
(4) (2003) 325–331.
ular spinal cord defect in adult rats. By embedding brain-derived [18] L. Klouda, A.G. Mikos, Thermoresponsive hydrogels in biomedical applica-
neurotrophic factor (BDNF) releasing microtubules in the scaffold, tions, Eur. J. Pharm. Biopharm. 68 (2007) 34–45.
neurite growth was successfully enhanced (Fig. 7). [19] A. Richter, G. Paschew, S. Klatt, J. Lienig, K.-F. Arndt, H.-J.P. Adler, Review
on hydrogel-based pH sensors and microsensors, Sensors 8 (1) (2008)
561–581.
7. Conclusions [20] R. Langer, J. Folkman, Polymers for the sustained release of proteins and other
macromolecules, Nature 263 (5580) (1976) 797–800.
[21] P. Edman, B. Ekman, I. Sjoholm, Immobilization of proteins in microspheres
This review briefly presented the basic principles of operation of biodegradable polyacryldextran, J. Pharm. Sci. 69 (7) (1980) 838–842.
and practical aspects of hydrogel devices in a wide variety of appli- [22] S.J. de Jong, B. van Eerdenbrugh, C.F. van Nostrum, J.J. Kettenes-van den Bosch,
cations. They are able to convert chemical and electrical energy W.E. Hennink, Physically crosslinked dextran hydrogels by stereocomplex
formation of lactic acid oligomers: degradation and protein release behavior,
into mechanical energy and therefore are considered to be chemo-
J. Control. Rel. 71 (3) (2001) 261–275.
mechanical transducers. [23] W.E. Hennink, S.J. De Jong, G.W. Bos, T.F.J. Veldhuis, C.F. van Nostrum,
Hydrogels are becoming widely applied into traditional fields Biodegradable dextran hydrogels crosslinked by stereocomplex formation
of science or medicine such as sensing and actuating. Their soft for the controlled release of pharmaceutical proteins, Int. J. Pharm. 277 (1–2)
(2004) 99–104.
nature makes them excellent candidates in applications where [24] C. Xiao, M. Yang, Controlled preparation of physical cross-linked starch-g-PVA
dust particles would pose a threat or reduced the efficiency of hydrogel, Carbohydr. Polym. 64 (1) (2006) 37–40.
other structures. Besides, their biocompatibility and optional bio- [25] W.E. Hennink, C.F. van Nostrum, Novel crosslinking methods to design hydro-
gels, Adv. Drug Deliv. Rev. 54 (1) (2002) 13–36.
degradability further increases their vast possibilities. Examples [26] P.J. Flory, J. Rehner, Statistical mechanics of cross-linked polymer networks.
described in the present review include drug delivery and nerve II. Swelling, J. Chem. Phys. 11 (1943) 521–526.
regeneration applications. One of the most significant disadvan- [27] N. Yui, R.J. Mrsny, K. Park, Reflexive Polymers and Hydrogels: Understanding
and Designing Fast Responsive Polymeric Systems, CRC Press, 2004.
tages is their slow response time, which makes them unsuitable [28] M. Fixman, R. Alben, Polymer conformational statistics. I. Probability, J. Chem.
for large-scale applications or in situations where fast response is Phys. 58 (1973).
required (e.g. when used as artificial muscles or sensors for fast- [29] M. Fixman, J. Kovac, Polymer conformational statistics. III. Modified Gaussian
models of stiff chains, J. Chem. Phys. 58 (1973).
changing parameters). [30] A.A. Dinerman, J. Cappello, H. Ghandehari, S.W. Hoag, Swelling behavior of a
It is evident that the complexity of the devices and applications genetically engineered silk–elastinlike protein polymer hydrogel, Biomateri-
is increasing while research in this field is mature enough to start als 23 (21) (2002) 4203–4210.
