Download as pdf or txt
Download as pdf or txt
You are on page 1of 49

Materials Science and Engineering R 93 (2015) 1–49

Contents lists available at ScienceDirect

Materials Science and Engineering R


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

Stimulus-responsive hydrogels: Theory, modern advances,


and applications
Michael C. Koetting a,d,1, Jonathan T. Peters a,d,2, Stephanie D. Steichen b,d,3,
Nicholas A. Peppas a,b,c,d,*
a
McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
b
Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
c
College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
d
Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, United States

A R T I C L E I N F O A B S T R A C T

Article history: Over the past century, hydrogels have emerged as effective materials for an immense variety of
Available online 16 May 2015 applications. The unique network structure of hydrogels enables very high levels of hydrophilicity and
biocompatibility, while at the same time exhibiting the soft physical properties associated with living
Keywords: tissue, making them ideal biomaterials. Stimulus-responsive hydrogels have been especially impactful,
Hydrogels allowing for unprecedented levels of control over material properties in response to external cues. This
Polymers enhanced control has enabled groundbreaking advances in healthcare, allowing for more effective
Stimulus-responsive treatment of a vast array of diseases and improved approaches for tissue engineering and wound healing.
Smart materials In this extensive review, we identify and discuss the multitude of response modalities that have been
Swelling
developed, including temperature, pH, chemical, light, electro, and shear-sensitive hydrogels. We
Drug delivery
discuss the theoretical analysis of hydrogel properties and the mechanisms used to create these
pH responsive
Temperature responsive
responses, highlighting both the pioneering and most recent work in all of these fields. Finally, we review
Chemically-responsive the many current and proposed applications of these hydrogels in medicine and industry.
Molecularly imprinted polymers ! 2015 Elsevier B.V. All rights reserved.
Photo-responsive
Electrically-responsive
Shear stress
Scaffolds
Tissue engineering
Biosensors

Contents

1. General hydrogel theory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3


1.1. Physical structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
1.2. Equilibrium swelling theory and network characteristics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
1.3. Dual responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
1.3.1. Homogeneous behavior assumption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
1.3.2. IPN Interactions during the swelling process . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
1.3.3. Independent network assumption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
1.3.4. Model development. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5

* Corresponding author at: Department of Biomedical Engineering, The University of Texas at Austin, 107W Dean Keeton Street, Stop C0800, Austin, TX 78712,
United States. Tel.: +1 512 471 6644; fax: +1 512 471 8227.
E-mail addresses: mkoetting@utexas.edu (M.C. Koetting), jonathan.peters@utexas.edu (J.T. Peters), sdsteichen@utexas.edu (S.D. Steichen),
peppas@che.utexas.edu (N.A. Peppas).
1
Tel.: +1 512 471 6910.
2
Tel.: +1 512 471 4757.
3
Tel.: +1 512 472 5681.

http://dx.doi.org/10.1016/j.mser.2015.04.001
0927-796X/! 2015 Elsevier B.V. All rights reserved.
2 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

1.4. Mechanical behavior. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6


2. Solute transport in hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3. pH-responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.1. Polymer composition and swelling behavior. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2. Swelling behavior and theoretical considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.3. pH responsive hydrogels for controlled drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3.1. Anionic hydrogel networks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3.2. Cationic hydrogel networks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
3.4. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4. Temperature responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.1. Swelling theory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.1.1. LCST . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.1.2. UCST . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.2. Common monomers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.2.1. LCST monomers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
4.2.2. UCST monomers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.3. Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.3.1. Oral drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.3.2. Particle based drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.3.3. Tissue culture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
4.3.4. Ocular drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
4.3.5. Membranes, microfluidics, and sensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
5. Chemically-responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
5.1. Glucose-responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
5.1.1. Glucose-oxidase gels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
5.1.2. Concanavalin A gels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
5.1.3. Phenylboronic acid gels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
5.2. Enzyme-responsive . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
5.3. Molecularly imprinted polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
5.3.1. Synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
5.3.2. Diagnostics/sensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
5.3.3. Drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5.3.4. Sorbents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
6. Photo-responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
6.1. Theory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
6.2. Common monomers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
6.2.1. Isomerization monomers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
6.2.2. Degradation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
6.2.3. Dimerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
6.3. Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
6.3.1. Tissue culture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
6.3.2. Microfluidics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
6.3.3. Drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
7. Electrically-responsive hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
7.1. Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
7.1.1. Artificial muscles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
7.1.2. Drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
8. Shear stress responsive hydrogels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
8.1. Theoretical considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
8.2. Shear-thinning hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
8.2.1. Injectable hydrogel systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
8.2.2. Other shear-thinning applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
8.3. Shear-thickening hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
8.4. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
9. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44

Hydrogels are three dimensional network structures consisting While Paul Flory set the basic theories for hydrogel analysis, it is
of polymeric chains joined by tie points or joints and swollen in interesting to note that hydrogels had been prepared long before
water up to thermodynamic equilibrium. This is a simple and quite his original theoretical treatments were established. Indeed, early
accurate definition of these materials, which have become so work on crosslinked polymers and networks first appeared in
popular in numerous applications over the past 50 years. While German literature in the mid-1930s. Meanwhile significant work
this general definition has been used to present and analyze on the behavior of ‘‘natural hydrocolloids’’ appeared in the late
swollen crosslinked hydrogels, especially as defined in our 1986– 1930s, but without structural insight. In addition, work in that
1987 books on ‘‘Hydrogels in Medicine and Pharmacy,’’ there have period and in the 1940s concentrated mostly on reaction kinetics
been variations of this basic definition and deviations from the and mechanical properties of the ensuing networks.
basic thermodynamic and structural equations that define their PJ Flory (1944–1952; Nobel prize 1974) set the main framework
performance. of analysis of gels with his thermodynamic theories, statistical
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 3

mechanical analysis, and the first analysis of critical miscibility Depending on the monomers incorporated into the hydrogel
characteristics of hydrogels. At the same time, pioneering work was and the conditions of the surrounding medium, hydrogels can
also done in the Soviet Union (Kargin, 1945–1960, Ushakov, 1943– exhibit a full range of charge properties, being neutral, cationic,
1953, Korshak 1952–1973), and in Japan (Sakurada, 1948–1965). anionic, or ampholytic. Much like other macromolecules, such as
Hydrogels possess numerous properties that make them ideal proteins or nucleic acids, hydrogels’ network charge depends
candidates for use as biomaterials, finding significant use in the strongly on pH of the surrounding medium, as this determines the
fields of drug delivery, tissue engineering, implants, and more. The protonation status of the incorporated pendant groups. Most ionic
main feature of hydrogels is their ‘‘soft’’ material nature, owing to gels exhibit some degree of charge in aqueous solvents. Carboxylic
their highly hydrophilic nature that encourages uptake of water, acid pendant groups are commonly used to impart anionic
leading to hydrated yet solid materials, much like cells in the body. properties to a hydrogel, while amine pendant groups are
Their hydrophilic and crosslinked nature also imparts excellent commonly used for imparting cationic properties.
biocompatibility, and many common hydrogels have found wide The physical structure of hydrogels can be characterized by
use both in laboratory studies and clinical uses. Some of the most standard mechanical testing methods. In addition to mechanical
commonly used hydrogels include synthetics like poly(ethylene properties such as the Young’s modulus (E), storage modulus (G0 ),
glycol) (PEG), poly(vinyl alcohol) (PVA), or poly(2-hydroxyethyl and loss modulus (G00 ) [3], the hydrogel structure can be
methacrylate) (PHEMA), as well as naturally occurring hydrogels quantitatively modeled using several parameters: v2,s is the polymer
like agarose, alginate, chitosan, collagen, fibrin, and hyaluronan. volume fraction in the swollen state, which describes the hydrogel’s
Hydrogels become especially useful when they are used as hydration level; M C is the average molecular weight between
‘‘smart’’ materials that can respond to changes in their environ- crosslinks, which describes the overall density of crosslinks in the
ment. In this extensive review, we discuss multiple responsive hydrogel; and j is the mesh size of the hydrogel, which reflects the
modalities—including responses to pH, temperature, chemicals, porosity of the gel (and is dependent on the hydration level and
light, electric fields, and shear stress—and discuss recent advances the crosslinking density). These parameters can be used to model
both in tailoring these responses and applying them. As the diverse the behavior of both nonionic [4] and ionic [5] hydrogels through
work reviewed herein attests, responsive hydrogels are currently equilibrium swelling theory and rubber elasticity theory [6].
transforming our world by improving healthcare and finding novel
benefits in industrial applications. 1.2. Equilibrium swelling theory and network characteristics

1. General hydrogel theory Flory–Rehner theory may be utilized for quantitative analysis of
nonionic hydrogels [7]. Flory–Rehner theory posits that hydrogel
1.1. Physical structure equilibrium is attained through a balance of enthalpic mixing, which
promotes swelling, and the elastic forces imposed by the crosslinked
Hydrogels are characterized by hydrophilic polymers that are hydrogel chains, which promotes contraction. Using Gibbs free
crosslinked into an insoluble, but highly hydrophilic structure. As energy, the theory’s basis may be stated as presented in Eq. (1).
suggested by such a basic definition, hydrogels are a broad class of
materials that can be prepared in many different ways and can
DGtotal ¼ DGelastic þ DGmixing (1)
exhibit significantly different behaviors. For instance, the cross- Direct application of Flory–Rehner theory to ionic hydrogels is
links that form the hydrogel network may take on many forms, incorrect, as additional forces result from ionic interactions within
such as covalent chemical links, ionic bonds, weak physical the hydrogel and with the surrounding medium that shift the
entanglements, hydrogen bonds, or other dipolar interactions hydrogel’s swelling equilibrium. This additional contribution is
[1]. Chemical crosslinking can be achieved through many methods. incorporated in an additional term, DGionic, as shown in Eq. (2).
For example, use of dimethacrylates as monomers in a chain
polymerization leads to crosslinking as both methacrylate DGtotal ¼ DGelastic þ DGmixing þ DGionic (2)
functionalities are incorporated into the polymer backbone at Beyond simply resulting in additional energy terms, ionic
two different locations, leading to crosslinking. Glutaraldehyde, on properties in the hydrogel also complicate analysis through the
the other hand, has been widely used as a post-polymerization mixing term, DGmixing. The mixing term describes the thermody-
reagent to promote crosslinking after the polymer chains have namic interactions occurring between the polymer and the
already been formed. Regardless of the strategy used, however, solvent, and is typically expressed through use of an empirical
hydrogels take many forms, including copolymers, blends, or polymer–solvent interaction parameter, x1. However, this param-
interpenetrating networks (IPNs). IPNs are often prepared by eter is explicitly defined for nonionic systems, and use with ionic
polymerization and crosslinking of one complete polymer systems yields approximate results, with potential for error of 30–
network, followed by polymerization and crosslinking of a second 40% from the correct values.
in the presence of the first network. If the two polymerizations Application of Eq. (1) to nonionic hydrogel systems prepared in
occur by significantly disparate methods, the network formation the absence of solvent yields an expression for determining the
may be performed simultaneously. average molecular weight between crosslinks, MC , as shown in
In addition to the variety of crosslinking methods used, hydrogels Eq. (3). Theoretical treatment of hydrogels prepared in solvent
may adopt significantly different final morphologies. Hydrogels may required additional modification by Peppas and Merrill that
be amorphous, semiscrystalline, supramolecular, or colloidally considered modifications to the elastic potential caused by
aggregated [1]. The variance in morphology reflects the wide range interaction with the solvent, leading to the expression shown in
of monomers that can comprise a hydrogel: monomers may be Eq. (4) [4].
natural or synthetic; copolymers of both natural and synthetic
2
components may be used; and the final characteristics are highly 1 2 ðȳ=V 1 Þ½lnð1 # y2;s Þ þ y2;s þ x1 y2;s '
¼ # (3)
dependent on the ratios of monomers incorporated into the network MC M¯N ½y
1=3
# ðy2;s =2Þ'
2;s
and their spatial ordering. As such, the structure of hydrogels
becomes strongly dependent on the synthesis procedures followed,
1 2 ðȳ=V 1 Þ½lnð1 # y2;s Þ þ y2;s þ x1 y22;s '
solvent used, monomers and their ratios, and even the degradative ¼ # (4)
and mechanical history of the polymer [2]. MC M¯N y2;r ½ðy2;s =y2;r Þ1=3 # ðy2;s =2y2;r Þ'
4 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

In these expressions, M¯N represents the average molecular weight swelling behavior of a hydrogel. Once experimentally deter-
of polymer chains as formed without crosslinks, V1 is the molar mined, quantification of the mesh size may proceed through
volume of the solvent, y is the specific volume of the polymer in the application of the Peppas–Merrill or Brannon-Peppas expression,
amorphous state, y2,r and y2,s represent the polymer volume as appropriate, for determining M C , which is then applied in
fractions in the ‘‘relaxed’’ and ‘‘swollen’’ states, respectively, and x1 Eq. (8).
is the Flory polymer–solvent interaction parameter. The ‘‘swollen’’
state refers to the equilibrium state of the polymer after exposure 1.3. Dual responsive hydrogels
to the solvent. The ‘‘relaxed’’ state is the state of the polymer
immediately following polymerization but before any swelling has Interpenetrating polymer networks (IPNs) are composed of
occurred. In the event that the polymer is formed in the absence of two or more polymers in a network and are formed when one
solvent, no solvent is incorporated in the newly formed network, polymer network is crosslinked in the presence of another
causing y2,r to a value of approximately 1, thus leading to previously crosslinked polymer network [10]. The benefit of IPNs,
equivalence between Eqs. (3) and (4). as compared to standalone polymer networks, is their ability to
It is also important to note that the term y2,s/2 appearing in the respond to a multitude of stimuli because each polymer network
preceding equations effectively incorporates the functionality of within the IPN can have a unique environmental responsiveness
the crosslinks—the number of bonds monomers may make. A more [11]. However, the modeling of the swelling behavior of these IPNs
rigorous treatment would replace y2,s/2 with 2y2,s/f, where w is can be quite complex because of the two or more independent
the functionality of the crosslinks, assumed to be 4 for most environmentally-sensitive networks involved. Zhang [12] has
crosslinked networks. Thus, the forms of Eqs. (3) and (4) are only proposed a model based on three primary assumptions: homoge-
explicitly valid for tetrafunctional crosslinks. Therefore, in the neous behavior, IPN interaction, and independent network
case of mixed multifunctional crosslinks, this analysis is again behavior.
approximate.
Hydrogel porosity is quantified through the mesh size 1.3.1. Homogeneous behavior assumption
parameter, j. Porosity is a very important characteristic of the In an IPN, the different polymer compositions and ratios can
hydrogel’s structure, as it determines the ability of solute to result in phase separation between the two polymers. Even
diffuse through the hydrogel matrix at various conditions—a though phase separation can be avoided by carefully tailoring the
phenomenon that is widely exploited in the many hydrogels polymer composition, it is still an important consideration as any
discussed in this review. The mesh size is a measure of the average observed phase separating behavior can impact the swelling of
linear distance between crosslinks, not a diameter or diagonal the hydrogel networks. When swelling, some minor phase
distance. The mesh size may be determined through either of two separation occurs in the skin layer of the swollen network;
possible methods. One method is direct application of Eqs. (5) and however, none is observed in the bulk. For this reason, during
1=2
(6), in which the term ðr o 2 Þ corresponds to the root-mean- theoretical treatment of IPN swelling, it is assumed that the IPNs
square end-to-end distance of the polymer chains between are homogeneous single-phase hydrogels. This assumption
crosslinks, also termed the unperturbed distance. The term a is allows for the determination of the polymer–solvent interaction
known as the extension ratio, which is determined from n2,s via parameter.
Eq. (6).
1.3.2. IPN Interactions during the swelling process
1=2
j ¼ aðr¯o 2 Þ (5) The Gibbs total free energy change during the swelling process
of a single polymer network under constant temperature and
#1=3
a ¼ y2;s (6) pressure is denoted as follows:

Because of the esoteric nature of the unperturbed distance, it is DGtot ¼ DGmix þ DGel (9)
frequently calculated rather than known directly. The unperturbed
where DGmix is the free energy contributions due to mixing, and
distance may be calculated by Eq. (7),
DGel is the free energy contribution due to the elastic retractile
! "1=2 forces in the network.
1=2 2C n M̄ c
ðr¯o 2 Þ ¼l (7) Within an IPN, free energy change due to the swelling of both
Mr
polymer networks must be considered. The total Gibbs free energy,
in which l is the bond length along the polymer backbone (often therefore, has contributions from each polymer system within the
1.54 Å for vinyl polymers), Cn is the Flory characteristic ratio— IPN, DG2 and DG3. Each polymer network contribution can be
defined as the ratio of the mean square unperturbed distance to the broken down further into both a mixing, DGmix, and elastic, DGel,
expected end-to-end distance for a freely jointed chain of the same contribution. Additional free energy interaction parameters
length—which is tabulated for many polymers, M C is again the include contributions from the solvent or swelling agent, DG1,
average molecular weight between crosslinks (calculable using the and from the ionic interactions of ionizable polymeric networks,
Peppas–Merrill or Brannon-Peppas equation, as appropriate), and DGion. Interestingly, because of the constraints of constant
Mr is the molecular weight of the repeat units. Combining this with temperature when calculating Gibbs free energy, temperature-
Eq. (5) leads to a more frequently useful calculation for the mesh responsive networks do not require the inclusion of a separate
size, as shown in Eq. (8) [8,9]. interaction term. With all of these parameters, the total Gibbs free
! "1=2 energy of an IPN can be written as
#1=3 2C n M̄ C
j ¼ y2;s l (8) DGtot ¼ DGmix;2 þ DGmix;3 þ DGel;2 þ DGel;3 þ DGion (10)
Mr

In order to accurately characterize the porosity of a hydrogel


for use in biomedical applications through the mesh size, n2,s 1.3.3. Independent network assumption
should be determined experimentally in the end-use solvent, as Assuming the networks are completely independent of one
many parameters such as ionic strength of the solution, pH, and another, the Flory–Huggins equation for DGmix and the rubber
interactions with other molecules may significantly affect the elasticity equation for DGel are still acceptable to use for each
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 5

network. Thus, the derivative of Eq. (10) with respect to the and ve,2 is the effective number of chains within the network
number of moles of swelling agent becomes structure. Upon taking the derivative with respect to number of
! " ! " moles, we obtain Eq. (18):
@DGmix;2 @DGmix;3
m1 # mo1 ¼ N þN ! " ! " ! "
@n1 T;P @n1 @DGel;2 @a2 ve;2 # 1=3 v2 $
! " ! " N ¼ RT v¯1 v2 # (18)
@DGel;2 @a2 @a2 T;P @n1 T;P V0 2
þN
@a2 T;P @n1 T;P The third term in Eq. (15) represents the contribution to the
! " ! "
@DGel;3 @a3 mixing equation of the second polymeric network in the IPN. It is
þN þ ðDm(1 Þion # ðDm1 Þion (11)
@a3 T;P @n1 T;P the polymer that is polymerized in the presence of the first
polymerized polymer. Therefore, it cannot be derived in the same
There are additional parameters in this equation: N is way as Eq. (18). The assumption that the networks are elastically
Avogadro’s number; n1 is the total number of moles of solvent; independent from one another, however, allows for the treatment
and a2 and a3 are linear chain deformation factors. ðDm(1 Þion # of the primary network as a micromolecular diluent that is present
ðDm1 Þion represents the difference in chemical potential inside and during crosslinking of the secondary network. The change of
outside the gel, respectively, due to ionic contributions. configurational entropy during swelling can be expressed by
Eq. (19):
1.3.4. Model development ! "! " #!
The final form of the mixing term for single component polymer 1 a33 3 a3 2
DSel;3 ¼ kve ln o3 # # 1 (19)
networks can be determined from the Flory–Huggins theory: 2 a3 2 ao3
! "
@DGmix 0 The superscript 0, here, refers to the unswollen IPN, and the
Dmmix ¼ N ¼ RTðlnð1 # vÞ þ v þ x1 v2 Þ (11 )
@n1 T;P corresponding linear deformation factor, ao3 , is represented as
x1 denotes the polymer–solvent interaction parameter, and v is the ! "1=3 ! "1=3
1 Vo
polymer volume fraction. In an IPN, each polymer has its own ao3 * ¼ o (20)
vo3 V # Vo
volume fraction, denoted v2 and v3, and the solvent/swelling agent
has a corresponding volume fraction, v1. All three fractions must And the swollen IPN linear deformation factor, a3, is defined as
therefore be expressed as ! "1=3 ! "1=3 ! o "
1 V V þ ðn1 v¯1 =NÞ
v1 þ v2 þ v3 ¼ 1 (12) a3 * ¼ o ¼ o (21)
v3 V # Vo V # Vo
The individual polymer–solvent interaction parameter, x, is Thermodynamically, it is known that
difficult to obtain for each individual polymer in an IPN.
Therefore, the total mixing term can be approximated by DGel ¼ T DSel;3 (22)
considering the two networks as one homogeneous network
Knowing this relationship and expressing ve,3 as moles/cm3, the
and including an average x factor, x̄. Thus, the total mixing term
following expression was derived:
can be written as follows:
! " ! " ! " ! "
! " ! " ! " @DGel;3 @a3 @T DSel;3 @a3
@DGmix @DGmix;2 @DGmix;3 N ¼ #N
N )N þN (13) @a3 T;P @n1 T;P @a3 T;P @n1 T;P
@n1 T;P @n1 T;P @n1 T;P
RT ve;3 v1 # v3 $
or, ¼ o vo2:3
3 v1=3
3 # (23)
ðV # V o Þ 2
! "
@DGmix An ionic interaction term has been expressed in terms of various
N ¼ RT½ln½1 # ðv2 þ v3 Þ' þ ðv2 þ v3 Þ
@n1 T;P ionic parameters:
þ x̄ðv2 þ v3 Þ2 ' ! "2
v¯1 RT #v2 þ v3 $2 Ka
2 ðDm(1 Þion # ðDm1 Þion ¼ (24)
¼ RT½lnv1 þ ð1 # v1 Þ þ x̄ð1 # v1 Þ ' (14) 4IM r v̄ 10#pH þ K a
Eq. (11) can be rewritten as follows by using Flory’s rubber This expression, developed by Brannon-Peppas and Peppas,
elasticity theory: will be discussed in more detail later in this review. Briefly, Mr is
! " ! " ! " the molecular weight of the repeat units of the polymer, I is
@Gmix @DGel;2 @a2
m1 # mo1 ¼ N þN the ionic strength of the solvent/swelling agent, V1 is the
@n1 T;P @a2 T;P @n1 T;P molar volume of the solvent/swelling agent, v̄ is the specific
! " ! "
@DGel;3 @a3 volume of the polymer, and the equilibrium constant of the
þN þ ðDm(1 Þion # ðDm1 Þion (15)
@a3 T;P @n1 T;P ionizable network is Ka for anionic systems and Kb for cationic
systems.
The free energy contributions from the elastic retractile forces Substituting Eqs. (17), (23) and (24) into Eq. (10) yields
of an isotropic network according to rubber elasticity theory are
given by Eq. (16).
%
! "
kT ve;2 m1 # mo1 ¼ RT ln½1 # ðv2 þ v3 Þ' þ ðv2 þ v3 Þ þ x̄ðv2 þ v3 Þ2
DGel;2 ¼ ð3a22 # 3 # lna32 Þ (16)
2 ! "! " ! "! "&
v̄1 ve;3 v2 v̄1 ve;3 v3
where,
þ v̄21=3 # þ o vo2:3
3 v1=3
3 #
V0 2 V # V0 2
! "2 ! "2
V V 0 þ ðn1 v¯1 =NÞ þ ðn3 v¯3 =NÞ v̄1 RT
v2 þ v3 Ka
a32 ¼ ¼ (17) þ (25)
V0 V0 4IMr v̄ 10# pH þ K a
In this expression, V0 is the initial unswollen volume of the Eq. (25) describes the chemical potential difference between pure
primary network, V is the total volume of the IPN when swollen, solvent and solvent in the swollen IPN. This is expressed in terms of
6 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

the volume fractions of the primary, v2, and secondary, v3, polymer Equilibrium swelling and elasticity theory have been modified for
networks. The term ve can be written as non-covalently crosslinked systems by treating junctions or entan-
! " glements as covalent crosslinks. Normally, the same equations apply,
V 2Mc
ve ¼ 1# (26) but M C is replaced by either molecular weight between junctions
v̄Mc M
ðM̄ j Þ or entanglements ðM̄ e Þ to indicate the gel is not covalently
Substituting Eq. (26) into Eq. (25) yields an expression for crosslinked. Applying these theories to semicrystalline networks is
overall chemical potential difference describing the swelling more difficult, and is only applicable as an approximation. This is
behavior of an IPN: because crystals are a larger size range; on the order of 10 nm, and
violates the Gaussian distribution that is assumed in original Flory–
%
Rehner, Peppas–Merrill, and Brannon-Peppas equations.
m1 # mo1 ¼ RT ln½1 # ðv2 þ v3 Þ' þ ðv2 þ v3 Þ þ x̄ðv2 þ v3 Þ2
! "! "! " 2. Solute transport in hydrogels
v̄1 2M c;2 v2
þ 1# v1=3
2
v̄2 Mc;2 M n;2 2
! "& Solute transport in hydrogels is crucial to understand when
v̄1 1=3 v 3
þ vo2:3 v # utilizing them as drug carriers, particularly as many therapeutics
v2 Mc;3 3 3 2
! " ! "2 have a narrow therapeutic range. In hydrogels, solute transport is
v̄1 RT v2 þ v3 2 Ka governed by Fick’s law, shown in vector form below [18]. In Eq. (29),
þ (27)
4IMr v̄ 10 # pH
þ Ka ci is the concentration of species i and Dig is the diffusion coefficient,
which is often a function of ci. Hydrogels have significant impact on
In Eq. (27), the parameters Mc,2 and Mc,3 are the molecular
the diffusion of loaded therapeutics, even though they have high
weight between crosslinks in the primary and secondary networks.
water content. Ende et al. examined the effect of various factors,
When solved, the expression gives insight into the swelling
including mesh size, pH, and temperature, and discovered that they
behavior of the IPN hydrogel network. Eq. (27) reduces to the
all significantly alter solute transport [19]. They concluded that
equation for a single ionizable hydrogel network when there is no
hydrogels could be tailored for the delivery of a specific solute.
secondary network present (i.e., v3 = 0).
Brannon-Peppas and Peppas took this a step further by developing
pH responsive hydrogels that exhibit near zero-order release [20].
1.4. Mechanical behavior
@ci
¼ r + ðDig ðci Þrci Þ (29)
The mechanical behavior of hydrogels is crucial in understand- @t
ing the effect they will have in biomedical applications. Hydrogels
Renkin’s early work into solute diffusion utilized one dimensional
have been known to exhibit a variety of physical behaviors, from
Fickian diffusion, Eq. (30), to examine the effects of solute diffusion
elastic recovery to the time-dependent recovery associated with
[21].
viscous behavior. These properties are largely dependent on the
crosslinking nature and density, and can be manipulated for @N i @c
different applications. For example, tissue culture substrates often ¼ #DA i (30)
@t @x
require stiffer hydrogels; whereas intravenously injected particles
that exhibit lower stiffness demonstrate better bioavailability and Here @Ni/@t is the diffusion rate for species i, D is the diffusion
improved circulation [13,14]. coefficient, A is the apparent area, and @ci/@x is the concentration
The viscoelasticity that is commonly associated with hydrogels gradient across the membrane. In Renkin’s experiments, the rate of
used for biomedical applications comes from being used above the diffusion of solutes through inert hydrogels was measured. His
glass transition temperature (Tg). This viscoelastic behavior comes results were in close agreement with theory proposed by
from the restricted rearrangement of polymer segments due to Pappenheimer [22]. This work demonstrated the impact of pore
deformation. The Tg of hydrogels is often significantly depressed size on the solute transport thorough porous media.
due to the plasticization caused by the presence of the solvent, The interactions between solute and polymer play a large role in
meaning the Tg of the hydrogels used in biomedical applications is biological systems. Charge and hydrophobic interactions can lead
well below physiological temperature when hydrated. In this to partitioning of solutes in hydrogel systems. Gudeman and
regime, stress-relaxation, creep, and dynamic loading are impor- Peppas studied these effects in ionic systems of IPNs of poly(vinyl
tant to consider throughout the lifetime of the polymer [15]. alcohol) and poly(acrylic acid). They varied the pH and ionic
Peppas and Merrill [16] modified Flory’s original theories on content of the hydrogel and tested the transport above and below
polymer elasticity [17], to account for the presence of solvent in the pKa of acrylic acid, observing a decrease in the rate of solute
hydrogels. This work is summarized by Eq. (2). Here t is the applied transport [23]. They further confirmed that permeation is driven
stress as a function of elongation, r is the polymer density, R is the by size exclusion and restrictions on the Brownian motion of
universal gas constant, and T is absolute temperature. This allows solutes through porous substrates [24]. They developed a one-
for determination of the molecular weight between crosslinks, M C , dimensional diffusion-convective theory to define the systems,
by measuring the applied stress, t. As with mesh size calculations, and even developed equations to account for the impact of the pore
the swollen polymer volume fraction refers to the solution in wall on solute-solvent drag.
which the gel is swollen. Peppas and Reinhart developed free volume theory based
model for a system of water, solute, and polymer [25]. This model
! "! "! "1=3
rRT 2M̄C 1 y2;s predicted the dependence of the diffusion coefficient on solute
t¼ 1# a# (28)
hydrodynamic radius (rs), mesh size, and degree of swelling, as
MC M¯N a
2 y2;r
well as other structural characteristics of the hydrogels.
Both equilibrium swelling and elasticity theory can also be ! ! "
applied to non-covalently crosslinked gels. These theories were DSM M C # M̄C( k2 rs2
¼ k1 exp # (31)
developed based on the assumptions of tetrafunctional crosslinks DSW M¯N # M̄C( QM # 1
and isotropic behavior. This is normally the case with covalent
crosslinks. Hydrogels often have physical entanglements, hydrogen- DSM and DSW are the diffusion coefficients of solutes in the
bonded structures, or microcrystallites that have different behaviors. hydrogel and water respectively. This ratio is the normalized
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 7

diffusion coefficient. k1 and k2 are parameters of the polymer–


water complex, M¯C ( is the average critical molecular weight
between crosslinks at which diffusion is precluded, and QM is the
degree of swelling of the membrane. This theory assumes diffusion
in highly swollen membranes. Characterization of the diffusion
through amorphous PVA membranes validated this theory [26].
Prausnitz based a theory on the statistical distribution of
network chains and used Monte Carlo simulations to develop a
modified size exclusion theory [27]. However, this theory does not
consider effects of pendant groups or ionic interactions. The
intention was to provide a general understanding focusing on
chains in free space to be built upon by others.
To describe the effect of ionic interactions, solute diffusion Fig. 1. Equilibrium swelling behaviors of anionic and cationic hydrogels. Behavior is
through PAA has been observed as a function of pH. Solute dependent on the ionic pendant groups.
diffusion through PVA/PAA membranes has been demonstrated to Reprinted with permission from Khare et al. [53].

be a function of ionic strength and pH of the solvent [23,28–31].

most common monomers used to introduce pH-responsive behavior


3. pH-responsive hydrogels include acrylic acid (AA), methacrylic acid (MAA), dimethylami-
noethyl methacrylate (DMAEMA), diethylaminoethyl methacrylate
pH-responsive hydrogels are a subset of stimuli-responsive (DEAEMA) and acrylamide (AAm) [44]. Table 2 shows the chemical
systems capable of responding to perturbations in the environ- structures of these common monomers as well as their anionic or
mental pH. These responses range from pH-induced deswelling/ cationic behavior.
swelling behavior [32]. Systems that can respond to a dynamic pH Natural polymers such as albumin [45], gelatin [46], alginate
environment are of particular interest for biomedical applications [47,48], and chitosan [49] can also exhibit pH-responsive behavior.
as several locations in the body exhibit substantial pH changes Specifically, proteins such as albumin and gelatin, when in a linear
during either normal function or as part of a disease state. These pH configuration, will form helices stabilized by hydrogen bonding at
variations exist within sites such as the gastrointestinal tract given pH and temperature conditions. These helices can then act as
[33,34], vagina [35], blood vessels, intracellular vesicles [36–38], the crosslinks in the hydrogel network. The isoelectric point, pI, of
inflamed tissue/wounds [39], and the extracellular tumor envi- the protein will then dictate the swelling behavior. When the pH
ronment [40,41] which can trigger a pH response. The particular of the solution is either lower or higher than the pI, the protein will
dynamic pH ranges are detailed in Table 1. accrue surface charge, which will result in electrostatic repulsion
and swelling of the network [32]. Polysaccharides such as chitosan
3.1. Polymer composition and swelling behavior and alginate undergo physical crosslinking due to hydrophobic or
charge interactions. Swelling occurs as a result of the ionization
The pH responsive behavior of the hydrogel network is of groups along the polysaccharide chain resulting in the buildup
imparted by the presence of ionizable pendant groups in the of charge and subsequent electrostatic repulsion and swelling
polymer backbone [32]. When exposed to an aqueous solution of [50]. The benefit of natural pH-responsive polymers, as compared
an appropriate pH and ionic strength, these pendant groups will to their synthetic counterparts is their ability to degrade within the
ionize and result in the buildup of a fixed charge along the polymer. body over time, which is ideal for implanted materials or
The generation of electrostatic repulsive forces results in the pH- circulating drug delivery vehicles [51].
dependent swelling and deswelling processes as the water is either
absorbed or expelled from the hydrogel network [32,42,43].
Two different families of pH-responsive hydrogel exist that differ 3.2. Swelling behavior and theoretical considerations
in their pendant group ionization and subsequent swelling behavior.
Anionic hydrogel networks contain pendant groups that are ionized The swelling behavior of ionic hydrogels is governed by the
in solutions at a pH greater than their acid dissociation constant, or properties of the polymer, the properties of the swelling medium,
pKa. Therefore, the hydrogel swells at pH > pKa because of the large and the polymer–solvent interactions. The composition of the
osmotic pressure generated by the presence of the ions. Conversely, swelling medium dictates the pH and ionic strength of the solution,
cationic pendant groups are ionized at a pH less than their pKa and which is governed by the primary counterions in solution and their
the corresponding hydrogel network is, therefore, swollen at valency [32,52]. The hydrogel network, in turn, acts as a
pH < pKa. These swelling behaviors are described in Fig. 1. The semipermeable membrane to those counterions. This localization
of charge influences the osmotic balance between the hydrogel and
external swelling solution and results in water imbibition. The ion
Table 1 exchange that causes this osmotic balance will obviously be
Locations within the body that exhibit a dynamic pH range
during normal function or as a response to a disease state.
impacted by the ionic interactions present in a charged gel,
specifically, the ionizable groups and degree of ionization [53–
Location pH 55]. Therefore, the ionic contribution to the overall swelling must
Blood 7.34–7.45 be considered and is represented by the ion osmotic swelling
Stomach 1.0–3.0 pressure, pion:
Upper small intestine 4.8–8.2
X
Colon 7.0–7.5
pion ¼ RT ðC i # Ci( Þ (32)
Tumor, extracellular 7.2–6.5
Early endosome 6.0–6.5
Late endosome 4.5–5.0
Ci and Ci* are the counterion concentrations inside and outside
Vagina 3.8–4.5 the gel, respectively. R is the universal gas constant and T is the
Inflamed tissue/wound 5.4–7.4 absolute temperature [55]. Additionally, the elasticity of the
Adapted from Schmaljohann [51]. polymer network and its hydrophilicity/hydrophobicity will impact
8 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Table 2
Structures of common anionic and cationic pH-responsive monomers.

