Sei sulla pagina 1di 27

Biomaterials 33 (2012) 3279e3305

Contents lists available at SciVerse ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Review

Alginate derivatization: A review of chemistry, properties and applications


Siddhesh N. Pawar, Kevin J. Edgar*
Macromolecules and Interfaces Institute, Department of Wood Science and Forest Products, Institute for Critical Technology and Applied Science, Virginia Tech, Blacksburg,
VA 24061, USA

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

Article history: Alginates have become an extremely important family of polysaccharides because of their utility in
Received 13 December 2011 preparing hydrogels at mild pH and temperature conditions, suitable for sensitive biomolecules like
Accepted 4 January 2012 proteins and nucleic acids, and even for living cells such as islets of Langerhans. In addition, the complex
Available online 26 January 2012
monosaccharide sequences of alginates, and our growing ability to create controlled sequences by the
action of isolated epimerases upon the alginate precursor poly(mannuronic acid), create remarkable
Keywords:
opportunities for understanding the relationship of properties to sequence in natural alginates (control of
Alginate
monosaccharide sequence being perhaps the greatest synthetic challenge in polysaccharide chemistry).
Bioarticial pancreas
Drug delivery
There is however a trend in recent years to create value-added alginates, by performing derivatization
Drug release reactions on the polysaccharide backbone. For example, chemical derivatization may enable alginates to
Hydrogel achieve enhanced hydroxyapatite (HAP) nucleation and growth, heparin-like anticoagulation properties,
Polysaccharide improved cellesurface interactions, degradability, or tuning of the hydrophobic-hydrophilic balance for
optimum drug release. The creation of synthetic derivatives therefore has the potential to empower the
next generation of applications for alginates. Herein we review progress towards controlled synthesis of
alginate derivatives, and the properties and applications of these derivatives.
2012 Elsevier Ltd. All rights reserved.

1. Introduction with control over monosaccharide sequence and nature, location


and quantity of substituents. This in turn enables the tailoring of
Alginates are unbranched polysaccharides consisting of 1/4 alginate derivative properties such as solubility, hydrophobicity,
linked b-D-mannuronic acid (M) and its C-5 epimer a-L-guluronic afnity for specic proteins, and many others. Such modications
acid (G). The natural copolymer is an important component of algae are complicated by key alginic acid properties including solubility,
such as kelp, and is also an exopolysaccharide of bacteria including pH sensitivity, and complexity (which can make both synthetic
Pseudomonas aeruginosa. It is comprised of sequences of M (M- control and analysis difcult). Both the promise and the difculty of
blocks) and G (G-blocks) residues interspersed with MG sequences alginate modication have attracted much effort towards
(MG-blocks). While it is possible to obtain alginates from both algal controlled derivatization.
and bacterial sources, commercially available alginates currently The signicance of alginates as natural polysaccharides in
come only from algae. The copolymer composition, sequence and biomedicine can hardly be overstated. Alginates are currently used
molecular weights vary with the source and species that produce as wound dressing materials for the treatment of acute or chronic
the copolymer. Due to the abundance of algae in water bodies, there wounds [2]. They also play a crucial part in the progression of cystic
is a large amount of alginate material present in nature. Industrial brosis, wherein bacterial biolms formed from alginate gels are
alginate production is approximately 30,000 metric tons annually, secreted by P. aeruginosa [3]. More importantly, the use of alginate
and is estimated to comprise less than 10% of the biosynthesized crosslinking to make hydrogels for cell encapsulation has proved to
alginate material [1]. Therefore there is signicant additional be most advantageous for biomedical applications [4e6]. Worthy of
potential to design sustainable biomaterials based on alginates. The mention is the role played by alginates gels in encapsulating islets
combination of chemical and biochemical techniques provides of Langerhans for diabetes treatment [7]. Chemical modication of
considerable potential for creating modied alginic acid derivatives alginates is used as a tool to attain one of two ends e (A) enhance
existing properties (example: improvement of ionic gel strength by
additional covalent crosslinking, increase hydrophobicity of the
* Corresponding author. Tel.: 1 540 231 0674. backbone, improve biodegradation, or achieve greater HAP nucle-
E-mail address: kjedgar@vt.edu (K.J. Edgar). ation and growth), or (B) introduce completely new properties

0142-9612/$ e see front matter 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biomaterials.2012.01.007
3280 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

otherwise not existing in unmodied alginates (example: afford matrix as gels containing sodium, calcium, magnesium, strontium
anticoagulant properties, provide chemical/biochemical anchors to and barium ions, such that the counterion composition is deter-
interact with cell surfaces or bestow temperature dependent mined by the ion-exchange equilibrium with seawater. A schematic
characteristics such as lower critical solution temperature). In of the alginate extraction procedure is represented in Fig. 2. The
short, alginate derivatization is the most convenient way to achieve rst step in the extraction process is removal of the counterions by
both inherent property enhancement and new property introduc- proton exchange using 0.1e0.2 M mineral acid. In the second step,
tion. We herein review the chemical routes to such modications, insoluble alginic acid is solubilized by neutralization with alkali
and the resulting properties achieved. such as sodium carbonate or sodium hydroxide to form sodium
alginate. Rigorous separation processes such as sifting, otation,
2. Description of alginates centrifugation and ltration follow in order to remove particulate
matter. Sodium alginate is then precipitated directly by alcohol,
2.1. Biosynthesis of alginates calcium chloride or a mineral acid. The product is dried and milled.
Alginates obtained using the described procedure contain several
Recent progress in biosynthesis of bacterial alginates has been mitogens and cytotoxic impurities making them unsuitable for
reviewed in several excellent articles [8e11]. Alginate biosynthesis biomedical applications. Ultrapure and amitogenic alginates that
(Fig. 1) involves the oxidation of a carbon source to acetyl-CoA, are suitable for biomedical purposes have therefore been prepared
which enters the TCA cycle to be converted to fructose-6- using more rigorous extraction processes. Free ow electrophoresis
phosphate via gluconeogenesis. Fructose-6-phosphate then was applied as one technique to remove mitogenic impurities from
undergoes a series of biosynthetic transformations to be eventually commercial alginates [18]. This method was however not suitable
converted to GDP-mannuronic acid, which acts as a precursor to for large scale processing because it was time consuming and
alginate synthesis. In general, the biosynthetic operation can be required expensive electrophoresis equipment. A chemical extrac-
broken down into four stages: (1) GDP-mannuronic acid precursor tion method was therefore described using Ba-alginate gels [19].
synthesis; (2) cytoplasmic membrane transfer and polymerization Ba2 ions show higher afnity towards alginates compared to Ca2
to polymannuronic acid; (3) periplasmic transfer and modication; ions. Ba-alginate gels are therefore stable in acidic and neutral pH
and (4) export through the outer membrane. Post-polymerization environments, but disintegrate under alkaline pH conditions.
modication of alginates occurs at stage (3), where poly- Mitogenic contaminants were rst eluted from Ba-alginate beads
mannuronic acid is acetylated at the O-2 and/or O-3 positions by by treatment with various solutions followed by ethanol extraction,
several transacetylases [12e14]. Epimerization is then performed after which the pure alginate beads were dissolved in alkaline
by a family of epimerase enzymes to convert some non-acetylated solutions. Finally the solution was dialyzed, Ba2 exchanged for Na
M residues to G residues [14e17]. Finally, alginate is released from ions and pure alginate precipitated from ethanol.
the cell through transmembrane porins.
2.3. Structure determination
2.2. Manufacture
The rst report on the chemical structure of alginates appeared
Current commercial production of alginates is based entirely on as early as 1966. Larsen et al. described in detail the partial
algal sources. Alginates occur in brown algae in the intracellular hydrolysis of alginates followed by fractionation to obtain alginates

Fig. 1. Bacterial alginate biosynthesis pathway [9]. With kind permission from Springer ScienceBusiness Media: Biotechnol Lett; Bacterial alginates: from biosynthesis to
applications; 28(21); 2006; 1701e1712; Remminghorst U., Rehm B.H.; Figure 3. Copyright Springer ScienceBusiness Media B.V. 2006.
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3281

Fig. 2. Schematic showing alginate extraction procedure from algae.

containing different copolymer compositions [20]. Fractionation 2.4. Physical properties


yielded a soluble (hydrolysable) fraction and an insoluble (resis-
tant) fraction. The resistant fractions consisted of molecules which 2.4.1. Solubility
had either mainly M rich or mainly G rich residues, while the The solubility of alginates in water is governed by three
hydrolysable fractions consisted of a high proportion of alternating parameters e a) pH of the solvent, b) ionic strength of the medium,
MG residues. A structure was therefore proposed which consisted and c) presence of gelling ions in the solvent. To make alginates
of M-blocks, G-blocks, and hydrolysable MG alternating blocks. A soluble it is essential that the pH be above a certain critical value
representative structure of the alginate backbone is shown in Fig. 3 and the carboxylic acid groups be deprotonated. Changing the ionic
where (a) shows the chain conformation and (b) shows the typical strength of the medium affects solution properties such as polymer
block distribution. Larsen et al. later employed free boundary conformation, chain extension, viscosity and therefore solubility.
electrophoresis to examine the hydrolysis products isolated by Alginates gel in the presence of divalent cations such as Ca2, Sr2
fractionation [21]. The results conrmed the presence of M-blocks, and Ba2. It is therefore necessary to have an aqueous solvent free
G-blocks and alternating MG blocks. Computer driven mathemat- of crosslinking ions to enable dissolution. The solubility of alginates
ical models were also used in understanding the alginate micro- in organic media requires formation of a tetrabutylammonium
structure [22e24]. The clearest understanding of alginate backbone (TBA) salt. We recently reported the complete dissolution of TBA-
structure was however achieved only using 1H and 13C NMR alginate in polar aprotic solvents containing tetrabutylammonium
spectroscopy. uoride (TBAF) [28]. The solubility of alginates depends strongly on
Penman et al. described a method for the determination of M/G the state of the backbone carboxylic acid groups. Alginic acid with
ratio in alginates using peak ratios obtained from the 1H NMR its carboxylic acid groups in their protonated form was not fully
spectrum [25]. Grasdalen et al. later showed that 1H NMR can also soluble in any solvent system examined, including water. Na-
yield fractions of the four dyads MM, MG, GM and GG [26]. Fig. 4 alginate dissolved in water, but was not entirely soluble in any
shows the anomeric region in the 1H NMR spectra of alginates organic medium examined. TBA-alginate was completely soluble in
with varying M contents. The monomer and dyad fractions were water, ethylene glycol and polar aprotic solvents containing TBAF,
obtained using the following equations, where IA, IB and IC are the but did not dissolve in any other solvent systems under consider-
intensities of the three peaks labeled A, B and C respectively. ation. Table 1 shows the results of a detailed solubility study [28].

IA IC
FG ; FGG ; FGG FGM FG ;
IB IC IB IC
FMM FMG FMG

Fig. 3. Representative alginate structure: (a) chain conformation and (b) block Fig. 4. Anomeric region in the 1H NMR spectra of alginates containing varying M
distribution. contents. Peak assignments as described by Grasdalen, H [27].
3282 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Table 1
Solubility of alginates in various solvents at a concentration of 15 mg/mL [28].

H2O EG DMAc DMF DMSO DMAc/LiCl DMF/TBAF DMSO/TBAF DMAc/TBAF DMI/TBAF


Alginic acid e e e e e e e e e e
Na-alginate e e e e e e e e e
TBA-alginate e e e e

() complete solubility and (-) partial or no solubility; EG ethylene glycol.