[31] T. Tanaka, D.J. Fillmore, Kinetics of swelling of gels, J. Chem. Phys. 70 (3) (1979)
experimenting with advanced structures. As a result, it can be safely
1214–1218.
predicted that hydrogel devices will continue to be highly involved [32] B.G. Kabra, S.H. Gehrke, Rate-limiting steps for solvent sorption and desorp-
in biomedical research and applications in the future to come. tion by microporous stimuli-sensitive absorbent gels, in: F.L. Buchholz, N.A.
However, specific directions for research focus cannot be given. Peppas (Eds.), Superabsorbent Polymers: Science and Technology, American
Chemical Society, Washington, DC, 1994, pp. 76–86.
Different applications demand hydrogels with different properties. [33] S.H. Gehrke, Synthesis, equilibrium swelling, kinetics permeability and appli-
Examples include bio-degradability (drug delivery) or not (artificial cations of environmentally responsive gels, in: K. Dusek (Ed.), Responsive
muscles), single or array elements, etc. Gels: Volume Transitions II, Springer, Berlin/Heidelberg, New York, 1993, pp.
81–144.
[34] S. Draxler, M.E. Lippitsch, pH sensors using fluorescence decay time, Sens.
References Actuators B 29 (1–3) (1995) 199–203.
[35] N.F. Sheppard, M.J. Lesho, P. McNally, A.S. Francomacaro, Microfabricated
[1] N.A. Peppas, Hydrogels in Medicine and Pharmacy, vol. 1, CRC Press, 1986. conductimetric pH sensor, Sens. Actuators B 28 (2) (1995) 95–102.
[2] H. Park, K. Park, Hydrogels in bioapplications, in: R.M. Ottenbrite, S.J. Huang, [36] S.K. Sahoo, T.K. De, P.K. Ghosh, A. Maitra, pH- and thermo-sensitive hydrogel
K. Park (Eds.), Hydrogels and Biodegradable Polymers for Bioapplications, nanoparticles, J. Colloid Interface Sci. 206 (2) (1998) 361–368.
American Chemical Society, Washington, DC, 1996, pp. 2–10. [37] K. Lee, S.A. Asher, Photonic crystal chemical sensors: pH and ionic strength, J.
[3] V. Kudela, Encyclopedia of Polymer Science and Engineering, Wiley, New Am. Chem. Soc. 122 (39) (2000) 9534–9537.
York, 1987. [38] L.H. Gan, Y.Y. Gan, G.R. Deen, Poly(N-acryloyl-N -propylpiperazine):
[4] T.R. Hoare, D.S. Kohane, Hydrogels in drug delivery: progress and challenges, a new stimuli-responsive polymer, Macromolecules 33 (21) (2000)
Polymer 49 (8) (2008) 1993–2007. 7893–7897.
[5] O. Wichterle, D. Lim, Hydrophilic gels in biological use, Nature 185 (1960) [39] L.H. Gan, G. Roshan Deen, X.J. Loh, Y.Y. Gan, New stimuli-responsive copoly-
117–118. mers of N-acryloyl-N -alkyl piperazine and methyl methacrylate and their
[6] K. Dusek, D. Patterson, Transition in swollen polymer networks induced by hydrogels, Polymer 42 (1) (2001) 65–69.
intramolecular condensation, J. Polym. Sci. Part A-2: Polym. Phys. 6 (7) (1968) [40] J. Varshosaz, M. Falamarzian, Drug diffusion mechanism through pH-sensitive
1209–1216. hydrophobic/polyelectrolyte hydrogel membranes, Eur. J. Pharm. Biopharm.
[7] T. Tanaka, I. Nishio, S.-T. Sun, S. Ueno-Nishio, Collapse of gels in an electric 51 (3) (2001) 235–240.
field, Science 218 (4571) (1982) 467–469. [41] R. Bashir, J.Z. Hilt, O. Elibol, A. Gupta, N.A. Peppas, Micromechanical can-
[8] S. Hirotsu, Electric-field-induced phase transition in polymer gels, J. Appl. tilever as an ultrasensitive pH microsensor, Appl. Phys. Lett. 81 (16) (2002)
Phys. 24 (1985) 388–396. 3091–3093.