its affinity and ability to imbibe water [55,56]. The crosslinking fluid. In the case of drug delivery applications, the swelling medium
density is another hydrogel property that has a significant influence is biological fluid which has a wide variety of different ionic species
on the overall swelling capability of the network as it impacts the and will, therefore, drastically impact not only the swelling behavior
extent to which the final hydrogel network can swell. but also solute diffusion into and out of the polymer network
Brannon-Peppas and Peppas have modeled the overall swelling [56,59,60]. Additionally, the ionic character of the solute/drug will
capacity of an ionic hydrogel network, which includes the also impact its diffusion into the hydrogel due to the interaction
dependencies of the swelling behavior on the ionic strength of between two charged species. The ionic strength of the solution
the surrounding swelling agent and the ions within the gel can shield the charge of the polymer network and decrease either
[54,57]. The expression is a modification of the original Peppas– the repulsion or attraction between the solute and the hydrogel,
Merrill equation (Eq. (4)) including the contributions of the ionic resulting in increased diffusion [61,62]. Therefore, the ionic
moieties to the free energy and chemical potential. The results character and other properties of the solute, solvent, and network
yield two separate but equivalent expressions for anionic and all function together to impact the overall swelling behavior of ionic
cationic hydrogels, Eqs. (33) and (34), respectively: hydrogels in complex fluids for biomedical applications.
! !2
V1 v22;s 10#pKa 3.3. pH responsive hydrogels for controlled drug delivery
4I v̄2 Mr2 10#pH þ 10#pKa
pH-responsive hydrogel systems have been widely used for the
¼ ½lnð1 # v2;s Þ þ v2;s þ x1 v22;s ' controlled drug delivery of a variety of therapeutics ranging from
! "! " "! " ! "#
V1 2M̄ c v2;s 1=3 v2;s proteins [63], to small molecule drugs [64], chemotherapeutics
þ 1# v2;r # (33) [65], and genetic material such as RNA and DNA [66]. These
v̄M̄c M̄ n v2;r 2v2;r
therapeutic molecules can be dissolved or encapsulated within the
! !2 hydrogel network [54], such as in protein loaded hydrogels [67], or
V1 v22;s 10#pK b electrostatically bound to the charged hydrogel network, as in
4I v̄2 Mr2 10#pH þ 10#pK b the case of genetic material [68]. The controlled drug release
from these loaded hydrogels are triggered by a change in the
¼ ½lnð1 # v2;s Þ þ v2;s þ x1 v22;s '
"! surrounding pH, for example, the change in pH during the transit
! "! " " ! "#
V1 2M̄ c v2;s 1=3 v2;s through the GI tract or intracellular trafficking pathways (Table 1)
þ 1# v2;r # (34)
[69]. This pH-response can be tailored for delivery to specific sites
v̄M̄c M̄ n v2;r 2v2;r
around the body by careful and intentional selection of the
Using these equations to calculate the average molecular monomer components. A brief overview of hydrogel formulations
weight between crosslinks, Mc , in addition to the previously and corresponding applications will be provided.
described parameters, also requires the ionic strength, I, and the
dissociation constants, Ka and Kb. Mr is the molecular weight of the 3.3.1. Anionic hydrogel networks
repeating unit and v̄ is the specific volume of the polymer. This Anionic hydrogel networks, as a reminder, remain collapsed at
complex expression, although somewhat cumbersome to use, low pH due to the presence of physical interactions (i.e., hydrogen
provides important insight into the equilibrium structure and bonding) that keep the network tightly complexed. Once the pH
behavior of ionic hydrogel networks at the molecular and increases above the pKa of the polymer, the complexes dissociate
macromolecular level [57]. due to changes in the ionic character and the polymer swells due to
The kinetics of hydrogel swelling is largely determined by mass a combination of electrostatic repulsion and water imbibition
transfer limitations. Ionic gel swelling kinetics also rely on ion [44]. This swelling behavior is triggered by an increase in the pH
exchange, ion interactions, and Donnan equilibrium consider- of the surrounding environment, which is observed in systems
ations [55,58]. The swelling behavior, both equilibrium and such as the gastrointestinal tract and the vaginal canal during
dynamic, is clearly dependent on the nature of the surrounding intercourse.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 9

3.3.1.1. Oral delivery. Anionic hydrogel network swelling is trig- in a concentrated protein solution. The insulin was loaded into the
gered by the substantial pH change in the gastrointestinal tract, hydrogel network at high efficiencies [79]. These insulin-loaded
ranging from as low as pH 1.0 in the stomach to upwards of 8 in particles were introduced to the gastrointestinal tract of male
certain segments of the upper small intestine and colon Sprague-Dawley rats and the overall bioavailability of the insulin
[33,34]. During transit through the GI tract, the anionic hydrogel was measured by the protein concentration in the bloodstream
network remains collapsed when in the stomach, thereby after time. The insulin-loaded hydrogels were capable of introduc-
protecting any delicate encapsulated therapeutic cargo from being ing insulin into the bloodstream of these rats at a 4.6–7.2%
denatured by the harsh pH conditions and digestive enzymes. bioavailability and the transported insulin was able to exert a
Based upon the composition of the polymer network, swelling can hypoglycemic effect [79–81]. The insulin-loaded hydrogel carriers
be initiated either in the highly absorptive upper small intestine or were also successful in mitigating the effects of food intake on
further down the GI tract, in the colon. glucose levels when orally administered three times per day with
The laboratory of Peppas et al. has focused substantial effort on food [82].
developing anionic hydrogel networks for the oral delivery of P(MAA-g-EG) has also shown some success for the delivery of
therapeutic proteins to the upper small intestine. The primary additional protein therapeutics, such as interferon-b [83] and
network of interest is a copolymer consisting of methacrylic acid calcitonin [83,84]. However, efforts in the lab have shifted toward
(MAA) polymer backbone and grafted poly(ethylene glycol) (PEG) the optimization of different complexation hydrogel networks for
chains. This network, henceforth designated as P(MAA-g-EG), the oral delivery of a variety of protein therapeutics. One such
obtains its pH-responsive behavior from the MAA, which has a example, developed by Carr et al., is P(MAA-co-NVP), a hydrogel
pendant carboxylic acid that is protonated below its pKa of 4.8 and network with a backbone composed of a copolymer of MAA and N-
deprotonates at pH > pKa. When MAA is protonated it undergoes vinyl pyrrolidone (NVP) [85]. N-vinyl pyrrolidone is a highly
hydrogen bonding, or complexation, with the etheric oxygen of the hydrophilic monomer that can be readily polymerized into the
PEG, which maintains the hydrogel in its collapsed configuration. backbone of the hydrogel network and possesses desirable
The negative charge present on the pendant carboxylic acid after properties such as mucoadhesion, minimal toxicity, hydrogen
deprotonation participates in the electrostatic repulsion that bonding groups, and a neutral charge. P(MAA-co-NVP) exhibits
initiates the swelling behavior, while the hydrophilic nature of the appropriate anionic swelling behavior needed for oral
PEG helps to increase the rate of water imbibition [70]. Upon delivery, in fact, remaining more tightly collapsed at low pH
swelling, the porous structure of the P(MAA-g-EG) expands than its P(MAA-g-EG) counterpart. High insulin loading efficien-
resulting in a corresponding increase of the mesh size, j, from cies were observed and the release profile (shown in Fig. 2) shows
70 Å during collapse to ,210 Å when swollen [71]. John Klier et al. no insulin release at low pH and complete insulin release within
were the first to investigate the P(MAA-g-EG) complexation 10 min after exposure to neutral pH [86]. Also promising is the fact
hydrogel for the oral delivery of protein therapeutics that the P(MAA-co-NVP) hydrogel microparticles appear to have
[72,73]. The swelling properties of this system were successfully no cytotoxic effect on two model cell lines, Caco-2 colon
tailored by modifying the ratio of hydrogen-bonding groups of adenocarcinoma cells and HT29-MTX mucus-secreting goblet
MAA to hydrogen-bonding groups of PEG and the molecular cells [86].
weight of the PEG tether. Specifically, when the hydrogen bonding The hydrogel network was also tested for its ability to load
groups of MAA and PEG are equivalent and the molecular weight of proteins of different isoelectric points and higher molecular
the PEG tether is 1000 (corresponding to 23 repeat ethylene glycol weights, such as calcitonin and growth hormone, respectively.
units/hydrogen bonding sites), the P(MAA-g-EG) exhibited the While P(MAA-co-NVP) was quite successful in the loading and
highest degree of complexation at low pH and an improved release of the higher molecular weight growth hormone
swelling response at high pH [70,71]. (MW , 22 kDa), even performing better than P(MAA-g-EG), the
The P(MAA-g-EG) systems have been optimized for the delivery calcitonin did not mirror that behavior [87]. High isoelectric point
of protein therapeutics to the upper small intestine, where the proteins, when compared to lower isoelectric point proteins, are
tight junctions of the intestinal epithelial layer are more conducive positively charged at neutral pH. Therefore, at neutral pH, or the
to the passive transport of bulkier macromolecules. This ‘‘absorp- pH during release, the negatively charged hydrogel network and
tion window’’ can be rather narrow as most material passes positively charged protein have favorable ionic interactions that
through the upper small intestine within 2–4 h after exiting the limit or completely inhibit release from the network. Koetting
stomach [74]. The incorporation of high molecular weight PEG et al. has focused on developing alternative anionic hydrogel
tethers into the P(MAA-g-EG) network is of utmost importance, not networks composed of itaconic acid (IA) and NVP copolymer
just to allow for swelling, but also because of the mucoadhesive specifically for the treatment and delivery of high isoelectric point
behavior of PEG. The tethers of PEG interact with the mucus lining proteins [62].
of the upper small intestine, penetrating into the polysaccharide The optimization of P(MAA-g-EG) and other complexation
matrix of the mucus and engaging in physical entanglements and hydrogel networks for the specific therapeutic of interest is
hydrogen bonding [75]. This behavior anchors the hydrogel incredibly important. Minor variations in the size, isoelectric
particle at the intestinal wall where it can remain for longer point, and hydrophobic/hydrophilic composition will drastically
periods of time to ensure complete diffusion of the therapeutic out affect the loading efficiencies, release kinetics, and overall value
of the matrix and also guarantees the close proximity of the of the hydrogel system. Many therapeutic molecules, particu-
therapeutic to the site of delivery for improved overall bioavail- larly those used to treat cancer, are incredibly hydrophobic in
ability [76,77]. Other mucoadhesive molecules have been incor- nature and do not partition well into the very polar, hydrophilic
porated into the P(MAA-g-EG) network including wheat germ environment within the hydrogel matrix [65]. However, the oral
agglutinin [78] and dextran (ongoing). delivery of chemotherapeutic agents, particularly for cases of
The P(MAA-g-EG) network has shown great promise for the oral cancers within the gastrointestinal tract, would be particularly
delivery of insulin, in particular. Insulin is a small protein with a desirable as it has the potential to limit the off-target effects
molecular weight of 5.8 kDa and a pI of 5.3. The insulin is loaded of chemotherapeutics [37,65]. Schoener et al. synthesized
into the hydrogel network in a post-synthesis loading scheme by hydrophilic P(MAA-g-EG) hydrogels with embedded hydropho-
swelling the hydrogel network, crushed into microparticles and bic nanoparticles composed of poly(methyl methacrylate)
sieved into narrow size ranges (<75 mm, 75–150 mm, >150 mm), (PMMA). The hydrophobic chemotherapeutic agents, modeled
10 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

3.3.1.2. Vaginal delivery. A healthy vagina will have a pH between


4-5 [35]. However, upon the introduction of sperm (pH ,7.5)
during intercourse, the pH of the vagina can easily increase to
above pH 7. In addition to raising the overall pH of the vaginal
environment, the ejaculatory fluid can carry a variety of sexually
transmitted diseases, including the HIV virus [94]. Several systems
currently exist to locally administer therapeutics designed to
prophylactically treat potential STD exposure, for example, vaginal
rings for the delivery of the HIV microbicide dapivirine [95] and
potential monoclonal antibody therapies [96], thin films adminis-
tering contraceptive antimicrobial agents [97], and a gel containing
tenofovir, an anti-HIV prodrug [98]. pH-responsive nanogels have
several unique advantages such as their small size, minimal site
irritation, and the ability to protect the loaded drug and both target
and control its release into the environment [94]. Unlike the other
systems, a pH-responsive nanogel system will only release the
therapeutic upon a pH shift induced by the introduction of
potentially infectious semen. Additionally, if engineered correctly,
the nanoparticles will slowly degrade over time into harmless
bioproducts requiring no removal of a spent film or ring. The
system proposed by T. Zhang et al. are nano-sized hydrogel
particles composed of pH-responsive MAA and degradable
Fig. 2. Insulin release from P(MAA-co-NVP) at both low pH (,3) and neutral pH (,7)
conditions.
poly(lactic-co-glycolic acid) (PLGA) loaded with either Tenofovir,
Reprinted with permission from Carr and Peppas [86]. TNF, or its prodrug TDF, tenofovir disoproxil fumarate [94]. Nano-
particles were synthesized by either freeze- or spray-drying
techniques [94,99]. These systems showed successful incorpo-
as fluorescein in these studies, could partition into the ration of either TNF or TDF and a clear pH-responsive drug release
hydrophobic or non-polar region of the hydrogel network and for most tested polymer formulations. When introduced to
associate with the PMMA nanoparticles. The incorporation of different vaginal cell types some minor cytotoxicity was observed,
hydrophobic moieties did have a detrimental effect on final but, more importantly, the nanoparticles were uptaken into the
equilibrium swelling capability of the hydrogel, but drastically vaginal endothelial cells, which is important for retention within
improved fluorescein loading and subsequent release and, the vaginal canal [94].
importantly, did not shift the dynamic pH-response [65]. The
customization of synthetic pH-responsive hydrogel networks 3.3.2. Cationic hydrogel networks
is clearly a very powerful tool for the oral delivery of sensitive Cationic hydrogel networks exhibit opposite swelling behavior
therapeutics such as proteins and hydrophobic chemotherapeu- to anionic hydrogels. Specifically, the hydrogels exist at a swollen
tic drugs. state at low pH (pH < pKa) and collapse upon exposure to a higher
A number of biologically derived polymers, including alginate pH environment (pH > pKa). The swelling behavior is, therefore,
and chitosan are attractive alternatives to synthetic polymers for triggered by a decrease in the pH of the surrounding area.
pH-responsive applications due to their inherent biocompatibility
and physicochemical properties. Alginate is a naturally derived 3.3.2.1. Gastrointestinal delivery. Chitosan is a naturally derived
polysaccharide-based biopolymer that is extracted from brown polysaccharide obtained from the deacetylation of chitin, an
algae (kelp) [88]. When exposed to calcium ions (Ca2+), the abundant polysaccharide extracted from marine crustacean. It is
polysaccharide crosslinks and forms a gel [88,89]. These mild linear in structure and composed of linear b-(1 ! 4)-glycosidic-
gelation conditions are highly favorable for biological applications linked 2-amino-2-deoxy-b-D-glycan monosaccharide units. The
because sensitive therapeutics, or even cells, could be encapsulat- primary amines in the chitosan structure result in the overall
ed in the hydrogel network during synthesis while still maintain- polysaccharide being positively charged and, therefore, inherently
ing activity and viability [88]. Alginate exhibits a pH-dependent mucoadhesive [100]. This, coupled with its inherent biocompati-
swelling/deswelling behavior, collapsing at low pH and swelling at bility and mild gelation conditions, makes chitosan an attractive
neutral pH [90]. When formed into calcium-crosslinked beads, system for a multitude of drug delivery applications [101,102]. The
alginate hydrogels have successfully administered both vaccines cationic character of the chitosan means that it is insoluble in
[91] and small molecule [92,93] drugs via the oral route, protecting water at neutral or basic pH due to the free amino groups present.
them throughout their transit in the GI tract and delivering them at When those amino groups undergo deionization at acidic pH, the
the sites of interest. More recently, a pH-responsive hydrogel structure becomes soluble in water. It retains this inherent pH-
system composed of a semi-IPN network of a chitosan derivate responsiveness when crosslinked into a hydrogel network either
(NOCC) and alginate has been developed for the oral delivery of by modification by synthetic polymer species or by ionic cross-
protein therapeutics. The NOCC and alginate associate with one linking with multivalent anions [100].
another due to favorable charge interactions and are then This pH-responsive behavior of chitosan-based hydrogels can
crosslinked by the presence of the naturally occurring crosslinking be harnessed for the targeted gastrointestinal delivery of a variety
agent, genipin [90]. The pH responsiveness of this system is driven of therapeutics. Without modifying the chitosan hydrogel net-
by the behavior of the anionic alginate hydrogel, remaining work, it would remain collapsed at the neutral pH in the mouth and
collapsed at gastric pH and swelling at intestinal pH. The only swell once reaching the acidic environment of the stomach. To
encapsulation of a model protein, bovine serum albumin (BSA), achieve this gastric-specific delivery, Patel et al. synthesized a
was successful with the hydrogel network protecting the cargo semi-IPN network composed of chitosan and poly(ethylene oxide)
during low pH exposure and releasing it only upon triggering by pH [103]. The polymer network swelled approximately 10 times more
change [90]. in the gastric fluid than in the intestinal fluid. When model
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 11

antibiotics, metronidazole and amoxicillin, were loaded into the


hydrogel network and then released at gastric and intestinal pH
conditions, both antibiotics were released to a far greater extent at
acidic pH than at neutral pH [103]. Upon the inclusion of another
ionic moiety, such as alginate, into the hydrogel network, the pH-
responsive behavior can be adjusted. Dai et al. crosslinked a
chemically modified chitosan, N-succinyl chitosan (Suc-Chi),
copolymerized with alginate by ionic gelation. The addition of
Ca2+ ions results in the ionic complexation of the network,
providing the tie points for crosslinking. The resulting system
remains collapsed at low pH and swells at neutral pH, much like
one would expect out of an anionic system. The therapeutic
nifedipine was encapsulated within the hydrogel network and its
release monitored over different pH conditions. As would be
expected, minimal release was observed at low pH when the gel
was collapsed and more significant release was seen once the gel
was swollen. Interestingly, the N-succinyl modification to the
chitosan improved its solubility at neutral pH. Although the ionic
character of the chitosan was not changed, the pH-response of the Fig. 3. Volume swelling ratio, Q, as a function of pH for Cationic PDBP nanogels of
network could be manipulated very readily simply by changing the various crosslinking ratios: *, 0.01; *, 0.025; ~, 0.05; ~, 0.1.
solubility of the chitosan material [104]. Reprinted with permission from Fisher and Peppas [106].

3.3.2.2. Intracellular delivery. Intracellular vesicle pH, particularly 3.3.2.3. Intracellular delivery of chemotherapeutics. The ability of
within those vesicles engaged in intracellular trafficking, decreases cells to preferentially uptake nanoparticles of specific size and
rapidly from neutral to acidic conditions [36–38]. Peppas et al. surface charge can be taken advantage of for the intracellular
have recently focused on the development of cationic nanogels delivery of a variety of therapeutics [112]. This is particularly
for the intracellular delivery of siRNA. The cationic nanogels advantageous for the treatment of cancer because nanoparticles
were composed of a copolymer of poly(2-(diethylamino)ethyl- containing chemotherapeutic agents can be administered directly
methacrylate) (DEAEMA) and t-butyl methacrylate with grafted into the cancerous tumor cells and initiate apoptosis. If active
poly(ethylene glycol) chains (PDBP). These nanogels were approx- targeting moieties are included on the surface of the nanoparticle,
imately 51 nm in diameter and exhibited the expected pH- preferential and specific delivery to tumor cells can be accom-
responsive swelling/deswelling behavior around the pKa of the plished, which leaves the healthy cells intact and significantly
gel (Fig. 3). The swelling response, loading efficiencies for protein, reduces off-target side effects [37].
cellular uptake and biocompatibility were all tailored by tuning the Both natural and synthetic cationic hydrogels have been
crosslinking density, hydrophobic polymer content, and polymer developed to deliver chemotherapeutic drugs into cancerous cells.
composition [105–107]. To further tune the swelling behavior and A hydrogel composed of a chitosan derivative, N-[(2-hydroxy-3-
to optimize the system for the intracellular delivery of siRNA, trimethylammonium)propyl] chitosan chloride (HTCC) was ioni-
Liechty et al. synthesized three separate cationic nanogel cally crosslinked by sodium tripolyphosphate to form cationic
formulations: (1) crosslinked 2-(diethylaminoethyl) methacry- chitosan-based nanogels [113]. These nanogels were further
late and poly(ethylene glycol) methyl ether methacrylate, functionalized by the surface conjugation of Apo-transferrin, an
P(DEAEMA-g-PEGMA), (2) inclusion of t-butyl methacrylate, iron shuttling protein that interacts with the overexpressed
P(DEAEMA-co-TBMA-g-PEGMA), and (3) inclusion of t-butylami- transferrin receptors on the surface of tumor cells and initiates
noethyl methacrylate P(DEAEMA-co-TBAEMA-g-PEGMA). The cell-mediated endocytosis. The cationic pH-dependent swelling
increase in hydrophobic content, i.e., the inclusion of TBMA and behavior results the gel swelling upon uptake into the endosome and
TBAEMA, resulted in a decreased onset of pH-dependent gel releasing its chemotherapeutic payload, methotrexate disodium
swelling, while the TBMA depressed the critical swelling pH from (MTX). When these systems were added to the immortalized HeLa
7.8 to 7.0 [108]. Forbes et al. were able to exhibit tighter control of cancer cell line, they were uptaken at a higher rate than unmodified
final cationic nanogel parameters by switching from a UV- nanoparticles and induced a higher rate of apoptosis [113].
initiated polymerization to ARGET-ATRP initiated polymerization A synthetic cationic gel was synthesized via an emulsion
[109,110]. Furthermore, these cationic nanogels were able to be copolymerization of 2-(N,N-diethylamino)ethyl methacrylate with
successfully complexed with siRNA, and promote cellular uptake a heterobifunctional PEG with a 4-vinylbenzyl group at the a-end
in RAW264.7 macrophages and HEK293T cells [66,110]. It is and a carboxylic acid group at the v-end. The chemotherapeutic
argued that the pH-triggered swelling of the nanogel in response agent doxorubicin was loaded into the hydrogel network via a
to the decrease in pH within the endosome results in the so-called solvent evaporation method and incorporated at up to 26 wt%. The
proton-sponge effect, which ends in the bursting of the vesicle due pH-responsive moieties within the hydrogel allowed for a burst
to excessive water imbibition due to an osmotic pressure release of doxorubicin to occur at endosomal pH, which improved its
imbalance [111]. Once the vesicle bursts, the siRNA is no longer cytotoxic effects on HuH-7 human hepatoma cells compared to
complexed with the now deionized cationic nanoparticle and is naked doxorubicin. The uptake of doxorubicin bearing pH-respon-
distribution into the cellular cytosol where it can exert sive hydrogels via endocytosis results in the accumulation and
therapeutic effect. The particles developed by Forbes et al. do delivery of the chemotherapeutic agent directly into the cellular
indeed accomplish siRNA-mediated gene silencing, which indi- cytosol where it can exert its most potent cytotoxic effect [114].
cates that the siRNA and associated nanogel undergo some extent
of endosomal escape [66,110]. These results have powerful 3.4. Concluding remarks
implications for the treatment of a variety of disorders that are
caused by gene overexpression including several autoimmune Both anionic and cationic hydrogels have demonstrated utility
disorders and types of cancer. in pH-responsive drug delivery applications ranging from oral
12 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

protein delivery to intracellular chemotherapeutic and gene hydrophobic segments requires the formation of water cages.
delivery. The wide variety of existing pH-responsive polymers, These water cages balance the insolubility of the hydrophobic
both of synthetic and natural origin, allow the researcher to fine backbone in water, allowing the hydration of the polymer. The
tune the swelling properties for the application and target of formation of these cages comes at a steep entropic cost, which is
interest. The inherent pH variability within the human body, only balanced out by the hydrogen bonds formed by the amide
especially during particular disease states, lends great applicability groups. As the temperature increases, the entropic costs become
to these pH-responsive hydrogel systems. too great, as seen in Fig. 4, coming to a peak at the LCST. This forces
the system to react by expelling the water from the hydrogel,
4. Temperature responsive hydrogels resulting in collapse.

Thermoresponsive materials for medical use have been of great 4.1.2. UCST
interest due to the relatively universal physiological temperature Although UCST systems are less common than LCST, the
of 37 8C and the development of a number of mechanisms to common UCST hydrogel is an interpenetrating network (IPN)
manipulate and control temperature in vivo. Thermoresponsive composed of interlocking networks of acrylamide (AAm) and
hydrogels fall into two primary categories, positively and acrylic acid (AA) [118]. The critical responsiveness of these
negatively responsive systems. Normally, these systems are materials is often described by what is commonly referred to as
identified by having an upper critical solution temperature (UCST) a ‘‘zipper’’ effect, illustrated in Fig. 5. Essentially, hydrogen bonds
or a lower critical solution temperature (LCST), respectively. The between the AA and AAm networks force the gel to collapse at low
expansion or collapse that correlates with the critical shift in temperatures. As the temperature increases, the enthalpic gains
aqueous solubility has been utilized as a mechanism for drug from these bonds are overpowered by the entropic loss of
delivery, membrane separation/cleaning, and recently, in situ separation of the water and polymer phases. The resulting
gelling scaffolds for tissue regeneration [115,116]. criticality unzips the networks, allowing the hydration of the
polymer networks.
4.1. Swelling theory Although this is a relatively simple explanation, it is often a
sufficient explanation for most UCST systems, even though the
4.1.1. LCST one-to-one alignment of AAm units to AA units is idealized.
The swelling response of reverse thermoresponsive biomater- However, the critical response depends on a number of other
ials has been extensively studied for poly(N-isopropylacrylamide) factors, in addition to the breaking of hydrogen bonds, to result in
(PNIPAAm) and other alkyl-substituted acrylamides. The LCST some of the drastic transitions observed [119]. Another important
response arises from a balance of hydrophobic and hydrophilic factor in the effective swelling of these UCST hydrogels comes in
groups, and the entropic and enthalpic costs required to solvate the form of ionic repulsions. Also, due to the presence of AA units,
these groups. Bae et al. demonstrated the effect of temperature on the swelling response can be greatly impacted by the ionic strength
the aqueous mixing of different N-alkyl substituted acrylamide and pH of the fluid. Due to the need of protonated acrylic acid
hydrogels, including PNIPAAm and other non-temperature sensi- groups, the optimal swelling response of the polymers has been
tive polymers [117]. By utilizing the following definitions of the shown to be approximately 4.6, about the pKa of acrylic acid.
polymer interaction parameter, x, and the enthalpic and entropic However, this can be overcome by the use of propyl acrylic acid,
contributions to this parameter, xH and xS respectively, they were which has a pKa of over 7 [120].
able to identify the enthalpic and entropic shifts throughout this These issues, along with the high degree of sensitivity to ionic
critical phase shift. species, lead to limitations of these technologies in vivo where pH
and ionic strength are constantly fluctuating both in and between
lnð1 # v2s Þ þ v2s þ y(e VS0 v2r fðv2s =v2r Þ1=3 # 1=2ðv2s =v2r Þg patients. This is the primary reason why UCST systems have been
x¼ (35)
v22s studied less than their LCST counterparts.

1 4.2. Common monomers


xH ¼ ¼ #Tð@x=@TÞ (36)
Tð@x=@ð1=TÞÞ
4.2.1. LCST monomers
0
xS ¼ x # xH (36 )
4.2.1.1. N-alkyl substituted monomers. There are a wide range of
The variables v2s , and v2r are the volume fraction of the synthetic and naturally occurring polymer systems that exhibit
polymer in the swollen and relaxed state, respectively, ye* is the LCSTs around physiological temperature [121]. The majority of the
crosslinking density, and VS0 is molar volume of the solvent. work done in LCST systems has centered around hydrogels
The results of these studies are seen in Fig. 4. There are two things composed of poly(N-isopropylacrylamide) (PNIPAAm) and its
to note from these studies. First, poly(acrylamide) (PAAm) is the copolymers. PNIPAAm exhibits an LCST around 32 8C, close to
only gel to demonstrate a mild positive swelling response. This is physiological temperature, making it an ideal target for biomedical
due to the lack of a hydrophobically modified amide group, applications. The LCST can be and has been modified by a number
meaning that the primary interaction is hydrogen bonding, not the of comonomers [121]. The inclusion of hydrophilic monomers
hydrophobic interactions that dictate N-alky substituted acryla- raises the LCST, while hydrophobic comonomers depress the
mides. Second is the sharp peaks in the PNIPAAm and poly(N0 N0 - critical point. However, there are limitations to the shift of
diethyl acrylamide) (PDEAAm) results. These two polymers are the the LCST, due primarily to the necessity of contiguous NIPAAm
only two to exhibit a critical phase shift; they are also the two units. Because of this requirement, the lower the NIPAAm content
polymers with the most hydrophobic alkyl side-groups, as is, the weaker the collapse.
determined by their inverse volume fraction in ethanol. In addition to PNIPAAm, there have been studies to identify
This means that the swelling responses exhibited by these LCST other N-alkyl substituted acrylamides that display LCST swelling
polymers are greatly aided by drastic differences in hydrophobic- behavior [122]. One common monomer that is looked to in order to
ity. The other major note is the large contribution of entropy to the shift the LCST above 37 8C is PDEAAm, exhibiting an LCST of
x parameter. This is due to the fact that the hydration of approximately 39 8C. However, the work by Plate and Okano has
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 13

Fig. 4. (a) Inverse polymer volume fraction as a function of time. (b) Polymer interaction parameter in ethanol. (c) polymer interaction parameter in water. (d) Enthalpic
contribution to the polymer interaction parameter in water. (e) Entropic contribution to the polymer interaction parameter in water. *poly(acrylamide), ~poly(dimethyl
acrylamide), & poly(ethyl acrylamide), & poly(acrylroylpyrolidine), ~ poly(diethylacrylamide), * poly(N-isoproylacrylamide).
Reprinted with permission from Bae et al. [117].
14 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

4.2.2. UCST monomers

4.2.2.1. AAm and AA. IPNs and other hydrogels that exhibit UCST
behavior generally focus around an acrylic acid based monomer.
Some random copolymers of AAm and AAc have also demonstrated
some unique UCST properties, though nothing that can be
identified as a critical temperature response [129]. Like their LCST
counterparts, the AAm and AA networks can be modified with
hydrophobic or hydrophilic comonomers to shift the critical
temperature, although the magnitude of collapse diminishes with
increasing comonomer concentration [118,130].