2.4.2. Ionic crosslinking alginate gels is a preferred technique for immunoprotection and
Alginate chelates with divalent cations to form hydrogels. Gel has been reviewed in several publications [4e6]
formation is driven by the interactions between G-blocks which
associate to form tightly held junctions in the presence of divalent 2.4.3. Alginic acid gels
cations [29]. In addition to G-blocks, MG blocks also participate, When the pH of alginate solutions is lowered below the pKa of
forming weak junctions [30]. Thus, alginates with high G contents the uronic acids in a highly controlled fashion, acid gels are formed.
yield stronger gels. The afnity of alginates towards divalent ions Such gels are stabilized by an intermolecular hydrogen bonding
decreases in the following order: Pb > Cu > Cd > Ba > Sr > Ca > network. Two methods are generally used to make acid gels [33]. In
Co, Ni, Zn > Mn [31]. Ca2, however, is the most commonly used the rst method, a slowly hydrolyzing lactone such as GDL is added
cation to induce alginate gel formation. A pictorial representation to a solution of Na-alginate. In the second method, pre-formed Ca-
of the three junction types possible in Ca-alginate gels is shown alginate gels are converted to acid gels by proton exchange.
in Fig. 5.
Calcium crosslinking of alginates can be performed by two 2.5. Chemical properties
methods. The rst is a diffusion method, wherein crosslinking
ions diffuse into the alginate solution from an outside reservoir. The Polysaccharides undergo hydrolytic cleavage under acidic
second is the internal setting method, where the ion source is conditions. The mechanism of acid hydrolysis of the glycosidic
located within the alginate solution and a controlled trigger bond has been described by Timell [34]. It involves three steps: (1)
(typically pH or solubility of the ion source) sets off the release of protonation of the glycosidic oxygen to give the conjugate acid; (2)
crosslinking ions into solution. The diffusion method yields gels heterolysis of the conjugate acid forming a non-reducing end group
having a Ca2 ion concentration gradient across the thickness, and a carbonium-oxonium ion; and (3) rapid addition of water to
while internal setting gives gels with uniform ion concentrations the carbonium-oxonium ion, forming a reducing end group. The
throughout [32]. Diffusion set gels are typically made by dropping acid hydrolysis mechanism for alginates is illustrated in Fig. 6.
a Na-alginate solution into a CaCl2 bath. Internal set gels typically Sodium alginate in the form of dry powder can be stored without
use insoluble calcium salts such as CaCO3 as a calcium source. degradation in a cool, dry place and away from sunlight for several
A change in the pH caused by a slowly hydrolyzing lactone such as months. The shelf life can be extended to several years by storing it
2
D-glucono-d-lactone (GDL) triggers the release of Ca ions inter- in the freezer. Alginic acid degrades more rapidly than the sodium
nally and leads to gel formation. The encapsulation of cells within salt form. The reason for this enhanced degradation rate is thought
to be intramolecular catalysis by the C-5 carboxyl groups [35]
The enzymatic degradation of alginates by lyase occurs by a b-
elimination mechanism resulting in unsaturated compounds
(Fig. 7) [36,37]. A similar degradation route is followed when they
are subjected to strongly alkaline environments. The rate of
degradation increases rapidly above pH 10.0 and below pH 5.0.
Above a pH of 10.0, the degradation arises mostly from the b-
elimination mechanism, while below 5.0 the degradation is mostly
due to acid catalyzed hydrolysis [38]. The mechanism of b-elimi-
nation involves abstraction of the proton at the C-5 position, which
is enhanced by the electron-withdrawing effect of the carbonyl
group at C-6 position. When the C-6 carboxyl group is ionized, the
electron-withdrawing effect is moderated and abstraction of the C-
5 proton is not as facile as when the carboxyl group is protonated
[39]. However, the rate of abstraction is still sufciently high to
cause relatively rapid degradation at high pH.
Alginates are susceptible to chain degradation not only in the
presence of acids or bases, but also at neutral pH values in the
presence of reducing compounds. Alginates derived from brown
algae contain varying amounts of phenolic compounds, depending
on the algal species [40]. The rate of degradation in species con-
taining a higher quantity of phenolics was shown to be much
greater [41]. A number of reducing compounds such as hydroqui-
none, sodium sulte, sodium hydrogen sulde, cysteine, ascorbic
acid, hydrazine sulfate and leuco-methylene blue also caused
Fig. 5. Possible junction points in alginates. (a) GG/GG junctions, (b) MG/MG junctions, degradation in alginates. The mechanism of degradation involves
and (c) GG/MG junctions [30]. Reprinted with permission from Donati I., Holtan S.,
Mrch Y.A., Borgogna M., Dentini M., Skjk-Brk G.; New hypothesis on the role of
the formation of a peroxide leading to free radical creation, which
alternating sequences in calcium-alginate gels; Biomacromolecules 2005; 6(2); eventually causes breakdown of the alginate chain [42]. In addition,
1031e1040. Copyright 2005 American Chemical Society. sterilization techniques such as heat treatment, autoclaving,
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3283

Fig. 6. Acid-catalyzed hydrolytic degradation of alginates.

ethylene oxide treatment and g-irradiation cause alginate degra- secondary eOH positions (C-2 and C-3) or the one eCOOH (C-6)
dation [43]. In short, degradation is an important concern when position. The difference in reactivity between the two functional
subjecting alginates to various modication reactions. The chem- group types can be easily used to selectively modify either one of
istry and reaction conditions employed must therefore be chosen the two types. Selective modication of either the C-2 or C-3
carefully so that rapid molecular weight loss does not occur during hydroxyl group is challenging due to their minor reactivity differ-
the synthesis of alginate derivatives. ences. In addition, the reaction may be controlled in terms of
selective modication of M or G residues. This may be achieved by
3. Alginate modication taking advantage of the selective chelation of G residues in Ca-
alginate gels, or by using the partial solubility properties of algi-
The derivatization and design strategies for alginates depend on nates in certain solvent systems. More importantly, the reactivity of
three important parameters: solubility, reactivity and character- alginates towards acids, bases and reducing agents cannot be
ization (Fig. 8). (A) Solubility: alginates may be dissolved in overlooked when performing derivatization reactions. Competitive
aqueous, organic or mixed aqueous-organic media for derivatiza- degradation reactions can cause rapid molecular weight loss in
tion. The choice of solvent system can dictate the type of reagents short time periods. (C) Characterization: in order to understand
that may be used for modication. In addition, the degree of algi- the substitution patterns obtained for the alginate derivatives, it is
nate solubility in the solvent system can impact derivative substi- often essential to have multiple alginate samples with a range of M/
tution pattern. (B) Reactivity: alginates can be modied at the two G ratios. In addition, derivatization of alginates enriched in M, G or

Fig. 7. Alkaline degradation of alginates by b-elimination.


3284 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Fig. 8. Important parameters governing alginate derivatization.

MG blocks may be required to achieve a detailed understanding of Wassermann reported the acetylation of alginic acid using
the substitution patterns. A lack of commercial availability of algi- ketene, a gaseous reagent [44,45]. The reactive small molecule
nates with controlled sequences may impede complete structural ketene was used as an acetylating agent, probably in order to avoid
characterization of the derivatives. Due to the complex nature of the alginate degradation caused by exposure to acetic acid or
the alginate copolysaccharide backbone, use of advanced analytical anhydride, and catalysts such as pyridine or strong acids. Acetyla-
techniques is often necessary. tion was performed by swelling alginic acid fully in acetone, and
In view of the aspects of alginate derivatization just described, then reacting with ketene at room temperature to form alginic acid
we herein review the progress made in modication of these useful acetate. The product was insoluble in water as well as in common
polysaccharides. We highlight important chemistries that have organic solvents. Alginic acid acetate was converted to its Na- or Ca-
been reported in the literature to date, and discuss the properties salt forms by treatment with sodium or calcium hydroxide solu-
attained and applications envisioned for the synthesized deriva- tions respectively. Alternatively, Na- or Ca-alginate was directly
tives. Our goal is to present a clear and complete picture of the reacted with ketene to form Na- or Ca-alginate acetates. Approxi-
available methods for alginate modication, enabling the creation mately one acetyl group was introduced per monosaccharide unit
of successful strategies for synthesis of target alginate derivatives, of the alginate chain, as determined by a titration method. Degra-
and pointing out the areas where synthetic advances are still dation during acetylation was assessed using viscosity measure-
needed, through a solid understanding of the chemistry of this ments. While Na-alginate acetate and alginic acid acetate showed
important polysaccharide. lower viscosities due to more rapid molecular weight loss, Ca-
alginate acetate did not degrade to a very large extent. Regiose-
3.1. Acetylation of alginates lectivity of the reaction and composition of the products were not
reported, most likely due to the limited analytical methods avail-
Alginates as biosynthesized by bacteria are partially acetylated able at that time.
[12e14]. The acetylation patterns govern both biosynthesis (by Schweiger reported the synthesis of both partially and fully
preventing epimerization of acetylated monosaccharides) and acetylated alginic acid derivatives using an acid catalyzed esteri-
biological functions of bacterial alginates. In vitro acetylation of cation technique [46,47]. The reaction was performed by sus-
alginates is therefore valuable to the understanding of structure- pending alginic acid in a mixture of acetic acid and acetic anhydride
property relationships of bacterial alginates. The earliest known containing perchloric acid. Products with DS values up to 1.85 were
report addressing the chemical modication of alginates was achieved at moderate temperatures without signicant degrada-
published by Chamberlain et al., wherein acetylation of the tion. While DS >1.85 was possible using higher temperatures, these
hydroxyl groups of alginic acid was described [44]. The hydroxyl conditions also led to signicant molecular weight loss. Corrobo-
groups present in alginic acid yarn could not be reacted with rating the phenomenon reported by Chamberlain et al. [44], the
acetic anhydride in the dry state due to strong H-bonding. presence of water was essential to the availability of hydroxyl
However when swollen with water, the hydroxyl groups were groups for acetylation. Strong H-bonds present in the dehydrated
available for reaction. Following the swelling process, solvent state prevented the hydroxyl groups from reacting. The viscosities
exchange was performed to replace water with glacial acetic acid. of alginate acetates plotted against their corresponding DS values
The alginic acid yarn swollen with glacial acetic acid was sub- are shown in Fig. 9.
jected to a mixture consisting of benzene, acetic anhydride and A hypothesis was presented based on the assumption that an
sulfuric acid catalyst. The yarn acetylated in this manner yielded increase in the solution viscosity is caused by the presence of free,
alginic acid with 97.3% diacetate, as determined from the total unsubstituted and non-hydrogen bonded hydroxyl groups owing to
acetyl content. However, severe degradation was reported to their ability to associate with water molecules. A decrease in the
occur during the reaction. solution viscosity on the other hand results from either (a) acid
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3285