[9] M. Zhai, F. Yoshii, T. Kume, K. Hashim, Syntheses of PVA/starch grafted hydro- [42] J.Z. Hilt, A.K. Gupta, R. Bashir, N.A. Peppas, Ultrasensitive biomems sen-
gels by irradiation, Carbohydr. Polym. 50 (3) (2002) 295–303. sors based on microcantilevers patterned with environmentally responsive
[10] Z. Zhao, Z. Li, Q. Xia, H. Xi, Y. Lin, Fast synthesis of temperature-sensitive hydrogels, Biomed. Microdev. 5 (3) (2003) 177–184.
PNIPAAm hydrogels by microwave irradiation, Eur. Polym. J. 44 (4) (2008) [43] C. Ruan, K. Zeng, C.A. Grimes, A mass-sensitive pH sensor based on a stimuli-
1217–1224. responsive polymer, Anal. Chim. Acta 497 (1–2) (2003) 123–131.
[11] J.C. Cuggino, C.I.A. Igarzabal, J.C. Rueda, L.M. Quinzani, H. Komber, M.C. [44] B.D. Johnson, D.J. Beebe, W.C. Crone, Effects of swelling on the mechanical
Strumia, Synthesis and characterization of new hydrogels through copoly- properties of a pH-sensitive hydrogel for use in microfluidic devices, Mater.
merization of N-acryloyl-tris-(hydroxymethyl)aminomethane and different Sci. Eng., C 24 (4) (2004) 575–581.
crosslinking agents, Eur. Polym. J. 44 (11) (2008) 3548–3555. [45] S. Herber, W. Olthuis, P. Bergveld, A. van den Berg, Exploitation of a pH-
[12] S.-E. Park, Y.-C. Nho, H.-I. Kim, Preparation of poly(polyethylene glycol sensitive hydrogel disk for CO2 detection, Sens. Actuators B 103 (1–2) (2004)
methacrylate-co-acrylic acid) hydrogels by radiation and their physical prop- 284–289.
erties, Radiat. Phys. Chem. 69 (3) (2004) 221–227. [46] V.K. Yadavalli, W.-G. Koh, G.J. Lazur, M.V. Pishko, Microfabricated protein-
[13] S. Benamer, M. Mahlous, A. Boukrif, B. Mansouri, S.L. Youcef, Synthesis and containing poly(ethylene glycol) hydrogel arrays for biosensing, Sens.
characterisation of hydrogels based on poly(vinyl pyrrolidone), Nucl. Instrum. Actuators B 97 (2–3) (2004) 290–297.
Methods Phys. Res., Sect. B 248 (2) (2006) 284–290. [47] Y. Zhang, H.-F. Ji, D. Snow, R. Sterling, G.M. Brown, A pH sensor based on a
[14] N.A. Peppas, J.J. Sahlin, Hydrogels as mucoadhesive and bioadhesive materi- microcantilever coated with intelligent hydrogel, Instrum. Sci. Technol. 32 (4)
als: a review, Biomaterials 17 (1996) 1553–1561. (2004) 361–369.
K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774 773

[48] G. Gerlach, M. Guenther, J. Sorber, G. Suchaneck, K.-F. Arndt, A. Richter, [78] C. Wartelle, W. Schuhmann, A. Blöchl, F. Bedioui, Integrated compact bio-
Chemical and pH sensors based on the swelling behavior of hydrogels, Sens. compatible hydrogel-based amperometric sensing device for easy screening
Actuators B 111–112 (2005) 555–561. of drugs involved in nitric oxide production by adherent cultured cells, Elec-
[49] S. Herber, J. Bomer, W. Olthuis, P. Bergveld, A. van den Berg, A miniaturized trochim. Acta 50 (25–26) (2005) 4988–4994.
carbon dioxide gas sensor based on sensing of pH-sensitive hydrogel swelling [79] B. Bahreyni, Fabrication and Design of Resonant Microdevices, William
with a pressure sensor, Biomed. Microdev. 7 (3) (2005) 197–204. Andrew, 2008.