Fig. 5. Zipper-like hydrogen bonding of PAA-PAAm IPN. Reprinted with permission 4.3. Applications
from Katono et al. [118]. Copyright 1991 Elsevier.
4.3.1. Oral drug delivery
The use of temperature sensitive hydrogels as drug delivery
turned up other monomers for these systems with a range of LCSTs. vehicles has been studied since their inception. The significant
However, they also exhibit different magnitudes of collapse, as the change between ambient temperature and physiological temper-
same alterations that shift the critical temperature lead to different ature has been targeted as a mechanism for the delivery of proteins
hydration levels, both in collapsed and swollen states [117]. and small molecules, such as ibuprofen [131] and insulin
[132]. However, this mechanism of administration was rendered
4.2.1.2. Polyethylene glycol. PEG is an interesting amphiphilic obsolete with the development of pH responsive hydrogels.
molecule that, in addition to providing effective stealthing
properties, demonstrates negative temperature response in 4.3.2. Particle based drug delivery
aqueous environments [123]. PEG methacrylate hydrogels de- Originally, it was postulated that thermally responsive hydrogel
crease in swelling ratio as temperature increases. The temperature nanoparticles could be actuated by the rise in temperature in the
response of these gels can be manipulated by changing the length body that accompanies a fever. However, the immune response to
of the PEG chains, as longer chains lead to lower solubility at the presence of these nanoparticles would result in a similar
lower temperatures. Although PEG hydrogels do exhibit a localized temperature increase. This problem can be neutralized by
temperature response, it is not a critical response, though their the stealthing of these particles with PEG tethers [133]. However,
inverse temperature response can be utilized in the backbone of the temperature sensitive particles still fail to respond to the
other macromolecular systems. relatively slight temperature changes that often accompany fevers.
One popular system is commonly referred to as pluronic F-127. This required investigation into other uses for temperature
This material is a triblock polymer consisting of poly(propylene responsive hydrogels. One of the largest fields of interest for
oxide) (PPO) sandwiched between two poly(ethylene oxide) blocks thermal responsive materials falls into the category of theranos-
that also demonstrates an LCST gelling response. These systems tics, or composites designed to deliver both therapeutic and
lack covalent crosslinks, and thus are not standard hydrogel diagnostic capabilities. These systems generally are developed as
networks containing chemical crosslinks. The gelation of these composites of an external stimulus, thermogenic, and traceable
systems relies heavily on the self-assembly of hydrophobic propyl particle, such as iron oxide or gold, coated in a thermally
oxide groups versus hydrophilic ethylene oxide units. Because of responsive, drug loaded hydrogel [134].
this, these systems have been investigated less for their effective- These systems will ideally provide for improved control and
ness as large scale gels, and more for particle based drug delivery localization of therapeutics that are highly toxic when delivered
[124]. The self-assembly of nanoparticles can be controlled by both systemically. One treatment where this technology is desperately
the block lengths and concentration in water. needed is in chemotherapeutic delivery, as the highly toxic drugs
show improved effectiveness when localized and concentrated at
4.2.1.3. Other LCST responsive monomers. The LCST response has the site of a tumor [135]. UCST and LCST hydrogels have been used
been observed in polymers of other naturally occurring and as the thermally responsive polymer, however no systems have
synthetic polymers. Chitosan, a naturally occurring cationic managed to reach clinical trials, primarily due to targeting
polymer, forms gels much in the way PEO-PPO-PEO systems do limitations and the complexity of the treatment.
[125]. When chitosan is dissolved in water, it shifts from a flowing
polymer solution to a gel at about 32 8C. Due to its natural properties, 4.3.3. Tissue culture
it has been included into a PNIPAAm backbone to develop Thermally responsive materials, particularly PNIPAAm, have
crosslinked structures without impacting the LCST, while still been utilized as cell substrates in an attempt to trap cells in a porous
demonstrating the positive biological properties of chitosan [126]. membrane at 37 8C. Galperin et al. [136] developed degradable
Poly(vinyl methyl ether) also collapses at 32 8C, and responds porous PNIPAAm hydrogels. Porosity was manipulated in two ways:
much in the same way PNIPAAm does [127]. These systems are first by the change in crosslink density, and second by the inclusion
polymerized and cross-linked by gamma radiation, and thus the of degradable micro-particles. The degradable microparticles
degree of crosslinking and the overall swelling response can be provide pores large enough to allow NIH 3T3 cells to pervade the
controlled by the level of irradiation. No results are present for the hydrogel matrix. They also included a thermally degradable linker
impact of copolymers on the swelling response of these systems. that allows for dissolution of the hydrogel matrix. This is crucial
This is similar to another vinyl thermoresponsive monomer that for in vivo applications, as the matrix needs to initially be a scaffold
has been recently utilized in hydrogel systems, poly(vinyl for tissue generation, while subsequently disappearing as the
caprolactone). For this polymer, the lack of study is due to the synthesized tissue incorporates into the target area.
sensitive nature of the pendant group [128]. The caprolactone side One area where these thermoresponsive substrates have
chains are sensitive to changes in pH as they tend to polymerize started to stand out versus other hydrogel substrates is as in situ
between the ester groups. gelling platforms. These are systems that can be injected into
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 15

damaged areas, and fill in missing tissue through the precipitation effects on the retinoid enzymatic profile, the temperature sensitive
and post-synthesis crosslinking of PNIPAAm based polymers. The material proved one of the least deleterious routes for cell growth
Mikos group developed a thermally and chemically gelling system and detachment. This and other substrate studies that utilize, at
composed of PNIPAAm, AAm, pentaerythritol diacrylate mono- least, a PNIPAAm based coating to remove substrates from retinal
stearate (PEDAS), and 2-hydroxy ethyl acrylate (HEA), as demon- cell regeneration have demonstrated the need for the thermal
strated in Fig. 6 [137]. These systems non-covalently gel at 37 8C, gelling material in the realm of ocular rehabilitation [136].
while providing a mechanism to covalently bond to provide a
substantial substrate for tissue growth. 4.3.5. Membranes, microfluidics, and sensors
Thermally responsive materials also offer a unique mechanism
4.3.4. Ocular drug delivery for the actuation of flow channels. The collapse that the LCST
Thermoresponsive materials are effective for retinal treatment materials, specifically PNIPAAm, are capable of has provided sharp
for many of the same reasons that they are effective for tissue on/off switching to control the release of therapeutics. Hoare, et al.
engineering. They provide substrate for cell growth, and gel at developed a magnetic particle loaded membrane for the restricted
physiological temperature [138]. Furthermore, the phase transition delivery of fluorescein, represented in the schematic in Fig. 7
provides a mechanism for detachment from a cell layer [126]. This [141]. By entrapping microgels of thermally responsive PNIPAAm,
allows for the attachment of a hydrogel contact lens, and as the they were able to actuate the hydrogel-based valve by alternating
temperature increases, these hydrogels can promote healing and magnetic field. Thermally switching polymers offer another
be peeled off after reaching equilibrium above their LCST [139]. interesting characteristic, as the hydrated–dehydrated shift has
Ocular treatments can take the form of either delivering a proven effective as a mechanism to remove fouling from
therapeutic or as a cell substrate loaded with growth promoters. membranes made of the hydrogels [139,142]. This functionality
Verestiuc et al. developed and characterized a system for the also lends itself to microsensors, providing a mechanism by which
delivery of pilocarpine hydrochloride and other ocular drugs adsorbed solutes can be cleared out for reusability [143].
utilizing AA functionalized chitosan, copolymerized with either
NIPAAm or hydroxyethyl methacrylate (HEMA) copolymers 5. Chemically-responsive hydrogels
[126]. They varied the concentration of ratio of chitosan:HEMA
and chitosan:NIPAAm, and measured drug delivery rates and lift Another broad class of hydrogels has been designed to exhibit
off forces. They found that not only did NIPAAm gels have higher swelling or degradation in response to individual target molecules.
adhesive strengths, but there was also a significant drop after Because of the wide array of chemicals which could act as practical
heating above the LCST. stimuli for achieving useful functions, this review focuses on only a
Studies performed by Von Recum et al. have investigated the few examples of chemically-responsive hydrogels, as an exhaus-
effect of PNIPAAm scaffolds on the enzymatic integrity of donor tive review would be a monumental undertaking. Nevertheless,
retinal cells [140]. They discovered that in addition to having no ill these few examples are demonstrative of the basic mechanism by

Fig. 6. Thermo-gelling schematic for injectable PNIPAAm based hydrogels. Reprinted with permission from Hacker et al. [137]. Copyright 2008 American Chemical Society.
16 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 7. Stimulus-responsive membrane: (a) temperature-triggering, comparison of nanogel particle size in suspension (blue data, right y-axis) and differential flux of sodium
fluorescein through the nanogel-loaded membranes (red data, left y-axis) as a function of temperature; (b) magnetic triggering, temperature profile in the sample chamber
and differential flux of sodium fluorescein out of membrane-capped devices as a function of time over four successive on/off cycles of the external magnetic field; (c) schema
of the proposed mechanism of membrane function. Reprinted with permission from Hoare et al. [141]. Copyright 2009 American Chemical Society. (For interpretation of the
references to color in this figure legend, the reader is referred to the web version of the article.)

which many chemically responsive hydrogels have been made. 5.1. Glucose-responsive hydrogels
The mechanisms either make use of the activity of the target
molecule itself, as with using peptide-based hydrogels that Glucose-responsive hydrogels respond to the presence of
degrade in response to the target enzyme, or use a transduction elevated levels of glucose and have obvious uses in the treatment
pathway to convert recognition of the target molecule into a pH, of diabetes. Diabetes affected approximately 171 million people
temperature, or electrical charge change that drives swelling, worldwide in 2000, and is expected to affect approximately
collapse, or degradation. 366 million by 2030 [144]. Diabetes can be effectively treated with
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 17

injection of insulin, but the high frequency of injection, the need 5.1.2. Concanavalin A gels
for frequent testing, and the inconvenience and pain associated The second main pathway for glucose-responsiveness relies on
with the injection route have led to researchers seeking improved the use of lectins, primarily concanavalin A (Con A). Hydrogels are
insulin delivery options. Hydrogels that are capable of acting as comprised of a glycosylated polymer backbone and physically
long-term insulin depots that respond to increased blood glucose immobilized Con A [159–161]. Con A binds with the glycosylated
levels and automatically release doses of insulin at appropriate moieties in the hydrogel, leading to a tightly complexed matrix
times are a promising development, and could obviate the need with small pore sizes, keeping insulin entrapped within the
for frequent injection and therefore provide a more convenient hydrogel. As glucose diffuses into the polymer matrix, the glucose
treatment option that would improve treatment efficacy and competitively binds with Con A; because Con A has greater affinity
quality of life for hundreds of millions of people. for glucose than for the glycosylated moieties, glucose displaces
the glycosylated polymer, causing the hydrogel to decomplex and
5.1.1. Glucose-oxidase gels either swell, releasing insulin by diffusion, or exhibit a gel–sol
Glucose-responsive hydrogels have typically relied on one of transition, releasing insulin by liberation from the matrix, depend-
three mechanisms for detecting glucose and responding appropri- ing on if the hydrogel is chemically crosslinked or not, respectively.
ately [145]. The first of these is to use glucose oxidase (GOx) as a This release mechanism is shown schematically in Fig. 8.
detecting enzyme and its reaction with glucose as a transduction Because the mechanism of these early Con A-based gels relied
pathway for stimulating response. Enzymes entrapped in hydro- on competitive displacement of Con A from the glycosylated
gels have been reported and used since 1963 when Bernfeld and polymer, they suffered from a relatively slow response time and
Wan reported successful immobilization of enzymes into acryl- leakage of Con A from the hydrogel that limited therapeutic
amide hydrogels [146]. Glucose oxidase was later immobilized in potential and repeatability. The stability of the hydrogel system
hydrogels comprised of N,N-dimethylaminoethyl methacrylate, has been improved through alteration of Con A and through
hydroxyethyl methacrylate, and tetraethylene glycol dimethacry- alternative linking strategies. Conjugation with poly(ethylene
late, making the first true glucose-responsive hydrogels using glycol) chains yielded a more hydrophilic, stable Con A that
glucose oxidase [147–150]. Since then, many studies have been exhibited improved solubility, stability, and affinity toward
performed seeking to improve upon these original hydrogels using glucose, which in turn improved sensitivity of the hydrogel and
the same GOx pathway. reduced lag time [162,163]. Furthermore, covalent immobilization
The basic mechanism by which these GOx-based gels work is by of Con A into the hydrogel matrix using carbodiimide crosslinking
GOx-catalyzed oxidation of glucose followed by pH-responsive prevents leaching of Con A out of the polymer, increasing stability
swelling or deswelling of the hydrogel. When glucose is present of the gel and allowing for repeatable swelling behavior [164–
in the blood (or other surrounding medium), it diffuses into the 166]. In addition to various experiments using modified insulin
hydrogel matrix, where GOx catalyzes reaction of glucose with [167–169], these improvements have led to Con A-based systems
oxygen per the following reaction [151]: for insulin delivery that have significant therapeutic potential.
GOx
Glucose þ O2 þ H2 O#!Gluconic acid þ H2 O2 5.1.3. Phenylboronic acid gels
The last main mechanism for glucose-responsive hydrogels has
One of the products of the reaction, gluconic acid, reduces the been based on use of phenylboronic acid (PBA) functionalized
local pH in the hydrogel, which then elicits a pH-dependent hydrogels. PBA has long been known to have ability to bind
response per one of the mechanisms described previously. saccharide, being first reported in 1954 by Kuivila et al. [170]. PBA
Therefore, the immobilized GOx acts both as the glucose sensor does this by forming reversible, covalently linked complexes with
and the transduction mechanism—the actual response is pH diols [171,172], as shown in Fig. 9. When glucose, with its diol
dependent, but the GOx-catalyzed oxidation reaction is responsi- functionality, enters the hydrogel, the PBA moieties reversibly
ble for converting glucose detection into the requisite pH shift. complex with glucose, shifting equilibrium toward cationically
Researchers have expanded on this basic idea in multiple charged boron residues. The phenylborate group ordinarily exists
ways. Multiple pH-responsive polymers have been used, such in aqueous environments at equilibrium between uncharged and
as poly(N-isopropylacrylamide-co-methacrylic acid) [152,153], cationically charged forms. However, reaction with glucose only
poly(o-phenylenediamine) [154], poly(diethylaminoethyl meth- occurs through the cationically charged form, the product of which
acrylate-g-ethylene glycol) [155], and poly(methacrylic acid-g- is also cationically charged. Therefore, the reaction shifts the
ethylene glycol) [156]. Additionally, use of hydrogels based on overall gel charge density toward cationically charged residues.
sulfonamide chemistry allowed for a pH transition to occur within This increased charge endows the hydrogel with greater hydro-
a narrow and physiological pH range of 6.5–7.5 [157]. philicity, which, together with ionic repulsion within the hydrogel,
Another important improvement was the introduction of leads to a distinct swelling response.
catalase into the hydrogels. Because the GOx-catalyzed reaction PBA-based hydrogels have become the most promising
is oxygen-dependent and consumes oxygen, the reaction is candidates for clinical use because they can offer sensitive glucose
oxygen limited within the local environment of the hydrogel. responsiveness, but do not have the same degradability and
To address this issue, catalase has been simultaneously stability concerns that adversely affect GOx and Con A-based gels.
immobilized in the hydrogel, which catalyzes the degradation While GOx and Con A are proteins susceptible to degradation by
of the hydrogen peroxide product back into oxygen and water enzymatic action or simple leakage out of the gel, PBA is a synthetic
[150,151,157,158]: functional group that is not readily degraded in vivo and is
incorporated into the polymer directly, thus providing the
Catalase 1
H2 O2 #! O2 þ H2 O potential for long acting drug delivery depots while also avoiding
2
extensive chemistry to immobilize proteins in the gel matrix.
This reaction helps keep oxygen available for the GOx-catalyzed PBA-based gels have therefore become the most prominent area
reaction, and also importantly removes the hydrogen peroxide of research in glucose-responsive hydrogels. Many researchers
product which can cause degradation of the GOx enzyme. As a have reported unique variations on the basic chemistry that yield
result, the rate of swelling and enzyme stability are both improved suitable drug delivery carriers or glucose sensors [173–181]. Al-
by introduction of catalase. though such systems are yet to replace insulin injection or insulin
18 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 8. Concanvalin A-based glucose-responsive hydrogel swelling mechanism.

pumps as the primary treatment option for diabetics, the advances the GFLG sequence is used as a side chain link between the drug
in glucose-responsive systems, whether based on glucose oxidase, and the HPMA backbone. The HPMA copolymer with GFLG side
concanavalin A, or phenylboronic acid, have shown that superior chains is made by polymerization of HPMA and methacryloyl-
options are available, and further improvements on the technolo- GFLG-nitrophenyl ester monomers, and the drug is conjugated
gies will hopefully lead to treatment options that improve the by aminolysis [186,189,192]. Drugs studied have included
quality of life for the hundreds of millions of people living with doxorubicin [185], insulin [193], and ampicillin [190]. The
diabetes worldwide. peptide sequence was selected because it is cleavable by
cathepsin B, releasing the drug payload into the lysosomal
5.2. Enzyme-responsive compartment of the target cell, but other peptides could
of course be used to target various other enzymes and thus
In addition to hydrogels like the glucose-responsive hydrogels other cell types or compartments of the body using this same
that respond upon detection of a target molecule, hydrogels can basic system.
also be made to respond by being directly acted upon by the target Peptides have also been incorporated into hydrogels as the
molecule. This is most notable in the design and use of enzyme- chemical crosslinker rather than as a pendant linker, thus causing
responsive hydrogels. Among the possible methods for making gel–sol degradation of the hydrogel upon cleavage by the target
enzyme-responsive hydrogels, one of the well-studied methods enzyme. Kim and Healy [194] synthesized poly(N-isopropylacry-
has been to use peptide chains as linkers, either as crosslinks lamide-co-acrylic acid) hydrogels that incorporated QPQGLAK
within the hydrogel or as links between the polymer backbone and peptides as crosslinkers for use as artificial extracellular matrix in
drug molecules. These peptides are typically made of short tissue engineering. The QPQGLAK sequence was selected to be
sequences chosen to be specifically digestible to certain enzymes, specifically degraded by matrix metalloproteinases (MMPs) such
causing degradation of the hydrogel in the presence of the targeted as MMP-13 that are produced by osteoblasts. The peptides were
enzyme. incorporated into the hydrogels directly during the polymeriza-
Kopecek and colleagues have for decades developed hydro- tion: first the sequence was functionalized to have acrylic end
philic polymers based on N-(2-hydroxypropyl)methacrylamide groups by reaction with acryloyl chloride, and then the product
(HPMA) with drugs attached via short oligopeptide sequences was used as a monomer along with N-isopropylacrylamide and
[182–191]. The oligopeptide sequence is chosen to target acrylic acid in a standard chain polymerization reaction. The
cleavage—and therefore release of the conjugated drug into hydrogel exhibits an LCST temperature response, as described
the target site—to a specific enzyme. For example, the GFLG previously, but also is degradable by MMPs. The result is a possible
sequence was selected for cathepsin B-based degradation, artificial ECM that is degradable in the body, but in response to a
thus enabling lysosomal drug delivery [188]. In these studies, biological signal that is found in remodeling bone instead of the

Fig. 9. Reaction of glucose with phenylboronic acid (PBA). Binding of glucose to PBA yields more negatively charged and thus hydrophilic hydrogel, leading to the observed
swelling response.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 19

random hydrolysis that PLA, PGA, or PLGA hydrogels exhibit, thus further responses using transduction pathways, as exemplified in
allowing the hydrogel to act as a scaffold for bone regeneration that the glucose-responsive hydrogels.
will not remain in the body and impede bone recovery, but will
remain sufficiently long for osteoblasts to migrate into the scaffold 5.3.1. Synthesis
and begin bone reformation. The basic synthesis procedure for MIPs is simply described. The
The use of peptide crosslinkers has also been extended to drug molecule to be recognized is incubated with one or more
delivery systems. Glangchai et al. [195] incorporated the GFLGK monomers, chosen to exhibit some form of interaction with the
sequence into nanofabricated hydrogel particles formed through structure of the template molecule, allowing the monomers to bind
step and flash imprint lithography (S-FIL). GFLGK was function- to the template. The bound monomers are then polymerized and
alized using acrylation by acryloyl chloride and then used in the crosslinked into a set polymer, while the monomer units are still
S-FIL photopolymerization along with poly(ethylene glycol) bound to the target molecule. Finally, the template is washed out of
diacrylate to form shape-specific, highly monodisperse nanopar- the polymer, leaving a porous polymer where the functional units
ticles. The GFLGK sequence was chosen to be specifically degraded of the monomers are bound in such a manner as to form a binding
by cathepsin B, and the resulting nanoparticles proved to be site for the target molecule upon further exposure [204]. This
effective at delivering encapsulated plasmid DNA or IgG antibodies forms the typical ‘‘lock and key’’ pairing often mentioned with
upon exposure to the enzyme. regard to antibodies, where the synthetic ‘‘lock’’ is built up around
West and Hubbell [196] had previously worked on similar the ‘‘key’’ of the template molecule.
systems also for use as an artificial extracellular matrix. Their The interaction between the monomer and the template may be
system consisted of telechelic block copolymers of the form BAB, covalent or non-covalent in nature. Non-covalent interactions—such
where A is a PEG chain and B represents a peptide sequence chosen as hydrogen bonding, pi-pi stacking, dipolar interactions, ion
to be degradable by a particular enzyme—APGL was chosen for pairing, and hydrophobic interactions—are more common for
degradation by collagenase, and VRN was chosen for degradation imprinting [205–207]. The use of non-covalent interactions offers
by plasmin. The triblock polymers were then acrylated by acryloyl easy synthesis: no chemical reactions are required, reducing the
chloride and made into hydrogels by UV-initiated polymerization. required amount of knowledge of the target molecule’s properties
The hydrogels were shown to be degradable by the expected and the required level of control over the synthesis, and a small
enzymes and thus potential candidates for assisting in wound number of widely-used and inexpensive compounds, such as
healing and tissue engineering. methacrylic acid, acrylic acid, methacrylamide, acrylamide, vinyl-
One of the primary limitations of using peptides for targeting is pyridine, or hydroxyethyl methacrylate, may be used to achieve
the difficulty making large quantities of synthetic peptides, which target recognition. However, because of the same non-specific
results in high cost. The use of peptides as crosslinkers, rather than as nature of non-covalent interactions that make synthesis straight-
an integral part of the hydrogel backbone, enables targeted forward, this method also yields MIPs that exhibit significant levels
degradability while greatly reducing the amount of peptide required of binding of molecules other than the target, thus limiting
since the bulk of the gel is comprised of some other, cheaper selectivity. This issue can be overcome by using more advanced
biomaterial. Therefore, because of the limiting cost of peptides, the monomer structures that provide better specificity for the target
crosslinking approach is more likely to be clinically relevant since it molecules, although this increases cost slightly [208–211].
provides degradation with limited amount of peptide. Furthermore, Multiple reviews have been published on the detailed
using other known biomaterials as the hydrogel backbone polymer synthesis and evaluation of MIPs [204–207,212–218], so this
allows for multiple response modalities, such as temperature or pH- section focuses on applications. However, it should be briefly
responsiveness along with the degradation pathway. mentioned that successful retention of the formed binding site
It is therefore important to note that many other crosslinking generally requires high levels of crosslinking. At low crosslinking
chemistries are available. While the above work all utilized density, once the template is removed and the covalent or non-
acrylation to directly incorporate the peptides in the polymeriza- covalent interactions are no longer present, the recognition sites
tion reaction, any suitable crosslinking chemistry may be utilized. are free to redistribute through entropic mixing as allowed by the
Michael addition [195], click chemistry, carbodiimide reactions, hydrogel structure, typically to a point where any observed
reductive amination, thiolation and maleimide chemistry, and binding is non-specific due to the loss of the binding-site
many other potential crosslinking chemistries will all serve the geometry [219]. High crosslinking density inhibits this entropic
purpose, although the ideal pathway will be system-specific mixing and thus keeps the geometric requirements for recogni-
depending on what unwanted cross-reactions could occur [197]. tion largely satisfied. Other than this constraint, however, the
synthetic procedures for MIPs are widely varied, and many
5.3. Molecularly imprinted polymers different chemistries can achieve significant target recognition
and specificity.
Molecularly imprinted polymers (MIPs) make use of hydrogels
formed in the presence of a template molecule in order to produce 5.3.2. Diagnostics/sensors
binding sites that will later recognize the target molecule, much The most commonly mentioned application of MIPs is for use as
like how antibodies or aptamers are capable of binding target inexpensive diagnostics or sensors. The need for quick and easy
molecules. MIPs are a promising method for yielding similar detection of biological signals for medicine and dangerous
results as antibodies or aptamers in multiple applications; chemicals for security purposes drives an estimated $52 billion
although they do not currently have as high of specificity and diagnostics market [220], which relies largely on use of antibodies
binding capability, because they are formed from synthetic for sufficient specificity [221]. However, because of the many
monomers, they do not have the disadvantages of high cost, current limitations of antibody selection and production, cheaper
limited ranges of stability (with respect to pH, temperature, and tests that are easier to make, and therefore quicker to market in
enzymatic environment), and limited shelf-life that limit anti- response to new health and safety concerns, are in high demand.
bodies and (to a lesser extent) aptamers [198–203]. MIPs therefore MIPs may offer exactly that.
bear mention as an important and expanding class of chemically- Because of their facile synthesis, MIPs can be quickly and
responsive hydrogels; although the response is at times limited to inexpensively made for most target molecules. MIPs have been
recognition and binding of the target molecule, this can lead to prepared for detection of many targets, such as hemoglobin,
20 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

trypsin [222], cholesterol [223], uric acid [224,225], Hev b1 latex The true capability of MIPs as drug delivery devices is still yet to
allergen [226], TNT [227], perfluorooctane sulfonate [228], theoph- be shown. Feedback-controlled devices would be ideal, where
ylline [229], diazepam [229], atrazine [230,231], morphine [232], highly-selective binding of target molecules would trigger or stop
corticosteroid [233], yohimbine [234], methyl-a-glucoside [235], the release of a molecule such that it is delivered only when needed.
and S-propranolol [236,237], to name a few. However, the advantages of MIPs will only be seen when more
In order to function as a sensor or diagnostic, some transduction traditional systems, like those seen in the glucose-responsive
mechanism is needed to convert the binding action into a hydrogels, are not sufficiently selective. Therefore, MIPs will likely
noticeable and/or quantifiable response. There have been multiple only see commercial use for drug delivery if they can be shown
proposed mechanisms for achieving this. The transducer may be an to offer improved delivery capability and/or control over more
added element to polymer matrix or can be incorporated as part of traditional responsive hydrogels, or in the very select cases where
the polymer itself. great specificity toward a drug over its structural analogs is needed.
Systems with incorporated transducers are capable of reagent-
less sensing, which generally results in faster results and simpler 5.3.4. Sorbents
assays due to the direct nature of generating a signal. Therefore, Along with their potential use as inexpensive, portable, and
integrated transducers are preferable, but unfortunately are easily storable sensors and diagnostics, MIPs will likely find wide
currently limited. Available reagentless technologies include utility as sorbents. The basic purpose of MIPs is to selectively bind a
electrochemical sensing via voltage, current, impedance, or particular molecule (or even cell), so they are inherently useful for
capacitance changes [238–244], fluorescent quenching [245], selective isolation or removal of particular components from a
infrared spectroscopy [246], Raman spectroscopy, and optical or mixture or solution. MIPs have already been used, both academi-
acoustic measurements of changes in mass or refractive index cally and commercially, for various purposes in solid phase
[247–250]. extraction, food processing and purification, biological sample
Addition of elements to the polymer can improve sensor treatment, and environmental analysis [268–272].
sensitivity by providing signal amplification. For electrochemical The primary benefit of using MIPs as opposed to more
detection methods, MIP nanoparticles have been attached to the traditional sorbents used in solid phase extraction is, once again,
surface of carbon nanotubes [225,251–253], gold nanoparticles the selectivity they show for a particular analyte. Traditional
[254], and silver nanoparticles [255], which increases binding sorbents may be chosen to show some selectivity, but inevitably
surface area and provides better electrical properties for stronger end up adsorbing at least trace amounts of various other
signal with lower levels of analyte. For optical methods, addition of components as well. González-Mariño et al. [273] demonstrated
quantum dots has allowed amplification of fluorescent detection, this selectivity by comparing adsorbing capability of a common,
leading to highly sensitive detection assays [256–261]. While these commercially-available hydrophilic balance sorbent (Oasis HLB), a
methods require additional synthesis complexity, the enhanced mixed-mode sorbent (Oasis MCX), and an MIP sorbent for analysis
sensitivity they provide will doubtlessly make the addition of of five amphetamines from wastewater samples. The hydrophilic
transducing elements the primary method of producing sensors balance sorbent performed worst—three of the five analytes could
and diagnostics. not even be measured by LC–MS following extraction—followed by
the mixed-mode sorbent. However, these were significantly
5.3.3. Drug delivery outperformed by the MIPs, which gave the cleanest extracts, the
Although not yet widely studied in drug delivery systems, some lowest matrix effects, the lowest limits of detection, and the
studies have been performed using MIPs to achieve high levels of greatest precision.
control over release of drug molecules [262,263]. Norell et al. [264] Because of this selectivity, MIPs have seen widespread use
first studied use of theophylline-imprinted MIPs for drug delivery through multiple industries [274]. In the field of food processing,
of theophylline. Using MIPs with pre-bound theophylline at levels MIPs have been made for extraction of a wide variety of analytes:
up to 50 mg/g polymer, they demonstrated complete release norfloxacin, enrofloxacin, and ciprofloxacine from milk [275,276];
within 6–10 h, but with only small differences between imprinted tetracycline from fish [277]; chloramphenicol from milk and
and non-imprinted polymers. Ciardelli et al. [265] followed up on shrimp [278]; zeralenone, simazine, atrazine, propazine, fenuron,
this work using nanospheres comprised of poly(methylmethacry- linuron, metoxuron, clortoluron, isoproturon, metobromuron, and
late-co-methacrylic acid) imprinted with theophylline to create tebuconazole from multiple vegetables [279–282]. Biological uses
nanoparticles capable of binding up to 1 mg theophylline/g have focused on primarily on either plasma or urine analysis, for
polymer. This system also allowed for sustained release of many various analytes: alfuzosin [283,284]; cefathiamidine [202];
theophylline—approximately 50% of theophylline was released enrofloxacine and ciprofloxacine [285]; tamoxifen [286]; amoxi-
within 3 h, while 80% was achieved over 7 days. Clearly, the cillin [287,288]; and dopamine [289]. Finally, environmental uses
specificity of binding to the target molecule is the main advantage have focused almost solely on treatment of water, whether from
of MIPs; however, in both of these simple, proof-of-concept lakes, rivers, or sewage, and have likewise focused on various
systems, nothing was demonstrated that could not have been analytes: 17-b-estradiol [290]; simazine, atrazine, propazine, and
achieved through more traditional drug delivery systems. terbutilazine [291]; phenobarbital, cyclobarbital, amobarbital, and
More advantageous use of MIPs has been shown in the phenytoin [292]; diclofenac [293]; and bisphenol A [294].
enantiomeric-specific delivery of drug compounds. The selectivity Although the compounds listed above are quite numerous, that
of MIPs allows for preferential binding of one enantiomeric form of is far from an exhaustive list of the multiple uses of MIPs as
a chiral molecule over the opposing enantiomer, which can be sorbents. Clearly, the selectivity and binding capability afforded by
exploited for selective delivery of selected enantiomers from MIPs as used in hydrogels makes them a highly useful chemically-
racemic mixtures of compounds [266]. For example, an S- responsive material. The ability to bind a particular analyte with
propanolol imprinted MIP made from methacrylic acid crosslinked high specificity using inexpensive materials that are readily stored
with ethylene glycol dimethacrylate was able to selectively deliver for long periods of time enables many practical uses, many of
the S-enantiomer over the R-enantiomer; since the S-isomer has which are yet to be fully realized. There remains much work to be
approximately 100-fold greater activity than the R-isomer, this done in advancing the technology of molecularly imprinted
selectivity can be used to deliver the superior medication without polymers, but such work will indubitably lead to widespread
requiring upstream separation in the manufacturing process [267]. commercial use of MIPs in the near future.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 21

6. Photo-responsive hydrogels unless the gels are collapsed. The relative response of these
hydrogels can generally be well characterized and standardized
The great advantage of photoresponsive polymers is the high to the intensity of light emitted (I0). The other simplification that
level of control that is granted by the stimulus. Photoresponsive hydrogel systems allow for photoreactive systems is the
polymers, commonly, respond to light energy by way of a reaction. relatively low diffusion rates of molecules through the system.
This reaction can be a variety of degradation and bonding This becomes important when considering additive photoreac-
reactions. However, they are unanimously governed by the tions and reversible systems. When a secondary molecule is
Planck–Einstein relation: involved, the rate of diffusion from the activated location can lead
hc to bleeding over in photochemical reactions. However, when the
E ¼ hy ¼ diffusional timescale is limited by the hydrogel’s structure, this
l
effect is limited [298].
where the energy of the light (E) is equal to Planck’s constant (h)
multiplied by the frequency (y). Photo-dependent reaction rates 6.2. Common monomers
have shown strong correlations to the energy and irradiance of
light, with little variation [295]. This, combined with the high Photoresponsive monomers fall into 3 primary categories:
degree of dimensional control of light exposure, makes photo- isomerization, degradation, and dimerization. These three mecha-
catalyzed reactions an attractive mechanism for hydrogel nisms can be incorporated as crosslinkers and as pendant groups.
manipulation. The major downfall with photo-induced changes
comes in in vivo models. Due to the inability of ultraviolet and 6.2.1. Isomerization monomers
visual electromagnetic wavelengths to penetrate tissue, these These materials can be further classified as either a cis–trans
mechanisms are only really viable for ex vivo systems and skin shift or cyclization. One of the more common structures that utilize
level treatments. However, recent advances with near infrared a cis–trans shift in order to exhibit drastic differences in polarity
(IR) wavelength sensitive reactions have opened up some and hydrophobicity are azobenzenes. Azobenzenes have been
possibilities. incorporated as both monomers and cross-linkers into hydrogels
to induce a photo-sensitive phase shift. The cis–trans shift leads to
6.1. Theory changes in stacking efficiency and, in response to polarized light,
can lead to orthogonal bending [299]. The azobenzene systems
Photochemical reaction kinetics depends on a number of also bring reversibility to their stacking, shifting from trans to cis
factors, including standard reaction parameters such as product forms and back in response to 360 and 440 nm light, respectively.
and reactant concentrations, temperature, etc. However, uniquely, The wavelength of light that these systems respond to can be
they depend on a concept of absorbed intensity (Iabs). This is altered by manipulation of the end groups attached to the benzene
defined by the Beer–Lambert law in Eq. (37), for individual and rings [300].
mixtures of absorbing species, respectively [296]. There are other cis–trans reactive groups, however very few of
! " them exist as covalently linked hydrogel networks, due to
I0
log ¼ eCd ¼ e1 C 1 d þ e2 C 2 d . . . (37) availability of double bonds to radical polymerizations. However,
Iabs
they do exist as sol–gel networks. For example, a modified fumaric
where C is concentration, d is depth of penetration, and e is a molar amide has been used to trigger a photo-switching from solution to
absorptivity constant. It is important to note that the molar a gel, and has been shown to contain enzymes and substrates
absorptivity (e) is wavelength and, at high levels, concentration separately until UV irradiation [301].
dependent. However, this does not account for scattering, meaning The other form of isomerization often utilized is cyclization. The
at high concentrations the relationship between absorbed and delicate balance of ring structures caused by ring strain makes
emitted light breaks down. This often translates to ineffectiveness these molecules prime targets for low energy photo-irradiation.
of photoresponsive reactions. The common functional group for these reactions is spirobenzo-
The other major determining factor for photochemical reac- pyran [302]. This group is also unique in that, in the non-cyclized
tions is the overall quantum yield (w). Quantum yield is the form it presents as zwitterionic, with the charged groups being
measure of efficiency of reaction, essentially measuring the rate of consumed during cyclization. This translates into a hydrophilic
reaction with respect to the absorbed intensity, defined in Eq. (38), linear polymer, and a hydrophobic polymer when cyclized. This
where A is the light reactive species. In practice, it is easier to functional group is also interesting, because instead of having on/
determine it from the kinetic constant (k), energy, emitted light off switching, the light source only acts as a catalyst to cyclization.
intensity, and proportionality constant of the wavelength of light. Since the cyclized product is unstable, the cessation of exposure
However, this breaks down in the rare condition of an absorbance will naturally reform the linear starting product. However, there
(Iabs/I0) well above 10% [297]. has been some success, through modification, of developing a
product with a mildly stable cyclized product [303].
#d½A'=dt kNA hc kNA hy
’* ¼ 106 ¼ 106 (38)
Iabs elI0 eI 0
6.2.2. Degradation
This quantity is compared to primary quantum yield (w1), Degradation depends on the breakdown of covalent bonds.
essentially the efficiency of reaction assuming no side reactions. A However, this can be utilized in a number of ways to present
primary quantum yield close to unity means that, ideally, the different functional groups or, in the form of a cross-linker, change
reactant converts absorbed energy entirely into product. This the mesh size of a polymer. The most common group utilized for
occurs only when the energy cannot be quenched by fluorescent photo-degradable moieties is the o-methoxy nitro-benzene family
emission, or converted into other energy forms. The ratio of overall of monomers. The delicate balance of the strong electron draw of
to primary quantum results in a value similar to kinetic chain the nitro group coupled with the electron-donating effect of the
length of polymerizations, as it is the ratio of rate of reactant methoxy group makes this a good leaving group, requiring little
consumption to the rate of initiation. energy to break down the bridge. This, combined with the ease of
In the realm of hydrogels, due to the high water content and, functionalization of the benzene ring, has resulted in a large
inversely, low reactant concentrations, these are rarely issues, number of monomers and cross-linkers that have been utilized for
22 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

photodegradable polymers [304]. The wavelength of light that demonstrates a much higher cell population, as can be seen in
these monomers respond to can also be manipulated by the Fig. 10. This can be used to pattern the cell growth, as the
functionalization of the benzene group, allowing for modification fibroblast cells preferentially proliferate on softer, light exposed,
of the photoresponse, attaining a range of reactivity to light with hydrogels.
wavelengths from 350 to 450 nm, even going as high as 872 nm for DeForest and Anseth developed systems with higher control by
two photon exposure processes [305]. developing hydrogels with dual photo-responses, in the form of a
By including two nitrobenzene groups in one monomer, a degradable cross-linker and a side group for conjugation with
hydrogel can be designed to respond to multiple wavelengths by bioactive groups [295]. The gel was formed with an azide-
the inclusion of a single monomer. In these instances, cleavage of difluorinated cyclooctyne click reaction. Then, by the careful
one nitrobenzene group leads to a significant shift of the inclusion of carbon-carbon double bonds and a nitro benzene
responsiveness of the second. This allows for, potentially, one groups, they were able to pattern in a desired thiolated biomarker
wavelength for the degradation of a cross-linker, and another to while also providing a mechanism to degrade the crosslinked
present a new pendant group with unique properties [304]. A network. The patterning of the thiolated biomarker allows the
number of different cross-linkers have taken advantage of this, hydrogel to express cell compatible biomarkers with high levels of
by combining different chemical bridges between the two control. The degradable nitrobenzene group provided a mecha-
nitrobenzene groups. These have focused mostly on alkyl and nism for mechanical alteration, giving two mechanisms to control
ethylene oxide chains, in order to provide significant space the cell growth in the hydrogel. As seen in Fig. 11, the hydrogel can
between the two reactive groups. However, some aryl ethers even be patterned in 3 dimensional space, providing high levels of
have been utilized in order to combine the two groups. Ethylene control over cell growth.
oxide chains are the most appealing for biological systems, as
they exhibit higher biocompatibilities than their alkyl and ether 6.3.2. Microfluidics
counterparts [304]. Another large field for phtoresponsive materials is in micro-
Other degradation reactions that have been developed are less fluidics, where the ability to actuate flow channels and switches
customizable, but still important. They usually deal with the are necessary for the control of mixing and washing procedures.
removal of a small side group, such as a hydroxyl. Regardless of Photo-reversible hydrogels are crucial for these roles, as they
size, the removal of these groups has been shown to yield in provide a swelling response to plug or open flow channels. Sugiura
significant changes to the chemical structure of the hydrogels. One et al. achieved this by developing spirobenzopyran-functionalized
such group that has been demonstrated to have such an effect is PNIPAAm gels, as described in Fig. 12 [302]. These hydrogel
triphenylmethane [306]. This group responds to light by the systems allowed for real-time pathway opening, allowing for
ejection of a hydroxyl group, forming a carbocation. enhanced flow control based on the irradiated area, as seen in
Fig. 13.
6.2.3. Dimerization
Dimerization and degradation are often both available in 6.3.3. Drug delivery
response to different wavelengths of light. In this sense, the Photo-actuated drug delivery runs into a major issue due to the
previous section was incomplete. Dimerization depends on limited penetration of visible and UV light through skin. However,
the conjoining of two units. Occasionally, these reactions require light controlled degradation does offer an interesting control
the presence of a third species, as a catalyst. A large number mechanism for a drug loaded nanoparticle. Azagarsamy et al.
of these reactions depend on cyclization of two double bonds. demonstrated the high level of on/off control that photo-
Coumarin is a common molecule that provides an effective degradable nanoparticles provide [312]. Griffin and Kasko took
dimerization route. Two coumarin molecules cyclize to form a this work a step further by including multiple therapeutics that
cyclo-butyl ring, and due to intense ring strain, they readily break released in response to different wavelengths of light [313]. They
up in response to ,350 and ,250 nm light respectively [307]. By included three alternatively modified nitrobenzene linkers that
polymerizing an acrylate form of this copolymer into a PNIPAAm connected the drugs into the backbone of the hydrogel. Although
backbone, He et al. managed to make a photo-crosslinkable and each drug does not exclusively release, there is enough difference
degradable hydrogel system [308]. This is a common mechanism in molar absorptivity, as shown in Table 3, to significantly affect
for reversible dimerization; it has also been achieved with other the rate of release of the different therapeutics. The different types
functional groups, such as cinnamylidene acetate [309] and of control are described in Fig. 14.
anthracene modified PEG chains [310].
7. Electrically-responsive hydrogels
6.3. Applications
It has long been known, since work both by and preceding Flory,
6.3.1. Tissue culture that many hydrogels also demonstrate stimuli-sensitivity toward
One of the major contributions that photoresponsive hydrogels electromagnetic fields. Whereas many of the previously-discussed
have made is the exact control they can yield over substrates for stimulus-responsive gels are quite limited in the number of
cell growth. Due to relative narrow ranges of sensitivity, high monomers available for producing the response, responses to an
conversion percent, and the exact three dimensional control that applied electric field can be observed using any polyelectrolyte gel.
can be achieved with lasers, they offer precise control for ex vitro Hydrogels of poly(2-(acrylamido)-2-methylpropanesulfonic acid)
substrate modification. Since cell differentiation depends greatly (PAMPS) [314,315], poly(acrylic acid) [316,317], poly(acrylamide)
on mechanical properties and chemical biomarkers, the high level [316], chitosan and poly(2-hydroxyethyl methacrylate) (PHEMA)
of control allowed for can drastically change the rate of cell [318], poly(dimethylsiloxane) (PDMS) with titanium dioxide
proliferation on a substrate. particles [319], and others have been studied and reported;
One method investigated altering mechanical properties however, stimuli-sensitivity can be observed in hydrogels based on
through the degradation of side groups. By copolymerization any ionizable monomer. Neutral hydrogels are unresponsive on
of 2-nitrobenzyl acrylate and hydroxyethyl acetate, a rigid their own [314], but can become electromagnetically-responsive if
polymer can be formed [311]. The degradation of the nitrobenzyl coupled with dielectric liquids or electrically-responsive particles
group to acrylic acid yields a softer, more wettable polymer that [317,320].
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 23

Fig. 10. Effect of exposure time and degradation on (a) Wettability of poly(Hydroxyethylacrylate-co-2-nitrobenzyl acrylate (p(HEA-co-2-NBA)). (b) Swelling ratio of P(HEA-
co-2-NBA). (c) Elastic modulus of P(HEA-co-2-NBA). (d) Cell concentration in P(HEA-co-2-NBA) with fluorescent imagages of (a) 0, (b) 15, and (c) 90 min exposure times.
Reprinted with permission from Ramanan et al. [311]. Copyright 2010 The Royal Chemical Society.