formation. Furthermore, alginates with DS 1.4 which contained


monoacetylated and diacetylated, but no non-acetylated saccha-
ride residues resisted gel formation. This suggested that the pres-
ence of non-acetylated saccharide units was a must for chelate
formation. It also indicated that the presence of hydroxyl groups
was essential for ionic crosslinking, and that carboxylate anions
were only partially responsible for gelation. Therefore, based on the
data acquired a chelate structure was proposed wherein a single
Ca2 ion co-ordinated with two carboxylate groups and two vicinal
hydroxyl groups belonging to the same sugar to form a gel. This
proposed chelate structure was further supported by the fact that
the tendency towards gelation was shown to be greater for algi-
nates containing a higher fraction of unreacted uronic acid sugar
residues.
While the early efforts to acetylate alginate were successful,
they were limited in terms of structural characterization of the
synthesized derivatives. These investigators were hampered by
a lack of detailed knowledge about the composition and sequence
Fig. 9. Viscosity of alginate acetate plotted against corresponding DS value (repre-
of the alginate backbone, which was not achieved until a series of
sented as degree of acetylation) [46]. Reprinted with permission from Schweiger R.G.;
Acetylation of alginic acid. I. Preparation and viscosities of algin acetates; J Org Chem fundamental papers published in the 1960s-1980s illuminated the
1962; 27(5); 1786e1789. Copyright 1962 American Chemical Society. alginate copolysaccharide structure [20e27,48,49]. Early studies
were also restricted by the absence of advanced techniques for
structural analysis. DS values in alginate acetates were determined
hydrolysis or (b) substitution of the free hydroxyls with acetyl predominantly in these early papers using deacylation and titra-
groups. The initial decrease in viscosity up to DSw0.2 is evident in tion methods to measure the acetyl content. Insight about the
Fig. 9, and was attributed to a combined effect of (a) and (b). acetyl substitution variation between M and G monosaccharide
However, as the reaction progressed, H-bonding between the residues, and the regioselectivity between the two secondary OH
vicinal hydroxyl groups was overcome and an alginate mono- groups on each, was possible only later through powerful char-
acetate product was formed. An increase in the total number of free acterization techniques such as NMR spectroscopy. Modern
hydroxyl groups occurred as a result of more vicinal H-bonds being researchers are also advantaged by the availability of alginates
cleaved, resulting in a spike in the solution viscosity. A viscosity with different M/G contents and sequences, by isolation from
maximum was reached at DSw0.7. Beyond this maximum, the various organisms and by enzymatic preparation in vitro. Alginate
monoacetate sugars were acetylated further to form diacetates. variants having different block structures and sequences were not
This led to a reduction in the number of free hydroxyls and a cor- available until the biochemistry of alginates was better under-
responding decrease in viscosity. The presence of vicinal H-bonds stood. [8e11]
was therefore thought to be an important factor causing alginate Acetylation of alginates coupled with a detailed analysis of the
hydroxyl groups to have low reactivity. However, the hydroxyl acetyl substitution pattern along the backbone was rst reported by
group reactivity in monoacetylated alginate products was higher Skjk-Brk et al. [50] The reaction was carried out by preparing Ca-
compared to unreacted alginates. This phenomenon was substan- alginate gel beads in aqueous media, then replacing water with
tiated by kinetic data which indicated a rapid increase in the pyridine via solvent exchange. The beads were then suspended in
reaction rate at DS values between 0.7 and 1.5. The increase in the a mixture of pyridine-acetic anhydride at 38  C to perform the
reaction rate was associated with increasing availability of non acetylation reaction. Fig. 10 shows the reaction scheme. Acetylated
H-bonded hydroxyl groups for acetyl substitution. Ca-alginate beads were washed thoroughly with water, Ca2 ions
The partially and fully acetylated alginates were used as tools to removed and exchanged with Na ions, and the nal product dia-
enhance understanding of the chelate structure formed in ionically lyzed and freeze dried. Alternatively, acetylation was also per-
crosslinked alginate gels [47]. The addition of divalent ions to formed using alginic acid. In all cases, the presence of water was
a solution of alginate diacetate (DS 2.0) did not lead to gel essential for reaction to occur and the DS was found to depend on

Fig. 10. Acetylation of alginate using pyridine/acetic anhydride. For acetylation in gel form, M Ca2 and M00 Na [50]. For homogeneous acetylation in DMSO/TBAF, M TBA
and M00 eH or Na [28].
3286 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

the amount of water present. The DS value reached maximum at


a water content of w20%. The reaction rate was fast for the rst
40 min of reaction time, and then dropped off over the next 20 h.
The alginate acetate derivatives so prepared were characterized
using 1H NMR spectroscopy. The anomeric and high eld reso-
nances overlapped, making accurate peak assignments difcult.
The splitting patterns resulting from the O-acetyl peaks around
w2.0e2.2 were therefore used for substituent analysis. In order to
make accurate peak assignments possible, it was essential to have
alginates enriched in certain block sequences and acetylate them to
identify the peaks arising from their respective structures. By
comparing the 1H NMR spectra of acetylated alginates enriched in
G-blocks and M-blocks, the resonances at 2.04e2.06 ppm were
assigned to the acetyl groups from monoacetylated G residues.
Using the relative changes in the peak intensities observed at
varying DS values, signals arising from monoacetylated G residues
were identied and distinguished from diacetylated G residues. 1H
NMR spectra of acetates synthesized from alginates containing
varying MG block sequences were compared, and the peaks arising
from acetylated G residues adjacent to G residues were distin-
guished from acetylated G residues adjacent to M residues.
The relative acetyl substitution ratios for M and G residues were
calculated and the results are summarized in Table 2. Alginate ob-
tained from L. hyperborea with FG 0.68 was acetylated to two
different DS values. At both DS values, the substitution favored M
residues over G. In addition, when two alginates obtained from
L. hyperborea and Ascophyllum. nodosum with varying FG values
were acetylated to the same DS values, substitution occurred
preferentially on M residues. Therefore, selective acetylation of M
residues was possible through the reaction of Ca-alginate beads Fig. 11. Dependence of the modulus of rigidity (G) on DS (represented as D.a.) in
with pyridine and acetic anhydride. acetylated Ca-alginate gels [51]. Reprinted from Carbohyd Res 1989; 185(1); Skjk-
Brk G., Zanetti F., Paoletti S.; Effect of acetylation on some solution and gelling
Skjk-Brk et al. also reported the effects of acetylation upon properties of alginates; 131e138; Copyright 1989, with permission from Elsevier.
alginate properties [51]. Molecular weight measurements showed
no signicant degradation taking place during gel acetylation. The
a heterogeneous mixture was formed with partial dissolution of
addition of acetyl groups to the backbone caused noticeable chain
TBA-alginate, as conrmed by 1H NMR and dynamic light scattering
extension at 0.1 M ionic strength. Even a small proportion of acetyl
(DLS). Fig. 12 shows an image of the two mixtures. In addition to
groups on the backbone played an important role in dening the
DMSO, dipolar aprotic solvents such as DMF, DMAc and DMI fully
polymer conformation. When Ca-alginate gels were formed, acet-
ylation was found to severely diminish the ability of Ca2 ions to
induce conformational ordering. Consequently, lower strength gels
resulted from acetylated alginates. On drying and re-swelling the
Ca-alginate beads, the degree of swelling increased 500-fold from
DS 0 to DS 0.65. This was a result of enhancement in the positive
osmotic pressure resulting from a higher number of dissociating
counterions per polymer chain, due to impairment of co-operative
binding by the added acetyl groups. A plot of the modulus of
rigidity (G) of gels made from acetylated alginates vs. DS is shown
in Fig. 11. Even at low DS values where M residues were acetylated
selectively, a large drop in modulus was recorded. Thus, to form
Ca2 junctions, it was important that both G- as well as M-residues
remained unsubstituted.
We recently reported a strategy for acetylation of alginates by
homogeneous dissolution of TBA-alginate in two-component
organic solvent systems [28]. TBA-alginate dissolved completely
in DMSO solutions containing TBAF. In the absence of TBAF,

Table 2
Relative acetyl substitution ratios on M and G residues for alginates [50].

Alginate source FG DS Acetylation (%)

M G Fig. 12. Partial dissolution of TBA-alginate in DMSO (left) and complete dissolution in
L. hyperborea 0.68 0.1 >90 <10 DMSO/TBAF (right) at a TBAF concentration of 1.1% (w/v). Alginate concentration in
L. hyperborea 0.68 0.40 80 20 both vials was 15 mg/mL [28]. Reprinted with permission from Pawar S.N., Edgar K.J.;
A. nodosum 0.40 0.40 63 36 Chemical modication of alginates in organic solvent systems; Biomacromolecules
2011; 12(11); 4095e4103. Copyright 2011 American Chemical Society.
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3287

dissolved TBA-alginate in the presence of TBAF. Partial acetylation Table 3


of alginates was achieved in all solvent systems using pyridine/ Homogeneous and heterogeneous acetylation of alginates and the corresponding DS
values [28].
acetic anhydride reagent under homogeneous conditions with DS
ranging between 0.74 and 0.85. A maximum DS of w1.0 was Alginate DSHomogeneous DSHeterogeneous
however achieved for acetylation under anhydrous conditions 100% M 0.80 0.91
wherein TBAF was synthesized in situ [52], or by adding a second 63% M 0.80 0.48
29% M 0.78 0.63
portion of acetic anhydride to the reaction mixture. The DS value
13% M 0.78 0.54
for alginate acetates could not be increased signicantly 0% M 0.82 0.72
beyond w1.0 under any attempted conditions. A potential expla-
DS values measured using 1H NMR. All reactions employed TBA alginate at 100 mg/
nation for this limited DS was based on monosaccharide ring mL DMSO using alginate monosaccharide unit:pyridine:acetic anhydride molar
electronic effects. The uronate ring in unreacted TBA-alginate ratio of 1:17:15; homogeneous reactions also employed 100 mg TBAF/mL DMSO.
contained one electron-withdrawing substituent at the C-5 posi-
tion. During acetylation, the monoacetate product was formed rst
by reaction at one of the two hydroxyl groups. The monoacetylated related to the partial solubility of TBA-alginate in DMSO. A possible
uronate ring now had two electron-withdrawing substituents; the explanation may be that the M-blocks have a higher afnity for
rst was the C-5 carboxylate and the second was the acetyl DMSO, thus facilitating the selective acetylation of M residues. A
substituent. It is likely that with two electron-withdrawing contrasting picture was seen for homogeneous reactions in DMSO/
substituents present, the reactivity of the second hydroxyl group TBAF, where all polymer chains were fully solubilized. Thus, there
was signicantly reduced. The DS value may therefore be limited was equal accessibility to G-blocks, M-blocks and MG blocks, which
to w1.0 when pyridine/anhydride is used as the acylating reagent resulted in a statistical distribution of acetyl groups.
system. As previously described in this review, alginates degrade via
An alternate theory was based on the stereochemistry of the b-elimination at higher pH values. [38,39]. Pyridine is a mild base
monosaccharide residues. In M residues, the 2-OH group is axial which nonetheless can degrade alginates. Furthermore, uoride
and can interact with the C-4 proton through 1,3-diaxial interac- from TBAF is also known to be a strong base [53]. Molecular weight
tions, thus favoring substitution at the 3-OH position. In G residues measurements were therefore performed on alginate acetates that
the 3-OH group is axial and therefore interacts with the C-5 proton were reacted for different time periods, and compared to unreacted
through 1,3-diaxial interactions. Substitution of the 2-OH position TBA-alginate. Table 4 shows the obtained results, which clearly
was hence favored on G units. Thus, in each monosaccharide, point to signicant chain degradation. The starting DP (0 min) of
stereochemistry favored reaction of one out of the two hydroxyl 144 is reduced to 66 within the rst 15 min of reaction. DP
groups, limiting the DS to w1.0. While these possible causes for an continues to decrease throughout the 2 h of reaction time. Fig. 13
upper DS limit were postulated, they were not corroborated shows the right-shift in the SEC chromatograms for the alginate
experimentally. In addition to acetates, propionylation of alginates acetate obtained using light scattering as degradation proceeds.
was also reported using propionic anhydride and pyridine to obtain Thus in order to preserve the alginate molecular weight, it was
a product with DS 0.31. desirable to minimize reaction times and temperatures.
To better understand the selectivity of alginate acetylation, the
reaction was performed using alginates containing varying M/G 3.2. Phosphorylation of alginates
ratios. Under homogeneous reaction conditions, the DS values ob-
tained were nearly identical (w0.8) across the entire range of The synthesis of phosphorylated alginate derivatives was
alginate M/G ratios. This proved that the reaction was not selective recently described to evaluate their ability to induce hydroxyapa-
for M vs. G, and that both M and G residues could be acetylated. 1H tite nucleation and growth [54]. Fig. 14 shows the reaction scheme.
NMR peak assignments showed that both 2- and 3-OH positions on Phosphorylation was performed using urea/phosphoric acid
M residues could be acetylated under homogeneous conditions. reagent by creating a suspension of alginate in DMF. A maximum DS
Regioselectivity between the 2-OH and 3-OH positions on G resi- of 0.26 was achieved using an alginate:H3PO4:urea mole ratio of
dues was however difcult to determine with certainty due to 1:20:70; the heterogeneous nature of the reaction apparently
severe overlap of signals in the 1H-NMR spectrum. limited the maximum DS attainable. Phosphoric acid as a strong
Acetylation of TBA alginates performed in DMSO without added acid is likely to cause alginate molecular weight degradation.
TBAF, that is, under partly heterogeneous conditions, afforded more Accordingly, a 2e4 fold reduction in the value of Mw was reported
variable DS values compared to homogeneous acetylation. The for phosphorylated alginates compared to unreacted alginates.
products of heterogeneous reactions yielded DS values that varied The regioselectivity of phosphorylation with respect to four
with the M/G ratios of the starting polymers. The 1H-NMR spectra possible substitution sites (2-OH and 3-OH positions of each
of heterogeneously acetylated alginates showed a predominance of residue, G and M) was studied using NMR spectroscopy. A combi-
two peaks at shifts of 5.08 and 4.47 ppm respectively. This nation of NMR techniques including one-dimensional 1H and 31P,
1
predominance was not observed in the case of homogeneous H-1H COSY, 1H-31P HMBC, 1H-31P HMQC-TOCSY and 1H-13C HSQC
reaction products. Previous assignments for alginates have shown
that the peak at w5.08 ppm arises from the H-1 signal of G residues
Table 4
in GGG, MGG and GGM triads. The peak centered at w4.47 ppm on Molecular weight degradation of TBA-alginate during acetylation [28].
the other hand arises from the H-5 signal of G residues in GGG
triads [27]. Thus, the predominance of the two peaks in heteroge- Reaction time DS Mn (kDa) Mw (kDa) PDI DP