[50] A.C. Mack, J. Mao, M.J. McShane, Transduction of pH and glucose-sensitive [80] Y. Zhang, H.-F. Ji, G.M. Brown, T. Thundat, Detection of CrO42—using a hydro-
hydrogel swelling through fluorescence resonance energy transfer, Sensors, gel swelling microcantilever sensor, Anal. Chem. 75 (18) (2003) 4773–4777.
IEEE (2005) 912–915. [81] N.I. Abu-Lail, M. Kaholek, B. LaMattina, R.L. Clark, S. Zauscher, Micro-
[51] Q.T. Trinh, G. Gerlach, J. Sorber, K.-F. Arndt, Hydrogel-based piezoresistive pH cantilevers with end-grafted stimulus-responsive polymer brushes for
sensors: design, simulation and output characteristics, Sens. Actuators B 117 actuation and sensing, Sens. Actuators B 114 (1) (2006) 371–378.
(1) (2006) 17–26. [82] L.G. Carrascosa, M. Moreno, M. Álvarez, L.M. Lechuga, Nanomechanical
[52] Y.-Y. Liu, X.-D. Fan, B.-R. Wei, Q.-F. Si, W.-X. Chen, L. Sun, pH-responsive biosensors: a new sensing tool, TrAC-Trends Anal. Chem. 25 (3) (2006)
amphiphilic hydrogel networks with IPN structure: a strategy for controlled 196–206.
drug release, Int. J. Pharm. 308 (1–2) (2006) 205–209. [83] H.P. Lang, M. Hegner, C. Gerber, Cantilever array sensors, Mater. Today 8 (4)
[53] S. Bhattacharya, F. Eckert, V. Boyko, A. Pich, Temperature-, pH-, and magnetic- (2005) 30–36.
field-sensitive hybrid microgels, Small 3 (4) (2007) 650–657. [84] I.S. Han, M.-H. Han, J. Kim, S. Lew, Y.J. Lee, F. Horkay, J.J. Magda, Constant-
[54] M George, T.E. Abraham, pH sensitive alginate-guar gum hydrogel for the volume hydrogel osmometer: a new device concept for miniature biosensors,
controlled delivery of protein drugs, Int. J. Pharm. 335 (1–2) (2007) 123–129. Biomacromolecules 3 (6) (2002) 1271–1275.
[55] J.X. Sun, Y.F. Luo, H. Peng, Z.W. Han, Design, synthesis and characterization of [85] Z.A. Strong, A.W. Wang, C.F. McConaghy, Hydrogel-actuated capacitive trans-
a novel pH-sensitive hydrogel, Chin. Chem. Lett. 19 (12) (2008) 1475–1478. ducer for wireless biosensors, Biomed. Microdev. 4 (2) (2002) 97–103.
[56] S. Lee, B.L. Ibey, G.L. Coté, M.V. Pishko, Measurement of pH and dissolved [86] R.C.H. Kwan, P.Y.T. Hon, K.K.W. Mak, R. Renneberg, Amperometric
oxygen within cell culture media using a hydrogel microarray sensor, Sens. determination of lactate with novel trienzyme/poly(carbamoyl) sulfonate
Actuators B 128 (2) (2008) 388–398. hydrogel-based sensor, Biosens. Bioelectron. 19 (12) (2004) 1745–1752.
[57] H. Maruyama, H. Matsumoto, T. Fukuda, F. Arai, Functionalized hydrogel [87] M. Guenther, G. Gerlach, C. Corten, D. Kuckling, J. Sorber, K.F. Arndt, Hydrogel-
surface patterned in a chip for local pH sensing, in: IEEE 21st Int. Conf. on based sensor for a rheochemical characterization of solutions, Sens. Actuators
Microelectromech. Syst., 2008. MEMS 2008, 2008. B 132 (2) (2008) 471–476.