When a polyelectrolyte gel is placed within an electric field the charge of the polymer hydrogel. This process is well-described
exhibiting a potential gradient, such as by placing between two by the Donnan equilibrium effect [322,323]. Upon application of an
electrodes with an applied voltage, the hydrogel will swell or contract external electric field, the charged network ions and counterions
depending on the charge of the hydrogel [314,321]. This responsive are attracted in opposite directions by electrophoretic forces.
behavior occurs through a combination of Coulombic, electropho- However, the network ions are restricted from individual
retic, piezoelectric, electroosmotic, and electrostrictive interactions. movement because of the crosslinked nature of the hydrogel.
As a result, the magnitude of the response varies based on the charge Therefore, although there is some contraction of the hydrogel
density of the hydrogel, the degree of crosslinking in the hydrogel, based on electrophoresis alone, the magnitude of the response is
the magnitude of the applied voltage, the dielectric properties of limited because the crosslinking prohibits long-range rearrange-
the surrounding medium, and the pH and ionic strength of the ment of the network ions into a more compact form. However, the
surrounding medium, all of which modulate the strength and transport of counterions leads to an osmotic potential that in turn
probability of electric interactions within the hydrogel [321]. leads to electroosmotic movement of water molecules carried
The mechanism of the swelling response has been described as along with the counterions [314]. The net result is an electrically-
follows. Within a polyelectrolyte hydrogel, there are fixed charges induced contraction of the polyelectrolyte hydrogel with the
on the polymer backbone of the hydrogel that, at equilibrium, polymer hydrogel and the water and counterions moving in
attract counterions in the surrounding medium that balance out opposite directions.
24 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 11. (a) exposure sensitivity, (b) 2 D patterning, and (c) 3 D patterning of (1) photo-modified and (2) photodegradeable hydrogels. Reprinted with permission from
DeForest and Anseth [295]. Copyright 2011 Macmillan Publishers Limited.

Many efforts have been made to model this responsiveness. Gong that there is a ‘‘minimum’’ value of W/Ww at which contraction
et al. [314] modeled this contractive behavior in a one-dimensional stops, possibly due to water bound to the macroions by attractive
model using Poisson–Boltzmann and Navier–Stokes equations to dipole forces at very close distance, and that the magnitude of
describe the transport of counterions and water. The modeling response is significantly different between experimental and
yields theoretical equations (Eqs. (39) and (40)) for calculating predicted results.
contraction efficiency (uav) and weight swelling ratio (W/Ww): The discrepancy between model and experiment results
nR o primarily from the simplifications of one-dimensional modeling,
ro
r i ½2pr=rðrÞ'dr which neglects the full impact that crosslinking has within the
uav ¼ (39) hydrogel, and from using free water values for the dielectric
pðro2 # ri2 Þ
constant and viscosity of water, rather than experimentally-
Z t determined values that would be more likely to be observed within
W LðtÞ vav ðtÞdt
¼ ¼ 1# (40) the charged network. Nevertheless, although using only a one-
Ww L0 0 L0
dimensional transport model, this early work highlighted several
where uav [m3/C] is the average gel contraction efficiency, ri [m] is salient features of hydrogel response to an electric field, provided
the radius of the cylindrically-shaped macroion, ro [m] is the support for the proposed mechanisms by which the response was
distance to the midpoint of two adjacent macroions, r(r) [C/m3] is thought to occur, and demonstrated the ability to understand
the local charge density in the gel, W is the weight of the gel at hydrogel swelling dynamics with basic application of electric
time t, Ww is the weight of the gel at t = 0, L(t) is the length of the theory and transport phenomena. Later theoretical studies have
polymer gel at t, L0 is the length of the gel at t = 0, and vav(t) is used similar theoretical treatments to obtain highly accurate
the average value of the water flow. models of the electrical swelling response [324,325].
The above model was compared against experimental data
taken by weight swelling measurements (gravimetry) using 7.1. Applications
poly(2-(acrylamide)-2-methylpropanesulfonic acid) (PAMPS)
gels. Upon application of an electric field, a large potential drop 7.1.1. Artificial muscles
is quickly seen due to the formation of a Helmholtz bilayer and the Ionized hydrogels which show electrically-responsive behavior
gel responds rapidly. As can be seen in Figs. 15 and 16, the model have many potential uses. One of the most exciting and frequently
agrees with experiment in showing that a significant and rapid studied potential applications is as artificial muscles. Because
contraction of the PAMPS gel should occur in response to the many hydrogels exhibit high biocompatibility and similar proper-
electric field, that contraction is faster with higher degree of ties to biological tissues, they are prime candidates for use in tissue
swelling (q), that the response is approximately linearly replacements. Those that exhibit electrically-stimulated swelling
proportional to the quantity of electricity, and that the contrac- or contraction are especially auspicious for use as muscle, since
tion efficiency is inversely proportional to the crosslinking they can mimic the response seen by muscle fibers in response to
density of the gel. Notable deviations from the model include the electrical voltage applied by neurons. Of course, for use as
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 25

Fig. 12. (a) Chemical schematic for swellable photoresponsive micro-well patterning. (b) Photoresponsive microchannel formation schematic. (c) Microvalve photo-actuation
schematic. Reprinted with permission from Sugiura et al. [302]. Copyright 2009 The Royal Chemical Sociery.
26 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 13. Time lapse of micrographs of photo-actuated microchannels. (a), (c), (e), and (g) are irradiation steps. (b), (d), (f), and (h) are subsequent visualizations of latex flow
through the formed microchannels. Reprinted with permission from Sugiura et al. [302]. Copyright 2009 The Royal Chemical Sociery.

artificial muscles, rapid kinetics are necessary, while simulta- immersed in a surfactant solution (n-dodecyl pyridinium chlo-
neously being repeatable, yet exhibiting sufficient material dride), it was demonstrated that the gel could be made to ‘‘walk’’
strength to handle high loads and stresses in repeated use. through a solution by alternately applying and reversing a 20 V
Furthermore, because the range of human movement is varied, at potential at 2 s intervals. The mechanism by which this occurred
times necessitating large movements and at others subtle ones, a was explained as ‘‘a reversible and cooperative complexing of
sensitive and tunable response is also needed. A significant amount surfactant molecules on the polymer gel in an electric field, causing
of work has been published looking to achieve these multiple goals. the gel to shrink’’ and bend, as shown schematically in Fig. 17 and
Hydrogels have been made that respond to the application of an visually in Fig. 18. The benefits of this system are that it performs
electric field with angular movements or linear deformation, or a work as an actuator in a rapid manner with reversible and tunable
mixture of both. Large ranges of angular movements have been response, all while retaining the desirable properties of a ‘‘soft’’
observed, ranging from a few degrees to over 3608 [326,327]. The material.
pioneering work on electrochemomechanical systems was per- Kim et al. [318] expanded on Osada et al. and reported on a
formed by Osada et al. [315]. Using a PAMPS hydrogel with low chitosan/P(HEMA) semi-IPN hydrogel system that exhibited
crosslinking density of N,N0 -methylene bisacrylamide that was controllable angular deformations ranging from 38 to 458,
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 27

Table 3
Molar absorptivities (e) for ortho-nitrobenzene linkers. Apparent rate constants of degradation (kapp, l = 365 nm) for their fluorescein conjugates. Reprinted with permission
from Griffin and Kasko [313]. Copyright 2012 American Chemical Society.

dependent approximately linearly on voltage applied and non- a basic actuator, a faster response would be preferable for use in an
linearly on ionic strength of the surrounding medium. Further- artificial muscle application.
more, reversing the polarity of the voltage reversed the direction of Otero and Cortés [327] reported the synthesis and characteri-
deformation. Unfortunately, the bending response occurred over a zation of a polypyrrole film surrounding a non-conducting film
long time period of 20–40 s. While this is sufficiently fast for use as that exhibited the ability to angularly deform with application of

Fig. 14. Fractional release of rhodamine, AMCA, and fluorescein from a hydrogel as a function of light exposure (l = 436 nm, I0 = 44.6 - 1.0 mW/cm2, t = 5 min; l = 405 nm,
I0 = 21.4 - 1.1 mW/cm2, t = 5 min; and then l = 365 nm, I0 = 5.53 - 0.14 mW/cm2, t = 5 min); solid lines depict predicted release, actual release shown as data points. Reprinted
with permission from Griffin and Kasko [313]. Copyright 2012 American Chemical Society.
28 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 15. Relative hydrogel weight change for various degrees of swelling, q. Top: Fig. 16. Relative hydrogel weight change as a function of quantity of electric flow.
simulated results; Bottom: experimental results with PAMPS hydrogel. ^, q = 25; Top: simulated results; Bottom: experimental results with PAMPS hydrogel.
*, q = 70; &, q = 100; ~, q = 200; *, q = 256; ~, q = 512; &, q = 750. Reprinted with Symbols are the same as in Figure MK3. Reprinted with permission from Gong et al.
permission from Gong et al. [314]. Copyright 1994 American Chemical Society. [314]. Copyright 1994 American Chemical Society.

an electric field while exhibiting tactile sensitivity in measuring Although acrylic copolymer elastomers have been reported for
the work required. The hydrogel achieved angular deformations such a response, exhibiting strains from 233 up to 380%, these
of up to 1088, and was capable of moving objects up to materials require relatively large voltages and often do not have
1200 times its weight. The response time was also rapid, with the softness (low Young’s modulus) expected of a biomaterial
measurements taken after only 10 s of response time. Such [335,336]. Hydrogels based on poly(vinyl alcohol)/dimethyl
systems, including many other systems exhibiting electrically- sulfoxide (PVA/DMSO) or poly(vinyl chloride)/di-n-butylphthalate
responsive angular deformation [328–334], show promise as have shown the ability to exhibit electrically-induced responses of
actuators, and the simplicity of the design is promising for up to 8% strain, as well as bending of up to 1808 within a time frame
application in biological uses. of only 90 ms [337–339]. In fact, Hirai et al. [338] were able to
The other typical response is a linear deformation of the utilize a PVA/DMSO gel to flap a 12.5 cm wing at 2 Hz by periodic
hydrogel—an expansion or contraction as described previously. application of an electric field, demonstrating the capability of

Fig. 17. Schematic of hydrogel bending mechanism by association of surfactant molecules in electric field. Reprinted with permission from Osada et al. [315]. Copyright 1992
Nature Publishing Group.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 29

for applications in drug delivery. Because of the predictable and


highly reproducible contraction, or deswelling, exhibited by
electrically-responsive hydrogels, these systems could make auspi-
cious drug carriers that enable highly targeted drug delivery with
excellent temporal and spatial control. The application of an
electrical field to sites within the body is a bit difficult, however,
and is likely an obstacle that will limit the use of such systems.
Nevertheless, it is not an impossible problem. Topical electric field
application through iontophoresis and electroporation has allowed
for transdermal drug delivery, and would enable electric responses
of hydrogels implanted subcutaneously, allowing for pulsed drug
release over long periods of time [340–343].
As an alternative to external electric field application, Santini
et al. [344] prepared a silicon microchip containing 34 separate
drug reservoirs sealed with a thin layer of gold that dissolves upon
application of a 1.2 V potential. The potential was applied directly
in experiments, but could potentially be performed by the device
itself with incorporation of a battery and an onboard controller
that regulates when each well is released. Because thousands of
wells could be incorporated into such a device, regular application
of a drug over a long period of time could be achieved without
needing an external field. One can imagine multiple potential
improvements that could allow this system to be even more
application specific: the onboard controller could potentially
receive wireless signals from an external device to know when to
release drug if irregular intervals are needed, or could incorporate
some kind of sensing capability to pulse only in response to some
bodily condition. Although this system did not make use of
electrically-responsive hydrogels, it demonstrates that electrical
responsiveness may still be used within the body; the gold films
used for response could easily have been replaced with electrical-
ly-responsive hydrogels to enable a different drug release profile.
Thus, while application of an external field may be a difficult—
though not intractable—challenge, there are potential ways of
using cleverly designed implantable systems with batteries to
enable regular drug delivery without needing externally-applied
electric fields. Further, one can imagine that future advancements
in wireless electricity could also enable simpler application of
external electric fields, opening up the ability to make use of this
well-characterized responsiveness.
The release of drug from electrically-responsive hydrogels can
be effected through multiple mechanisms: diffusion out of a
hydrogel matrix following electrically-induced swelling, which
changes pore size to alter diffusion kinetics; syneresis following
electrically-induced contraction of the hydrogel; electrophoresis
of charged drugs through the hydrogel; electrostatic partitioning of
Fig. 18. PAMPS hydrogel bending and movement from application of electric field. charged drugs; or erosion of the hydrogel complex stimulated by
The front hook and rear hooks can only slide forward along the rail, as prevented by
an electric field [340,345]. In the first of these mechanisms, the
ratchet teeth. Thus, with repeated on/off application of a 20 V electric field and the
bending mechanism shown in Figure MK5, the hydrogel slides unidirectionally porosity of the hydrogel is enhanced in response to the application
forward. Reprinted with permission from Osada et al. [315]. Copyright 1992 Nature of the electric field. This behavior has been reported in ultrafiltra-
Publishing Group. tion membranes by Pasechnik et al. [346], in poly(pyrrole)
membranes by Burgmayer and Murray [347], and in liquid
crystalline membranes by Bhaskar et al. [348].
using such gels as artificial muscles. Although the strain observed More common than drug release by enhanced permeability is
was just 8% in length, this was hundreds of times greater response the forced convection of drug out of the hydrogel following
than observed in conventional ferroelectric materials. Liang et al. electrically-induced deswelling. The drug is carried out of the
[336] expanded on this by demonstrating use of a PVA/DMSO gel to hydrogel with the syneresis of water out of the matrix. Kim and Lee
achieve motility of up to 14.4 mm/s in a path-controlled manner in [349] used this behavior to achieve approximately pulsatile release
air by application of a 400 V/mm electric field. Although this is a of cefazoline and theophylline from anionic poly(vinyl alcohol)/
high potential to be applied, the combination of rapid speed poly(acrylic acid) interpenetrating polymer networks. The ionic
and well-controlled path of the gel shows great promise for use in properties of the drug were shown to have significant effect on the
small bionics. release rate, with the ionic cefazoline showing greater release rates
and greater responsiveness to the magnitude of the applied electric
7.1.2. Drug delivery field than the nonionic theophylline. Therefore, the observed
As with the multiple other responsive hydrogels discussed in this release of the uncharged theophylline indicates that the deswel-
review, electrically-responsive hydrogels have also been explored ling of the hydrogel was responsible for drug release, while the
30 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

greater release of the charged cefazoline shows that electrophore- molecules are more prone to uptake by the convective force and
sis and electroosmosis of charged drugs can also be used for or easily transport through the pores of the hydrogel, while larger
contribute to drug release. molecules require larger forces to move and can be excluded by
Similar results have been obtained in many other studies. small pore sizes [356].
Ramanathan and Block [350] used acetylated chitosan to achieve Hydrogels that erode in response to an applied electric field
electrically stimulated delivery of anionic benzoic acid, neutral have also been synthesized [357,358]. Kwon et al. [357] formed a
hydrocortisone, and cationic lidocaine, in order of delivery rate. complex comprised of poly(ethyloxazoline) and poly(methacrylic
Sawahata et al. [351] used poly(methacrylic acid) hydrogels to acid), which exhibit hydrogen bonding between the oxazoline and
deliver pilocarpine hydrochloride, raffinose, and insulin with well- carboxylic acid groups. Upon application of an electric field,
controlled on-off capability. Importantly, a threshold voltage was electrolysis of water produces hydroxyl ions at the cathode,
observed, below which no drug release was measured; although increasing the pH near the cathode which in turn causes
this value was only about 1.1 V/cm, it will likely prevent small deprotonation of the carboxylic acid groups and resultant loss of
voltages that can occur in vivo from causing premature drug hydrogen bonding capability. The complex therefore disintegrates
release, as sufficient voltage to effect the conformational change into two separate water-soluble polymers, releasing any encapsu-
must be achieved. Sutani et al. [352] used anionic gels made from lated drug as the surface dissolves away. Using insulin-loaded
acrylamide or HEMA, along with hyaluronic acid, to achieve complexes formed into disks and applying a 5 mA current in
electrically responsive delivery of the model drug D-chlorphenir- pulses, this system showed the ability to deliver insulin at a
amine maleate. Yuk et al. [353] used calcium alginate gels with constant rate in pulsatile fashion. This zero-order release profile
poly(acrylic acid) to deliver hydrocortisone, observing enhanced from the disk is characteristic of surface erosion in a slab geometry,
release with increased poly(acrylic acid) content, since the ionized which the disks approximate [359]. Therefore, surface erosion of
carboxylic acid groups allow for enhanced deswelling. the hydrogel enables well-controlled drug delivery by liberating
It is commonly observed that increasing the magnitude of the the encapsulated drug upon erosion.
electric field increases the rate of drug release [350,351,354,355]. The benefit of these erodible systems is that the erosion of the
The trend is generally nonlinear, as can be seen in Fig. 19 [354], hydrogel liberates the drug completely into the surrounding
owing to the ‘‘saturation’’ of the deswelling response, as repulsive medium, rather than requiring transport by diffusion or convection
forces in the gel and entropic mixing forces eventually match the through hydrogel pores, which can severely limit the rate of release
applied forces once the gel is sufficiently collapsed. of large molecules. Defects in the gel matrix may make the release
Another interesting observation is the common occurrence of rate susceptible to wide variations, however. Nevertheless, such
back-flow of the released drug into the gel after release when systems show promise as potential drug delivery depots for long-
attempting pulsatile delivery [351,356]. Typically, the drug is term, pulsatile release of a wide range of both large and small
released when an electric field is applied, per the hydrogel’s molecule drugs.
deswelling response and the resulting syneresis of water and drug In vivo tests of electrically responsive hydrogels have been
out of the hydrogel. Once the electric field is turned off, the performed. Kagatani et al. [360] demonstrated a poly(dimethyla-
hydrogel will return to its equilibrium state. The expansion of minopropylacrylamide) gel loaded with insulin as a subcutane-
the hydrogel creates an osmotic pressure that convectively draws ously implanted drug delivery depot in rats. A schematic of the
the surrounding medium into the hydrogel until equilibrium is electrode system used is shown in Fig. 20. The anode was applied to
established. During this process, some of the drug present in the the abdomen of the rats, and consisted of an acrylic casing with
surrounding medium, after having previously been released from silicon gum, stainless foil, and an electrical wire attached to the
the hydrogel, is brought in with the water (or other medium) by inner surface and saline filling the casing. The cathode was applied
the convective current. Because the process is convective, it is seen to the back of the rats, and was the same setup as the anode with
to occur whether the drug concentration inside the hydrogel is the addition of a foam rubber sponge and cotton in the casing to
greater or lesser than the concentration in the surrounding ensure contact with the skin to maintain a complete circuit. The
medium [340,351]. Furthermore, the size of the drug is important drug-loaded gels were inserted under the abdominal skin by
in determining to what extent this back-flow occurs. Small surgical incision. Direct current of 1.0 mA was applied for 1 min
initially, and for 10 min after 2 h to give pulsed doses of insulin.
The results of Kagatani et al., shown in Fig. 21, showed pulsatile
release of insulin only when current was applied which led to a
noticeable decrease in plasma glucose concentrations [360]. Re-
lease was rapid and well-controlled, with no discernible lag time in
response and no observed leakage of insulin between pulses.
Furthermore, bioavailability calculations showed that the gel
system used in the rats carried sufficient insulin for up to
1670 individual pulses, although this assumes ideal total release,
which could be affected by insulin aggregation or binding within
the gel. Rats in this experiment did not show any visible skin
irritation from the applied electric field, as low current densities
were used. The results of this experiment are therefore quite
encouraging as implantable drug delivery depots for enabling
externally-controlled, pulsatile release of drug compounds over a
long time period, especially with more recent improvements to
iontophoretic devices.

8. Shear stress responsive hydrogels


Fig. 19. Release rate of model drug (edrophonium chloride) as a Function of Electric
Current. Edrophonium chloride was released from a 14 mm P(AMPS/BMA) hydrogel
disk with various electric currents applied. Reprinted with permission from Kwon Stimulus-responsive hydrogels capable of responding to
et al. [354]. Copyright 1991 Elsevier. mechanical perturbations in the environment are classified as
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 31

Fig. 20. Schematic of electrode system used for In Vivo test of electrically-responsive PDMAPAA hydrogel insulin delivery system in rats. Reprinted with permission from
Kagatani et al. [360]. Copyright 1997 Wiley-Liss, Inc. and the American Pharmaceutical Association.

shear-responsive hydrogels. Upon exposure to shear-stress these applications, specific polymer compositions, and the theoretical
hydrogels undergo some conformational change that imparts considerations behind their behaviors will be explored.
either shear-thinning or shear-thickening behaviors to the
polymer network [361,362]. These characteristic responses to 8.1. Theoretical considerations
shear can be harnessed for a wide variety of biomedical
applications ranging from injectable hydrogel networks for drug The mechanical properties of the hydrogel networks will dictate
delivery and tissue engineering to wound repair [363–366]. These their response to applied shear-stress and the applications to
which they are suitable [367]. Therefore, a great deal of emphasis
has been placed on elucidating the mechanisms behind polymeric
and hydrogel network properties and responses to mechanical
perturbation [368–371].
Polymeric systems, such as hydrogels, typically exhibit a
viscoelastic mechanical behavior upon deformation, i.e., a behavior
that is an intermediate between a viscous (liquid) and elastic
(solid) material [370]. When stress is applied, viscous materials
will strain linearly with time and resist shear flow. Elastic materials
will strain with applied stress and return quickly to the original
state once that stress is removed [372,373]. Viscoelastic materials,
exhibiting a combination of viscous and elastic properties, will
strain in a time-dependent manner upon application of a stress.
This time-dependency is due to a complex rearrangement of
molecules that occurs upon a macroscopic mechanical deforma-
Fig. 21. Plasma Glucose Concentration Profile with Insulin Administration by tion [368]. In polymeric systems, the application of stress will
PDMAPAA Electrically-Responsive Hydrogel. Current of 1.0 mA was applied for result in the molecular rearrangement of a portion of the polymer
1 min at t = 0 h and for 10 min at t = 2 h. A noticeable decrease in glucose levels is chain. The resultant movement is called creep. The overall polymer
observed at each time, indicating pulsatile release. *, PBS; &, PDMAPAA gel only; network will remain solid even while the polymer chains
&, PDMAPAA gel with current; ~, insulin-loaded PDMAPAA gel; ~, insulin-loaded
PDMAPAA gel with current. Reprinted with permission from Kagatani et al.
rearrange, or creep, due to the applied stress. This results in an
[360]. Copyright 1997 Wiley-Liss, Inc. and the American Pharmaceutical accumulation of back stress in the material. When the applied
Association. stress and internal back stress reach equilibrium, the material will
32 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

no longer creep. Upon removal of the applied stress, the back stress described by Eq. (44) below:
causes the polymer to recover. This combination of polymer creep,
a viscous behavior, and material recovery, an elastic behavior, deðtÞ
s ðtÞ ¼ EeðtÞ þ h (44)
results in the viscoelastic properties exhibited by polymer dt
networks [368,369]. When exposed to constant stress, the model predicts that the
In addition to the described creep behavior, viscoelastic strain will tend to a constant value, s/E, as time approaches
polymers will also exhibit stress relaxation, wherein the material, infinity. This represents the material deforming at a decreasing
upon being subjected to constant strain, will relax and stress will rate and reaching a steady-state strain. The material will recover to
gradually decrease [374]. These viscoelastic behaviors can be its original undeformed state once the stress is removed. This
modeled using a linear combination of elastic and viscous model, while capable of accurately describing creep, is much less
components, modeled as springs and dashpots, respectively. The accurate at predicting stress relaxation [368,369].
Maxwell, Kelvin–Voigt, and Standard Linear Solid models use The standard linear solid model is a combination of the Maxwell
different combinations of these components to predict the stress model and a spring in parallel. The schematic is shown in Fig. 22
and strain interactions and temporal dependencies in response to and described by Eq. (45) below:
different loading conditions [369].
The elastic component of the polymer network can be modeled de ðE2 =hÞððh=E2 Þðds =dtÞ þ s # E1 eÞ
¼ (45)
as a spring with the elastic modulus, E. The modulus represents the dt E1 þ E2
energy stored in the spring. The corresponding relationship
Upon application of a stress, the material will deform
between stress, s, and strain, e, can then be described by Eq. (41):
instantaneously due to the elastic portion of the strain. After this
s ¼ Ee (41) initial response, deformation will continue and approach a steady-
state strain, which represents the viscous portion of the strain. This
The viscous components of the polymer network are modeled as
model, although more accurate than both the Maxwell and Voigt
dashpots, or mechanical dampers that resist perturbation due to
models, inadequately predicts the strain under certain loading
friction from a viscous force. The dashpot dissipates energy at a
conditions. Furthermore, it is much more difficult to handle
rate relative to the viscosity of the modeled system and can be
mathematically than the other models [369,374].
modeled by Eq. (42):
Experimental determination of the creep and stress relaxation
de moduli can be readily obtained. For creep determination, the
s¼h (42) material of interest is exposed to steady uniaxial stress s0 and the
dt
time-dependent strain e(t) = d(t)/L0 is measured [375]. The ratio of
where h is the viscosity of the material, s is the stress, and de/dt is
time-varying strain to applied constant stress provides the creep
the strain rate.
compliance:
The Maxwell model is represented by an elastic spring and
viscous dashpot connected in series, as shown in Fig. 22 and eðtÞ
described by Eq. (43) below: C cr p ðtÞ ¼ (46)
s0
detotal s 1 ds For stress relaxation determination, a steady strain is applied to
¼ þ (43)
dt h E dt the material and the time-dependent stress is measured [375]. The
The model, under constant stress, has two strain components. relaxation modulus is the ratio of time-varying stress to the
The first is attributed to the elastic behavior of the polymer system applied constant strain as described below:
and occurs instantaneously upon application of stress. The second s ðtÞ
is due to the viscous behavior of the polymer, and increases in a Erel ðtÞ ¼ (47)
e0
time-dependent manner after onset of stress. In constant strain
conditions, the model accurately predicts that stress decreases While insights into the creep and stress relaxation of polymeric
exponentially with time when put under constant strain. However, species are incredibly important, the limitation is on the time scale
it does not capture creep behavior accurately, as it incorrectly of analysis. Creep and stress relaxation tests study the material
predicts that strain increases with time instead of the observed responses at longer time scales, ranging from minutes to days.
trend of strain rate decreasing with time [369,372]. Dynamic tests, which assess the response of the polymeric network
The alternative Voigt model places the viscous dashpot and to sinusoidally applied stress or strain, allow for elucidation of
elastic spring in parallel, instead of in series (Fig. 22), and is material responses at much shorter time scales [375]. This
experimental method, known as small amplitude oscillatory shear
(SAOS), is a small deformation rheological technique carried out
within the linear region of a polymer network [376]. In this linear
viscoelastic region, the hydrogel properties are independent of the
magnitude of applied stress or strain [371,377,378].
In practice, either a sinusoidal shear strain or shear stress
is applied at an angular frequency, v, and described by
g(t) = g0(sin(vt + d)) or t(t) = t0(sin(vt + d)), respectively. The mea-
sured shear stress or shear strain is a phase-shifted sine wave,
described by either t(t) = t0(sin(vt + d)) or g(t) = g0(sin(vt + d)). In a
purely elastic material, the stress and strain curves are completely
in-phase with one another, i.e., the phase angle, d, is zero. Conversely,
in a purely viscous material, the two curves are out of phase by
908. As expected, a viscoelastic material, having intermediate
Fig. 22. Models for linear viscoelastic polymer systems (top) Maxwell (bottom left)
properties between a purely elastic and viscous material, will result
Kelvin–Voigt and (bottom right) Standard linear solid. Springs represent the elastic in an observed phase angle somewhere between 08 and 908
component and dashpots represent the viscous components of the polymer system. [367,370,371,377].
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 33