neously acetylated alginates suggested that GGG triad sequences 0 min 0 56.37 98.18 1.74 144
15 min 0.65 20.36 48.88 2.40 66
remained relatively unreacted. G-blocks were therefore preserved,
30 min 0.75 13.23 33.32 2.52 43
resulting in selective acetylation of M residues. Table 3 shows the 1h 0.51 8.13 19.61 2.41 28
DS values obtained for alginates with various M contents, under 2h 0.90 2.78 10.91 3.93 9
both homogeneous as well as heterogeneous conditions. DS values measured using 1H NMR. All reactions were performed in 1% (w/v) DMSO/
The difference in DS values and distribution observed between TBAF using M063 sample at 40  C for 2 h Ac2O was used as the acyl reagent with an
homogeneous and heterogeneous reaction products was likely Alg:Pyr:Acyl ratio of 1:17:15.
3288 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Fig. 13. Right-shift in the SEC chromatograms obtained using light scattering for the alginate acetates shown in Table 4, recorded over increasing reaction time [28]. Reprinted with
permission from Pawar S.N., Edgar K.J.; Chemical modication of alginates in organic solvent systems; Biomacromolecules 2011; 12(11); 4095e4103. Copyright 2011 American
Chemical Society.

was used to carry out detailed peak assignments. The analysis to prevent blood from clotting [56]. It is therefore of interest to see
pointed towards phosphate substitution on M residues such that whether alginate sulfates would possess similar anticoagulation
a higher degree of phosphorylation occurred on the 3-OH group properties. Yumin et al. rst reported the sulfation of sodium
compared to the 2-OH group. This regioselectivity was a result of alginate using chlorosulfonic acid in formamide [57]. The reaction
the higher reactivity of the equatorial 3-OH. The equatorial sites scheme is depicted in Fig. 15(a). Sulfates with DS up to 1.41 were
were more accessible compared to axial during phosphorylation. obtained, and the values were measured using the %C to %S ratio
NMR spectroscopy also showed evidence for substitution occurring from elemental analysis. The anticoagulant activity of alginate
on G residues. However, the regioselectivity between 3-OH and 2- sulfates was measured using activated partial thrombosis time
OH groups for G residues could not be cleanly measured because (APTT), thrombin time (TT) and prothrombin time (PT). Using APTT,
the correlations observed for G residues in a 1H, 31P-HMQC-TOCSY anticoagulant activity of alginate sulfate was found to be compa-
experiment were much weaker compared to those for M residues. rable to heparin. But using PT, the activity was very low. Since APTT
Interestingly, these phosphorylated alginates were found to be is based on an intrinsic coagulation pathway and PT is based on an
incapable of forming Ca-crosslinked ionic gels. The reduction of external coagulation pathway, alginate sulfates were shown to have
molecular weight during reaction in addition to conformational a greater inuence on the intrinsic coagulation pathway.
changes resulting from phosphorylation were cited as causes for While sulfation is benecial in many cases, oversulfation is
the inability of these alginate derivatives to form gels. However, Ca- undesirable since it can cause side effects. Therefore to decrease the
crosslinked gels could be formed by blending unreacted alginate anticoagulant activity, alginate sulfates were reacted with 2,3-
with phosphorylated alginate. Such gels formed using alginate epoxypropyl trimethylammonium chloride to attach pendent
blends showed higher resistance to calcium extraction compared to quaternary amine groups (Fig. 15(b)). The number of quaternary
gels formed using only unreacted alginates. The authors pointed ammonium groups attached was controlled by the moles of the
out that the phosphate groups were likely to have participated in epoxy reagent added. Both unsulfated hydroxyl and carboxylate
the chelation process, thus making the gels more stable. groups added to the epoxy compound to append the quaternary
ammonium salts. While the sulfation step required use of chlor-
3.3. Sulfation of alginates osulfonic acid, most likely causing molecular weight loss, the
epoxide condensation did not cause degradation. The anticoagulant
Sulfation of polysaccharides, both enzymatically in nature as activities of alginate sulfates were greatly reduced by attachment of
well as by chemical methods is known to provide blood- the quaternary amines. Furthermore, the reduction of activity was
compatibility and anticoagulant activity [55]. Heparin is the most proportional to the number of quaternary ammonium groups
widely used naturally sulfated polysaccharide because of its ability attached.

Fig. 14. Phosphorylation of alginate [54].


S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3289

Fig. 15. (a) Alginate sulfation using chlorosulfonic acid and (b) quaternization of alginate sulfate using 2,3-epoxypropyl trimethylammonium chloride.

Cohen et al. reported the synthesis of alginate sulfates using groups were added to either one or both hydroxyl positions. The
carbodiimide coupling chemistry [58]. The hydroxyl groups were use of a strong acid reagent caused substantial molecular weight
sulfated using a DCC-sulfuric acid reagent system such that the loss. The average molecular weight decreased from 100 to 10 kDa
carboxylate groups remained unreacted. The reaction mechanism during reaction, but was not affected by the M/G ratio.
(Fig. 16) is marked by the formation of a DCC-H2SO4 adduct which The binding of alginate sulfates to both heparin-binding and
reacts with the hydroxyl nucleophile. The products were charac- non-binding proteins was compared to unmodied alginates.
terized using 13C NMR, and the spectra revealed that the C-1 and While the sulfates bound strongly to heparin-binding proteins,
C-6 peak shifts did not change upon sulfation. The C-2 and C-3 unmodied alginates did not bind. Furthermore, both sulfates and
peaks however shifted downeld, thus proving that the sulfate unmodied alginates showed no afnity for heparin non-binding

Fig. 16. Sulfation of alginates using DCC-sulfuric acid route [58].


3290 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

proteins. In nature, specic binding of growth factors to heparin et al. attempted the hydrophobic modication of sodium alginate
and heparan sulfate protects them from denaturation and proteo- by covalent attachment of short polyether chains to the alginate
lytic degradation. The ability of alginate sulfates to provide backbone [62]. Sodium alginate was rst activated using sodium
protection to growth factors and sustain their release was therefore metaperiodate, which oxidized the two secondary hydroxyl groups
studied. Ca-alginate microspheres were formed by blending algi- to aldehyde groups. The aldehydes were then reacted with a-
nate sulfates with unreacted alginates. A much more sustained amino, u-benzyloxy tetraoxyethylene (C6H5(OCH2CH2)4NH2,
release of bFGF growth factor was achieved from alginate/alginate abbreviated as BzlO-TEG-NH2) by reductive amination to yield
sulfate microspheres compared to that from pure alginate micro- a tetraoxyethylene functionalized amine derivative of alginate
spheres. The sustained in vivo release of bFGF afforded enhanced (BzlO-TEG-NH-Alg; Fig. 18). Through oxidization, alginate hydroxyl
angiogenic activity compared to non-specically bound alginate. groups having low reactivity were converted to aldehydes having
Fan et al. reported the sulfation of alginates by an uncommon signicantly higher reactivity. The carboxylate groups were
reagent prepared using sodium bisulte and sodium nitrite in an preserved in the process, thus retaining the ability of the modied
aqueous solvent [59]. Traditional sulfation reagents such as sulfuric alginate to form ionic gels.
acid, chlorosulfonic acid, sulfuryl chloride, sulfur trioxide and sul- The amount of periodate reagent used for the reaction was
famic acid cause hydrolytic degradation of alginates. The new calculated to be sufcient to oxidize 20% of the available uronate
synthetic route was therefore used to overcome this limitation of residues. A signicant reduction in the viscosity was recorded on
traditional reagents. The reaction was performed in water, wherein completion of this step due to molecular weight degradation and
the sulfation reagent was rst adjusted to the desired pH followed extension of chain conformation by ring opening. Breakdown of the
by addition of sodium alginates (Fig. 17). DS values for the deriva- alginate molecular weight in solutions containing periodate, as
tives were measured using barium sulfate nephelometry. An previously mentioned, depends on the sequence and distribution of
optimum DS value of 1.87 was achieved at pH 9.0, 40  C and 2:1 M and G residues [63]. Alginates containing longer MG block
mole ratio of reagent:uronic acid residue. Anticoagulant activities sequences were found to degrade more rapidly than alginates
for the sulfated alginate derivatives were measured and the DS, containing shorter MG blocks. Partial oxidation of alginates may be
molecular weight and concentration were found to be inuential advantageous for biomedical applications because they degrade in
factors. aqueous media, while unmodied alginates do not [64].
While the sulfation chemistries described thus far yield alginate The oxidized alginate derivative was reductively aminated with
sulfates, control over the placement of sulfate groups along the excess BzlO-TEG-NH2 and sodium cyanoborohydride (NaCNBH3) in
alginate backbone has not yet been achieved. It is known that most phosphate buffer (pH 7). Due to the difcult removal of residual
biological functions in sulfated polysaccharides are related to their BzlO-TEG-NH2, BzlO-TEG-NH-Alg was converted to its Ca-gel form
sulfation pattern and sequence [60,61]. Therefore, advances in and washed with ethanol. After the removal of residual BzlO-TEG-
controlled placement of sulfate groups along the alginate backbone NH2, the product was transformed back to its Na-salt form and
may eventually translate into greater understanding and perhaps characterized using 1H and 13C NMR. The DS value was however
control of structure-property relationships in alginate sulfates. quantied using 13C NMR, since the 1H NMR spectrum showed
complex overlap occurring in both anomeric and high eld regions.
3.4. Hydrophobic modication Based on the integration ratios of the benzyl aromatic carbons and
the carboxylic carbon, the DS was calculated to be 0.12.
Alginates are hydrophilic polysaccharides. In addition to As the charged (anionic) alginate backbone is decorated with
hydrophilicity resulting from the two hydroxyl groups, the hydrophobic entities, two competing forces emerge - (a) repulsive
carboxylate ion enhances water solubility at pH 5 and above. The interactions resulting from the charged carboxylate groups
motivation for hydrophobic modication of alginates is therefore to promote chain extension, and (b) attractive interactions resulting
transform the polysaccharide from its predominantly hydrophilic from the hydrophobic groups promote chain collapse. The result of
nature to a molecule with amphiphilic or hydrophobic character- these forces is the formation of high molecular weight aggregates
istics. The most straightforward way to achieve this transformation in solution when (b) dominates (a). For BzlO-TEG-NH-Alg the
is by covalent attachment of hydrophobic moieties such as long electrostatic repulsive forces offset the hydrophobic attractive
alkyl chains or aromatic groups to the polymer backbone. Hubert forces, preventing the formation of high molecular weight