[58] C. Zhao, X. Zhuang, P. He, C. Xiao, C. He, J. Sun, X. Chen, X. Jing, Synthesis [88] D.P. O’Neal, M.A. Meledeo, J.R. Davis, B.L. Ibey, V.A. Gant, M.V. Pishko, G.L. Cote,
of biodegradable thermo- and pH-responsive hydrogels for controlled drug Oxygen sensor based on the fluorescence quenching of a ruthenium complex
release, Polymer 50 (18) (2009) 4308–4316. immobilized in a biocompatible poly(ethylene glycol) hydrogel, Sens. J. IEEE
[59] D.P. Huynh, M.K. Nguyen, B.S. Kim, D.S. Lee, Molecular design of novel 4 (6) (2004) 728–734.
pH/temperature-sensitive hydrogels, Polymer 50 (12) (2009) 2565–2571. [89] K. Liu, H.-F. Ji, Detection of Pb2+ : using a hydrogel swelling microcantilever
[60] H. Liu, M. Liu, L. Ma, J. Chen, Thermo- and pH-sensitive comb-type grafted sensor, Anal. Sci. 20 (1) (2004) 9–11.
poly(N,N-diethylacrylamide-co-acrylic acid) hydrogels with rapid response [90] A.R. Thete, T. Henkel, R. Göckeritz, M. Endlich, J.M. Köhler, G.A. Groß, A hydro-
behaviors, Eur. Polym. J. 45 (7) (2009) 2060–2067. gel based fluorescent micro array used for the characterization of liquid
[61] J. Chen, L. Rong, H. Lin, R. Xiao, H. Wu, Radiation synthesis of pH-sensitive analytes, Anal. Chim. Acta 633 (1) (2009) 81–89.
hydrogels from ␤-cyclodextrin-grafted PEG and acrylic acid for drug delivery, [91] L. Jongpaiboonkit, W.J. King, G.E. Lyons, A.L. Paguirigan, J.W. Warrick, D.J.
Mater. Chem. Phys. 116 (1) (2009) 148–152. Beebe, W.L. Murphy, An adaptable hydrogel array format for 3-dimensional
[62] J. Zhang, R. Xie, S.-B. Zhang, C.-J. Cheng, X.-J. Ju, L.-Y. Chu, Rapid cell culture and analysis, Biomaterials 29 (23) (2008) 3346–3356.
pH/temperature-responsive cationic hydrogels with dual stimuli-sensitive [92] G. Ghosh, A.G. Lee, S.P. Palecek, Hydrogel-based protein array for quantifying
grafted side chains, Polymer 50 (11) (2009) 2516–2525. epidermal growth factor receptor activity in cell lysates, Anal. Biochem. 393
[63] M.J. Mc Gann, C.L. Higginbotham, L.M. Geever, M.J.D. Nugent, The synthe- (2) (2009) 205–214.
sis of novel pH-sensitive poly(vinyl alcohol) composite hydrogels using a [93] A. Richter, D. Kuckling, S. Howitz, G. Thomas, K.F. Arndt, Electronically con-
freeze/thaw process for biomedical applications, Int. J. Pharm. 372 (1–2) trollable microvalves based on smart hydrogels: magnitudes and potential
(2009) 154–161. applications, J. Microelectromech. Syst. 12 (5) (2003) 748–753.
[64] Y. Xiao, W. Xu, Q. Zhu, B. Yan, D. Yang, J. Yang, X. He, S. Liang, X. Hu, Preparation [94] D.J. Beebe, J.S. Moore, J.M. Bauer, Q. Yu, R.H. Liu, C. Devadoss, B.-H. Jo, Func-
and characterization of a novel pachyman-based pharmaceutical aid. II: A pH- tional hydrogel structures for autonomous flow control inside microfluidic
sensitive, biodegradable and biocompatible hydrogel for controlled release of channels, Nature 404 (6778) (2000) 588–590.
protein drugs, Carbohydr. Polym. 77 (3) (2009) 612–620. [95] A. Baldi, G. Yuandong, P.E. Loftness, R.A. Siegel, B. Ziaie, A hydrogel-actuated
[65] H. Li, Y.K. Yew, Simulation of soft smart hydrogels responsive to pH stimu- environmentally sensitive microvalve for active flow control, J. Microelec-
lus: ionic strength effect and case studies, Mater. Sci. Eng. C 29 (7) (2009) tromech. Syst. 12 (5) (2003) 613–621.