The relationship between the oscillating stress and strain can be hydrogels that have utility for drug delivery and tissue engineering
described by the complex modulus, G, which can be resolved into applications shear-thin for injectable delivery [387,388]. Therefore,
two components, G0 and G00 , it is important to consider the polymer properties in the non-linear
regions.
G ¼ G0 þ iG00 (48) There are a number of experiments that can probe the non-
linear viscoelastic response of polymer networks; two powerful
G0 , the storage modulus, is the real or in-phase component of techniques are the step shear rate test and large amplitude
the complex modulus. It measures the stored deformation energy oscillatory shear (LAOS).
during applied shear and, therefore, represents the stiffness or The step shear rate experiment measures the viscosity, h, of a
elastic contribution of the material. The loss modulus, G00 , is the polymer network as a function of shear rate, ġ , by applying a
imaginary or out-of-phase component of the complex modulus. specific shear rate in a step-wise manner until the system reaches
This modulus captures the energy dissipation during applied shear, steady state. With the provided information, the polymer network
i.e., the liquid-flow or viscous response. The magnitudes of G0 and can be deemed as shear-thickening, where dynamic viscosity
G00 , and the ratio between them, describe the elastic versus increases with shear rate, or shear-thinning, where dynamic
viscous character of a viscoelastic material. Specifically, if G0 > G00 viscosity decreases with strain rate. Shear thinning is predomi-
the material behaves more like an elastic solid. Conversely, if nately attributed to the alignment of polymer chains or micro-
G00 > G0 , the material will behave more like a viscous liquid structures under flow, ultimately leading to decreased drag or
[370,371,374,378]. friction, and a reduced viscosity [382]. Shear-thickening, con-
The gelation kinetics of hydrogel systems can be determined by versely, occurs when an ordered system, such as a colloidal
monitoring the temporal dependence of G0 and G00 . For most solution, becomes disordered in response to applied shear. This
systems, the curves for G0 and G00 intersect as time increases and the disorder results in an increase in viscosity, hence the shear
magnitude of G0 surpasses that of G00 . This indicates the transition of thickening behavior [389,390]. Shear-thinning or thickening
the material into a robust gel from an initial liquid-like state behaviors affect how rapidly a polymer solution can be processed,
[367,379]. This so-called ‘‘cross-over’’ point can also be observed which is essential for industrial processing techniques.
for temperature-responsive gels, where the sol–gel transition is However, there are a number of limitations of this experimental
captured at the temperature, not time, where the G0 and G00 curves technique that limit its utility. Specifically, it is unable to capture
intersect [380]. the polymer response to high deformation at short time scales
After gelation, gel stiffness can be observed over long and short because the system has yet to reach a steady state. The elastic
timescales by modulating the frequency of the oscillating shear response of the viscoelastic material is also lost because the
stress or strain and measuring the corresponding G0 and G00 values. polymer cannot be observed in its dynamic state. Finally, and most
The gel may act very differently at high frequency, or short detrimentally to the investigation of the material properties of
timescales, versus low frequency, or long timescales [367]. To hydrogel systems, the step shear rate experiment will not work for
elaborate this point, the structures within a gel at short times (high chemically crosslinked networks or networks held together by
frequency) have not had time to respond to applied force and, delicate hydrogen bonds, which are responsible for a significant
therefore, appear stiff and elastic. That same gel at long times (low portions of physically crosslinked gels [8,388]. To overcome this
frequency) has had time to respond and adjust, resulting in a particular limitation, a strain- or stress-controlled rheometer
viscous material that can flow and rearrange [381]. The crossover operated with the motor in steady mode can capture the viscosity
frequency denotes at which frequency or timescale the gel as a function of shear rate for crosslinked materials, but the
transitions from being predominately elastic (G0 > G00 ) to predomi- response to high deformation at short time scales is still lost [391].
nately viscous (G00 > G0 ). The final gel stiffness will depend on the LAOS is very similar to the previously described SAOS
ratio between G0 and G00 at long time scales [382]. experiments used to obtain G0 and G00 in the linear viscoelastic
As emphasized by Yan et al., both the gelation kinetics and the region of polymer systems. However, instead of delivering small
final gel stiffness are essential material parameters when amplitude oscillations to the system in order to exclusively probe
determining the ultimate success and utility of a hydrogel the linear region, the amplitudes are increased and the polymer
formulation for biomedical application. Specifically, these param- moves into the non-linear region of its behavior. Compared to the
eters will impact the ability of the hydrogel matrix to homo- step shear rate experiment, LAOS requires no sudden or drastic
geneously suspend therapeutic cargo, promote cellular migration, change in speed and does not need the polymer system to settle to
maintain cell populations, and preserve its mechanical properties a steady-state position before obtaining the data of interest.
[365,367,383,366,384]. Additionally, and most importantly, both the strain amplitude and
Exploration of hydrogels and polymers within the linear frequency can be controlled to investigate different properties and
viscoelastic region, where the properties are independent of the systems of interest [382,386,388]. The imposed shear strain and
magnitude of shear amplitude, clearly provides valuable insight the corresponding shear rate in LAOS are the sinusoidal curves
into polymer behavior. This linearity, at a molecular level, is g(t) = g0sin(vt) and ġ ðtÞ ¼ g 0 cosðvtÞ, respectively. Due to the
attributed to the presence of molecular entanglements whose periodic nature of the applied shear strain, Fourier analysis can be
destruction by flow is balanced by their formation due to Brownian completed on the collected data, which allows for the filtering of
motion. When the net change in the number of entanglements is noise and artifacts [385]. The Fourier decomposition of LAOS non-
zero, the material exhibits linear viscoelastic behavior. Once this sinusoidal stress data provides a series containing only odd
balance is eliminated, the material will exit the linear region and harmonics (shown in Eq. (49)):
enter into non-linearity [381,385].
These non-linearities in polymer response typically appear X
1
t 12 ¼ s ðtÞ ¼ s j sinð jvt þ d j Þ (49)
when the amplitude of applied shear is large or when material j¼1
properties change due to deformation, such as in shear or strain-
thinning and shear or strain-thickening systems, i.e., in cases Only odd harmonics are considered in this analysis because the
where the net change in molecular entanglements no longer equals even harmonics provide no valuable information to the stress
zero [386]. In industrial applications, polymers are typically output having been attributed to noise, secondary flows, slip, and
exposed to large deformations during processing and many transient responses [387]. The higher odd harmonics, j = 3 and
34 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

above, contribute to the non-sinuosidal stress responsive curve in of the stress response curve to a sinusoidal shear strain
non-linear viscoelastic materials [385,387]. For the linear case, the deformation (as shown in Fig. 23) [387,388,392]. Therefore,
amplitudes of these higher harmonics are all zero and the above instead of defining the bulk material viscoelasticity, G0 and G00
expression will still hold true. Furthermore, s1 is linear for a given are specific to the particular experiment that was performed
strain amplitude g0 and the parameters G0 , G00 , and d1 are all [387]. If measured carefully at a fixed frequency, however, G0 (g0)
independent of g0 [385]. and G00 (g0), can provide useful information about the character-
In the nonlinear viscoelastic case, the stress response, sj(g0,v), istics of a gel within the non-linear region [385,388].
and the phase difference between the stress response and applied Four different types of non-linear strain-dependent behavior
shear strain, dj(go,v), are determined from discrete Fourier emerge upon exploration of G0 (g0) and G00 (g0): (1) strain thinning,
transforms. Notably, they both depend on the strain amplitude (2) strain hardening, (3) weak strain overshoot, and (4) strong
and frequency of the applied strain, which solidifies their non- strain overshoot. Strain thinning occurs when G0 and G00 decrease
linearity [382]. From the first harmonic of the stress response with increasing strain amplitude. This behavior is commonly
curve obtained from discrete Fourier transform, the non-linear observed in polymeric systems and can be attributed to the
viscoelastic properties can be adequately described [388]. The alignment of polymeric chains in the direction of flow, much like
viscoelastic moduli, G0 and G00 , specifically, can be defined by the shear thinning [381,382,388,393]. Strain hardening, or shear
following expression: thickening, behaves in the exact opposite manner, with G0 and
X G00 increasing with increasing strain amplitude. The resistance to
s ðtÞ ¼ g 0 ½G0n ðv; g 0 ÞsinðnvtÞ þ G00n ðv; g 0 ÞcosðnvtÞ' (50) flow alignment occurs due to the formation of microscale
n;odd
complexes which prevent the orientation of the polymer chains
This expression captures the portions of the stress response that with the applied flow [382,388–390]. Weak strain overshoot
are in phase (sin) and out of phase (cos) with the applied large describes a system whose loss modulus, G00 , experiences a local
amplitude sinusoidal strain. While viscoelastic parameters G0 and maximum before decreasing along with the storage modulus, G0 .
G00 are readily calculable and fully describe the material response in This behavior is likely due to polymers with weak complexes that
the linear region, this does not hold true in the non-linear case associate upon the initiation of strain (initial increase in G00 ) before
[382]. This breakdown occurs due to the dependence of G0 and G00 being destroyed at higher strain amplitudes (subsequent decrease
on strain amplitude, G0 (g0) and G00 (g0), which results in a distortion in G00 ) [382]. This behavior is extremely common in soft glassy

Fig. 23. Within the linear viscoelastic region (bottom left) the storage and loss moduli, G0 and G00 , respectively are independent of the applied strain amplitude. The resulting shear
stress curve is, therefore, sinusoidal (top left). However, once entering the non-linear region (bottom right), G0 and G00 become dependent on shear amplitude, i.e. G0 (g0) and G00 (g0),
and the shear stress curve is no longer sinusoidal (top right). This illustrates the different behaviors expected from small amplitude oscillatory shear (SAOS) and large amplitude
oscillatory shear (LAOS) experiments. As a note, the distortion of the stress curve in the non-linear region can be attributed to the higher harmonics of G0 and G00 .
Reprinted with permission from Hyun et al. [388].
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 35

systems, block copolymers, and xanthan gum solutions, and, (1) Biocompatibility of the hydrogel network, its precursors, and
therefore, of interest with hydrogel systems [382,388]. The final any degradation products.
type, strong strain overshoot, exhibits local maxima for both G0 and (2) Reasonable force required to induce shear flow.
G00 . Thus, it is an intermediate between strain-hardening, where G0 (3) Ex vivo gelation and in vivo recovery after shear must occur
and G00 both increase, and weak strain overshoot, where G00 has a rapidly and under physiological conditions.
maximum but G0 exclusively decreases. This response is attributed (4) Hydrogel network structure must be tunable for the desired
to polymers with stronger associations than those seen in type (3), biological functionality.
but weaker than those observed in type (2): most likely, (5) Mechanical stability for the entire duration of its use.
hydrophobic interactions or micellar clusters [388]. (6) Controlled biodegradability.
Numerous mathematical and experimental methods are used
to calculate these parameters and have been thoroughly reviewed The benefits of injectable hydrogels, as opposed to surgically
by Hyun et al. [388]. However, it has been argued in literature that inserted systems, are numerous and include their non-invasive
the first harmonics of the storage and loss moduli at a given strain implantation, ability to tightly and completely form to a cavity, and
amplitude, G1’(g0) and G100 (g0), are sufficient to describe the an ability to easily and homogeneously incorporate therapeutic
material [394]. This is because the non-linear stress response curve molecules and even cells [365,366,384,400]. In situ forming
deviates no more than 15% from its purely sinusoidal linear hydrogels are injected and undergo a sol–gel transition upon
counterpart due to contributions from higher harmonics [388,394]. entering the body cavity due to a change in pH [401–404],
So, while complex calculations of the non-linear viscoelastic temperature [403,405,406], or ion concentration [407–409].
properties, G0 (g0) and G00 (g0), can indeed be useful, it is more Shear-thinning hydrogels, when compared to in situ forming
important to investigate the polymer network at a variety of strain hydrogel systems, have several advantages. The shear-thinning
amplitudes outside of the linear range to gain valuable insight into hydrogels undergo a sol–gel transition ex vivo, which allows for
its rheology and overall behavior. complete and meaningful characterization of the system proper-
Both linear and non-linear viscoelasticity are necessary to ties prior to injection [365]. Furthermore, the gelation conditions
consider when completing a rheological analysis of a hydrogel can be tightly controlled and are not subject to the variability
system, particularly those that exhibit any type of shear-responsive experienced in vivo [410]. Additionally, the shear-thinning
behavior. The ability and extent to which a hydrogel responds to an hydrogels have been shown to regain their physical properties
applied shear can affect not only the application of, but also the after injection more rapidly than in situ forming gels and are far less
ultimate success of an engineered hydrogel system. The remainder likely to leach into neighboring tissues [404,411–413]. The shear-
of this section will be dedicated to providing several examples of thinning hydrogels appropriate for injectable delivery for drug
shear-responsive hydrogel systems, with special focus on the delivery and tissue engineering can be composed of a multitude of
mechanism by which the shear-response is conferred and the components ranging from naturally derived proteins to engineered
utilization of viscoelastic parameters to characterize the response. peptides to synthetic polymer species and colloidal systems.

8.2. Shear-thinning hydrogels 8.2.1.1. Natural protein-based hydrogels. Naturally derived protein-
based hydrogels are potentially excellent candidates for biomedical
Shear-thinning hydrogels exhibit decreased viscosity as a applications due to their intrinsic biocompatibility, biodegradabili-
function of increased applied shear force. These systems must be ty, and desirable mechanical properties [414,415]. Fibrous proteins,
robust prior to the introduction of shear, flow or thin during the such as silk, possess mechanical properties far surpassing other
application of shear, and recover their mechanical properties, i.e., naturally- and synthetically-derived polymers due to repetition in
elastic moduli, after the cessation of shear [365,367]. This their primary structure resulting in a very consistent and homoge-
behavior is predominately observed in self-assembled polymeric neous secondary structure that yields incredibly strong yet flexible
systems of both natural and synthetic origin. In self-assembled materials [416–418]. Silk fibroin, the main protein component in
systems, the forces that favor assembly (hydrophobic interac- silk, will, at high enough concentrations, self-assemble into a
tions, hydrogen bonding, and electrostatic interactions) overcome hydrogel that is composed of b-sheet-rich fibrils [419,420]. The
the forces opposing assembly (electrostatic repulsion and presence of b-sheets in the hydrogel network directly impacts the
solvation) to form stable, physically cross-linked networks material strength and biodegradation profile resulting in a strong
[395]. However, these forces are inherently weak and can be and tough hydrogel capable of being degraded over long time
disrupted during the application of a substantial shearing force, periods by enzymatic activity [419,421,422]. Additionally, silk
which allows the network to flow or thin. Once the external shear fibroin, its resultant silk hydrogel, and degradation by-products have
is removed, the hydrogel network can reassemble and recover been shown to be cytocompatible and non-immunogenic [421,423].
its structure, i.e., self-heal [365,395]. The kinetics of the shear- Modulating temperature, pH, protein concentration, and ion
thinning and self-healing processes, as well as the mechanical concentration can control the gelation kinetics and mechanical
properties both before and after shear, will determine the ability properties of the silk fibroin hydrogels. Specifically, gels with
of the hydrogel to be utilized for biomedical applications such higher compressive strength and moduli were achieved at
as injectable hydrogel systems for drug delivery and tissue increased temperature and initial protein concentration. Increased
engineering, wound healing, osteoarthritis treatment, and bio- gelation rate is triggered by increased temperature, decreased pH,
film disruption on implanted biomaterials [365,383,366,384, increased Ca2+ concentration and increased silk fibroin concentra-
396–398]. The various applications and types of shear-thinning tion [419,424,425]. However, the gelation kinetics achieved by
hydrogels, as well as the respective mechanisms by which they modulating the previously mentioned parameters were still not
shear thin, will be discussed in detail. rapid enough to achieve homogeneous cell encapsulation at
physiological conditions. A sonication technique developed by X.
8.2.1. Injectable hydrogel systems Wang et al. resulted in silk fibroin hydrogels capable of gelling
The ability of shear-thinning hydrogels to flow under applied within 30 min to 2 h by initiating the formation of b-sheet-
shear stress makes them attractive candidates for injectable containing fibers via spinning. This acceleration of the gelation
systems. The requirements of injectable systems are as follows process resulted in human bone marrow derived mesenchymal
[365,384,399,400]: stem cells (hMSCs) being successfully and homogeneously
36 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

incorporated into the hydrogel matrix and capable of being [367]. An alternative to naturally derived proteins is the utilization
sustained for 21+ days [426]. of synthetic polypeptides that mimic the structure and function of
Yucel et al. expanded upon this previously developed sonication the natural materials. These recombinant protein hydrogels can be
technique to form silk fibroin hydrogels capable of exhibiting engineered to not only include the necessary primary sequences to
shear-thinning behavior. Briefly, silk fibroin was vortexed, instead induce the desired secondary structural characteristics, such as a-
of sonicated, which initiated the formation of b-sheet-containing helices and b-sheets, but also to contain functional epitopes to
fibers, and enhanced the self-assembly of the hydrogel network. incorporate biological functionality and activity. The peptide
The gelation kinetics were controlled by varying the duration of sequence can be readily manipulated to form hydrogel networks
vortexing, assembly temperature, and/or the initial protein with the properties required for the specific applications of interest
concentration. Most importantly, these hydrogel networks exhib- [367,414,429].
ited shear thinning at high enough shear strain amplitudes and Protein hydrogels composed of engineered polypeptidic
recovered their G0 to near pre-strain values immediately after sequences based on leucine zippers have shown promise as
cessation of shear (Fig. 24). The shear-thinning behavior was shear-thinning materials. Leucine zippers are responsible for DNA,
attributed to the temporary release of intercluster dangling chain protein, and transcription factor linking within the cell [365,430].
entanglements, which allows large b-sheet-rich clusters to slide They contain a seven-residue heptad peptide sequence, (abcdefg)
past one another. This mechanism is proposed as an alternative to motif, which repeats over the length of the polypeptide. The ‘a’ and
the rupture of permanent, intermolecular b-sheet crosslinks, ‘d’ positions of the repeating sequence are hydrophobic in nature,
which was deemed unlikely due to the very rapid recovery of typically leucine, while the ‘e’ and ‘g’ residues are charged species.
material post-shear [427]. These shear-thinning silk fibroin gels The amino acids assemble into a helical conformation (Fig. 25) with
have great promise as injectable tissue engineering scaffolds for the hydrophobic moieties relegated to one side of the helix
the homogeneous administration of viable cells [427,428]. [415,429,431]. The coiled-coil dimers are formed as a result of the
hydrophobic interactions between the two (abcdefg) helices but
8.2.1.2. Synthetic protein-based hydrogels. While naturally derived are stabilized due to the pH-dependent interaction of the charged
protein-based hydrogels have shown great utility for tissue species [365,415,430,432]. The inclusion of responsive moieties
engineering and drug delivery applications, there are some into the a, b, e or g positions can confer environmental-
limitations associated with their use. Specifically, they are derived responsiveness to the peptidic structures, resulting in the self-
from natural sources and, therefore, suffer from a lack of batch-to- assembly of the sequences into nanostructures under desired
batch consistency with regards to their material properties physiological conditions [367,431]. These nanostructures can

Fig. 24. (a) and (b) depict strain-sweeps from vortex-induced fibroin hydrogels with varying silk concentrations. The arrows depict yielding/thinning behavior. (c) and (d)
show frequency sweeps before (open symbols) and immediately after shear-thinning by injection (closed symbols).
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 37

block composed of either poly(L-lysine) or poly(L-glutamic acid)


salts and a hydrophobic block consisting of either poly(L-leucine)
or poly(L-valine) [442]. The self-assembly was governed by
the hydrophobic and electrostatic interactions between the
different blocks on the copolymer. Parameters including sol–gel
transition behavior and elastic modulus could be controlled by the
composition of the charged block and hydrophobic to hydrophilic
ratio, respectively. Upon undergoing shear deformation, the
Fig. 25. A schematic of a coiled–coiled dimer containing the (abcdefg) motif. The ‘a’ hydrogels were capable of recovering their original properties to
and ‘d’ residues are typically hydrophobic amino acids, while the ‘e’ and ‘g’ residues
80–90% within a rapid period of time. The shear-thinning behavior
are charged moieties. The dashed lines represent hydrophobic and charge
interactions resulting in the coiled-coil structural motif. was attributed to the transient disruption of the interconnections
Obtained and modified from Jonker et al. [415]. between gel domains, as opposed to the complete disassociation of
the hydrophobic and electrostatic associations [442–444]. The
high concentration of a-helices within the polymer network due to
further assemble into hydrogel network materials upon the the presence of leucine also aided in the shear-thinning and self-
inclusion of appropriate amino acids into positions ‘b’, ‘c’, and ‘f’ healing behaviors, as the associations between the a-helices could
or by forming block copolymers containing a coiled-coil motif such be readily disrupted and reformed with rapid kinetics [445,446].
as the leucine zipper [413,415,432–439]. The amphiphilic diblock copolypeptide hydrogels have shown
B.D. Olsen et al. formed a triblock copolymer composed of promise as tissue engineering scaffolds for tissues in the central
coiled-coil end blocks (P) joined by a nonapeptide linker of various nervous system. They readily undergo shear-thinning during
length (Cx), forming PC10P with 10 nonapeptide repeat units or injection, reform rapidly in vivo and, most importantly, show
PC30P with 30 nonapeptide repeat units (Fig. 26). Either PC10P or minimal cytotoxicity and immunogenicity while still encouraging
PC30P protein was hydrogelated at pH 7.6 and 4 8C for 2–4 h. For interaction with the existing brain tissue [447].
gelation to occur, the coiled-coil domains formed into pentameric Another class of synthetic polypeptide hydrogels are the so-
bundles via hydrophobic and ionic interactions, which function as called MITCH, or mixing-induced, two-component hydrogel sys-
physical crosslinks to maintain the integrity of the hydrogel network tems [448]. The two separate components assemble via molecular
structure. Upon exposure to shear force, these systems reveal strong recognition after mixing at physiological conditions. The MITCH
shear-thinning behavior and very rapid self-healing kinetics. When system developed by Foo et al. consists of
sheared in physiological conditions, the original modulus was
recovered in less than a minute. Changing the midblock length and (1) WW domain consisting of preserved tryptophan residues
the endblock composition modulated the final gel properties and linked together with an RGD-rich hydrophilic spacer [449,450].
shear-thinning capabilities [440]. The PC10P hydrogel recovered to (2) Proline-rich protein domain linked together by a hydrophilic
98% of its original elastic modulus within 1 min, whereas the PC30P spacer.
exhibited a small permanent decrease in the final modulus but did
recover after shear. This shear thinning behavior is attributed to the The intracellular WW domain interacts with the proline-rich
coiled-coil end block interactions and the yielding that occurs within protein with micromolar binding affinities resulting in a physically
the bulk of the material [413,441]. CHO K1 cells were encapsulated crosslinked gel (Fig. 27) [451,452]. The binding strength and
within the hydrogel network prior to gelation and subsequently specificity can be modified by adjusting the frequency of the
injected through a 22-gauge needle to assess the utility of this repeated domains (either tryptophan or proline) within each
network as an injectable system. Not only did the hydrogel shear segment. This adjustment of binding strength at the molecular
thin while undergoing injection, but over 95% of the cells survived level can dramatically impact the bulk viscoelastic properties of the
post-shear [413]. hydrogel. For example, the G0 values can range from ,9 to 50 Pa.
Another type of synthetic poly-peptide hydrogel capable of Furthermore, by simply increasing the binding affinities between
undergoing shear thinning are di-block copolymers. These the two components, the duration required for the polymer to self-
copolymers are amphiphilic in nature, with a charged hydrophilic heal decreases from ,30 min to less than 5 min [448].
Unlike many of the other protein-derived self-assembling
systems, the MITCH systems undergo a sol–gel transition without
the need of environmental changes in pH, temperature, or ion
concentration. For this reason, they are particularly attractive for
tissue engineering applications that require cell encapsulation, due
to the ability to gel under physiological conditions. Thus far, adult
neural stem cells (NSC), PC-12, HUVEC, and adipose-derived stem
cells have been successfully encapsulated within the MITCH gels
and remain physiologically active [365,415,453].

8.2.1.3. Peptide-based hydrogels. Peptide-based hydrogels are


composed of short amino-acid sequences that can self-assemble
into a hydrogel network. The mode of self-assembly, material
properties and biomaterial functionalities can be engineered into
the peptide sequence by using both naturally- and synthetically-
derived amino acids. These amino acids impart the desired
behaviors through an array of non-covalent interactions such as
hydrogen bonding, electrostatic interactions, and hydrophobic
interactions. Certain amino acids are even capable of inducing
Fig. 26. Network formed with a tri-block copolymer consisting of coiled-coil
forming end blocks and a polyanionic linker. temperature or electrochemical responsive behaviors into the
Reprinted with permission from Olsen et al. [413]. peptide sequence of interest [415,454,455].
38 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 27. MITCH schematic showing (top left) the WW association domains, CC43 and Nedd.3, and the proline peptide (PPxY) and (bottom left) hydrophilic spacers linking
repeat units of either the WW domains or proline peptides. (Right) The two-component gel results from the mixing of the WW association domains and the respective proline
peptide chains.
Reprinted with permission from Foo et al. [448].

One such family of peptides, called ‘MAX’ peptides, self- rigid, fibrillar hydrogels. The presence of physical crosslinks in the
assembles into b-hairpin structures that form hydrogel networks form of hydrophobic interactions and hydrogen bonding stabilize
capable of shear thinning behavior. Developed by Pochan and the hydrogel but also allow for shear-thinning upon the introduc-
Schneider, these MAX peptides have been studied for well over a tion of a shearing force (Fig. 28) [411,458,462,463]. The manipula-
decade [455–461]. The first generation peptide, dubbed MAX1, tion of the peptide sequence, concentration, ionic strength, and
consisted of two b-strands composed of alternating hydrophilic temperature can result in hydrogels exhibiting vastly different
lysine (K) and hydrophobic valine (V) residues linked by a tetra- porosities, moduli, and degradation kinetics [410]. For example,
peptide type II’ b-turn spacer, i.e., (VK)4-ValD-Pro-Pro-Tyr-(VK)4 MAX8, a newer generation peptide in the MAX family, substitutes a
[455]. The lysine residues, when protonated, prevent any lysine residue at position 15 with a glutamic acid residue. This
aggregation or self-assembly due to electrostatic repulsion simple modification results in a much more rapid gelation process
[455,458]. The lysine charge can be neutralized by changing the and a stiffer gel structure [411]. Because of the easily modifiable
pH of the environment [455], shielded by increasing salt peptide sequence, the gel properties can be tailored for a given
concentration [458], or collapsed by increasing temperature application, such as ensuring homogeneous distribution of a
[456]. The neutralization, collapse or shielding of the lysine charge therapeutic for drug delivery or cell cargo for tissue engineering
results in a conformational change within the peptide sequence [367,410,464].
and triggers gelation. The process of gelation starts by the MAX1 and MAX8 peptides and those self-assembled systems
rearrangement of the peptide sequence into b-hairpin structures that rely on b-hairpin and fibril formation are highly susceptible to
exhibiting a hydrophilic and hydrophobic face. These b-hairpins shearing forces [365]. Interestingly, when the MAX1 and MAX8
then assemble into fibrils, which interact and entangle to form hydrogel networks were probed during steady shear flow, there

Fig. 28. (Top) Self-assembly of b-hairpin peptide and their subsequent assembly into fibrillar hydrogels. Also pictured is the reaction of the network to the application of shear
force. (Bottom) Proposed mechanism of the network behavior during shear-thinning and recovery, or self-healing.
Reprinted with permission from Guvendiren et al. [365] (Top) Haines-Butterick et al. [411]. (Bottom) Yan et al. [464].
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 39

was minimal alignment of the fibril nanostructures. Thus, the shear systems may be able to act as anti-fouling surfaces for biomaterials
thinning of the gel is not due to fibril alignment and slipping under as they would disrupt any biofilm that attempts to form around the
applied shear. Instead, the shear-thinning behavior is a result of implanted material [397].
the gel fracturing into microdomains (as shown in Fig. 29). These MAX1 and MAX8 hydrogel networks have been used as injectable
microdomains can flow past one another under flow conditions; drug depots for the delivery of therapeutic molecules. The release
however, once shear stops, they immediately percolate and the gel kinetics of the peptide hydrogels was explored using model
stiffens (G0 > G00 ). These domains will relax over time and therapeutics of various molecular weight and charge. Specifically,
interpenetrate with one another, further stiffening the gel and dextran at 20, 70 or 150 kDa or lactoferrin protein (,77 kDa) was
contributing to the self-healing properties of the system [410]. incorporated into the peptide hydrogel prior to gelation. The
Both MAX1 and MAX8 peptide hydrogels are considered mixture was then sheared and allowed to re-solidify before being
cytocompatible and noninflammatory [465]. Specifically, these monitored for 30 days to measure bulk molecule release. It was
hydrogels were found to exhibit minimal-to-no cytotoxicity to a determined that mesh size and electrostatic interactions between
variety of cell lines including NIH3T3 murine fibroblasts, C3H10T1/ the macromolecule and the hydrogel had the greatest impact on
2 mesenchymal stem cells, hepatocytes, MG63 osteoblast progen- release kinetics. As molecular weight increased, the interactions
itor cells, and primary articular chondrocytes [410,411,466,467]. between the therapeutic and the hydrogel mesh hampered diffusive
The homogeneous distribution C3H10T1/2 cells in MAX8 and capability. Similarly, negatively charged species were more likely to
MAX1 hydrogels is dependent on the gelation kinetics. As shown in interact with the mesh and release much slower than the positively
Fig. 29, the MAX8 gels have a much more homogeneous charged molecules [469]. MAX8 peptide hydrogels were further
distribution, which can be attributed to their increased gelation explored as drug delivery vehicles for curcumin, a therapeutic
speed when compared to the MAX1 system [411]. Upon cessation with anti-inflammatory, anti-tumorgenic, and antioxidant abilities.
of shear, the MAX8 gels quickly recover their initial G0 values. This Altunbas et al. determined that curcumin could be homogeneously
rapid recovery preserves the homogeneous distribution of cells suspended in the peptide hydrogel matrix and be released over
within the polymer matrix [411]. Interestingly, these hydrogel extended periods of time (,15 days) and exert cytotoxic effect over
networks possess inherent anti-microbial activity against several DAOY human medulloblastoma tumor cells [470].
bacterial strains including Staphylococcus epidermidis, Staphylococ- Alternative peptide sequences have also shown promise in the
cus aureus, Streptococcus pyogenes, Klebsiella pneumoniae, and formation of shear-thinning hydrogel networks. Decapeptides
Escherichia coli. The hydrogels have been shown to disrupt the composed of an alternating charged/neutral amino acid pattern,
cellular membrane of these bacterial cells, which causes bacterial were synthesized by Yu et al. KVW10 and EVW10 consist of
death. Intriguingly, the hydrogels appear to exhibit selective hydrophobic/neutral valine residues alternated with cationic
toxicity to bacterial cells but spare mammalian cells [468]. These lysine or anionic glutamic acid residues, respectively [471]. Either

Fig. 29. (Top) Distribution of fluorescently labeled C3H10t1/2 cells in a MAX1 (left) and MAX8 (right) peptide hydrogel. (Bottom left) G0 is being measured as a function of time
to observe the properties of the MAX8 hydrogel before, during and after the introduction of shear flow. The cells were stained for viability before (bottom middle) and after
(bottom left) being injected through a syringe.
Reprinted with permission from Haines-Butterick et al. [411].
40 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

end of the decapeptide chains was capped by a functional group to


remove any additional charged moieties that could impede self-
assembly. Specifically, an acetyl group capped the N-terminus,
while the C-terminus was capped by an amide. The self-assembly
of the decapeptides were driven by the mutually attractive but
self-repulsive behavior exhibited by the KVW10 and EVW10
decapeptide sequences. Namely, when alone in solution, the
species will not gel due to self-repulsion. However, when mixed
together, self-assembly will occur because the mutual attraction
between the charged residues of the peptides drives physical
interaction. This behavior is independent of pH and ionic-strength,
which is attractive for in vivo and in vitro applications, and will also
occur at very low peptide concentrations [471,472]. Similar to the
MITCH systems, these peptide hydrogels are capable of shear
thinning because the physical interactions between the two
charged peptide chains can be temporarily disrupted with applied
force. The hydrogel is able to withstand repeated shear thinning
and still regain up to 90% of its original pre-shear rigidity.
Furthermore, adjusting the hydrophobicity of the neutral amino
acid species in the peptide sequence easily controls the gel
modulus [471,473].
Multi-domain peptides are peptide sequences with coded
Fig. 30. Shear-thinning behavior of MDP hydrogels self-assembled in various ionic
regions that either encourage or prevent assembly. They exhibit an
solutions. Shear is applied at t = #1 and released at t = 0. G0 is measured as a function
ABA motif, where the B block is composed of hydrophilic Gln/Ser of time and as a response to applied shear.
residues and hydrophobic Leu residues, and the A block consists of Reprinted with permission from Aulisa et al. [395].
either positively charged Lys or negatively charged Glu. The B block
residues self-assemble in such a way that the hydrophilic and
hydrophobic residues reside on opposite faces, flanked by the 8.2.1.4. Synthetic hydrogels. Synthetic hydrogels can be composed
terminal A blocks [474–479]. This self-assembly can be triggered of a wide variety of polymeric materials and have the benefit of
by the protonation or deprotonation of the terminal A blocks by pH allowing the researcher to exert complete control over the
titration or charge shielding by increasing the environmental ionic composition, structure, and responsiveness of the synthesized
strength [395]. This self-assembly stemming from each ABA block hydrogel. One such system is a blend of hyaluronic acid (HA) and
arrangement promotes fibril formation and hydrogel gelation methylcellulose (MC). Methylcellulose is a thermally responsive
[415]. These hydrogels shear-thin upon application of shear force biocompatible material that will form a gel at or above 37 8C in
and recover their properties very rapidly, within ,20 s, after water. This weak gel is held together by hydrophobic interactions
cessation of shear (Fig. 30) [395,474]. These MDP gels have been [484,485]. Hyaluronic acid is a non-immunogenic and biocompat-
shown to exhibit no cytotoxicity and can induce the migration and ible material that exhibits shear-thinning behavior due to the
differentiation of MSCs from human exfoliated teeth (SHED) alignment of polymeric chains under flow [486–489]. However,
[478,480]. Additionally, the incorporation of degradable peptidic unless the HA is crosslinked, it is highly soluble and will disperse
sequences, such as those sensitive to matrix metalloproteinases, after injection [396]. Therefore, blending it with a material such as
into the MDP domains confers system biodegradability [474,478]. methylcellulose results in a robust gel. The gelation time of the
Another application of MDP hydrogel systems are as ‘drug HAMC gels can be controlled by varying the HA and MC
sponges’ to provide renal protection. Wang et al. have shown that compositions with the 2% HA/7% MC formulation gelling within
when human H9 embryonic stem cells are cultured in the presence 2 min. The shear-thinning behavior of the HA is maintained within
of an MDP system, the secretome of the stem cells, i.e., their the HAMC hydrogel as it flows upon the application of shear force
particular blend of growth factors cytokines, chemokines, etc., is and recovers its properties after shear is removed [490]. When
imbibed by the hydrogel network [476,481]. These hydrogels, once added to the intrathecal space of rats there was no apparent
perfused with the stem cell secretome had renoprotective effects cytotoxicity or immunogenicity in reaction to the injected HAMC
both in vitro and in vivo by mediated lipopolysaccharide-induced hydrogel [490].
renal trauma [476]. Similar protective effects were observed in Both cellular and therapeutic cargo can be incorporated into the
cardiac tissue when exposed to MDPs containing the secretomes of HAMC hydrogel and homogeneously suspended during gelation.
primary mouse MSCs [482]. The hydrogel can be administered into the interthecal space via
MDPs have also been used as one component of a multi-step injection due to the shear thinning properties. Human umbilical
temporally controlled release matrix for tissue engineering tissue-derived cells were successfully incorporated into the HAMC
applications. MDP hydrogels self-assemble around liposomes hydrogel without any loss of mechanical rigidity. The cells were
containing either a growth factor, such as placental growth successfully maintained over the course of three days [491]. Fi-
factor-1 (PlGF-1), vascular endothelial growth factor (VEGF) or a broblast growth factor 2, FGF2, and poly(ethylene glycol)-modified
chemokine such as monocyte chemoattractant protein-1 (MCP-1). FGF2, PEG-FGF2 were incorporated into HAMC hydrogels. The
A second growth factor or chemokine is passively imbibed by the incorporation process did not adversely impact the bioactivity of
already assembled MDP hydrogel containing liposomes. These the FGF2 or PEG-FGF2 as confirmed by the dose dependent
multi-component systems still exhibit shear-thinning and subse- proliferation of Balb/3T3 cell. The therapeutic cargo was delivered
quent shear-recovery behaviors and the overall rigidity is not over the course of 25 h in an in vitro setting. However, it was shown
impacted by liposome inclusion. A bimodal drug release profile is that PEG-FGF2 entered the injured spinal tissue at much higher
observed and shown in Fig. 31, which emphasizes the ability of the concentrations than the unmodified growth factor [492]. HAMC is
multi-component system of MDPs and encapsulated liposomes to a viable option for both tissue engineering and drug delivery
exert a temporally-controllable drug release [483]. applications [493,494].
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 41

Fig. 31. Bimodal release of EGF-FITC and PlGF-1-TAMRA from hydrogel alone (blue) or liposome (red). Both drugs show ,100% release over a 15 day period. However, the
drug from the hydrogel alone is released much more rapidly, i.e. burst release, than the drug that had been encapsulated within the liposome. These systems exhibit temporal
control over drug release. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)
Reprinted with permission from Wickremasinghe et al. [483].