Fig. 17. Sulfation of alginates using sodium bisulte-sodium nitrite reagents [59].
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3291

Fig. 18. Synthesis of BzlO-TEG-NH-Alg by periodate oxidation of sodium alginate followed by reductive amination using BzlO-TEG-NH2.

aggregates. This was contrary to the observation found in similar screening effect there was an enhancement of hydrophobic inter-
dextran derivatives (BzlO-TEGeNH-Dex), where high molecular actions with increase in ionic strength. The increase in viscosity
weight aggregates were freely formed [65]. Dextran is an with salt concentration was therefore found to be signicantly
uncharged polysaccharide, and the absence of repulsive electro- higher for PGA-C12 compared to PGA. Fig. 20(b) shows the plot for
static forces in BzlO-TEG-NH-Dex caused hydrophobic interactions viscosity vs. NaCl concentration for both PGA and PGA-C12. An
to dominate. increase in the hydrophobicity of the environment was further
In addition to intramolecular effects, interactions in hydro- created by increasing the PGA-C12 concentration. A similar
phobically modied alginates are also present at the intermolecular concentration dependent increase in hydrophobicity did not occur
level. Hubert et al. later presented evidence for this through the with PGA.
synthesis of C12-linked alginate amide derivatives [66]. Propylene The synthesis of alginate amide derivatives with varying length
glycol esters of alginate (PGA) with w30% carboxylic acid groups hydrocarbon chains - PGA-C8, PGA-C14 and PGA-C12 has been
esteried (DSPG w0.3) were substrates for nucleophilic displace- described [68,69]. Clear evidence for the increase in hydrophobic
ment by dodecylamine (Fig. 19) to form the corresponding amides character of the polymer solution for long alkyl chain lengths was
(PGA-C12) with a DSAmide w 0.09 (calculated based on nitrogen observed. The rheological properties of PGA, PGA-C12 and PGA-C14
content analysis). The reaction was performed in the heteroge- derivatives were studied in the semi-dilute aqueous regime, and
neous state by making a suspension of PGA in DMF. a marked change was observed for the hydrophobically modied
The change in viscosity of PGA solutions after C12 modication derivatives when compared to parent PGA [70]. The intermolecular
was studied. Fig. 20(a) shows the viscosity vs. concentration plots hydrophobic interactions between long alkyl chains were disrupted
for both PGA and PGA-C12. A large increase in PGA-C12 viscosity reversibly under high shear.
with concentration was observed compared to that in PGA. This Hubert et al. also reported the synthesis of hydrophobically
increase in PGA-C12 viscosity was attributed to a combined effect of modied alginate derivatives starting from Na-alginate [71].
chain entanglements and intermolecular interactions between the Dodecyl chains were covalently linked to the carboxylate groups on
hydrophobic dodecyl chains. Interactions at an intermolecular level the backbone. TBA-alginate was rst synthesized by converting Na-
lead to the formation of physically crosslinked domains. This effect alginate to alginic acid and subsequent neutralization with TBA
was similar to that observed in hydrophobically modied hydrox- hydroxide. TBA-alginate was reported to be soluble in DMSO, but
yethyl cellulose (HMHEC) derivatives, wherein hydrophobically experiments in our laboratory have shown that TBA-alginate
driven association between individual molecules leads to the synthesized using a similar procedure was only partially soluble
formation of a three-dimensional network [67]. The viscosity in DMSO [28]. The reaction was carried out using dodecyl bromide
increase in unmodied PGA resulted only from chain entangle- reagent via nucleophilic displacement (Fig. 21). The product (Alg-
ments. The absence of intermolecular interactions and physically C12) was puried by dialysis and subsequently freeze dried, with
crosslinked domains prevented a large viscosity spike. The pres- hydrophobe DS 0.12 as determined by gas chromatography after
ence of intermolecular hydrophobic interactions in PGA-C12 was alkaline hydrolysis of the sample.
further conrmed when the ionic strength of the medium was The surface tension isotherms for Alg-C12 and unreacted Na-
varied. An increase in the salt concentration had an effect of alginate were compared by measuring the change in surface
screening the electrostatic repulsive forces leading to a more tension (s) with concentration. The s value for Na-alginate was
compact coil and higher viscosity. For PGA-C12, in addition to the surprisingly unchanged after C12 modication. It was proposed that

Fig. 19. Reaction scheme for the synthesis of PGA-C12.


3292 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Fig. 22. (a) Alg-C12 adsorption at the air water interface and (b) Alg-C12 DTAB
adsorption at the air water interface [71]. Reprinted from J Colloid Interface Sci 2000;
225(2); Babak V.G., Skotnikova E.A., Lukina I.G., Pelletier S., Hubert P., Dellacherie E.;
Hydrophobically associating alginate derivatives: Surface tension properties of their
mixed aqueous solutions with oppositely charged surfactants; 505e510; Copyright
2000, with permission from Elsevier.

collapsed micellar structures formed due to intramolecular


Fig. 20. (a) Viscosity vs. concentration (C) plot for PGA (empty squares) and PGA-C12
hydrophobic interactions. The dodecyl chains were buried inside
(half-lled squares) at a shear rate of 0.06 (1/s) and (b) Viscosity vs. NaCl concentration
(Cs) plot for PGA (empty squares) and PGA-C12 (half-lled squares) at a xed polymer
the micelles, protecting them from exposure to the adsorption
concentration of 1.2 g/dL and shear rate of 0.06 (1/s) [66]. Reprinted with permission interface. In addition, the charged alginate backbone made it
from Sinquin A., Hubert P., Dellacherie E.; Amphiphilic derivatives of alginate: evidence incapable of folding at short distances and restricted orientation of
for intra- and intermolecular hydrophobic associations in aqueous solution; Langmuir the hydrophobic chains at the surface. Fig. 22(a) shows a schematic
1993; 9(12); 3334e3337. Copyright 1993 American Chemical Society.
of the adsorption of Alg-C12 at the air water interface in the

Fig. 21. Reaction scheme for the synthesis of Alg-C12.


S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3293

presence of a high concentration of covalently attached long alkyl dissolve. The authors further cited that no BSA release occurred
chains. from Alg-C12 microspheres into the surrounding aqueous envi-
While Alg-C12 did not show markedly enhanced adsorption ronment due to the stability of hydrophobic junctions within the
activity, the addition of a cationic surfactant, dodecyl- gels.
trimethylammonium bromide (DTAB) with Na-alginate showed Nystrm et al. reported the synthesis of hydrophobically
a remarkable improvement in the surface adsorption. The surfac- modied alginate (Alg-CONH-C8) by coupling n-octylamine to the
tant possesses a C12 hydrophobic tail and a cationic head group backbone carboxylic acid groups using 1-ethyl-3-(3-
which interacted ionically with the negative charges on the alginate dimethylaminopropyl)carbodiimide hydrochloride (EDC-HCl) [74]
backbone. Therefore, while the hydrophobic C12 chain was cova- (Fig. 23). A DS value up to 0.30 was measured using 1H NMR,
lently linked in Alg-C12, the C12 chain was bonded ionically to the using the ratio of methyl (from the C8 chain) to anomeric protons,
alginate backbone in a mixture of Na-Alg DTAB. At identical C12 and was conrmed by elemental analysis. The effect of the M/G
molar concentrations, the ionically linked Na-Alg DTAB showed ratio and the sequence of the two monosaccharides on substitution
almost a thousand-fold higher surface adsorption activity pattern were however not reported. The mechanism of carbodii-
compared to the covalently linked Alg-C12. In other words, the mide mediated coupling of carboxylic acids to amines is depicted in
electrostatically linked C12 alkyl chains in Alg DTAB were more Fig. 24.
densely aligned at the airewater interface than the covalently The hydrophobic interactions occurring in Alg-CONH-C8 were
attached C12 chains in Alg-C12. Fig. 22(b) shows a schematic of reversible in the presence of b-cyclodextrin (b-CD). b-CD was
Alg DTAB adsorption at the airewater interface. The labile nature shown to have a noticeable effect on the Alg-CONH-C8 chain length.
of electrostatically attached C12 alkyl chains allowed a free re- Small-Angle Neutron Scattering (SANS) was used to calculate two
organization at the interface, while such re-organization was not physical parameters e persistence length (L) and correlation length
possible when the chains were covalently linked to the alginate (x). Persistence length is a measure of the stiffness of long polymer
backbone. Dellacherie et al. later reported the physico-chemical chains, whereas correlation length denes the average spatial
properties of hydrophobically modied alginates in aqueous solu- extent of polymer concentration uctuations resulting from
tion [72]. Fluorescence spectroscopy studies were performed which thermal changes. When b-CD was added to a solution of Alg-CONH-
showed an increase in the hydrophobic character with increasing C8, the value of L increased while that of x decreased. The truncated
alkyl chain lengths. cone structures in b-CD encapsulated the hydrophobic C8 chains on
Production of microspheres based on hydrophobically modied Alg-CONH-C8 causing a decrease in hydrophobic interactions. This
alginates by a dispersion gelation method was reported by Leo- in turn led to an increase in the net repulsive electrostatic forces,
nard et al. [73]. Gelation was induced in an aqueous solution of resulting in local stretching of polymer chains and higher L values.
Alg-C12 by the addition of NaCl. A dispersive shear stress was rst The decrease in x was a result of re-organization of the network,
applied to overcome the initially high solution viscosity of Alg-C12. whereby the hydrophobic interactions fell apart to yield a homo-
The solution was then dispersed in aq. NaCl to afford gel micro- geneous system devoid of junction points. Fig. 25 shows a sche-
particles. Microparticle size was controlled by varying the applied matic illustrating the encapsulation of hydrophobic C8 moieties on
shear stress. While the particles were stable in aqueous solutions Alg-CONH-C8 by the b-CD truncated cone.
for several days, they aggregated upon centrifugation. A small Nystrm et al. later reported complex formation between Alg-
amount of CaCl2 was therefore used during particle formation to CONH-C8 and hydroxypropyl-b-CD (HP-b-CD) or polymerized b-CD
strengthen the physical network by ion crosslinking. Increasing (poly-b-CD) [75]. On addition of HP-b-CD to a solution of Alg-
the polymer concentration in the starting mixture caused a slight CONH-C8, a reduction in viscosity was observed. This viscosity
increase in the particle size. Furthermore, at high DS values, small reduction was an effect of encapsulation of the hydrophobic
particle sizes could be achieved at low salt concentrations; while moieties by the HP-b-CD truncated cone, thus deactivating the
at low DS values, high salt concentrations were necessary to hydrophobic interactions. Addition of poly-b-CD to a solution of
achieve the same size. Bovine serum albumin (BSA) was encap- Alg-CONH-C8 showed more complex behavior, where the solution
sulated within the Alg-C12 gel microspheres. In water, the protein viscosities depended on both the poly-b-CD and Alg-CONH-C8
was released from unmodied alginate within a few hours, but concentrations.
BSA encapsulated in Alg-C12 microspheres was not released for up
to 5 days. In citrate solutions, no release of BSA encapsulated in 3.5. Attachment of cell signaling molecules
Alg-C12 microspheres was observed, whereas BSA encapsulated in
unmodied alginate gel was released instantaneously by dissolu- Alginates are important biomaterials in the area of bioengi-
tion of Ca-alginate beads. Sodium citrate is a Ca2 chelating agent, neering [76]. A critical requirement of such biomaterials is their
and likely caused a rapid Na-Ca2 exchange causing the beads to ability to provide an environment that is both physically and

Fig. 23. Reaction scheme for the synthesis of Alg-CONH-C8.