2261–2269. [96] N.A. Dzulkefli, M. Tehranirokh, B.Y. Majlis, Preparation of glucose hydrogel as
[66] Y. Yue, X. Sheng, P. Wang, Fabrication and characterization of microstructured actuator in microvalve, in: IEEE Int. Conf. on Semicond. Electron., 2008. ICSE
and pH sensitive interpenetrating networks hydrogel films and application 2008, 2008.
in drug delivery field, Eur. Polym. J. 45 (2) (2009) 309–315. [97] M.-C. Chen, H.-W. Tsai, C.-T. Liu, S.-F. Peng, W.-Y. Lai, S.-J. Chen, Y. Chang,
[67] Y.H. Bae, T. Okano, R. Hsu, S.W. Kim, Thermo-sensitive polymers as on–off H.-W. Sung, A nanoscale drug-entrapment strategy for hydrogel-based sys-
switches for drug release, Die Makromolekulare Chemie, Rapid Commun. 8 tems for the delivery of poorly soluble drugs, Biomaterials 30 (11) (2009)
(10) (1987) 481–485. 2102–2111.
[68] S.K. Li, A. D’Emanuele, On–off transport through a thermoresponsive hydrogel [98] G. Orive, A.R. Gascon, R.M. Hernandez, M. Igartua, J. Luis Pedraz, Cell
composite membrane, J. Control. Rel. 75 (1–2) (2001) 55–67. microencapsulation technology for biomedical purposes: novel insights and
[69] R. Gottlieb, T. Schmidt, K.-F. Arndt, Synthesis of temperature-sensitive hydro- challenges, Trends. Pharmacol. Sci. 24 (5) (2003) 207–210.
gel blends by high-energy irradiation, Nucl. Instrum. Methods Phys. Res., Sect. [99] G. Orive, R.M. Hernandez, A.R. Gascon, R. Calafiore, T.M.S. Chang, P.D. Vos, G.
B 236 (1–4) (2005) 371–376. Hortelano, D. Hunkeler, I. Lacik, A.M.J. Shapiro, J.L. Pedraz, Cell encapsulation:
[70] H. Katono, A. Maruyama, K. Sanui, N. Ogata, T. Okano, Y. Sakurai, promise and progress, Nat. Med. 9 (1) (2003) 104–107.
Thermo-responsive swelling and drug release switching of interpenetrating [100] G. Orive, M. De Castro, H.J. Kong, R.M. Hernandez, S. Ponce, D.J. Mooney, J.L.
polymer networks composed of poly(acrylamide-co-butyl methacrylate) and Pedraz, Bioactive cell-hydrogel microcapsules for cell-based drug delivery, J.
poly(acrylic acid), J. Control. Rel. 16 (1–2) (1991) 215–227. Control. Rel. 135 (3) (2009) 203–210.
[71] Y.H. Bae, T. Okano, S.W. Kim, Temperature dependence of swelling of [101] Y. Hu, X. Zhou, Y. Lu, C. Hu, X. Hu, Novel biodegradable hydrogels based on
crosslinked poly(N,N -alkyl substituted acrylamides) in water, J. Polym. Sci., pachyman and its derivatives for drug delivery, Int. J. Pharm. 371 (1–2) (2009)
Part B: Polym. Phys. 28 (6) (1990) 923–936. 89–98.
[72] L.D. Taylor, L.D. Cerankowski, Preparation of films exhibiting a balanced tem- [102] Y. Ishii, T. Nakae, F. Sakamoto, K. Matsuo, K. Matsuo, Y.-S. Quan, F. Kamiyama,
perature dependence to permeation by aqueous solutions—a study of lower T. Fujita, A. Yamamoto, S. Nakagawa, N. Okada, A transcutaneous vaccination
consolute behavior, J. Polym. Sci.: Polym. Chem. 13 (11) (1975) 2551–2570. system using a hydrogel patch for viral and bacterial infection, J. Control. Rel.