In addition to being blended in with MC, hyaluronic acid and b- networks. Block copolymers, in particular, contain both hydro-
cyclodextrin, can be formed into shear thinning hydrogels via phobic and hydrophilic moieties, which can induce polymer
guest–host mediated self-assembly. Rodell et al. modified threading into cyclodextrin cavity and subsequent stabilization
hyaluronic acid with either an adamantane functionality (Ad- [365]. This polymer threading is a transient behavior and can be
HA) or with b-cyclodextrin (CD-HA) [495]. Adamatane and its disrupted by the application of shear force and then self-heal
derivatives are widely used in the pharmaceutical industry for the [503].
prophylaxis or treatment of viral diseases as well as Parkinson’s One such triblock copolymer system is composed of a PEO-PPO-
[496]. Cyclodextrins are cyclic oligosaccharides of D-glucose units PEO, poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene
linked by a-1,4-linkages. The a-, b- and g-cyclodextrins contain oxide), linear polymer and an a-cyclodextrin [503]. The same
5, 7, or 8 anyhydroglucose units, respectively. These components complexation behavior is observed for PEO-PCL, poly(ethylene
have been used in the pharmaceutical, food and cosmetic oxide)-b-poly(e-caprolactone) diblock copolymers [503,504]. The
industries [497–500]. Together, adamantane-derivatives and b- incorporation of biodegradable linear polymers such as poly(lactic
cyclodextrin form a complex with a relatively high association acid) (PLA) and PEG which interact with a-cyclodextrin, can form
constant, ,1 . 105 M. This association is known as a guest–host biodegradable hydrogel systems for drug delivery applications
assembly, which occurs when two chemical species interact via [500]. A model protein therapeutic, FITC-BSA has been incorporat-
non-covalent associations, complexing together and driving ed into hydrogels composed of a-CD/PEO and a-CD/PEO-PHB-PEO
molecular assembly [501]. The guest–host complexation is a and exhibited release profiles that were dependent upon the
reversible association. Fig. 32 details the guest–host assembly, the erosion rate of the hydrogels [503,505]. The shear-thinning
synthesis of CD- and Ad-HA macromolecules, a visual assessment hydrogels exhibit self-healing kinetics are much slower than other
of the gelation and the reversible physical crosslinks that result in injectable shear-thinning systems, ,20 min [503]. Further inves-
shear thinning. tigation will be necessary to optimize these systems for in vivo
Network properties are controllable by adjusting macromolec- applications.
ular concentration and guest–host stoichiometry. Shear-thinning
behavior is observed and, once force is removed, the gel exhibits 8.2.1.5. Synthetic colloidal systems. Colloidal gels are composed of
near-immediate recovery. A model therapeutic, FITC-BSA, was suspended colloidal particles that exhibit a reversible sol–gel
incorporated into the gel and its release was monitored over transition that allows the gels to shear-thin [410,506,507]. The
60 days. The FITC-BSA exhibited a sustained release curve over the formation of colloidal gels is driven by the mixing of two different
course of the experiments. The release was readily controllable by colloidal particles that have favorable surface interactions. The
adjusting hydrogel parameters. Additionally, the hydrogel can interaction and self-assembly of these particles results in the
disassemble, or biodegrade, via a linear erosion mechanism observed sol–gel transition [363]. Colloidal gel shear-thinning
making it a promising candidate for injectable drug delivery and occurs when the surface interactions are disrupted under applied
tissue engineering systems [495]. shear, while self-healing is the reinstatement of those favorable
Cyclodextrins (CDs) have also shown great promise in forming interactions [363,508–512]. A colloidal system based on charged
shear-thinning hydrogel networks when associated with linear poly(D,L-lactic-co-glycolic acid) (PLGA) nanoparticles was devel-
block copolymers, such as poly(ethylene oxide) (PEO). This oped by Shi et al. One set of PLGA nanoparticles was coated with a
association is attributed to the truncated cone configurations positively charged polyvinylamine while the other set is coated
the CDs adapt where the hydrophilic hydroxyl groups form the with a negatively charged poly(ethylene-co-maleic acid). Their
outer surface and the inner cavity of the cone is hydrophobic self-assembly is dictated by electrostatic attraction between the
[499,500]. The linear triblock copolymers can penetrate into the oppositely charged nanoparticle structures [511,512]. The result-
hydrophobic cavity and form complexes [502]. The complex ing network is porous and rigid. The mechanical properties can be
formation is dictated by the size of the cavity, i.e., a-, b- and g- adjusted by the ratio of positively- to negatively-charged species.
variants, and the diameter of the polymer chain [502]. When the Both protein and hUCMSCs (human umbilical cord matrix stem
polymer chain enters into the hydrophobic cavity of the cells) can be encapsulated within the nanoparticles. Protein will be
cyclodextrins the complex is stabilized by hydrophobic interac- released as a function of the PLGA degradation rate, while stem
tions and hydrogen bonding. The combination of the polymer cells can encourage osteoconductive bone formation when
threading and stabilization results in the gelation of the hydrogel injected into bone defects [363,508,509].
42 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

Fig. 32. (a) Schematic of CD, Ad and the guest host complex, either when alone or when Cd or Ad are joined to HA. (b) Synthesis of Ad-HA and CD-HA. (c) Inversion test to
observe gelation between Ad-HA and CD-HA. (d) Schematic of guest–host interactions forming reversible physical crosslinks.
Reprinted with permission from Chen and Jiang [501].

8.2.2. Other shear-thinning applications hydroxyl groups and overall net charge. At a concentration
In addition to injectable hydrogels for drug delivery and tissue of 20 mg/mL, all of the peptides formed fibrillar hydrogels at
engineering scaffolds, shear-thinning gels have a host of other physiological conditions. Upon rheological examination, all of the
applications including for arthritis treatment, biofilm disruption, etc. peptide formulations exhibited shear-thinning behavior similar to
Hyaluronic acid (HA) occurs naturally within the body and that of native HA [364].
exists as a major component of the synovial fluid that lubricates Shear-thinning hydrogels have also exhibited some inherent
joints during motion. The viscoelastic properties that it imparts to anti-microbial activity, as was discussed in more detail earlier
the synovial fluid allows joints to be lubricated by a more viscous [397,468]. It has been hypothesized that the bacterial-specific
fluid during normal motion, but upon impact, the gel becomes membrane disruption upon encountering polymeric biocides is
more elastic in nature and cushions the joint [364,382,396]. A 50% due to the long-range interstatic reaction between the two that far
crosslinked hyaluronic acid hydrogel was developed by R. Barbucci surpasses that of mammalian cells [513]. Both the MAX8 and
et al. to treat osteoarthritis. Specifically, the crosslinked HA gel MAX1 self-assembled hydrogel systems inhibit both gram-positive
could be injected into a bone defect or in between the joint and and gram-negative bacterial growth by disrupting their mem-
persist longer than the uncrosslinked HA gels. Additionally, the branes. Synthetic polymer sequences such as biodegradable
mechanical properties were preserved both after shear and after poly(L-lactide)-b-poly(ethylene glycol)-b-poly(L-lactide) (PLLA-
ethylene oxide (EtO) and g-ray sterilization procedures. These PEG-PLLA) and a polycarbonate triblock copolymer (PLDA-CPC-
crosslinked gels were capable of promoting the healing of bone PLDA) can stereocomplex and form shear-thinning gels that also
defects in a rabbit in vivo model [396]. prevent gram-postive and gram-negative microbial growth [397].
Engineered peptide sequences exhibiting shear-thinning beha- Another system is based on an ABA triblock copolymer composed
viors were developed for use to promote joint lubrication for of a hydrophilic PEG middle block and vitamin E-containing
osteoarthritis treatment. These self-assembled hydrogel materials polycarbonate copolymers on either end of the copolymer. These
were composed of four different peptide sequences, P11-4, P11-8, systems exhibited desirable mechanical stiffness upon the
P11-9, and P11-12, which are differentiated by the number of induction of gelation but also underwent shear thinning upon
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 43

application of increased shear rate. They were examined for their (Cys-ELP) undergoes gelation upon exposure to hydrogen peroxide
ability to eradicate biofilms and microbial killing efficiencies. To due to disulfide linkages forming between the cysteine residues. The
this end, these systems kill more than 99.9% of bacteria and fungi it hydrogels post-gelation strain thicken under increasing shear rate
contacts. Additionally, these systems eradicate biomass and also until reaching a threshold shear rate upon which they shear-thin.
greatly disrupt established biofilms. The ability to have hydrogel This thickening is attributed to nonlinear tension along the stretched
materials that can disrupt bacterial membranes is a powerful tool polymer chains outside of the Gaussian range. Once the system
that could ultimately be used to prevent bio-film formation on reaches high enough shear, the network fractures and the tension is
implanted biomaterials and for the prevention and treatment of relieved resulting in the shear-thinning behavior [519]. Another
skin infections [513]. such system is a telechelic polyelectrolyte consisting of a poly(-
The inherent viscoelastic properties of shear-thinning hydro- dimethyl amino ethyl methacrylate) (PDMAEMA) middle block
gels are attractive for the development of topical treatments for flanked by two poly(methyl methacrylates) (PMMA). These poly-
skin disorders such as infections and burns. They will shear thin mers form flower-like micelles above a given concentration with a
during application and then gel for sustained contact with the hydrophobic core and looping hydrophilic polymer chains. When a
wound area. One such system is a cubosome based hydrogel critical concentration is reached, the hydrophobic ends can
imbibed with silver sulfadiazine for burn treatment [398]. An transiently disassociate from the micelle core and engage with
emulsion between the lipid phase monoolein and Polaxamer 407, a other hydrophobic ends forming a network (Fig. 33). Adjusting
nonionic surfactant forms the hydrogel network. Options for parameters such as pH and ionic strength controls this network
modification include the concentration of lipid and the inclusion of formation. The shear-thickening polymer behavior has multiple
polyvinyl alcohol during emulsion. The cubosome dispersions are theoretical causes: (1) non-linear stretching of the polymeric chains,
stable and undergo shear-thinning behavior. Furthermore, they are (2) incomplete relaxation of the disassociated hydrophobic chain
capable of releasing the imbibed silver sulfadiazine, which is used ends or (3) shear-induced increased density of elastic chains
for burn treatment. In vivo studies showed that this hydrogel had [518,520–522]. The dual shear-thickening and shear-thinning
great promise in the treatment of severe burns with minimal side behavior of the hydrogel confers toughness to the material at low
effects [398]. shear stresses but yields at higher shear stresses, which makes them
compatible for injectable delivery [523]. These hydrogel systems
8.3. Shear-thickening hydrogels have the ability to complex DNA by the electrostatic interactions
and, therefore, have potential for biological applications [518]. Other
Polymers that undergo increased viscosity as a function of examples of shear-thickening hydrogels include clay-polymer
increased shear rate are deemed shear thickening [389,390]. Ceram- nanocomposites [524,525], graphene-poly(vinyl alcohol) compo-
ic-based systems such as hydroxyapatite [514] and self-curing sites [526] and crosslinked guar gum [527]. However, the biomedical
poly(methymethacrylate) cements [515] used for bone tissue applications of shear-thickening systems, particularly those that
engineering are biomaterials that exhibit strain thickening beha- also exhibit shear-thinning at high shear rates is not as obvious and
viors. In both ceramics and bone cement, this behavior is attributed will need to be investigated in the future.
to the colloidal nature of powder suspensions, specifically when the
powder is at a high concentration and a fine enough particle 8.4. Concluding remarks
diameter [516]. As described by Reynolds et al., the particles in
suspension are efficiently packed with minimal fluid-filled space Shear-thinning hydrogel systems are versatile platforms for a
between them. When shear is minimal, the liquid between the number of biomedical applications. They are formed via intermo-
particles acts as lubricant keeping the viscosity low. Once shear is lecular interactions that lead to self-assembled structures. They all
increased, the distance between the particles also increases and the shear-thin upon application of shear force and self-heal with
volume of liquid is insufficient to fill the void. This loss of fluid varying kinetics after the cessation of shear. Varying the
contact results in dramatically increased shear stress and subse- composition and gelation conditions controls the mechanical
quent increase in viscosity [515]. Titanium oxide (TiO2) nanopar- properties of the hydrogel networks. These systems can be used for
ticles suspended in acetylacetone also shear-thicken and show injectable hydrogel depots for drug delivery and tissue engineering
promise as protective coatings on metallic and other substances for as well as for osteoarthritic treatment, biofilm disruption and skin
use in implanted biomaterials [517]. Hydrogels that exhibit shear- wound healing. Shear-thickening hydrogels typically exhibit
thickening behaviors tend to do so only within a certain range of shear-thickening behaviors at low shear rates and will then
shear stresses. Outside of those ranges, the hydrogel behaves as a shear-thin as the shear rate increases. While their biomedical
linear viscoelastic material or can even shear-thin at high enough applications are not as apparent as in the shear-thinning systems,
shear rates [427,518]. An elastin-like cysteine-containing hydrogel their shear-thickening behavior confers hydrogel toughness at low

Fig. 33. The concentration-dependent formation of a network of the cationic telechelic polymer, MMA-DMAEMA-MMA in a salt-free environment. Concentration of polymer
increases from left to right.
Reprinted with permission from Bossard et al. [518].
44 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

shear rates, such as those experienced in vivo, but their shear- [29] B. Amsden, Mech. Models Macromol. 31 (1998) 8382–8395, http://dx.doi.org/
10.1021/ma980765f.
thinning capabilities at high shear means their use can be extended [30] A.H. Muhr, J.M.V. Blanshard, Polymer 23 (1982) 1012–1026, http://dx.doi.org/
to injectable systems. 10.1016/0032-3861(82)90402-5.
[31] N.A. Peppas, D.L. Meadows, J. Membr. Sci. 16 (1983) 361–377, http://dx.doi.org/
10.1016/S0376-7388(00)81321-6.
9. Summary [32] P. Gupta, K. Vermani, S. Garg, Drug Discov. Today. 7 (2002) 569–579.
[33] J. Fallingborg, Dan. Med. Bull. 46 (1999) 183–196.
[34] A.C. Guyton, J.E. Hall, Textbook of Medical Physiology, 11th ed., Elsevier,
Hydrogels are an extremely diverse group of materials that
Philadelphia, 2006.
have a wide variety of uses. In recent decades, a boom in research [35] W.R. Lang, Obstet. Gynecol. Surv. 10 (1955) 546–560.
on ‘‘smart’’ hydrogels that exhibit responsiveness to a variety of [36] J.L. Wike-Hooley, Eur. J. Cancer Clin. Oncol. 21 (1985) 785–787.
responses has led to many novel applications of hydrogels. With an [37] S.D. Steichen, M. Caldorera-Moore, N.A. Peppas, Eur. J. Pharm. Biopharm. 48
(2013) 416–427.
array of triggering mechanisms—including pH, temperature, [38] P. Vaupel, F. Kallinowski, P. Okunieff, Cancer Res. 49 (1989) 6449–6465.
external chemicals, light, electrical fields, and shear stresses— [39] J. Dissemond, M. Witthoff, T.C. Brauns, D. Haberer, M. Goos, Hautarzt Z. Derma-
these hydrogels enable precise control over fundamental material tol. Venerol. Verwandte Geb. 54 (2003) 959–965.
[40] M. Grabe, G. Oster, J. Gen. Physiol. 117 (2011) 329–344.
properties, such as swelling, porosity, physical structure, and [41] A. Asokan, M.J. Cho, J. Pharm. Sci. 91 (2002) 903–913.
modulus. With this level of external control, numerous applica- [42] L.A. Sharpe, A.M. Daily, S.D. Horava, N.A. Peppas, Expert Opin. Drug Deliv. 11
tions within medical and industrial fields have suddenly moved (2014) 901–915.
[43] N.A. Peppas, P. Bures, W. Leobandung, H. Ichikawa, Eur. J. Pharm. Biopharm. 50
into the realm of possibility: well-controlled drug delivery; (2000) 27–46.
inexpensive and accurate biosensors; artificial muscles; or [44] A.M. Lowman, N.A. Peppas, Encyclopedia of Controlled Drug Delivery, John
effective scaffolds for tissue engineering. As research continues Wiley & Sons, 1999, pp. 397–418.
[45] H.Y. Park, I.H. Song, J.H. Kim, W.S. Kim, Int. J. Pharm. 175 (1998) 231–236.
toward finding novel mechanisms for stimuli-responsiveness and
[46] M.M. Welz, C.M. Ofner III, J. Pharm. Sci. 81 (1992) 85–90.
innovative applications of the responsive hydrogels, we will [47] A.W. Chan, R.A. Whitney, R.J. Neufield, Biomacromolecules 9 (2008) 2536–2545.
indubitably see significant advances in the coming decades in both [48] A.W. Chan, R.J. Neufield, Biomaterials 30 (2009) 6119–6129.
[49] S.Y. Kim, S.M. Cho, Y.M. Lee, S.J. Kim, J. Appl. Polym. Sci. 78 (2000) 1381–1391.
healthcare and industry that arise directly from stimuli-responsive
[50] X. Qu, A. Wirsen, A.-C. Albertsson, Polymer 41 (2000) 4589–4598.
hydrogels and tangibly improve the world. [51] D. Schmaljohann, Adv. Drug Deliv. Rev. 58 (2006) 1655–1670.
[52] I.C. Kwan, Y.H. Bae, T. Okano, S.W. Kim, B. Berner, Macromol. Symp. 33 (1990).
References [53] A.R. Khare, N.A. Peppas, G. Massimo, P. Colombo, J. Control. Release 22 (1992)
239–244.
[54] N.A. Peppas, J.Z. Hilt, A. Khademhosseini, R. Langer, Adv. Mater. 18 (2006)
[1] N.A. Peppas, Hydrogels in Medicine and Pharmacy, CRC Press, Boca Raton, FL, 1345–1360.
1987. [55] A.R. Khare, N.A. Peppas, Biomaterials 16 (2009) 559–567.
[2] B.V. Slaughter, S.S. Khurshid, O.Z. Fisher, A. Khademhosseini, N.A. Peppas, Adv. [56] N.A. Peppas, B.D. Barr-Howell, Hydrogels in Medicine and Pharmacy:
Mater. 21 (2009) 3307–3329, http://dx.doi.org/10.1002/adma.200802106. Fundamentals, CRC Press, 1986, pp. 27–54.
[3] K.S. Anseth, C.N. Bowman, L. Brannon-Peppas, Biomaterials 17 (1996) 1647–1657. [57] L. Brannon-Peppas, N.A. Peppas, J. Controll. Release 16 (1991) 319–330.
[4] N.A. Peppas, E.W. Merrill, J. Appl. Polym. Sci. 21 (1977) 1763–1770. [58] B.A. Firestone, R.A. Siegel, J. Appl. Polym. Sci. 43 (1991) 901–914.
[5] L. Brannon-Peppas, N.A. Peppas, Chem. Eng. Sci. 46 (1991) 715–722. [59] R. Bettini, P. Colombo, N.A. Peppas, J. Control. Release 37 (1995) 105–111.
[6] A.M. Lowman, in: N.A. Peppas (Ed.), Advances in Chemical Engineering, Elsevier, [60] T. Canal, N.A. Peppas, J. Biomed. Mater. Res. A 23 (1989) 1183–1193.
2004, p. 248. [61] J.E. Elliot, M. Macdonald, J. Nie, C.N. Bowman, Polymer 45 (2004) 1503–1510.
[7] P.J. Flory, J. Rehner Jr., J. Chem. Phys. 11 (1943) 512. [62] M.C. Koetting, N.A. Peppas, Int. J. Pharm. 471 (2014) 83–91.
[8] N. Peppas, J.Z. Hilt, A. Khademhosseini, R. Langer, Adv. Mater. 18 (2006) [63] N.A. Peppas, K.M. Wood, J.O. Blanchette, Expert Opin. Biol. Ther. 4 (2004) 881–887.
1345–1360. [64] T.K. Bronich, S.V. Vinogradov, A.V. Kabanov, Nano Lett. 1 (2001) 535–540.
[9] F.W. Billmeyer, Textbook of Polymer Science, John Wiley & Sons, Inc., New York, [65] C.A. Schoener, H.N. Hutson, N.A. Peppas, J. Biomed. Mater. Res. A 101A (2013)
1984. 2229–2236.
[10] L.H. Sperline, V. Mishra, Polym. Adv. Technol. 7 (1996) 197–208. [66] D.C. Forbes, N.A. Peppas, Macromol. Biosci. 14 (2014) 1096–1105.
[11] L. Verestiuc, C. Ivanov, E. Barbu, J. Tsibouklis, Int. J. Pharm. 269 (2004) (n.d.). [67] A.M. Lowman, M. Morishita, M. Kajita, T. Nagai, J. Pharm. Sci. 88 (1999) 933–937.
[12] J. Zhang, Structure and morphology of poly(methacrylic acid) poly(N-isopropyl [68] Y. Hu, P.U. Atukorale, J.J. Lu, J.J. Moon, S.H. Um, E.C. Cho, Biomacromolecules 10
acrylamide) interpenetrating polymeric networks with pH and temperature (2009) 756–765.
sensitivity, (Ph.D.), Purdue University, 2000 http://search.proquest.com/ [69] N.A. Peppas, R. Langer, AIChE J. 50 (2004) 536–546.
docview/304654073/abstract?accountid=7118 (accessed 16.12.14). [70] C.L. Bell, N.A. Peppas, J. Control. Release 39 (1996) 201–207.
[13] T.J. Merkel, S.W. Jones, K.P. Herlihy, F.R. Kersey, A.R. Shields, M. Napier, J.C. Luft, [71] A.M. Lowman, N.A. Peppas, Macromolecules 30 (1997) 4959–4965.
H. Wu, W.C. Zamboni, A.Z. Wang, J.E. Bear, J.M. DeSimone, Proc. Natl. Acad. Sci. [72] J. Klier, A.B. Scranton, N.A. Peppas, Macromolecules 23 (1990) 4944–4949.
108 (2011) 586–591, http://dx.doi.org/10.1073/pnas.1010013108. [73] N.A. Peppas, J. Klier, J. Control. Release 16 (1991) 203–214.
[14] J.L. Drury, D.J. Mooney, Biomaterials 24 (2003) 4337–4351, http://dx.doi.org/ [74] S.S. Davis, Drug Discov. Today 10 (2005) 249–257.
10.1016/S0142-9612(03)00340-5. [75] N.A. Peppas, Y. Huang, Adv. Drug Deliv. Rev. 56 (2004) 1675–1687.
[15] K.S. Anseth, C.N. Bowman, L. Brannon-Peppas, Biomaterials 17 (1996) 1647– [76] H. Park, J.R. Robinson, Pharm. Res. 4 (1987) 457–464.
1657, http://dx.doi.org/10.1016/0142-9612(96)87644-7. [77] N.A. Peppas, K.B. Keys, M. Torres-Lugo, A.M. Lowman, J. Control. Release 62
[16] N.A. Peppas, E.W. Merrill, J. Appl. Polym. Sci. 21 (1977) 1763–1770, http:// (1999) 81–87.
dx.doi.org/10.1002/app.1977.070210704. [78] K.M. Wood, G.M. Stone, N.A. Peppas, Biomacromolecules 9 (2008) 1293–1298.
[17] P.J. Flory, Principles of Polymer Chemistry, Cornell University Press, 1953. [79] J.E. Lopez, N.A. Peppas, Drug Dev. Ind. Pharm. 30 (2004) 497–504.
[18] J. Crank, The Mathematics of Diffusion, Academic Press, New York, 1975. [80] K. Nakamura, R.J. Murray, J.I. Joseph, N.A. Peppas, M. Morishita, A.M. Lowman, J.
[19] M.T. am Ende, D. Hariharan, N.A. Peppas, React. Polym. 25 (1995) 127–137, Control. Release 95 (2004) 589–599.
http://dx.doi.org/10.1016/0923-1137(94)00040-C. [81] B. Kim, N.A. Peppas, Int. J. Pharm. 266 (2003) 29–37.
[20] L. Brannon-Peppas, N.A. Peppas, J. Control. Release 8 (1989) 267–274, http:// [82] M. Morishita, T. Goto, K. Nakamura, A.M. Lowman, K. Takayama, N.A. Peppas, J.
dx.doi.org/10.1016/0168-3659(89)90048-5. Control. Release 110 (2006) 587–594.
[21] E.M. Renkin, J. Gen. Physiol. 38 (1954) 225–243, http://dx.doi.org/10.1085/ [83] N. Kamei, M. Morishita, H. Chiba, N.J. Kavimandan, N.A. Peppas, K. Takayama, J.
jgp.38.2.225. Control. Release 134 (2009) 98–102.
[22] J.R. Pappenheimer, J. Physiol. Rev. 1953 (1953) 387–423. [84] M. Torres-Lugo, N.A. Peppas, Biomaterials 21 (2000) 1191–1196.
[23] L.F. Gudeman, N.A. Peppas, J. Membr. Sci. 107 (1995) 239–248, http://dx.doi.org/ [85] D.A. Carr, N.A. Peppas, Macromol. Biosci. 9 (2009) 497–505.
10.1016/0376-7388(95)00120-7. [86] D.A. Carr, N.A. Peppas, J. Biomed. Mater. Res. A 92A (2010) 504–512.
[24] J.L. Anderson, J.A. Quinn, Biophys. J. 14 (1974) 130–150, http://dx.doi.org/ [87] D.A. Carr, M. Gomez-Burgaz, M.C. Boudes, N.A. Peppas, Ind. Eng. Chem. Res. 49
10.1016/S0006-3495(74)70005-4. (2010) 11991–11995.
[25] N.A. Peppas, C.T. Reinhart, J. Membr. Sci. 15 (1983) 275–287, http://dx.doi.org/ [88] W.R. Gambotz, S.F. Wee, Adv. Drug Deliv. Rev. 64 (2012) 194–205.
10.1016/S0376-7388(00)82304-2. [89] T. Yotsuyanagi, T. Ohkubo, T. Ohhashi, K. Ikeda, Chem. Pharm. Bull. (Tokyo) 35
[26] C.T. Reinhart, N.A. Peppas, J. Membr. Sci. 18 (1984) 227–239, http://dx.doi.org/ (1987) 1555–1563.
10.1016/S0376-7388(00)85036-X. [90] S.-C. Chen, Y.-C. Wu, F.-L. Mi, Y.-H. Lin, L.-C. Yu, H.-W. Sung, J. Control. Release 96
[27] A.P. Sassi, H.W. Blanch, J.M. Prausnitz, J. Appl. Polym. Sci. 59 (1996) 1337–1346, (2004) 285–300.
http://dx.doi.org/10.1002/(SICI)1097-4628(19960222)59:8<1337::AID- [91] J.L. Romalde, A. Luzardo-Alvarez, C. Ravelo, A.E. Toranzo, J. Blanco-Mendez,
APP18>3.0.CO;2-2. Aquaculture 236 (2004) 119–129.
[28] M.T. am Ende, N.A. Peppas, J. Control. Release 48 (1997) 47–56, http://dx.doi.org/ [92] B.-J. Lee, G.-H. Min, Int. J. Pharm. 144 (1996) 37–46.
10.1016/S0168-3659(97)00032-1.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 45