3294 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Fig. 24. Mechanism of carbodiimide mediated coupling of carboxylic acids to amines.

chemically favorable to the presence of biological species such as alginates capable of ionic gelation were therefore synthesized to be
living cells. In order to enhance the chemical interactions of algi- used as biomaterials for improved encapsulation and adhesion of
nate matrices with cells, they are functionalized with cell-specic hepatocytes.
ligands or extracellular signaling molecules. In addition to The synthesis was performed by reaction of the alginate
enhancement of the cellular interactions, functionalization may carboxylic acid groups with 1-amino-1-deoxy-b-D-galactose (Gal-
also play a role in controlling the growth, differentiation and 1-NH2) using aqueous carbodiimide chemistry (Fig. 26). An EDC/N-
behavior of cells in culture. Alginates as biomaterials offer several hydroxysulfosuccinimide (NHS) coupling reagent was used. The
advantages including hydrophilicity, biocompatibility and non- EDC/NHS coupling mechanism is similar to that depicted in Fig. 24,
immunogenicity [77]. The ability of alginates to form gels capable except the O-acylated urea derivative formed in the rst step is
of encapsulating cells, drugs and other biological entities is another stabilized by conversion into an amine reactive sulfo-NHS ester,
important advantage for biological applications. However, thus increasing the efciency of EDC mediated coupling.
a possible drawback is that cells do not adhere to alginates natu- The maximum achievable DS (measured by 1H NMR) was 0.36
rally. For that reason Mooney et al. have recently described algi- using 1.5 equivalents of Gal-1-NH2 per monosaccharide unit at pH
nates as blank slates [6]. Chemical functionalization with cell 4.5. 1H NMR spectroscopy was also used to identify the positions at
signaling moieties is therefore crucial in order to overcome the low which galactose substitution occurred on the alginate backbone.
afnity of alginates to cell surfaces. For accurate assignments of the new peaks that appeared in the
Skjk-Brk et al. reported the covalent attachment of galactose anomeric region after derivatization, it was essential to obtain 1H
moieties to the alginate backbone for enhancement of hepatocyte NMR spectra of polymannuronate, G-enriched alginate and poly-
cell recognition [78]. Hepatocytes perform a variety of metabolic alternating MG, and their corresponding galactose substituted
functions in the normal liver. However, outside the liver they lose derivatives. Three new signals appeared in the anomeric region
their function and are viable for only short time periods. The upon galactosylamine modication; signal (A) was assigned to the
presence of an anchor material that provides mechanical support H-1 proton of galactosylamine linked to G residues, signal (B) was
and immunoprotection to the cells prevents such loss of function- assigned to the H-1 proton of galactosylamine substituents linked
ality and cell death. Alginates with their unique gel forming ability to M residues neighboring G residues, and signal (C) resulted from
are considered very good matrix materials, providing a suitable a non-anomeric proton which experienced a peculiar environment
environment for sustaining hepatocytes outside the organism. The like that of galactose moieties linked to diaxial GG dyads. The areas
surface of hepatocyte cell membranes possesses an asialoglyco- of these peaks corresponded to the fraction of galactosylamine
protein receptor (ASGPR), which binds and internalizes glycopro- linked to G residues (A), the fraction of galactosylamine linked to M
teins with terminal b-galactosyl residues. Galactose modied residues in alternating sequences (B), and the amount of gal-
actosylamine residues linked to G residues neighboring G residues
(C). The difference between peaks A and C was then used to
calculate the fraction of substitution on G residues in alternating
sequences. The fraction of substituted M residues neighboring M
residues could not be calculated due to overlapping signals in the
4.60e4.80 ppm region.
The difcult nature of the analytical problem at hand is clearly
evident through this example [78]. The alginate derivatives studied
in this case were modied only at the carboxylic acid position on
the backbone. Increasing complexity arises in the NMR spectra
when substitution occurs on the two hydroxyl groups in addition to
the carboxyl group. For the galactosylamine substituted alginates,
the positional selectivity present at various DS values is shown in
Fig. 25. Schematic illustration of the encapsulation of the hydrophobic moieties on Table 5. Based on the data, it was concluded that at low DS values,
Alg-CONH-C8 by the b-CD truncated cone [74]. Reprinted with permission from Galant the reaction was more selective to G residues; as higher DS values
C., Kjoniksen A.L., Nguyen G.T.M., Knudsen K.D., Nystrom B.; Altering associations in
were attained; M residues participated in the reaction. In addition,
aqueous solutions of a hydrophobically modied alginate in the presence of beta-
cyclodextrin monomers; J Phys Chem B 2006; 110(1); 190e195. Copyright 2006 G residues neighboring M residues were more reactive than G
American Chemical Society. residues neighboring G residues.
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3295

Fig. 26. Reaction scheme for the synthesis of galactose substituted alginate.

The ability of galactose substituted alginate derivatives to form fashion - AlgE4 and AlgE6. The function of the AlgE4 enzyme was to
gel beads was studied. Substitution of galactose sugar residues on introduce long alternating MG sequences, while the function of
the alginate backbone caused a reduction in the number of avail- AlgE6 was to introduce G-block sequences within the galactosy-
able carboxylic acid groups, and impacted the bead size. Having lated polymannuronan backbone. Both enzymes catalyzed the
galactose side chains caused conformational disordering within epimerization of only those M residues that were not previously
gels, whereby the junctions formed were held less tightly galactosylated. Therefore, all G residues introduced by epimeriza-
compared to unmodied alginates. A result of this effect was that tion were unsubstituted, and thus available for gel formation.
1
larger bead volumes were achieved due to a net gain in hydration. H NMR spectroscopy was used to analyze the substitution
Galactose moieties also interfered with the polymer chain packing patterns on the alginate backbone and to calculate the DS. Gal-
process during gel formation. When the gel beads were dried and actosylation of polymannuronan yielded a product with DS of 0.12.
rehydrated, the kinetics and thermodynamics of swelling were On introducing alternating MG sequences, a new signal at
affected. Larger volume increases during rehydration resulted at w5.07 ppm appeared from the anomeric protons of the newly
high DS values. An increase in the DS was also found to decrease the introduced G units. Relative peak area calculations for this peak
afnity of alginates towards Ca2 ions. Cytotoxicity studies were showed that the FG value was 0.33. While M residues neighboring
performed which showed that all of these Gal-substituted alginate unsubstituted M residues were epimerized to G residues, those
derivatives were non-toxic. neighboring galactosylated M residues did not epimerize. Enzyme
The conformational effects resulting from the introduction of activities were determined for both epimerization reactions using
galactose moieties on the alginate backbone were studied using ratios of the experimentally achieved FG, and theoretically calcu-
intrinsic viscosity, radius of gyration and chiro-optical property lated maximum achievable FG values. Enzyme activity for the rst
measurements [79]. Galactose substitution on the alginate back- step (AlgE4) was 86%, but dropped to 21% for the second epimeri-
bone led to a complex conformational change in solution. The zation (AlgE6) step. The monomer and dyad fractions calculated for
presence of a fairly large moiety such as galactose in close proximity all three derivatives are shown in Table 6.
to the alginate backbone altered the conformational space available To better understand the effect of the presence of galactose
to the backbone. Additionally, when G residues in alternating moieties on the epimerization reaction, galactosylated poly-
sequences were substituted, the chain adopted a less extended mannuronan with a spacer group present between the galactose
conformation compared to the substitution of G residues in homo- unit and mannuronan residue was synthesized. Carboxylic acid
polymeric sequences. A decrease in the hydrodynamic radius was groups of mannuronic acid were coupled to p-aminophenyl-b-D-
therefore observed at low DS values, when G residues neighboring M galactopyranoside (pNH2PhbGal) using EDC/NHS coupling. The
residues were substituted. As the DS value increased, G residues rigid spacer group did not signicantly alter the epimerization of
neighboring other G residues were substituted, and an increase in unmodied M residues. In addition, the amide bond was intact
the hydrodynamic properties occurred. after exposure to both enzymes AlgE4 and AlgE6. Gel studies were
Although galactose moieties were covalently attached to the performed with the galactosylated alginates. When the only source
alginate backbone, substitution on both M and G units was disad- of G residues was from MG alternating sequences, the gels formed
vantageous due to the loss of mechanical properties for the alginate were based on the formation of interchain junctions. However,
gels. Skjk-Brk et al. therefore reported a method combining when the source of G residues was both MG alternating sequences
a chemical and enzymatic approach to achieve selective substitu- and G-block sequences, egg-box (Fig. 5(A)) structures were
tion on M residues [80]. Since G units were not galactosylated, the formed. The introduction of galactose units selectively on M resi-
mechanical properties of the ionotropic gels formed were not dues caused faster gelling and higher gel strength compared to
altered. A reaction scheme similar to that shown in Fig. 26 was used when galactose units were attached to both G and M residues. For
wherein polymannuronan (with no G residues) was galactosylated gels that contained galactose units only on M residues, the alginate
rst. Galactosylated polymannuronan was then subjected to two chains associated laterally with each other. This was contrary to
different epimerase enzyme catalyzed reactions in a sequential gels made from alginates bearing galactosyl groups on both M and

Table 5
Substitution selectivity for alginates at various DS values, measured using 1H NMR spectroscopy [78].