[73] H. van der Linden, W. Olthuis, P. Bergveld, An efficient method for the fabri- 131 (2) (2008) 113–120.
cation of temperature-sensitive hydrogel microactuators, Lab Chip 4 (2004) [103] H. Suzuki, T. Tokuda, K. Kobayashi, A disposable “intelligent mosquito” with
619–624. a reversible sampling mechanism using the volume-phase transition of a gel,
[74] D. Gan, L.A. Lyon, Fluorescence nonradiative energy transfer analysis of Sens. Actuators B 83 (1–3) (2002) 53–59.
crosslinker heterogeneity in core–shell hydrogel nanoparticles, Anal. Chim. [104] M. Suzuki, An artificial muscle by PVA hydrogel can generate high power
Acta 496 (1–2) (2003) 53–63. close to living skeletal muscles, in: Proceedings of the Annual International
[75] F. Yan, S. Asher, Cation identity dependence of crown ether photonic crystal Conference of the IEEE Engineering in Medicine and Biology Society, 1989.
Pb2+ sensing, Anal. Bioanal. Chem. 387 (6) (2007) 2121–2130. Images of the Twenty-First Century, 1989.
[76] J.H. Holtz, S.A. Asher, Polymerized colloidal crystal hydrogel films as intelli- [105] M. Bassil, J. Davenas, M. El Tahchi, Electrochemical properties and actua-
gent chemical sensing materials, Nature 389 (6653) (1997) 829–832. tion mechanisms of polyacrylamide hydrogel for artificial muscle application,
[77] M.J. Lesho, N.F. Sheppard, Adhesion of polymer films to oxidized silicon and Sens. Actuators B 134 (2) (2008) 496–501.
its effect on performance of a conductometric pH sensor, Sens. Actuators B [106] J. Silver, J.H. Miller, Regeneration beyond the glial scar, Nat. Rev. Neurosci. 5
37 (1–2) (1996) 61–66. (2) (2004) 146–156.
774 K. Deligkaris et al. / Sensors and Actuators B 147 (2010) 765–774

[107] P. Caroni, T. Savio, M.E. Schwab, Central nervous system regeneration: oligo- Wouter Olthuis was born in Apeldoorn, the Netherlands, on October 23, 1960. He
dendrocytes and myelin as nonpermissive substrates for neurite growth, Prog. received the MSc degree in electrical engineering from the University of Twente,
Brain Res. 78 (1988) 363–370. Enschede, The Netherlands in 1986 on the subject of thermally excited resonating
[108] S.A. Busch, J. Silver, The role of extracellular matrix in CNS regeneration, Curr. silicon membrane pressure sensors. In that year, he joined the Center for Micro-
Opin. Neurobiol. 17 (1) (2007) 120–127. Electronics, Enschede (CME) doing research on inorganic electret materials for
[109] J. Zuo, D. Neubauer, K. Dyess, T.A. Ferguson, D. Muir, Degradation of chon- subminiature silicon microphones. In 1987 he started his Ph.D.-project and received
droitin sulfate proteoglycan enhances the neurite-promoting potential of the Ph.D. degree from the Biomedical Engineering Division of the Faculty of Electri-
spinal cord tissue, Exp. Neurol. 154 (2) (1998) 654–662. cal Engineering, University of Twente, in 1990. The subject of his dissertation was
[110] A. Hejcl, P. Lesný, M. Prádný, J. Michálek, P. Jendelová, J. Stulík, E. Syková, the use of Iridium oxide in ISFET-based coulometric sensor–actuator devices. Since
Biocompatible hydrogels in spinal cord injury repair, Physiol. Res. 57 (2008) 1991 he has been working as an Assistant Professor in the Laboratory of Biosensors,
121–132. part of the MESA+ Research Institute, of the University of Twente and as such co-
[111] E. Syková, P. Jendelová, L. Urdzíková, P. Lesný, A. Hejčl, Bone marrow stem cells supervising many projects on both physical and (bio)chemical sensors and sensor
and polymer hydrogels—two strategies for spinal cord injury repair, Cell. Mol. systems for medical and environmental applications. Currently, he is Associate Pro-
Neurobiol. 26 (7) (2006) 1111–1127. fessor in the BIOS Lab-on-Chip group of the MESA+ Institute of Nanotechnology and
[112] D.R. Nisbet, K.E. Crompton, M.K. Horne, D.I. Finkelstein, J.S. Forsythe, Neural is as such responsible for the theme Electrochemical sensors and Sensor systems.