[93] S. Sugawara, T. Imai, M. Otagiri, Pharm. Res. 11 (1994) (n.d.). [140] H.V. Recum, A. Kikuchi, M. Yamato, Y. Sakurai, T. Okano, S.W. Kim, Tissue Eng. 5
[94] T. Zhang, T.F. Sturgis, B.-B.C. Youan, Eur. J. Pharm. Biopharm. 79 (2011) 526–536. (1999) 251–265, http://dx.doi.org/10.1089/ten.1999.5.251.
[95] A.D. Woolfson, M.L. Umrethia, V.L. Kett, R.K. Malcolm, Int. J. Pharm. 388 (2010) [141] T. Hoare, J. Santamaria, G.F. Goya, S. Irusta, D. Lin, S. Lau, R. Padera, R. Langer,
136–143. D.S. Kohane, Nano Lett. 9 (2009) 3651–3657, http://dx.doi.org/10.1021/
[96] R.J. Morrow, A.D. Woolfson, L. Donnelly, R. Curran, G. Andrews, D. Katinger, R.K. nl9018935.
Malcolm, Eur. J. Pharm. Biopharm. 77 (2011) 3–10. [142] R.M. Gant, Y. Hou, M.A. Grunlan, G.L. Coté, J. Biomed. Mater. Res. A 90A (2009)
[97] S. Garg, K. Vermani, A. Garg, R.A. Anderson, W.B. Rencher, L.J.D. Zaneveld, Pharm. 695–701, http://dx.doi.org/10.1002/jbm.a.32135.
Res. 22 (2005) 584–595. [143] N.I. Abu-Lail, M. Kaholek, B. LaMattina, R.L. Clark, S. Zauscher, Sens. Actuators B:
[98] U.M. Parikh, C. Dobard, S. Sharma, M. Cong, H. Jia, A. Martin, C.-P. Pau, D.L. Chem. 114 (2006) 371–378, http://dx.doi.org/10.1016/j.snb.2005.06.003.
Hanson, P. Guenthner, J. Smith, E. Kersh, J.G. Garcia-Lerma, F.J. Novembre, R. [144] WHO, Country and Regional Data on Diabetes, WHO, 2014 (n.d.). http://www.
Otten, T. Folks, W. Heneine, J. Virol. 83 (2009) 10358–10365. who.int/diabetes/facts/world_figures/en/ (accessed 13.12.14).
[99] T. Zhang, C. Zhang, V. Agrahari, J.B. Murowchick, N.A. Oyler, B.-B.C. Youan, [145] Q. Wu, L. Wang, H. Yu, J. Wang, Z. Chen, Chem. Rev. 111 (2011) 7855–7875,
Antivir. Res. 97 (2013) 334–346. http://dx.doi.org/10.1021/cr200027j.
[100] S.A. Agnihotri, N.N. Mallikarjuna, T.M. Aminabhavi, J. Control. Release 100 (2004) [146] P. Bernfeld, J. Wan, Science 142 (1963) 678–679, http://dx.doi.org/10.1126/
5–28. science.142.3593.678.
[101] A. Bernkop-Schnürch, S. Dünnhaupt, Eur. J. Pharm. Biopharm. 81 (2012) 463–469. [147] T.A. Horbett, J. Kost, B.D. Ratner, in: S.W. Shalaby, A.S. Hoffman, B.D. Ratner, T.A.
[102] R. Hejazi, M.M. Amiji, J. Control. Release 89 (2003) 151–165. Horbett (Eds.), Polymers as Biomaterials, Springer US, 1984, pp. 193–207,
[103] V.R. Patel, M.M. Amiji, Pharm. Res. 13 (1996) 588–593. http://link.springer.com/chapter/10.1007/978-1-4613-2433-1_14 (accessed
[104] Y.-N. Dai, P. Li, J.-P. Zhang, A.-Q. Wang, Q. Wei, Biopharm. Drug Dispos. 29 (2008) 13.12.14).
173–184. [148] T.A. Horbett, B.D. Ratner, J. Kost, M. Singh, in: J.M. Anderson, S.W. Kim (Eds.),
[105] W.B. Liechty, D.R. Kryscio, B.V. Slaughter, N.A. Peppas, Annu. Rev. Chem. Biomol. Recent Advances in Drug Delivery Systems, Springer US, 1984, pp. 209–220,
Eng. 1 (2010) 149–173. http://link.springer.com/chapter/10.1007/978-1-4613-2745-5_14 (accessed
[106] O.Z. Fisher, N.A. Peppas, Macromolecules 42 (2009) 3391–3398. 30.11.14).
[107] O.Z. Fisher, T. Kim, S.R. Dietz, N.A. Peppas, Pharm. Res. 26 (2009) 51–60. [149] G. Albin, T.A. Horbett, B.D. Ratner, J. Control. Release 2 (1985) 153–164, http://
[108] W.B. Liechty, R.L. Scheuerle, N.A. Peppas, Polymer 54 (2013) 3784–3795. dx.doi.org/10.1016/0168-3659(85)90041-0.
[109] D.C. Forbes, M. Creixell, H. Frizzell, N.A. Peppas, Eur. J. Pharm. Biopharm. 84 [150] L.A. Klumb, T.A. Horbett, J. Control. Release 18 (1992) 59–80, http://dx.doi.org/
(2013) 472–478. 10.1016/0168-3659(92)90212-A.
[110] D.C. Forbes, N.A. Peppas, ACS Nano 8 (2014) 2908–2917. [151] K. Podual, F.J. Doyle III, N.A. Peppas, Polymer 41 (2000) 3975–3983, http://
[111] W.B. Liechty, Eur. J. Pharm. Biopharm. 80 (2012) 241–246. dx.doi.org/10.1016/S0032-3861(99)00620-5.
[112] K. Raemdonck, J. Demeester, S.D. Smedt, Soft Matter. 5 (2009) 707–715. [152] K. Zhang, X.Y. Wu, J. Control. Release 80 (2002) 169–178, http://dx.doi.org/
[113] H. Zhang, S. Mardyani, W.C.W. Chan, E. Kumacheva, Biomacromolecules 7 (2006) 10.1016/S0168-3659(02)00024-X.
1568–1572. [153] C.R. Gordijo, A.J. Shuhendler, X.Y. Wu, Adv. Funct. Mater. 20 (2010) 1404–1412,
[114] M. Oishi, H. Hayashi, M. Iijima, Y. Nagasaki, J. Mater. Chem. 17 (2007) 3720–3725. http://dx.doi.org/10.1002/adfm.200901581.
[115] L. Klouda, A.G. Mikos, Eur. J. Pharm. Biopharm. 68 (2008) 34–45, http:// [154] C. Malitesta, F. Palmisano, L. Torsi, P.G. Zambonin, Anal. Chem. 62 (1990) 2735–
dx.doi.org/10.1016/j.ejpb.2007.02.025. 2740, http://dx.doi.org/10.1021/ac00223a016.
[116] S. Purushotham, R.V. Ramanujan, Acta Biomater. 6 (2010) 502–510, http:// [155] K. Podual, F.J. Doyle III, N.A. Peppas, J. Control. Release 67 (2000) 9–17, http://
dx.doi.org/10.1016/j.actbio.2009.07.004. dx.doi.org/10.1016/S0168-3659(00)00195-4.
[117] Y.H. Bae, T. Okano, S.W. Kim, J. Polym. Sci. B: Polym. Phys. 28 (1990) 923–936, [156] C.M. Hassan, F.J. Doyle, N.A. Peppas, Macromolecules 30 (1997) 6166–6173,
http://dx.doi.org/10.1002/polb.1990.090280609. http://dx.doi.org/10.1021/ma970117g.
[118] H. Katono, A. Maruyama, K. Sanui, N. Ogata, T. Okano, Y. Sakurai, J. Control. [157] S.I. Kang, Y.H. Bae, J. Control. Release 86 (2003) 115–121, http://dx.doi.org/
Release 16 (1991) 215–227, http://dx.doi.org/10.1016/0168-3659(91)90045-F. 10.1016/S0168-3659(02)00409-1.
[119] D.E. Owens, Y. Jian, J.E. Fang, B.V. Slaughter, Y.-H. Chen, N.A. Peppas, Macro- [158] D.-Y. Jung, J.J. Magda, I.S. Han, Macromolecules 33 (2000) 3332–3336, http://
molecules 40 (2007) 7306–7310, http://dx.doi.org/10.1021/ma071089x. dx.doi.org/10.1021/ma992098b.
[120] D.E. Owens, Thermally-Responsive Interpenetrating Polymer Network Nano- [159] T. Miyata, A. Jikihara, K. Nakamae, A.S. Hoffman, Macromol. Chem. Phys. 197
particles as Intelligent Therapeutic Systems, The University of Texas at Austin, (1996) 1135–1146, http://dx.doi.org/10.1002/macp.1996.021970330.
2006 http://gradworks.umi.com/32/66/3266943.html (accessed 14.12.14). [160] R. Ballerstadt, J.S. Schultz, Sens. Actuators B: Chem. 46 (1998) 50–55, http://
[121] R. Liu, M. Fraylich, B.R. Saunders, Colloid Polym. Sci. 287 (2009) 627–643, http:// dx.doi.org/10.1016/S0925-4005(97)00327-4.
dx.doi.org/10.1007/s00396-009-2028-x. [161] G.G. Adams, Y. Cui, J.H. Mitchell, M.J. Taylor, Rheol. Acta 45 (2006) 611–620,
[122] N.A. Platé, T.L. Lebedeva, L.I. Valuev, Polym. J. 31 (1999) 21–27, http://dx.doi.org/ http://dx.doi.org/10.1007/s00397-005-0013-y.
10.1295/polymj.31.21. [162] J.J. Kim, K. Park, J. Control. Release 77 (2001) 39–47, http://dx.doi.org/10.1016/
[123] S.A. Meenach, K.W. Anderson, J.Z. Hilt, J. Polym. Sci. A: Polym. Chem. 48 (2010) S0168-3659(01)00447-3.
3229–3235, http://dx.doi.org/10.1002/pola.24087. [163] J.J. Kim, K. Park, Pharm. Res. 18 (2001) 794–799, http://dx.doi.org/10.1023/
[124] J.J. Escobar-Chávez, M. López-Cervantes, A. Naik, Y. Kalia, D. Quintanar-Guerrero, A:1011084312134.
A. Ganem-Quintanar, J. Pharm. Pharm. Sci. 9 (2006) 339–358. [164] S. Tanna, T. Sahota, J. Clark, M.J. Taylor, J. Pharm. Pharmacol. 54 (2002) 1461–
[125] R.R. Bhattacharjee, M. Chakraborty, T.K. Mandal, J. Phys. Chem. B 110 (2006) 1469, http://dx.doi.org/10.1211/00223570290.
6768–6775, http://dx.doi.org/10.1021/jp056675b. [165] T. Miyata, A. Jikihara, K. Nakamae, A.S. Hoffman, J. Biomater. Sci. Polym. Ed. 15
[126] L. Verestiuc, O. Nastasescu, E. Barbu, I. Sarvaiya, K.L. Green, J. Tsibouklis, J. (2004) 1085–1098, http://dx.doi.org/10.1163/1568562041753061.
Biomed. Mater. Res. A 77A (2006) 726–735, http://dx.doi.org/10.1002/jbm.- [166] M.J. Taylor, S. Tanna, T.S. Sahota, B. Voermans, Eur. J. Pharm. Biopharm. 62 (2006)
a.30668. 94–100, http://dx.doi.org/10.1016/j.ejpb.2005.07.005.
[127] K.-F. Arndt, T. Schmidt, R. Reichelt, Polymer 42 (2001) 6785–6791, http:// [167] K. Makino, E.J. Mack, T. Okano, S.W. Kim, J. Control. Release 12 (1990) 235–239,
dx.doi.org/10.1016/S0032-3861(01)00164-1. http://dx.doi.org/10.1016/0168-3659(90)90104-2.
[128] M. Beija, J.-D. Marty, M. Destarac, Chem. Commun. 47 (2011) 2826, http:// [168] S.W. Kim, C.M. Paii, M. Kimiko, L.A. Seminoff, D.L. Holmberg, J.M. Gleeson, D.E.
dx.doi.org/10.1039/c0cc05184e. Wilson, E.J. Mack, J. Control. Release 11 (1990) 193–201, http://dx.doi.org/
[129] C. Echeverria, D. López, C. Mijangos, Macromolecules 42 (2009) 9118–9123, 10.1016/0168-3659(90)90132-D.
http://dx.doi.org/10.1021/ma901316k. [169] F. Liu, S.C. Song, D. Mix, M. Baudyš, S.W. Kim, Bioconjug. Chem. 8 (1997) 664–
[130] X.-C. Xiao, L.-Y. Chu, W.-M. Chen, S. Wang, Y. Li, Adv. Funct. Mater. 13 (2003) 672, http://dx.doi.org/10.1021/bc970128e.
847–852, http://dx.doi.org/10.1002/adfm.200304513. [170] H.G. Kuivila, A.H. Keough, E.J. Soboczenski, J. Org. Chem. 19 (1954) 780–783,
[131] R. Francis, D.K. Baby, D.S. Kumar, J. Appl. Polym. Sci. 124 (2012) 5079–5088, http://dx.doi.org/10.1021/jo01370a013.
http://dx.doi.org/10.1002/app.35644. [171] J.P. Lorand, J.O. Edwards, J. Org. Chem. 24 (1959) 769–774, http://dx.doi.org/
[132] C.M. Nolan, M.J. Serpe, L.A. Lyon, Biomacromolecules 5 (2004) 1940–1946, 10.1021/jo01088a011.
http://dx.doi.org/10.1021/bm049750h. [172] S.A. Barker, A.K. Chopra, B.W. Hatt, P.J. Somers, Carbohydr. Res. 26 (1973) 33–40,
[133] D. Gan, L.A. Lyon, Macromolecules 35 (2002) 9634–9639, http://dx.doi.org/ http://dx.doi.org/10.1016/S0008-6215(00)85019-3.
10.1021/ma021186k. [173] T. Yang, R. Ji, X.-X. Deng, F.-S. Du, Z.-C. Li, Soft Matter. 10 (2014) 2671, http://
[134] M.A. Phillips, M.L. Gran, N.A. Peppas, Nano Today 5 (2010) 143–159, http:// dx.doi.org/10.1039/c3sm53059k.
dx.doi.org/10.1016/j.nantod.2010.03.003. [174] T. Hoare, R. Pelton, Biomacromolecules 9 (2008) 733–740, http://dx.doi.org/
[135] A. Gabizon, H. Shmeeda, Y. Barenholz, Clin. Pharmacokinet. 42 (2003) 419–436, 10.1021/bm701203r.
http://dx.doi.org/10.2165/00003088-200342050-00002. [175] K. Kataoka, H. Miyazaki, M. Bunya, T. Okano, Y. Sakurai, J. Am. Chem. Soc. 120
[136] A. Galperin, T.J. Long, B.D. Ratner, Biomacromolecules 11 (2010) 2583–2592, (1998) 12694–12695, http://dx.doi.org/10.1021/ja982975d.
http://dx.doi.org/10.1021/bm100521x. [176] S. Kitano, Y. Koyama, K. Kataoka, T. Okano, Y. Sakurai, J. Control. Release 19
[137] M.C. Hacker, L. Klouda, B.B. Ma, J.D. Kretlow, A.G. Mikos, Biomacromolecules 9 (1992) 161–170, http://dx.doi.org/10.1016/0168-3659(92)90073-Z.
(2008) 1558–1570, http://dx.doi.org/10.1021/bm8000414. [177] T.D. James, K.R.A.S. Sandanayake, R. Iguchi, S. Shinkai, J. Am. Chem. Soc. 117
[138] S.B. Turturro, M.J. Guthrie, A.A. Appel, P.W. Drapala, E.M. Brey, V.H. Pérez-Luna, (1995) 8982–8987, http://dx.doi.org/10.1021/ja00140a013.
W.F. Mieler, J.J. Kang-Mieler, Biomaterials 32 (2011) 3620–3626, http:// [178] A.M. Horgan, A.J. Marshall, S.J. Kew, K.E.S. Dean, C.D. Creasey, S. Kabilan, Biosens.
dx.doi.org/10.1016/j.biomaterials.2011.01.058. Bioelectron. 21 (2006) 1838–1845, http://dx.doi.org/10.1016/j.bios.2005.11.028.
[139] H.E. Canavan, X. Cheng, D.J. Graham, B.D. Ratner, D.G. Castner, J. Biomed. Mater. [179] D. Wang, T. Liu, J. Yin, S. Liu, Macromolecules 44 (2011) 2282–2290, http://
Res. A 75A (2005) 1–13, http://dx.doi.org/10.1002/jbm.a.30297. dx.doi.org/10.1021/ma200053a.
46 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

[180] B.G. De Geest, A.M. Jonas, J. Demeester, S.C. De Smedt, Langmuir 22 (2006) 5070– [219] M.J. Whitcombe, I. Chianella, L. Larcombe, S.A. Piletsky, J. Noble, R. Porter, A.
5074, http://dx.doi.org/10.1021/la053368o. Horgan, Chem. Soc. Rev. 40 (2011) 1547, http://dx.doi.org/10.1039/c0cs00049c.
[181] D. Shiino, Y. Murata, A. Kubo, Y.J. Kim, K. Kataoka, Y. Koyama, A. Kikuchi, M. [220] RNCOS, Global In Vitro Diagnostic Market Analysis, 2014 http://www.rncos.
Yokoyama, Y. Sakurai, T. Okano, J. Control. Release 37 (1995) 269–276, http:// com/Market-Analysis-Reports/
dx.doi.org/10.1016/0168-3659(95)00084-4. Global-In-Vitro-Diagnostic-Market-Analysis-IM103.htm (accessed 14.12.14).
[182] J. Drobnı́k, J. Kopeček, J. Labský, P. Rejmanová, J. Exner, V. Saudek, J. Kálal, [221] N. Lavignac, C.J. Allender, K.R. Brain, Anal. Chim. Acta 510 (2004) 139–145,
Makromol. Chem. 177 (1976) 2833–2848, http://dx.doi.org/10.1002/ http://dx.doi.org/10.1016/j.aca.2003.12.066.
macp.1976.021771003. [222] S.M. Reddy, Q.T. Phan, H. El-Sharif, L. Govada, D. Stevenson, N.E. Chayen, Bioma-
[183] J. Kopeček, I. Cı́fková, P. Rejmanová, J. Strohalm, B. Obereigner, K. Ulbrich, cromolecules 13 (2012) 3959–3965, http://dx.doi.org/10.1021/bm301189f.
Makromol. Chem. 182 (1981) 2941–2949, http://dx.doi.org/10.1002/ [223] A. Aghaei, M.R. Milani Hosseini, M. Najafi, Electrochim. Acta 55 (2010) 1503–
macp.1981.021821102. 1508, http://dx.doi.org/10.1016/j.electacta.2009.09.033.
[184] J. Kopeček, P. Kopečková, T. Minko, Z.-R. Lu, Eur. J. Pharm. Biopharm. 50 (2000) [224] P.-Y. Chen, R. Vittal, P.-C. Nien, G.-S. Liou, K.-C. Ho, Talanta 80 (2010) 1145–1151,
61–81, http://dx.doi.org/10.1016/S0939-6411(00)00075-8. http://dx.doi.org/10.1016/j.talanta.2009.08.041.
[185] T. Minko, P. Kopečková, J. Kopeček, Int. J. Cancer 86 (2000) 108–117, http:// [225] P.-Y. Chen, P.-C. Nien, C.-W. Hu, K.-C. Ho, Sens. Actuators B: Chem. 146 (2010)
dx.doi.org/10.1002/(SICI)1097-0215(20000401)86:1<108::AID-IJC17>3.0.CO;2-8. 466–471, http://dx.doi.org/10.1016/j.snb.2009.11.035.
[186] V. Omelyanenko, P. Kopečková, C. Gentry, J.-G. Shiah, J. Kopeček, J. Drug Target. 3 [226] C. Sontimuang, R. Suedee, F. Dickert, Anal. Biochem. 410 (2011) 224–233, http://
(1996) 357–373, http://dx.doi.org/10.3109/10611869608996827. dx.doi.org/10.1016/j.ab.2010.11.043.
[187] P. Rejmanová, J. Kopeček, R. Duncan, J.B. Lloyd, Biomaterials 6 (1985) 45–48, [227] T. Alizadeh, M. Zare, M.R. Ganjali, P. Norouzi, B. Tavana, Biosens. Bioelectron. 25
http://dx.doi.org/10.1016/0142-9612(85)90037-7. (2010) 1166–1172, http://dx.doi.org/10.1016/j.bios.2009.10.003.
[188] P. Rejmanová, J. Kopeček, J. Pohl, M. Baudyš, V. Kostka, Makromol. Chem. 184 [228] T.T. Tran, J. Li, H. Feng, J. Cai, L. Yuan, N. Wang, Q. Cai, Sens. Actuators B: Chem.
(1983) 2009–2020, http://dx.doi.org/10.1002/macp.1983.021841006. 190 (2014) 745–751, http://dx.doi.org/10.1016/j.snb.2013.09.048.
[189] P. Rejmanová, J. Labský, J. Kopeček, Makromol. Chem. 178 (1977) 2159–2168, [229] G. Vlatakis, L.I. Andersson, R. Müller, K. Mosbach, Nature 361 (1993) 645–647,
http://dx.doi.org/10.1002/macp.1977.021780803. http://dx.doi.org/10.1038/361645a0.
[190] M.V. Solovsky, K. Ulbrich, J. Kopecek, Biomaterials 4 (1983) 44–48, http:// [230] M.T. Muldoon, L.H. Stanker, J. Agric. Food Chem. 43 (1995) 1424–1427, http://
dx.doi.org/10.1016/0142-9612(83)90069-8. dx.doi.org/10.1021/jf00054a002.
[191] V. Šubr, J. Kopeček, J. Pohl, M. Baudyš, V. Kostka, J. Control. Release 8 (1988) 133– [231] M. Siemann, L.I. Andersson, K. Mosbach, J. Agric. Food Chem. 44 (1996) 141–145,
140, http://dx.doi.org/10.1016/0168-3659(88)90039-9. http://dx.doi.org/10.1021/jf950233n.
[192] M. Št’astný, B. Řı́hová, J. Strohalm, K. Ulbrich, J. Control. Release 42 (1996) 229– [232] L.I. Andersson, R. Müller, G. Vlatakis, K. Mosbach, Proc. Natl. Acad. Sci. 92 (1995)
236, http://dx.doi.org/10.1016/0168-3659(96)01463-0. 4788–4792.
[193] V. Chytrý, A. Vrána, J. Kopeček, Makromol. Chem. 179 (1978) 329–336, http:// [233] O. Ramström, L. Ye, K. Mosbach, Chem. Biol. 3 (1996) 471–477, http://dx.doi.org/
dx.doi.org/10.1002/macp.1978.021790207. 10.1016/S1074-5521(96)90095-2.
[194] S. Kim, K.E. Healy, Biomacromolecules 4 (2003) 1214–1223, http://dx.doi.org/ [234] J. Berglund, I.A. Nicholls, C. Lindbladh, K. Mosbach, Bioorg. Med. Chem. Lett. 6
10.1021/bm0340467. (1996) 2237–2242, http://dx.doi.org/10.1016/0960-894X(96)00406-4.
[195] L.C. Glangchai, M. Caldorera-Moore, L. Shi, K. Roy, J. Control. Release 125 (2008) [235] A.G. Mayes, L.I. Andersson, K. Mosbach, Anal. Biochem. 222 (1994) 483–488,
263–272, http://dx.doi.org/10.1016/j.jconrel.2007.10.021. http://dx.doi.org/10.1006/abio.1994.1521.
[196] J.L. West, J.A. Hubbell, Macromolecules 32 (1998) 241–244, http://dx.doi.org/ [236] L.I. Andersson, Anal. Chem. 68 (1996) 111–117, http://dx.doi.org/10.1021/
10.1021/ma981296k. ac950668+.
[197] G.T. Hermanson, in: G.T. Hermanson (Ed.), Bioconjugate Techniques, 2nd ed., [237] H. Bengtsson, U. Roos, L.I. Andersson, Anal. Commun. 34 (1997) 233–235, http://
Academic Press, New York, 2008, pp. 213–233, http://www.sciencedirect.com/ dx.doi.org/10.1039/A703977H.
science/article/pii/B9780123705013000035 (accessed 14.12.14). [238] A. Merkoçi, S. Alegret, TrAC Trends Anal. Chem. 21 (2002) 717–725, http://
[198] R.J. Ansell, J. Chromatogr. B 804 (2004) 151–165, http://dx.doi.org/10.1016/ dx.doi.org/10.1016/S0165-9936(02)01119-6.
j.jchromb.2004.02.022. [239] M.C. Blanco-López, M.J. Lobo-Castañón, A.J. Miranda-Ordieres, P. Tuñón-Blanco,
[199] B. Hock, M. Rahman, S. Rauchalles, A. Dankwardt, M. Seifert, S. Haindl, K. Kramer, TrAC Trends Anal. Chem. 23 (2004) 36–48, http://dx.doi.org/10.1016/S0165-
J. Mol. Catal. B: Enzym. 7 (1999) 115–124, http://dx.doi.org/10.1016/S1381- 9936(04)00102-5.
1177(99)00036-3. [240] J. Okuno, K. Maehashi, K. Kerman, Y. Takamura, K. Matsumoto, E. Tamiya,
[200] J.E. Butler, Methods 22 (2000) 4–23, http://dx.doi.org/10.1006/meth.2000.1031. Biosens. Bioelectron. 22 (2007) 2377–2381, http://dx.doi.org/10.1016/j.bi-
[201] C. Esen, M. Andac, N. Bereli, R. Say, E. Henden, A. Denizli, Mater. Sci. Eng. C 29 os.2006.09.038.
(2009) 2464–2470, http://dx.doi.org/10.1016/j.msec.2009.07.012. [241] T.L. Panasyuk, V.M. Mirsky, S.A. Piletsky, O.S. Wolfbeis, Anal. Chem. 71 (1999)
[202] Y. Tang, Z. Huang, T. Yang, X. Hu, X. Jiang, Anal. Lett. 38 (2005) 219–226, http:// 4609–4613, http://dx.doi.org/10.1021/ac9903196.
dx.doi.org/10.1081/AL-200045122. [242] T.L. Delaney, D. Zimin, M. Rahm, D. Weiss, O.S. Wolfbeis, V.M. Mirsky, Anal.
[203] Z. Zeng, Y. Hoshino, A. Rodriguez, H. Yoo, K.J. Shea, ACS Nano 4 (2009) 199–204, Chem. 79 (2007) 3220–3225, http://dx.doi.org/10.1021/ac062143v.
http://dx.doi.org/10.1021/nn901256s. [243] S. Suwansa-ard, P. Kanatharana, P. Asawatreratanakul, B. Wongkittisuksa, C.
[204] M.J. Whitcombe, E.N. Vulfson, Adv. Mater. 13 (2001) 467–478, http://dx.doi.org/ Limsakul, P. Thavarungkul, Biosens. Bioelectron. 24 (2009) 3436–3441, http://
10.1002/1521-4095(200104)13:7<467::AID-ADMA467>3.0.CO;2-T. dx.doi.org/10.1016/j.bios.2009.04.008.
[205] K. Mosbach, Trends Biochem. Sci. 19 (1994) 9–14, http://dx.doi.org/10.1016/ [244] G.M. Birnbaumer, P.A. Lieberzeit, L. Richter, R. Schirhagl, M. Milnera, F.L. Dickert,
0968-0004(94)90166-X. A. Bailey, P. Ertl, Lab. Chip. 9 (2009) 3549–3556, http://dx.doi.org/10.1039/
[206] K. Haupt, K. Mosbach, Trends Biotechnol. 16 (1998) 468–475, http://dx.doi.org/ B914738A.
10.1016/S0167-7799(98)01222-0. [245] J. Li, C.E. Kendig, E.E. Nesterov, J. Am. Chem. Soc. 129 (2007) 15911–15918,
[207] P.A.G. Cormack, K. Mosbach, React. Funct. Polym. 41 (1999) 115–124, http:// http://dx.doi.org/10.1021/ja0748027.
dx.doi.org/10.1016/S1381-5148(99)00024-3. [246] M. Jakusch, M. Janotta, B. Mizaikoff, K. Mosbach, K. Haupt, Anal. Chem. 71 (1999)
[208] G. Wulff, R. Schönfeld, Adv. Mater. 10 (1998) 957–959, http://dx.doi.org/ 4786–4791, http://dx.doi.org/10.1021/ac990050q.
10.1002/(SICI)1521-4095(199808)10:12<957::AID-ADMA957>3.0.CO;2-4. [247] D.-F. Tai, M.-H. Jhang, G.-Y. Chen, S.-C. Wang, K.-H. Lu, Y.-D. Lee, H.-T. Liu, Anal.
[209] G. Wulff, T. Gross, R. Schönfeld, T. Schrader, C. Kirsten, Molecular and Chem. 82 (2010) 2290–2293, http://dx.doi.org/10.1021/ac9024158.
Ionic Recognition with Imprinted Polymers, American Chemical Society, [248] O. Hayden, C. Haderspöck, S. Krassnig, X. Chen, F.L. Dickert, Analyst 131 (2006)
1998, pp. 10–28, http://dx.doi.org/10.1021/bk-1998-0703.ch002 (accessed 1044–1050, http://dx.doi.org/10.1039/B608354B.
09.12.14). [249] T.-Y. Lin, C.-H. Hu, T.-C. Chou, Biosens. Bioelectron. 20 (2004) 75–81, http://
[210] D. Spivak, K.J. Shea, J. Org. Chem. 64 (1999) 4627–4634, http://dx.doi.org/ dx.doi.org/10.1016/j.bios.2004.01.028.
10.1021/jo982118s. [250] M. Piliarik, H. Vaisocherová, J. Homola, in: A. Rasooly, K.E. Herold (Eds.),
[211] C. Lübke, M. Lübke, M.J. Whitcombe, E.N. Vulfson, Macromolecules 33 (2000) Biosensors and Bioelectronics, Humana Press, 2009, pp. 65–88, http://link.
5098–5105, http://dx.doi.org/10.1021/ma000467u. springer.com/protocol/10.1007/978-1-60327-567-5_5 (accessed 11.12.14).
[212] A. Poma, A.P.F. Turner, S.A. Piletsky, Trends Biotechnol. 28 (2010) 629–637, [251] C.-L. Choong, J.S. Bendall, W.I. Milne, Biosens. Bioelectron. 25 (2009) 652–656,
http://dx.doi.org/10.1016/j.tibtech.2010.08.006. http://dx.doi.org/10.1016/j.bios.2008.11.025.
[213] Y. Lv, T. Tan, F. Svec, Biotechnol. Adv. 31 (2013) 1172–1186, http://dx.doi.org/ [252] H.-Y. Lee, B.S. Kim, Biosens. Bioelectron. 25 (2009) 587–591, http://dx.doi.org/
10.1016/j.biotechadv.2013.02.005. 10.1016/j.bios.2009.03.040.
[214] E. Verheyen, J.P. Schillemans, M. van Wijk, M.-A. Demeniex, W.E. Hennink, C.F. [253] Z. Zhang, Y. Hu, H. Zhang, L. Luo, S. Yao, J. Electroanal. Chem. 644 (2010) 7–12,
van Nostrum, Biomaterials 32 (2011) 3008–3020, http://dx.doi.org/10.1016/ http://dx.doi.org/10.1016/j.jelechem.2010.03.015.
j.biomaterials.2011.01.007. [254] M. Riskin, R. Tel-Vered, O. Lioubashevski, I. Willner, J. Am. Chem. Soc. 131 (2009)
[215] M.E. Byrne, V. Salian, Int. J. Pharm. 364 (2008) 188–212, http://dx.doi.org/ 7368–7378, http://dx.doi.org/10.1021/ja9001212.
10.1016/j.ijpharm.2008.09.002. [255] D. Du, S. Chen, J. Cai, Y. Tao, H. Tu, A. Zhang, Electrochim. Acta 53 (2008) 6589–
[216] J. Steinke, D.C. Sherrington, I.R. Dunkin, Synthesis and Photosynthesis, Springer, 6595, http://dx.doi.org/10.1016/j.electacta.2008.04.027.
Berlin, Heidelberg, 1995, , pp. 81–125http://link.springer.com/chapter/10.1007/ [256] H.-F. Wang, Y. He, T.-R. Ji, X.-P. Yan, Anal. Chem. 81 (2009) 1615–1621, http://
3-540-58908-2_3 (accessed 09.12.14). dx.doi.org/10.1021/ac802375a.
[217] A.G. Mayes, K. Mosbach, TrAC Trends Anal. Chem. 16 (1997) 321–332, http:// [257] H.-Y. Lin, M.-S. Ho, M.-H. Lee, Biosens. Bioelectron. 25 (2009) 579–586, http://
dx.doi.org/10.1016/S0165-9936(97)00037-X. dx.doi.org/10.1016/j.bios.2009.03.039.
[218] T. Takeuchi, J. Haginaka, J. Chromatogr. B: Biomed. Sci. App. 728 (1999) 1–20, [258] M.-H. Lee, Y.-C. Chen, M.-H. Ho, H.-Y. Lin, Anal. Bioanal. Chem. 397 (2010) 1457–
http://dx.doi.org/10.1016/S0378-4347(99)00057-2. 1466, http://dx.doi.org/10.1007/s00216-010-3631-x.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 47

[259] C.I. Lin, A.K. Joseph, C.K. Chang, Y.D. Lee, J. Chromatogr. A 1027 (2004) 259–262, [300] Y. Kageyama, N. Tanigake, Y. Kurokome, S. Iwaki, S. Takeda, K. Suzuki, T. Sugawara,
http://dx.doi.org/10.1016/j.chroma.2003.10.037. Chem. Commun. 49 (2013) 9386, http://dx.doi.org/10.1039/c3cc43488e.
[260] C.I. Lin, A.K. Joseph, C.K. Chang, Y.D. Lee, Biosens. Bioelectron. 20 (2004) 127– [301] S. Matsumoto, S. Yamaguchi, S. Ueno, H. Komatsu, M. Ikeda, K. Ishizuka, Y. Iko,
131, http://dx.doi.org/10.1016/j.bios.2003.10.017. K.V. Tabata, H. Aoki, S. Ito, H. Noji, I. Hamachi, Chem. Eur. J. 14 (2008) 3977–3986,
[261] S.E. Diltemiz, R. Say, S. Büyüktiryaki, D. Hür, A. Denizli, A. Ersöz, Talanta 75 http://dx.doi.org/10.1002/chem.200701904.
(2008) 890–896, http://dx.doi.org/10.1016/j.talanta.2007.12.036. [302] S. Sugiura, A. Szilágyi, K. Sumaru, K. Hattori, T. Takagi, G. Filipcsei, M. Zrinyi, T.
[262] C. Alvarez-Lorenzo, A. Concheiro, J. Chromatogr. B 804 (2004) 231–245, http:// Kanamori, Lab. Chip. 9 (2009) 196, http://dx.doi.org/10.1039/b810717c.
dx.doi.org/10.1016/j.jchromb.2003.12.032. [303] S. Wang, M.-S. Choi, S.-H. Kim, J. Photochem. Photobiol. Chem. 198 (2008) 150–
[263] S.A. Zaidi, Drug Deliv. (2014) 1–10, http://dx.doi.org/10.3109/10717544. 155, http://dx.doi.org/10.1016/j.jphotochem.2008.03.002.
2014.970297. [304] D.R. Griffin, A.M. Kasko, J. Am. Chem. Soc. 134 (2012) 13103–13107, http://
[264] M.C. Norell, H.S. Andersson, I.A. Nicholls, J. Mol. Recognit. 11 (1998) 98–102, dx.doi.org/10.1021/ja305280w.
http://dx.doi.org/10.1002/(SICI)1099-1352(199812)11:1/6<98::AID- [305] N. Fomina, C. McFearin, M. Sermsakdi, O. Edigin, A. Almutairi, J. Am. Chem. Soc.
JMR399>3.0.CO;2-Y. 132 (2010) 9540–9542, http://dx.doi.org/10.1021/ja102595j.
[265] G. Ciardelli, B. Cioni, C. Cristallini, N. Barbani, D. Silvestri, P. Giusti, Biosens. [306] M.G. Kodzwa, M.E. Staben, D.G. Rethwisch, J. Membr. Sci. 158 (1999) 85–92,
Bioelectron. 20 (2004) 1083–1090, http://dx.doi.org/10.1016/j.bios.2004.06.028. http://dx.doi.org/10.1016/S0376-7388(99)00008-3.
[266] R. Suedee, Pharm. Anal. Acta 04 (2013), http://dx.doi.org/10.4172/2153- [307] Y. Chujo, K. Sada, T. Saegusa, Macromolecules 23 (1990) 2693–2697, http://
2435.1000264. dx.doi.org/10.1021/ma00212a017.
[267] R. Suedee, T. Srichana, G.P. Martin, J. Control. Release 66 (2000) 135–147, http:// [308] J. He, L. Tremblay, S. Lacelle, Y. Zhao, Soft Matter. 7 (2011) 2380, http://
dx.doi.org/10.1016/S0168-3659(99)00261-8. dx.doi.org/10.1039/c0sm01383h.
[268] L.A. Berrueta, B. Gallo, F. Vicente, Chromatographia 40 (1995) 474–483, http:// [309] F.M. Andreopoulos, C.R. Deible, M.T. Stauffer, S.G. Weber, W.R. Wagner, E.J.
dx.doi.org/10.1007/BF02269916. Beckman, A.J. Russell, J. Am. Chem. Soc. 118 (1996) 6235–6240, http://
[269] Y. Lv, Z. Lin, W. Feng, X. Zhou, T. Tan, Biochem. Eng. J. 36 (2007) 221–229, http:// dx.doi.org/10.1021/ja954117c.
dx.doi.org/10.1016/j.bej.2007.02.023. [310] Y. Zheng, M. Micic, S.V. Mello, M. Mabrouki, F.M. Andreopoulos, V. Konka, S.M.
[270] Y. Lv, Z. Lin, T. Tan, W. Feng, P. Qin, C. Li, Sens. Actuators B: Chem. 133 (2008) 15– Pham, R.M. Leblanc, Macromolecules 35 (2002) 5228–5234, http://dx.doi.org/
23, http://dx.doi.org/10.1016/j.snb.2008.01.067. 10.1021/ma012263z.
[271] J. Matsui, M. Okada, M. Tsuruoka, T. Takeuchi, Anal. Commun. 34 (1997) 85–87, [311] V.V. Ramanan, J.S. Katz, M. Guvendiren, E.R. Cohen, R.A. Marklein, J.A. Burdick, J.
http://dx.doi.org/10.1039/A700103G. Mater. Chem. 20 (2010) 8920, http://dx.doi.org/10.1039/c0jm00734j.
[272] B. Sellergren, Anal. Chem. 66 (1994) 1578–1582, http://dx.doi.org/10.1021/ [312] M.A. Azagarsamy, D.L. Alge, S.J. Radhakrishnan, M.W. Tibbitt, K.S. Anseth, Bioma-
ac00081a036. cromolecules 13 (2012) 2219–2224, http://dx.doi.org/10.1021/bm300646q.
[273] I. González-Mariño, J.B. Quintana, I. Rodrı́guez, R. Rodil, J. González-Peñas, R. [313] D.R. Griffin, A.M. Kasko, ACS Macro Lett. 1 (2012) 1330–1334, http://dx.doi.org/
Cela, J. Chromatogr. A 1216 (2009) 8435–8441, http://dx.doi.org/10.1016/j.chro- 10.1021/mz300366s.
ma.2009.09.069. [314] J.P. Gong, T. Nitta, Y. Osada, J. Phys. Chem. 98 (1994) 9583–9587, http://
[274] A. Beltran, F. Borrull, R.M. Marcé, P.A.G. Cormack, TrAC Trends Anal. Chem. 29 dx.doi.org/10.1021/j100089a036.
(2010) 1363–1375, http://dx.doi.org/10.1016/j.trac.2010.07.020. [315] Y. Osada, H. Okuzaki, H. Hori, Nature 355 (1992) 242–244, http://dx.doi.org/
[275] G. Qu, A. Wu, X. Shi, Z. Niu, W. Xie, D. Zhang, Anal. Lett. 41 (2008) 1443–1458, 10.1038/355242a0.
http://dx.doi.org/10.1080/00032710802119566. [316] Z. Liu, P. Calvert, Adv. Mater. 12 (2000) 288–291, http://dx.doi.org/10.1002/
[276] H. Yan, M. Tian, K.H. Row, J. Sep. Sci. 31 (2008) 3015–3020, http://dx.doi.org/ (SICI)1521-4095(200002)12:4<288::AID-ADMA288>3.0.CO;2-1.
10.1002/jssc.200800315. [317] F. Liu, M.W. Urban, Prog. Polym. Sci. 35 (2010) 3–23, http://dx.doi.org/10.1016/
[277] Y. Xiong, H. Zhou, Z. Zhang, D. He, C. He, Analyst 131 (2006) 829–834, http:// j.progpolymsci.2009.10.002.
dx.doi.org/10.1039/B606779B. [318] S.J. Kim, H.I. Kim, S.R. Shin, S.I. Kim, J. Appl. Polym. Sci. 92 (2004) 915–919, http://
[278] X.-L. Shi, M.-Q. Feng, J. Shi, Z.-H. Shi, J. Zhong, P. Zhou, Protein Expr. Purif. 54 dx.doi.org/10.1002/app.20059.
(2007) 24–29, http://dx.doi.org/10.1016/j.pep.2007.02.008. [319] M. Zrı́nyi, J. Fehér, G. Filipcsei, Macromolecules 33 (2000) 5751–5753, http://
[279] J.L. Urraca, M.D. Marazuela, M.C. Moreno-Bondi, Anal. Bioanal. Chem. 385 (2006) dx.doi.org/10.1021/ma000253c.
1155–1161, http://dx.doi.org/10.1007/s00216-006-0343-3. [320] C. He, S.W. Kim, D.S. Lee, J. Control. Release 127 (2008) 189–207, http://
[280] C. Cacho, E. Turiel, A. Martı́n-Esteban, D. Ayala, C. Pérez-Conde, J. Chromatogr. A dx.doi.org/10.1016/j.jconrel.2008.01.005.
1114 (2006) 255–262, http://dx.doi.org/10.1016/j.chroma.2006.02.051. [321] H. Li, Smart Hydrogel Modelling, Springer Science & Business Media, 2010.
[281] F.G. Tamayo, A. Martin-Esteban, J. Chromatogr. A 1098 (2005) 116–122, http:// [322] F.G. Helfferich, Ion Exchange, Courier Corporation, 1962.
dx.doi.org/10.1016/j.chroma.2005.08.068. [323] A.R. Khare, N.A. Peppas, Biomaterials 16 (1995) 559–567, http://dx.doi.org/
[282] M. Hu, M. Jiang, P. Wang, S. Mei, Y. Lin, X. Hu, Y. Shi, B. Lu, K. Dai, Anal. Bioanal. 10.1016/0142-9612(95)91130-Q.
Chem. 387 (2007) 1007–1016, http://dx.doi.org/10.1007/s00216-006-1004-2. [324] Y.K. Yew, T.Y. Ng, H. Li, K.Y. Lam, Biomed. Microdevices 9 (2007) 487–499, http://
[283] F. Chapuis, J.-U. Mullot, V. Pichon, G. Tuffal, M.-C. Hennion, J. Chromatogr. A 1135 dx.doi.org/10.1007/s10544-007-9056-4.
(2006) 127–134, http://dx.doi.org/10.1016/j.chroma.2006.09.076. [325] H. Li, T.Y. Ng, Y.K. Yew, K.Y. Lam, Biomacromolecules 6 (2005) 109–120, http://
[284] F. Hugon-Chapuis, J.U. Mullot, G. Tuffal, M.-C. Hennion, V. Pichon, J. Chroma- dx.doi.org/10.1021/bm0496458.
togr. A 1196–1197 (2008) 73–80, http://dx.doi.org/10.1016/j.chroma.2008. [326] S.J. Kim, M.S. Kim, S.I. Kim, G.M. Spinks, B.C. Kim, G.G. Wallace, Chem. Mater. 18
04.038. (2006) 5805–5809, http://dx.doi.org/10.1021/cm060988h.
[285] E. Caro, R.M. Marcé, P.A.G. Cormack, D.C. Sherrington, F. Borrull, Anal. Chim. Acta [327] T.f. Otero, M.t. Cortés, Adv. Mater. 15 (2003) 279–282, http://dx.doi.org/10.1002/
562 (2006) 145–151, http://dx.doi.org/10.1016/j.aca.2006.01.080. adma.200390066.
[286] B. Claude, P. Morin, S. Bayoudh, J. de Ceaurriz, J. Chromatogr. A 1196–1197 [328] R.H. Baughman, L.W. Shacklette, R.L. Elsenbaumer, E. Plichta, C. Becht, in: J.L.
(2008) 81–88, http://dx.doi.org/10.1016/j.chroma.2008.05.022. Brédas, R.R. Chance (Eds.), Conjugated Polymeric Materials: Opportunities in
[287] A. Beltran, R.M. Marcé, P.A.G. Cormack, D.C. Sherrington, F. Borrull, J. Sep. Sci. 31 Electronics, Optoelectronics, and Molecular Electronics, Springer, Netherlands,
(2008) 2868–2874, http://dx.doi.org/10.1002/jssc.200800085. 1990 , pp. 559–582, http://link.springer.com/chapter/10.1007/978-94-009-
[288] A. Beltran, N. Fontanals, R.M. Marcé, P.A.G. Cormack, F. Borrull, J. Sep. Sci. 32 2041-5_44 (accessed 14.12.14).
(2009) 3319–3326, http://dx.doi.org/10.1002/jssc.200900268. [329] R.H. Baughman, Synth. Met. 78 (1996) 339–353, http://dx.doi.org/10.1016/
[289] R. Suedee, V. Seechamnanturakit, B. Canyuk, C. Ovatlarnporn, G.P. Martin, J. 0379-6779(96)80158-5.
Chromatogr. A 1114 (2006) 239–249, http://dx.doi.org/10.1016/j.chro- [330] G.G. Wallace, P.R. Teasdale, G.M. Spinks, L.A.P. Kane-Maguire, Conductive Elec-
ma.2006.02.033. troactive Polymers: Intelligent Materials Systems, 2nd ed., CRC Press, 2002.
[290] M. Le Noir, F. Plieva, T. Hey, B. Guieysse, B. Mattiasson, J. Chromatogr. A 1154 [331] T.W. Lewis, G.M. Spinks, G.G. Wallace, A. Mazzoldi, D. De Rossi, Synth. Met. 122
(2007) 158–164, http://dx.doi.org/10.1016/j.chroma.2007.03.064. (2001) 379–385, http://dx.doi.org/10.1016/S0379-6779(00)00397-0.
[291] R. Carabias-Martı́nez, E. Rodrı́guez-Gonzalo, E. Herrero-Hernández, J. Chro- [332] A. Mazzoldi, D. De Rossi, in: Y. Bar-Cohen (Ed.), Conductive-Polymer-Based
matogr. A 1085 (2005) 199–206, http://dx.doi.org/10.1016/j.chroma.2005. Structures for a Steerable Catheter, 2000, 273–280, http://dx.doi.org/10.1117/
05.084. 12.387786.
[292] K. Hoshina, S. Horiyama, H. Matsunaga, J. Haginaka, J. Chromatogr. A 1216 (2009) [333] J.D. Madden, P.G. Madden, I.W. Hunter, Conducting Polymer Actuators as
4957–4962, http://dx.doi.org/10.1016/j.chroma.2009.04.071. Engineering Materials, 2002, 176–190, http://dx.doi.org/10.1117/12.475163.
[293] Z. Sun, W. Schüssler, M. Sengl, R. Niessner, D. Knopp, Anal. Chim. Acta 620 (2008) [334] W. Takashima, M. Kaneko, K. Kaneto, A.G. MacDiarmid, Synth. Met. 71 (1995)
73–81, http://dx.doi.org/10.1016/j.aca.2008.05.020. 2265–2266, http://dx.doi.org/10.1016/0379-6779(94)03252-2.
[294] D.K. Alexiadou, N.C. Maragou, N.S. Thomaidis, G.A. Theodoridis, M.A. Koupparis, [335] S.M. Ha, W. Yuan, Q. Pei, R. Pelrine, S. Stanford, Adv. Mater. 18 (2006) 887–891,
J. Sep. Sci. 31 (2008) 2272–2282, http://dx.doi.org/10.1002/jssc.200700643. http://dx.doi.org/10.1002/adma.200502437.
[295] C.A. DeForest, K.S. Anseth, Nat. Chem. 3 (2011) 925–931, http://dx.doi.org/ [336] S. Liang, J. Xu, L. Weng, L. Zhang, X. Guo, X. Zhang, J. Phys. Chem. B 111 (2007)
10.1038/nchem.1174. 941–945, http://dx.doi.org/10.1021/jp0673821.
[296] R.P. Wayne, Photochemistry, 1st ed., The Butterworth Group, 1970. [337] T. Hirai, H. Nemoto, T. Suzuki, S. Hayashi, M. Hirai, J. Intell. Mater. Syst. Struct. 4
[297] M.W. Tibbitt, A.M. Kloxin, K.S. Anseth, J. Polym. Sci. A: Polym. Chem. 51 (2013) (1993) 277–279, http://dx.doi.org/10.1177/1045389X9300400219.
1899–1911, http://dx.doi.org/10.1002/pola.26574. [338] T. Hirai, H. Nemoto, M. Hirai, S. Hayashi, J. Appl. Polym. Sci. 53 (1994) 79–84,
[298] B.J. Adzima, Y. Tao, C.J. Kloxin, C.A. DeForest, K.S. Anseth, C.N. Bowman, Nat. http://dx.doi.org/10.1002/app.1994.070530109.
Chem. 3 (2011) 256–259, http://dx.doi.org/10.1038/nchem.980. [339] T. Hirai, M. Hirai, in: P.Y. Osada, P.D.E.D. Rossi (Eds.), Polymer Sensors and
[299] C.J. Barrett, J. Mamiya, K.G. Yager, T. Ikeda, Soft Matter. 3 (2007) 1249, http:// Actuators, Springer, Berlin, Heidelberg, 2000, pp. 245–258, http://link.springer.
dx.doi.org/10.1039/b705619b. com/chapter/10.1007/978-3-662-04068-3_8 (accessed 14.12.14).
48 M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49