DS Total substitution on G (%) Total substitution Substitution on G adjacent Substitution on G adjacent Substitution on M Substitution on M adjacent
on M (%) to M (%) to G (%) adjacent to G (%) to M (%)
0.07 7 e 7 e e e
0.19 19 e 7.2 11.8 e e
0.36 29 7 7.3 21.7 7 e
3296 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Table 6 cells did not aggregate. Hepatocytes are anchorage sensitive cells,
Monomer and dyad compositions of galactosylated polymannuronan derivatives which means that their functions in vitro and in vivo are regulated
after epimerization [80].
by cell anchorage and cell aggregation [82]. GA thus enhanced liver
Alginate FG FM FGG FGM/MG FMM related functions in encapsulated hepatocyte cells. Gels made from
Galactosylated polymannuronan (MGal) 0 1 0 0 1 GA also showed higher cell viability compared to unmodied
AlgE4 Treated MGal (MGalE4) 0.33 0.67 0 0.33 0.34 alginates.
AlgE6 Treated MGalE4 (MGalE4E6) 0.45 0.55 0.16 0.29 0.26
Mooney et al. reported the linking of GRGDY cell adhesion
peptide to alginates for enhancement of their interactions with
mouse skeletal myoblasts [83]. Amine functional peptides were
G residues, where the bulky galactose units on G residues sterically attached to the alginate carboxylic acid groups using EDC/NHS
hinder lateral association among chains. Selective galactosylation coupling (Fig. 28). The interaction of C2C12 mouse skeletal
of M residues also provided gels that were more stable to saline myoblasts with GRGDY containing alginates was studied. Myoblast
solutions, critical to in vivo performance. cells seeded on to GRGDY-coupled alginate hydrogels attached and
Akaike et al. reported a strategy to galactosylate alginate using spread on the surface, while no attachment was observed on
lactobionic lactone and ethylenediamine [81] (Fig. 27). Lactobionic unmodied alginate control surfaces. The observed myoblast
acid was dehydrated to give lactobionic lactone in the rst step. adhesion was a result of specic interactions with the GRGDY
Then the lactone was reacted with excess ethylenediamine to form peptide sequences. Even though signicant cell spreading occurred,
the monoamine terminated lactobionic lactone (L-NH2). The free the number of cells did not increase within the rst 24 h of seeding.
primary amine groups of L-NH2 were then coupled with the At day 3, the number of cells increased slightly and reached an upper
carboxylic acid groups on the alginate backbone mediated by EDC/ limit. Thereafter the number of cells decreased. This decrease was
NHS. This afforded galactosylated alginate (GA) with DS 0.26 a result of the fusion of myoblasts into multinucleated microbrils,
(elemental analysis). an important step in the pathway to skeletal muscle differentiation.
The attachment of hepatocytes to GA coated polystyrene (PS) Mooney et al. also reported the effect of variation of GRGDY
surfaces was studied. Hepatocyte attachment to GA was 55%, ligand density and alginate M/G ratio on proliferation and differ-
compared to 3% for PS coated with unmodied alginate. This clearly entiation of C2C12 skeletal myoblast cells [84]. With an increase in
demonstrated that enhancement in hepatocyte attachment was the alginate G-content, cell proliferation increased signicantly. In
caused by the linking of galactose residues to alginate. Further- addition, alginates with high G-contents promoted extensive fusion
more, the number of hepatocytes attached was greater when the of the myoblasts causing a corresponding increase in the levels of
density of galactose residues on the GA coated surface was higher. muscle creatine kinase (MCK) activity over time. Cell proliferation
Specic interaction of hepatocyte surfaces with galactose moieties and differentiation was also dependent on the ligand density
was veried by poly-N-p-vinylbenzyl-D-lactoneamide (PVLA) present on the alginate backbone. As the ligand density increased,
competitive binding assay. The hepatocyte cells were encapsulated the proliferation and spreading of myoblast cells improved and an
within calcium crosslinked GA gels. In order to maintain cell increase in the cell count was recorded.
stability and liver-specic functions, the formation of three-
dimensional hepatocyte spheroids within the anchoring matrix is
essential. For calcium alginate gels formed using GA, more than 80% 3.6. Covalent crosslinking of alginates
of the cells were found to aggregate forming uniformly distributed
multicellular spheroids. In addition, aggregation increased as Ionic crosslinking of alginates in the presence of divalent cations
epidermal growth factor (EGF) and insulin were added. For calcium is well known [31]. Such physical gels are used in a variety of areas
alginate gels formed using unmodied alginates, the hepatocyte including textile printing, paper coating, wound dressing, food

Fig. 27. Reaction scheme for the synthesis of galactosylated alginate (GA) using lactobionic lactone.
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3297

Fig. 28. Reaction scheme for the synthesis of GRGDY containing alginates.

additives and biotechnology [6,85]. While the ionic gels are of great swelling due to the presence of ionic solutes was signicantly lower
interest, covalent crosslinking of alginates yields matrices with in magnitude compared to synthetic superswelling materials like
improved mechanical properties. Ionic alginate gels are known to crosslinked polyacrylic acid.
have limited stability in vivo. In physiological media, ion exchange Skjk-Brk et al. also studied the swelling behavior of cova-
with monovalent ions causes destabilization and rupture of the gel lently crosslinked alginate beads in the presence of monovalent and
[86]. Chemical crosslinking is an attractive strategy for overcoming divalent cations [92]. On swelling the beads in water, ion concen-
this drawback by creating more stable and robust networks. The tration in the interior of the bead was high compared to the exte-
methods used for covalent crosslinking of alginates and their rior. When the beads were swelled with monovalent salt solutions,
applications are therefore reviewed. the difference in ion concentration between the exterior and
Cascone et al. reported chemical crosslinking of alginates to interior was lower, and caused a marked decrease in bead volume
make beads for afnity and ion exchange chromatography [87]. In with increasing salt concentrations. Swelling with salt solutions of
an ion exchange material, availability of the carboxylic acid groups divalent (crosslinking) ions like Ca2, Sr2 and Ba2 showed
on the alginate backbone is necessary. The carboxylic acid groups in hysteresis in the swelling curves - a feature displayed due to cation
Ca-alginate gels participate in ion crosslinking and are thus are not binding in alginate gels. This proved that alginates retain their ion-
available for ion exchange. Covalent crosslinking by reaction of the binding properties in the covalently linked state. By introducing
alginate hydroxyl groups for network formation is therefore divalent cations, physical crosslink sites were introduced in addi-
essential for this application. This was achieved by reacting Ca- tion to the existing covalent linking sites, making stronger gels.
alginate beads with epichlorohydrin in the presence of aqueous Furthermore, the bead volume decreased as the pH of the swelling
NaOH. Calcium was later removed by multiple treatments with solution was reduced. Protonation of the alginate carboxylate
sodium citrate solutions. Covalent polysaccharide network forma- groups at lower pH values reduced the difference between interior
tion using epichlorohydrin is well known, and has been reported and exterior ion concentrations. This led to gel shrinkage. The
for starch and cellulose [88e90]. The proposed mechanism for change in gel volume as a function of ethanol addition showed that
alginate crosslinking using epichlorohydrin is shown in Fig. 29. The above a certain ethanol concentration, the gel transformed from
crosslinked alginate beads were stable between a pH range of 1e13 a dissolved voluminous state to a solid compact state. A large
and from 0 to 100  C. They were also stable in high ionic strength volume decrease at high ethanol concentrations occurred due to
media and polar solvents. In addition, large improvements in precipitation-like behavior.
mechanical and chemical resistance properties were observed for Yeom et al. reported the use of covalently crosslinked sodium
covalently crosslinked alginates compared to Ca-alginate gels. alginate membranes for pervaporation separation [93]. Alginates
Skjk-Brk et al. reported base catalyzed covalent crosslinking are considered to be good membrane materials for dehydration of
of alginates with epichlorohydrin to form superswelling materials ethanol-water mixtures due to their ability to incorporate water
[91]. Ca-alginate beads were rst prepared in water and the solvent into the hydrophilic membrane structure [94]. However, excess
was exchanged with 96% ethanol. The crosslinking reaction was hydrophilicity may be disadvantageous as it can cause low selec-
performed by suspending the Ca-alginate beads in ethanol con- tivity and poor membrane stability. Thus, chemical modication via
taining epichlorohydrin and NaOH. The synthesized beads were covalent crosslinking was performed for membrane property
able to swell up to 100 times their dry volume without mass loss. enhancement. The cis-oriented vicinal hydroxyl groups at the C-2
Volume increase during re-swelling of dried beads was unaffected and C-3 positions can react with glutaraldehyde to form acetal-
by the presence of non-ionic solutes such as glucose and glycerol. linked alginate networks. Fig. 30(a) shows the mechanism for
Ionic solutes such as NaCl and Na-galacturonate slightly reduced acid catalyzed acetal formation, while Fig. 30(b) shows the reaction
the extent of volume increase. However, the reduction in degree of scheme for glutaraldehyde crosslinking of alginates. Na-alginate
lms were rst cast from aqueous solutions. The lms were dried
and reacted in an acetone solution containing glutaraldehyde and
HCl. Crosslinking density in alginate lms was controlled using the
concentration of glutaraldehyde present in the reactive solution.
Swelling measurements for the crosslinked lms as well as Na-
alginate lms were performed in solutions containing varying
ethanol:water ratios. The liquid volume fractions in Na-alginate
lms were higher compared to crosslinked lms. In addition,
crosslinked alginate lms contained a higher fraction of ethanol
compared to Na-alginate after absorption.
Several reports on glutaraldehyde crosslinking of alginates for
pervaporation separation have been described, wherein inorganic
compounds such as zeolite, silicotungstic acid, aluminum-
Fig. 29. Mechanism for alginate crosslinking using epichlorohydrin. containing mesoporous silica and molecular sieves have been
3298 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Fig. 30. (a) Mechanism of acid catalyzed acetalization of vicinal hydroxyl groups, (b) reaction scheme for covalent crosslinking of Na-alginate using glutaraldehyde.

incorporated into the crosslinked network [95e99]. Ko et al. intermolecular distances of the molecules and makes larger chiral
demonstrated the use of glutaraldehyde crosslinked alginates to spaces. Such large chiral spaces are unsuitable as they allow the
form superabsorbent bers for use in disposable diapers and isomers to pass through without interaction with the chiral envi-
sanitary napkins [100]. Alginates crosslinked with divalent cations ronment, resulting in low separation efciency.
such as Ca2 have low absorbency due to tightly packed junction Riyajan et al. reported the use of a glutaraldehyde crosslinked
points. Chemical crosslinking using glutaraldehyde enables high alginate gel for the encapsulation and controlled release of neem
absorbency because linkers of adjustable lengths can be used to Azadirachtin-A (Aza-A) [102]. Aza-A, an insecticidal tetranor-
create crosslink junctions allowing more liquid to be absorbed. triterpenoid, is highly unstable and degrades or isomerizes when
Saline absorption showed that an increase in the glutaraldehyde exposed to light. One way to protect Aza-A from degradation is
concentration caused a decrease in saline absorbency. Saline through microencapsulation within crosslinked alginate. The Aza-A
absorbency values were also dependent on the number of ionic entrapment efciency was found to be a function of crosslinking
groups present on the alginate. Deprotonation of the alginate acid time, wherein the efciency decreased with increasing time. Beads
groups increased saline absorbency by creation of a more hydro- crosslinked for shorter times swelled faster compared to the longer
philic environment. An important factor for application as super- crosslinking times. The higher degree of crosslinking achieved by
absorbent materials is the extraction of uncrosslinked or partially longer reaction times resulted in better control over the release
crosslinked alginate chains into solution causing mass loss and properties of Aza-A. Entrapped Aza-A was released completely
reduction in absorbency. Reduced mass loss in synthetic urine within 5, 10 and 25 h in the case of beads crosslinked for 10, 20 and
solutions was reported compared to saline. Synthetic urine 30 min respectively. Controlled release from covalently crosslinked
contains Ca2 ions which result in the formation of ionic crosslinks alginate matrices has been reported for other active molecules such
in alginates. Ionic crosslinking in addition to the already exisiting neem seed oil (NSO) [103]
covalent crosslinking helps in preventing the loss of alginate chains Neufeld et al. studied the reaction kinetics of alginate cross-
into solution. linking by glutaraldehyde using equilibrium swelling measure-
Jegal et al. described the use of glutaraldehyde crosslinked Na- ments [104]. An equilibrium swelling model was used to calculate
alginate membranes to separate the optical isomers of water- median pore sizes for the gels. The reaction rate was zero-order
soluble a-amino acids (for example, mixtures of D-tryptophan and with respect to the alginate concentration, and independent of
L-tryptophan) [101]. Two important characteristics are required to the alginate M/G composition and sequences. Thus, both M and G
create a membrane material for effective isomer separation e were equally reactive towards glutaraldehyde. However, the reac-
hydrophilicity and the presence of a large number of chiral centers. tion rate increased with an increase in the alginate molecular
Alginates satisfy both requirements and are therefore suitable for weight. Higher molecular weights caused lower mobility of chains,
isomer separation. The degrees of alginate crosslinking and thus improving the probability of collision with glutaraldehyde.
swelling govern the separation efciency. Swelling increases the Reaction rate was second-order with respect to glutaraldehyde
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3299