tissue engineering of the CNS using hydrogels, J. Biomed. Mater. Res. Part B Since 2006 he is also the Director of the Educational Programme of Electrical Engi-
87B (1) (2008) 251–263. neering at the Faculty of Electrical Engineering, Mathematics and Computer Science
[113] J.A. Burdick, M. Ward, E. Liang, M.J. Young, R. Langer, Stimulation of neurite at the University of Twente.
outgrowth by neurotrophins delivered from degradable hydrogels, Biomate-
rials 27 (3) (2006) 452–459. Albert van den Berg received his Ph.D. at the University of Twente in 1988 on the
[114] S.R. Van Tomme, G. Storm, W.E. Hennink, In situ gelling hydrogels for phar- topic of chemically modified ISFETs. From 1988 to 1993 he worked in Neuchatel,
maceutical and biomedical applications, Int. J. Pharm. 355 (1–2) (2008) 1–18. Switzerland, at the CSEM and the University (IMT) on miniaturized chemical sen-
[115] A. Jain, Y.-T. Kim, R.J. McKeon, R.V. Bellamkonda, In situ gelling hydrogels for sors. From 1993 until 1999 he was research director micro-total analysis systems
conformal repair of spinal cord defects, and local delivery of BDNF after spinal (␮TAS) at MESA, University of Twente, a topic that was extended to Miniaturized
cord injury, Biomaterials 27 (3) (2006) 497–504. Chemical Systems (MiCS) in 1999. In 1998 he was appointed as part-time professor
“Biochemical Analysis Systems”, and later in 2000 as full professor on Miniaturized
Systems for (Bio)Chemical Analysis in the faculty of Electrical Engineering, embed-
Biographies ded the MESA+ Institute for Nanotechnology. In 2002 he received the Simon Stevin
Master award from the Dutch Technical Science foundation (STW). In 2003 he was
appointed as captain of the Nanofluidics Flagship within the national nanotech-
Kosmas Deligkaris was born in Thessaloniki, Greece, on March 25, 1983. He received nology program Nanoned. In 2005 he stayed for 6 months San Diego (USA) at the
his BSc. In Electronic Engineering from the Alexander Technological Educational La Jolla Institute for Allergy and Immunology (LIAI, group Green) during a sabbat-
Institute of Thessaloniki in 2008. He is currently pursuing his MSc Degree at the Uni- ical leave, while he received an Advanced Research Grant from ERC in 2008. In
versity of Twente, Enschede, The Netherlands, working in the Biomedical Systems 2009 he received the Spinoza prize, the most prestigious Dutch scientific award,
and Signals group. His research interests include neurotechnology, microsystems for his achievements in lab-on-a-chip research. His current research interests focus
and optimization algorithms. on microanalysis systems and nanosensors, nanofluidics and single cells on chips,
with applications in health care and environment. Albert van den Berg is member
Tadele Shiferaw Tadele completed his BSc in Electrical Engineering from Mekelle of the Royal Dutch Academy of Sciences (KNAW), the Dutch Health council, board
University, Ethiopia in 2007. Currently, he is pursuing his MSc degree in Elec- member of the Chemical and Biological Microsystems Society, member of the Dutch
trical Engineering in the specialization of Measurement and Control systems chemical society (KNCV) and deputy chair of the journal Lab on a Chip. He has co-
at the University of Twente, Enschede, The Netherlands. His research interests authored over 180 papers (H = 33) and over 10 patents, and has been involved in >5
include biomechatronics, structured agent-based control systems, intelligent con- spin-off companies.
trol, robotics, and embedded systems.

Potrebbero piacerti anche