[340] S. Murdan, J. Control. Release 92 (2003) 1–17, http://dx.doi.org/10.1016/S0168- [394] L. Heymann, S. Peukert, N. Aksel, J. Rheol. 46 (2002) 93–112.
3659(03)00303-1. [395] L. Aulisa, H. Dong, J.D. Hartgerink, Biomacromolecules 10 (2009) 2694–2698.
[341] M.B. Delgado-Charro, R.H. Guy, STP Pharma Sci. 11 (2001) 404–414. [396] R. Barbucci, S. Lamponi, A. Borzacchiello, L. Ambrosio, M. Fini, P. Torricelli, R.
[342] R. Vanbever, V. Préat, Adv. Drug Deliv. Rev. 35 (1999) 77–88, http://dx.doi.org/ Giardino, Biomaterials 23 (2002) 4503–4513.
10.1016/S0169-409X(98)00064-7. [397] Y. Li, K. Fukushima, D.J. Coady, A.C. Engler, S. Liu, Y. Huang, J.S. Cho, Y. Guo, L.S.
[343] J.E. Riviere, M.C. Heit, Pharm. Res. 14 (1997) 687–697, http://dx.doi.org/10.1023/ Miller, J.P.K. Tan, P.L.R. Ee, W. Fan, Y.Y. Yang, J.L. Hedrick, Angew. Chem. 52
A:1012129801406. (2013) 674–678.
[344] J.T. Santini, M.J. Cima, R. Langer, Nature 397 (1999) 335–338, http://dx.doi.org/ [398] N.M. Morsi, G.A. Abdelbary, M.A. Ahmed, Eur. J. Pharm. Biopharm. 86 (2014)
10.1038/16898. 178–189.
[345] P.E. Grimshaw, A.J. Grodzinsky, M.L. Yarmush, D.M. Yarmush, Chem. Eng. Sci. 44 [399] L. Yu, J. Ding, Chem. Soc. Rev. 37 (2008) 1473–1481.
(1989) 827–840, http://dx.doi.org/10.1016/0009-2509(89)85256-X. [400] J.S. Temenoff, A.G. Mikos, Biomaterials 21 (2000) 2405–2412.
[346] V.A. Pasechnik, A.N. Cherkasov, V.P. Zhemkov, V.V. Chechina, R.S. Alimardanov, [401] W.S. Shim, J. Kim, H.K. Park, K.M. Kim, I.C. Kwan, D.S. Lee, Biomaterials 27 (2006)
Effect of Direct-Current on the Permeability of Ultrafiltration Membranes, 5178–5185.
Plenum Publ. Corp. Consultants Bureau, New York, NY, 1979. [402] J.C. Garbern, E. Minami, P.S. Stayton, C.E. Murry, Biomaterials 32 (2011) 2407–2416.
[347] P. Burgmayer, R.W. Murray, J. Am. Chem. Soc. 104 (1982) 6139–6140, http:// [403] W.S. Shim, J.S. Yoo, Y.H. Bae, D.S. Lee, Biomacromolecules 6 (2005) 2930–2934.
dx.doi.org/10.1021/ja00386a061. [404] Y.-L. Chiu, S.-C. Chen, C.-J. Su, C.-W. Hsiao, Y.-M. Chen, H.-L. Chen, H.-W. Sung,
[348] R.K. Bhaskar, R.V. Sparer, K.J. Himmelstein, J. Membr. Sci. 24 (1985) 83–96, Biomaterials 30 (2009) 4877–4888.
http://dx.doi.org/10.1016/S0376-7388(00)83210-X. [405] E. Ruel-Garièpy, J.-C. Leroux, Eur. J. Pharm. Biopharm. 58 (2004) 409–426.
[349] S.Y. Kim, Y.M. Lee, J. Appl. Polym. Sci. 74 (1999) 1752–1761, http://dx.doi.org/ [406] B. Jeong, S.W. Kim, Y.H. Bae, Adv. Drug Deliv. Rev. 54 (2002) 37–51.
10.1002/(SICI)1097-4628(19991114)74:7<1752::AID-APP18>3.0.CO;2-H. [407] A. Rozier, C. Mazuel, J. Grove, B. Plazonnet, Int. J. Pharm. 57 (1989) 163–168.
[350] S. Ramanathan, L.H. Block, J. Control. Release 70 (2001) 109–123, http:// [408] E. Westerhaus, P.B. Messersmith, Biomaterials 22 (2001) 453–462.
dx.doi.org/10.1016/S0168-3659(00)00333-3. [409] S. Suri, R. Banerjee, J. Biomed. Mater. Res. A 79A (2006) 650–664.
[351] K. Sawahata, M. Hara, H. Yasunaga, Y. Osada, J. Control. Release 14 (1990) 253– [410] C. Yan, A. Altunbas, T. Yucel, R.P. Nagarkar, J.P. Schneider, D.J. Pochan, Soft
262, http://dx.doi.org/10.1016/0168-3659(90)90165-P. Matter. 6 (2010) 5143–5156.
[352] K. Sutani, I. Kaetsu, K. Uchida, Radiat. Phys. Chem. 61 (2001) 49–54, http:// [411] L. Haines-Butterick, K. Rajagopal, M. Branco, D. Salick, R. Rughani, M. Pilarz, M.S.
dx.doi.org/10.1016/S0969-806X(00)00381-9. Lamm, D.J. Pochan, J.P. Schneider, Proc. Natl. Acad. Sci. 104 (2007) 7791–7796.
[353] S.H. Yuk, S.H. Cho, H.B. Lee, Pharm. Res. 9 (1992) 955–957, http://dx.doi.org/ [412] K.L. Niece, C. Czeisler, V. Sahni, V. Tysseling-Mattiace, E.T. Pashuck, J.A. Kessler,
10.1023/A:1015821504229. S.I. Stupp, Biomaterials 29 (2008) 4501–4509.
[354] I.C. Kwon, Y.H. Bae, T. Okano, S.W. Kim, J. Control. Release 17 (1991) 149–156, [413] B.D. Olsen, J.A. Kornfield, D.A. Tirrell, Macromolecules 43 (2010) 9094–9099.
http://dx.doi.org/10.1016/0168-3659(91)90054-H. [414] Z. Sui, W.J. King, W.L. Murphy, Adv. Funct. Mater. 18 (2008) 1824–1831.
[355] C.-S. Hsu, L.H. Block, Pharm. Res. 13 (1996) 1865–1870, http://dx.doi.org/ [415] A.M. Jonker, D.W.P.M. Löwik, J.C.M. van Hest, Chem. Mater. 24 (2012) 759–773.
10.1023/A:1016045427545. [416] G.H. Altman, F. Diaz, C. Jakuba, T. Calabro, R.L. Horan, J. Chen, H. Lu, J. Richmond,
[356] M. Jensen, P. Birch Hansen, S. Murdan, S. Frokjaer, A.T. Florence, Eur. J. Pharm. Sci. D.L. Kaplan, Biomaterials 24 (2003) 401–416.
15 (2002) 139–148, http://dx.doi.org/10.1016/S0928-0987(01)00193-2. [417] J. Perez-Riguerio, C. Viney, J. Llorca, M. Elices, J. Appl. Polym. Sci. 70 (1998)
[357] I.C. Kwon, Y.H. Bae, S.W. Kim, Nature 354 (1991) 291–293, http://dx.doi.org/ 2439–2447.
10.1038/354291a0. [418] D.L. Kaplan, Polym. Degrad. Stab. 59 (1998) 25–32.
[358] I.C. Kwon, Y.H. Bae, S.W. Kim, J. Control. Release 30 (1994) 155–159, http:// [419] A. Matsumoto, J. Chen, A.L. Collette, U.-J. Kim, G.H. Altman, P. Cebe, D.L. Kaplan, J.
dx.doi.org/10.1016/0168-3659(94)90262-3. Phys. Chem. B 110 (2006) 21630–21638.
[359] H.B. Hopfenberg, Control. Release Polym. Formul. (1976) 26–32, http:// [420] S. Inoue, K. Tanaka, F. Arisaka, S. Kimura, K. Ohtomo, S. Mizuno, J. Biol. Chem. 275
dx.doi.org/10.1021/bk-1976-0033.ch003, American Chemical Society (accessed (2000) 40517–40528.
27.11.14). [421] Y. Cao, B. Wang, Int. J. Mol. Sci. 10 (2009) 1514–1524.
[360] S. Kagatani, T. Shinoda, Y. Konno, M. Fukui, T. Ohmura, Y. Osada, J. Pharm. Sci. 86 [422] R.L. Horan, K. Antle, A.L. Collette, Y. Wang, J. Huang, J.E. Moreau, V. Volloch, D.L.
(1997) 1273–1277, http://dx.doi.org/10.1021/js9700762. Kaplan, G.H. Altman, Biomaterials 26 (2005) 3385–3393.
[361] A.S. Hoffman, Adv. Mater. 65 (2013) 10–16. [423] Y. Wang, H.-J. Kim, G. Vunjak-Novakovic, D.L. Kaplan, Biomaterials 27 (2006)
[362] Y. Qiu, K. Park, Adv. Drug Deliv. Rev. 53 (2001) 321–339. 6064–6082.
[363] Q. Wang, L. Wang, M.S. Detamore, C. Berkland, Adv. Mater. 20 (2008) 236–239. [424] A.H. Zuglul, A. Mitsuo, H. Kiyoshi, Biosci. Biotechnol. Biochem. 57 (1993) 1910–
[364] C.J. Bell, L.M. Carrick, J. Katta, Z. Jin, E. Ingham, A. Aggeli, N. Boden, T.A. Waigh, J. 1912.
Fisher, J. Biomed. Mater. Res. A 78A (2006) 236–246. [425] C. Vepari, D.L. Kaplan, Prog. Polym. Sci. 32 (2007) 991–1007.
[365] M. Guvendiren, H.D. Lu, J.A. Burdick, Soft Matter. 8 (2012) 260–272. [426] X. Wang, J.A. Kluge, G.G. Leisk, D.L. Kaplan, Biomaterials 29 (2008) 1054–1064.
[366] A. Gutowska, B. Jeong, M. Jasionowski, Anat. Rec. 263 (2001) 342–349. [427] T. Yucel, P. Cebe, D.L. Kaplan, Biophys. J. 97 (2009) 2044–2050.
[367] C. Yan, D.J. Pochan, Chem. Soc. Rev. 39 (2010) 3528–3540. [428] M.K. Sah, K. Pramanik, Afr. J. Biotechnol. 10 (2013) 7878–7892.
[368] J.D. Ferry, Viscoelastic Properties of Polymers, 3rd ed., John Wiley & Sons, 1980. [429] S. Banta, I.R. Wheeldon, M. Blenner, Annu. Rev. Biomed. Eng. 12 (2010) 167–186.
[369] N.G. McCrum, C.P. Buckley, C.B. Bucknall, Princples of Polymer Engineering, 2nd [430] W.H. Landschulz, P.F. Johnson, S.L. McKnight, Science 240 (1988) 1759–1764.
ed., Oxford Science Publications, 1977. [431] B. Ciani, E.G. Hutchinson, R.B. Sessions, D.N. Woolfson, J. Biol. Chem. 227 (2002)
[370] P.C. Hiemenz, T.P. Lodge, Polymer Chemistry, 2nd ed., Taylor & Francis, 2007. 10150–10155.
[371] T.G. Mezger, Rheology Handbook, 2nd ed., Vincentz Network, Hannover, 2006. [432] S.B. Kennedy, E.R. deAzevedo, W.A. Petka, T.P. Russell, D.A. Tirrell, M. Hong,
[372] M.A. Meyers, K.K. Chawla, Mechanical Behavior of Materials, 2nd ed., Cam- Macromolecules 34 (2001) 8675–8685.
bridge University Press, 2008. [433] L. Mi, S. Fischer, B. Chung, S. Sundelacruz, J.L. Harden, Biomacromolecules 7
[373] H. Sundaram, B. Voigts, K. Beer, M. Meland, Dermatol. Surg. 36 (2010) 1859–1865. (2006) 38–47.
[374] W.N. Findley, J.S. Lai, K. Onaran, Creep and Relaxation of Nonlinear Viscoelastic [434] S.B. Kennedy, K. Littrell, P. Thiyagarajan, D.A. Tirrell, T.P. Russell, Macromole-
Materials, Dover Publications, 1976. cules 38 (2005) 7470–7475.
[375] D. Roylance, Engineering Viscoelasticity, 2001 http://web.mit.edu/course/3/3. [435] H.-A. Klok, Macromol. Perspect. 42 (2009) 7990–8000.
11/www/modules/visco.pdf. [436] C. Wang, J. Kopeček, R.J. Stewart, Biomacromolecules 2 (2001) 912–920.
[376] S.B. Ross-Murphy, Polym. Gels Netw. 2 (1994) 229–237. [437] C. Wang, R.J. Stewart, J. Kopeček, Nature 397 (1999) 417–420.
[377] G.M. Kavanagh, S.B. Ross-Murphy, Prog. Polym. Sci. 23 (1998) 533–562. [438] A. Tang, C. Wang, R.J. Stewart, J. Kopeček, J. Control. Release 72 (2001) 57–70.
[378] C.W. Macosko, Rheology: Principles, Measurements, and Applications, Wiley- [439] S. Fischer, X. Liu, H.-Q. Mao, J.L. Harden, Biomaterials 28 (2007) 3325–3337.
VCM, 1994. [440] W. Shen, J.A. Kornfield, D.A. Tirrell, Soft Matter. 3 (2007) 99–107.
[379] Y. Fang, K. Nishinari, Biopolymers 73 (2004) 44–60. [441] P.J. Skrzeszewska, J. Sprakel, F.A. de Wolf, R. Fokkink, M.A.C. Stuart, J. van der
[380] K. Pal, A.K. Banthia, D.K. Majumdar, Des. Monomers Polym. 12 (2009) 197–220. Gucht, Macromolecules 43 (2010) 3542–3548.
[381] H.A. Barnes, J. Non-Newton. Fluid Mech. 70 (1997) 1–33. [442] A.P. Nowak, V. Breedveld, L. Pakstis, B. Ozbas, D.J. Pine, D. Pochan, T.J. Deming,
[382] K. Hyun, S.H. Kim, K.H. Ahn, S.J. Lee, J. Non-Newton. Fluid Mech. 107 (2002) 51–65. Nature 417 (2002) 424–428.
[383] A.J. Engler, S. Sen, H.L. Sweeney, D.E. Discher, Cell 126 (2006) 677–689. [443] L. Pakstis, B. Ozbas, K.D. Hales, A.P. Nowak, T.J. Deming, D.J. Pochan, Biomacro-
[384] J.D. Kretlow, L. Klouda, A.G. Mikos, Adv. Drug Deliv. Rev. 59 (2007) 263–273. molecules 5 (2004) 312–318.
[385] R.S. Jeyaseelan, J. Giacomin, J. Non-Newton. Fluid Mech. 148 (2008) 24–32. [444] V. Breedveld, A.P. Nowak, J. Sato, T.J. Deming, D.J. Pine, Macromolecules 37
[386] R.H. Ewoldt, A.E. Hosoi, G.H. McKinley, Integr. Comp. Biol. 49 (2009) 40–50. (2004) 3943–3953.
[387] K. Tan, S. Cheng, L. Juge, L.E. Bilston, J. Biomech. 46 (2013) 1060–1066. [445] Z. Li, T.J. Deming, Soft Matter. 6 (2010) 2546–2551.
[388] K. Hyun, M. Wilhelm, C.O. Klein, K.S. Cho, J.G. Nam, K.H. Ahn, S.J. Lee, R.H. Ewoldt, [446] T.J. Deming, Soft Matter. 1 (2005) 28–35.
G.H. McKinley, Prog. Polym. Sci. 36 (2011) 1697–1753. [447] C.-Y. Yang, B. Song, Y. Ao, A.P. Nowak, R.B. Abelowitz, R.A. Korsak, L.A. Havton, T.J.
[389] W.H. Boersma, J. Laven, H.N. Stein, AIChE J. 36 (1990) 321–332. Deming, M.V. Sofroniew, Biomaterials 30 (2009) 2881–2898.
[390] I.D. Morrison, S. Ross, Colloidal Dispersions: Suspensions, Emulsions, and Foams, [448] C.T.S.W.P. Foo, J.S. Lee, W. Mulyasasmita, A. Parisi-Amon, S.C. Heilshorn, Proc.
Wiley-Interscience, New York, 2002. Natl. Acad. Sci. 106 (2009) 22067–22072.
[391] P.N. Patel, C.K. Smith, C.W. Patrick Jr., Biomed. Mater. Res. A 73A (2005) 313–319. [449] H.I. Chen, A. Einbond, S.-J. Kwak, H. Linn, E. Koepf, S. Peterson, J.W. Kelly, M.
[392] H.F. Brinson, L.C. Brinson, Polymer Engineering Science and Viscoelasticity: An Sudol, J. Biol. Chem. 272 (1997) 17070–17077.
Introduction, Springer, 2007. [450] M. Sudol, H.I. Chen, C. Bougeret, A. Einbond, P. Bork, FEBS Lett. 369 (1995) 67–71.
[393] G. Harrison, G.V. Franks, V. Tirtaatmadja, D.V. Boger, Korea–Aust. Rheol. J. 11 [451] J.R. Pires, F. Taha-Nejad, F. Toepert, T. Ast, U. Hoffmüller, J. Schneider-Mergener,
(1999) 197–218. R. Kühne, M.J. Macias, H. Oschkinat, J. Mol. Biol. 314 (2001) 1147–1156.
M.C. Koetting et al. / Materials Science and Engineering R 93 (2015) 1–49 49

[452] V. Kanelis, D. Rotin, J.D. Forman-Kay, Nat. Struct. Mol. Biol. 8 (2001) 407–412. [484] L. Li, P.M.T. Hangamathesvaran, C.Y. Yue, K.C. Tam, X. Hu, Y.C. Lam, Langmuir 17
[453] A. Parisi-Amon, W. Mulyasasmita, C. Chung, S.C. Heilshorn, Adv. Healthc. Mater. (2001) 8062–8068.
2 (2013) 428–432. [485] M.C. Tate, D.A. Shear, S.W. Hoffman, D.G. Stein, M.C. LaPlaca, Biomaterials 22
[454] R.J. Mart, R.D. Osborne, M.M. Stevens, R.V. Ulijn, Soft Matter. 2 (2006) 822–835. (2001) 1113–1123.
[455] J.P. Schneider, D.J. Pochan, B. Ozbas, K. Rajagopal, L. Pakstis, J. Kretsinger, J. Am. [486] K.P. Vercruysse, G.D. Prestwich, Crit. Rev. Ther. Carr. Syst. 15 (1998) 513–555.
Chem. Soc. 124 (2002) 15030–15037. [487] E.A. Balazs, Medical Applications of Hyaluronan and Its Derivatives, Plenum
[456] D.J. Pochan, J.P. Schneider, J. Kretsinger, B. Ozbas, K. Rajagopal, L. Haines, J. Am. Press, New York, 1991.
Chem. Soc. 125 (2003) 11802–11803. [488] K. Moriyama, T. Ooya, N. Yui, J. Control. Release 59 (1999) 77–86.
[457] L. Haines, K. Rajagopal, B. Ozbas, D. Salick, D.J. Pochan, J.P. Schneider, J. Am. [489] C. Weiss, New Frontiers in Medical Sciences: Redefining Hyaluronan, Weigel
Chem. Soc. 127 (2005) 17025–17029. GAaPH, 2000p. 89.
[458] B. Ozbas, K. Rajagopal, J.P. Schneider, D.J. Pochan, Phys. Rev. Lett. 93 (2004) [490] D. Gupta, C.H. Tator, M.S. Shoichet, Biomaterials 27 (2006) 2370–2379.
268106. [491] M.J. Caicco, T. Zahir, A.J. Mothe, B.G. Ballios, A.J. Kihm, C.H. Tator, M.S. Shoichet, J.
[459] D. Salick, D.J. Pochan, J.P. Schneider, Adv. Mater. 21 (2009) 4120–4123. Biomed. Mater. Res. A 101A (2012) 1472–1477.
[460] K. Rajagopal, M.S. Lamm, L. Haines-Butterick, D.J. Pochan, J.P. Schneider, Bio- [492] C. Kang, C.H. Tator, M.S. Shoichet, J. Control. Release 144 (2010) 25–31.
macromolecules 10 (2009) 2619–2625. [493] M.S. Shoichet, C.H. Tator, P. Poon, C. Kang, M.D. Baumann, Prog. Brain Res. 161
[461] R. Rughani, D. Salick, M.S. Lamm, T. Yucel, D.J. Pochan, J.P. Schneider, Bioma- (2007) 385–392.
cromolecules 10 (2009) 1295–1304. [494] Y. Wang, Y. Lapitsky, C. Kang, M.S. Shoichet, J. Control. Release 140 (2009) 218–223.
[462] B. Ozbas, J. Kretsinger, K. Rajagopal, J.P. Schneider, D.J. Pochan, Macromolecules [495] C.B. Rodell, A.L. Kaminski, J.A. Burdick, Biomacromolecules 14 (2013) 4125–4134.
37 (2004) 7331–7337. [496] A.A. Spasov, T.V. Khamidova, L.I. Bugaeva, I.S. Morozov, Pharm. Chem. J. 34
[463] T. Yucel, C.M. Micklitsch, J.P. Schneider, D.J. Pochan, Macromolecules 41 (2008) (2000) 1–7.
5763–5772. [497] J. Szejtli, Chem. Rev. 98 (1998) 1743–1753.
[464] C. Yan, M.E. Mackay, K. Czymmek, R.P. Nagarkar, J.P. Schneider, D.J. Pochan, [498] A. Hashidzume, A. Harada, Polym. Chem. 2 (2011) 2146–2154.
Langmuir 28 (2012) 6076–6087. [499] J. Li, X.J. Loh, Adv. Drug Deliv. Rev. 60 (2008) 1000–1017.
[465] L. Haines-Butterick, D. Salick, D.J. Pochan, J.P. Schneider, Biomaterials 29 (2008) [500] J. Araki, K. Ito, Soft Matter. 3 (2007) 1456–1473.
4164–4169. [501] G. Chen, M. Jiang, Chem. Soc. Rev. 40 (2011) 2254–2266.
[466] R. Rughani, J.P. Schneider, MRS Bull. 33 (2008) 530–535. [502] A. Harada, J. Li, M. Kamachi, Nature 356 (1992) 325–327.
[467] J. Kretsinger, L. Haines, B. Ozbas, D.J. Pochan, J.P. Schneider, Biomaterials 26 [503] J. Li, X. Ni, Z. Zhou, K.W. Leong, J. Am. Chem. Soc. 125 (2003) 1788–1795.
(2005) 5177–5186. [504] A. Harada, K. Kataoka, Prog. Polym. Sci. 31 (2006) 949–982.
[468] D. Salick, J. Kretsinger, D.J. Pochan, J.P. Schneider, J. Am. Chem. Soc. 129 (2007) [505] J. Li, X. Li, X. Ni, X. Wang, H. Li, K.W. Leong, Biomaterials 27 (2006) 4132–4140.
14793–14799. [506] M.J. Solomon, P.T. Spicer, Soft Matter. 6 (2010) 1391–1400.
[469] M. Branco, D.J. Pochan, N. Wagner, J.P. Schneider, Biomaterials 30 (2009) [507] E. Zaccarelli, J. Phys. Condens. Matter. 19 (2007) 323101.
1339–1347. [508] Q. Wang, J. Wang, Q. Lu, M.S. Detamore, C. Berkland, Biomaterials 31 (2010)
[470] A. Altunbas, S.J. Lee, S.A. Rajasekaran, J.P. Schneider, D.J. Pochan, Biomaterials 32 4980–4986.
(2011) 5906–5914. [509] Q. Wang, S. Jamal, M.S. Detamore, C. Berkland, J. Biomed. Mater. Res. A 96A
[471] S. Ramachandran, Y. Tseng, Y.B. Yu, Biomacromolecules 6 (2005) 1316–1321. (2011) 520–527.
[472] S. Ramachandran, P. Flynn, Y. Tseng, Y.B. Yu, Chem. Mater. 17 (2005) [510] M.E. Keegan, J.L. Falcone, T.C. Leung, W.M. Saltzman, Macromolecules 37 (2004)
6583–6588. 9779–9784.
[473] S. Ramachandran, J. Trewhella, Y. Yiider Tseng, Bruce Yu, Chem. Mater. 18 (2006) [511] L. Shi, C. Berkland, Adv. Mater. 18 (2006) 2315–2319.
6157–6162. [512] L. Shi, C. Berkland, Macromolecules 40 (2007) 4635–4643.
[474] E.L. Bakota, Y. Wang, F.R. Danesh, J.D. Hartgerink, Biomacromolecules 12 (2011) [513] A.L.Z. Lee, V.W.L. Ng, W. Wang, J.L. Hedrick, Y.Y. Yang, Biomaterials 34 (2013)
1651–1657. 10278–10286.
[475] M.S. Lamm, K. Rajagopal, J.P. Schneider, D.J. Pochan, J. Am. Chem. Soc. 127 (2005) [514] L.A. Cyster, D.M. Grant, S.M. Howdle, F.R.A.J. Rose, D.J. Irvine, D. Freeman, C.A.
16692–16700. Scotchford, K.M. Shakesheff, Biomaterials 26 (2005) 697–702.
[476] Y. Wang, E.L. Bakota, B.H.J. Chang, M. Entman, J.D. Hartgerink, F.R. Danesh, J. Am. [515] A. Nzihou, L. Attias, P. Sharrock, A. Ricard, Powder Technol. 99 (1998) 60–69.
Soc. Nephrol. 22 (2011) 704–717. [516] J.A. Lewis, J. Am. Ceram. Soc. 83 (2000) 2341–2359.
[477] K. Pagel, S.C. Wagner, K. Samedov, H. von Berlepsch, C. Böttcher, B. Koksch, J. Am. [517] M.J. Santillan, F. Membrives, N. Quaranta, A.R. Boccaccini, J. Nanopart. Res. 10
Chem. Soc. 128 (2006) 2196–2197. (2008) 787–793.
[478] K.M. Galler, L. Aulisa, K.R. Regan, R.N. D’Souza, J.D. Hartgerink, J. Am. Chem. Soc. [518] F. Bossard, T. Aubry, G. Gotzamanis, C. Tsitsilianis, Soft Matter. 2 (2006) 510–516.
132 (2010) 3217–3223. [519] D. Xu, D. Asai, A. Chilkoti, S.L. Craig, Biomacromolecules 13 (2012) 2315–2321.
[479] E.L. Bakota, O. Sensoy, B. Ozgur, M. Sayar, J.D. Hartgerink, Biomacromolecules 14 [520] S.T. Milner, T.A. Witten, Macromolecules 25 (1992) 5495–5503.
(2013) 1370–1378. [521] K.C. Tam, R.D. Jenkins, M.A. Winnik, Macromolecules 31 (1998) 4149–4159.
[480] M.K. Kang, J.S. Colombo, R.N. D’Souza, J.D. Hartgerink, Biomacromolecules 15 [522] G. Marrucci, S. Bhargava, S.L. Cooper, Macromolecules 26 (1993) 6483–6488.
(2014) 2004–2011. [523] T. Tominaga, V.R. Tirumala, E.K. Lin, J.P. Gong, H. Furukawa, Y. Osada, W. Wu,
[481] D. Drago, C. Cossetti, N. Iraci, E. Gaude, G. Musco, A. Bachi, S. Pluchino, Biochimie Polymer 48 (2007) 7449–7454.
95 (2013) 2271–2285. [524] M. Shibayama, Encyclopedia of Polymer Nanocomposites, Springer-Verlag,
[482] M. Webber, X. Han, S.N.P. Murthy, K. Rajangam, S.I. Stupp, J.W. Lomasney, J. Berlin, Heidelberg, 2013, pp. 1–12.
Tissue Eng. Regen. Med. 4 (2010) 600–610. [525] J.E. Martin, A.J. Patil, M.F. Butler, S. Mann, Adv. Funct. Mater. 21 (2011) 674–681.
[483] N.C. Wickremasinghe, V.A. Kumar, J.D. Hartgerink, Biomacromolecules 15 [526] J.E. Kim, H.S. Lee, Polymer 55 (2014) 287–294.
(2014) 3587–3595. [527] Y.T. Hu, J. Rheol. 58 (2014) 1789–1807.

You might also like