concentration and rst-order with respect to the acid catalyst alginate hydrogel materials were examined as potential materials
concentration. Lower temperatures slowed the rate of network for NP replacement. Na-alginate was rst converted to TBA-alginate
formation due to the high activation energy associated with acid and then dissolved in DMF for reaction. 2-Chloro-N-methyl pyr-
catalyzed acetalization reactions. idinium iodide (CMPI) was used to activate the carboxyl groups. A
Neufeld et al. later reported the synthesis of thermodynamically reactive diamine was then used along with triethylamine catalyst
controlled alginate network gels having pH-responsive properties to yield crosslinked alginates. Fig. 31 shows the reaction mecha-
[105]. Thermodynamic gels were synthesized by allowing the nism for synthesis of the alginate amide derivative using CMPI as an
crosslinking reaction to reach equilibrium. The reaction rate was activating agent, and (b) shows the crosslinking reaction using
zero-order with respect to the initial alginate concentration, but the a diamine.
critical gel time decreased at higher alginate concentrations. Higher DS values were controlled using the CMPI stoichiometry. Water
alginate fraction in mixtures led to a corresponding increase in the uptake and hydration kinetics revealed that the alginate hydrogel
hydroxyl group concentration. This improved the probability of swelled up to 250%, while NP swelled up to 200%. Rheology studies
collision with the crosslinking reagent to form the gel. The reaction showed a reversible sol-gel transition at a stress of 1270 Pa. This
kinetics were also affected by the addition of a cosolvent to the made the gels suitable for injection applications. Additionally, the
aqueous media. Up to 10e20% water-miscible organic solvents modulus values for crosslinked alginate hydrogels were very close
were added as cosolvents without precipitation. Dipolar aprotic to those found in NP. Chondrocyte cells proliferated and maintained
cosolvents such as acetone, dioxane and DMSO increase the rate of their ability to produce type II collagen within the alginate hydrogel
acetalization compared to pure water. The rate increase was an matrix.
effect of the differential solubility of glutaraldehyde between Johnson et al. reported the synthesis of crosslinked alginate
external solvent and the solvent present in the interior of gel beads. hydrogels using water soluble carbodiimide chemistry [107].
Dipolar protic cosolvents such as 1-propanol, ethanol and methanol Carboxylic acid groups on the alginate backbone reacted with
decreased the rate of acetalization compared to pure water. Alcohol hydroxyl groups to form a covalently crosslinked matrix. Na-alginate
groups from the cosolvent reacted with glutaraldehyde, resulting in membranes were rst cast from aqueous solutions, dried and then
a competitive side reaction and causing a decrease in crosslinking swollen in ethanol-water mixtures containing carbodiimide
reaction rate. The pH-responsive properties of the crosslinked coupling reagent. An optimum water:ethanol ratio was essential for
alginate gels were studied by subjecting them to stepwise pH- the reaction. At very low ethanol concentrations, sodium alginate
stimuli, and measuring the corresponding dynamic swelling membranes dissolved in the solvent mixture. At very low water
response. At pH 7.8, the carboxylic acid groups were deproto- concentrations, the carbodiimide coupling reagent was insoluble.
nated leading to increased hydrophilicity and electrostatic repul- The pH of the crosslinking medium had a signicant effect on nal
sion. This caused expansion of the network into a swollen state. At membrane swelling properties. At low pH values the carboxylic acid
pH 1.2 the carboxylic acid groups were protonated leading to groups were protonated. Since the carbodiimide mediated coupling
charge neutralization and limited solubility. This caused a rapid mechanism involves a protonated carboxylic acid, the crosslinking
chain rearrangement and gel contraction by the expulsion of water reaction was most efcient at low pH values. The degree of cross-
from the network. linking increased with the carbodiimide concentration of the
Barbucci et al. reported covalent crosslinking of alginate by mixture and reached a stable value above 60 mM. The heats of fusion
amide bond formation [106]. Covalently crosslinked hydrogels thus (DHm) and melting points (Tm) of membranes crosslinked at various
formed were used for the treatment of traumatic disorders of the carbodiimide concentrations were measured. The DHm values for
intervertebral disc (IVD). An important component of IVD is crosslinked membranes were considerably larger than those of non-
nucleus pulposus (NP), a type II collagen matrix consisting of crosslinked membranes. Thus, crosslinking increased the crystalline
chondrocyte cells along with proteoglycans such as versican and character of the membranes.
hyaluronan. The typically encountered hyal-based hydrogels have Mooney et al. reported the synthesis of covalently crosslinked
a disadvantage due to rapid enzymatic degradation. Therefore, alginates by partial oxidation of the vicinal hydroxyl groups at the C-

Fig. 31. (a) Mechanism for the synthesis of alginate amide derivative using CMPI activating agent, and (b) alginate crosslinking reaction using bifunctional amine reagent.
3300 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

2 and C-3 positions [108]. Low molecular weight poly(guluronate) reagent, another redox initiator system for grafting synthetic
segments were partially oxidized using sodium periodate to form polymers onto alginates [110,111]. An argument was presented
poly(aldehyde guluronate) [PAG], which was subsequently cross- wherein H-atom abstraction on the alginate backbone was reported
linked using adipic dihydrazide (Fig. 32). The acyl hydrazone bonds to have occurred by cleavage of the Alg-O-H bond to form Alg-O
linking the alginate chains were cleaved hydrolytically under radical. It may however be equally or more likely for H-atom
aqueous conditions, making the gel degradable. abstraction to occur by cleavage of the ring C-H bonds giving rise to
The degree of oxidation of alginate was controlled by varying carbon centered radicals. It should be noted that for alginates as
the molar ratio of sodium periodate to uronate monosaccharides. with other polysaccharides, grafting using a free radical initiator
With increasing dihydrazide concentration, a higher number of like CAN or Fentons reagent is rather unselective, since all C-H
crosslinks were formed. However, the crosslinks involved both bonds of the monosaccharides are labile due to the attached oxygen
intermolecular and intramolecular junctions. At an adipic dihy- functionality. Therefore it can be expected that graft initiation
drazide concentration of 150 mM, an optimum number of inter- points will occur at all monosaccharide carbons.
molecular crosslink junctions was formed which maximized Singh et al. described the grafting of polyacrylamide (PAAm)
modulus value. The pH of the crosslinking mixture had a signicant onto alginates using a ceric-induced system [112,113]. The advan-
effect on the mechanical properties of the nal gels. Crosslinking at tage of using acrylamide (AAm) compared to AN, MA or MMA for
pH 4.5 yielded gels with higher compressive moduli compared to radical grafting using CAN was that the homopolymer PAAm did
pH 7.4. This was a result of optimal hydrazide reactivity towards not form in the mixture. In the case of AAm, radical formation takes
aldehydes at low pH values. Compressive modulus values of the place only on the alginate backbone leading exclusively to grafted
nal gels were also controlled using the degree of oxidation of PAG, products. PAAm grafted alginate (PAAm-g-Alg) was evaluated for
where an increased number of aldehyde groups on the PAG back- its use as a commercial occulant. The occulation efciency of the
bone yielded gels with higher modulus values. PAAm-g-Alg was studied for suspensions of kaolin, iron ore and
coal nes, and its performance was found to be marginally better
3.7. Graft copolymerization of alginates than that of commercial occulants. Alginates containing longer
PAAm grafts performed better than short grafts. Hydrolysis of the
Graft copolymerization is a powerful method for modication of amide groups of PAAm-g-Alg was described through aqueous KOH
the physical and chemical properties of alginates. Grafting treatment [114]. When in contact with KOH under certain
synthetic polymer chains to the alginate backbone can introduce controlled conditions, the amide groups of PAAm-g-Alg can
hydrophobicity and steric bulkiness, which help protect the poly- hydrolyze without causing alginate degradation. Hydrolyzed
saccharide backbone from rapid dissolution and erosion. This in PAAm-g-Alg possessed enhanced occulation and thickening
turn translates into a sustained release of active molecules from characteristics vs. commercial occulants.
alginate matrices. Trivedi et al. reported the ceric ammonium Sa et al. reported the preparation of alginate based sustained
nitrate (CAN)-induced grafting of poly(acrylonitrile) (PAN), poly(- release interpenetrating network (IPN) tablets [115]. IPNs are
methyl acrylate) (PMA) or poly(methyl methacrylate) (PMMA) onto formed when one polymer is crosslinked in the presence of another
alginates [109]. Homopolymer by-products were separated from one. Such networks impart additional rigidity to simple crosslinked
the grafted alginates by solvent extraction. A comparison of the polymer matrices. Alginate based IPNs were formed by mixing
three different acrylate monomers showed that the grafting ef- together Na-alginate with PAAm-g-Alg followed by Ca-crosslinking.
ciency was highest for AN followed by MA followed by MMA. The The release of Diltiazem-HCl (DTZ) (a model water soluble drug)
homopolymer fractions formed showed a reverse trend where from the IPNs was studied. DTZ is a calcium channel blocker that is
MMA polymerized to a greater extent compared to MA and AN. widely used for the treatment of angina pectoris, arrhythmia and
Similarly, Trivedi et al. reported alginate grafting using Fentons hypertension. The DTZ release rate decreased as the ratio of PAAm-
g-Alg:Na-alginate increased. Reduced swelling of the matrix and
increased gel viscosity caused slow drug diffusion and hence sus-
tained release.
Aminabhavi et al. described the glutaraldehyde crosslinking
of PAAm-g-Alg to form membranes for pervaporation separation
of water-isopropanol mixtures as well as water-tetrahydrofuran
mixtures [116,117]. Li et al. reported the use of PMMA-g-Alg in
forming composites with hydroxyapatite for the study of properties
of the interface between the organic polymer and the inorganic
material [118]. Mollah et al. described an approach where 3-(tri-
methoxysilyl)propyl methacrylate was grafted onto Na-alginate to
form lms through casting. The lms were soaked in a silane
monomer mixture and were subsequently photocrosslinked to
study the physico-mechanical properties of the networks formed
[119]. Sa et al. described the synthesis of electrically responsive
alginate based hydrogels by grafting PAAm to the backbone and
subsequently hydrolyzing the amide [120]. Similar electro-
responsive hydrogels were also described by Yang et al. by graft-
ing poly(acrylic acid) (PAA) onto the alginate backbone [121]. The
derivatives were used as transdermal drug delivery systems for
ketoprofen, a model non-steroidal anti-inammatory drug. On
applying an electrical stimulus across the hydrogel, the positive
counterion moved towards the negative electrode, but the negative
carboxylate groups remained immobile. Consequently, an increased
Fig. 32. Sodium alginate crosslinking using adipic dihydrazide via reaction with PAG. osmotic pressure near the positive electrode led to deswelling of the
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3301

Fig. 33. Schematic describing the synthesis of Amylose-g-Alg [128]. Reprinted from Carbohyd Polym 2010; 82(2); Omagari Y., Kaneko Y., Kadokawa J-i.; Chemoenzymatic synthesis
of amylose-grafted alginate and its formation of enzymatic disintegratable beads; 394e400; Copyright 2010, with permission from Elsevier.

hydrogel, causing release of ketoprofen from the matrix. Kulkarni their ability to show lower critical solution temperature (LCST)
et al. also reported the use of hydrolyzed PAAm-g-Alg in forming behavior in aqueous solutions at 32  C. Alginates on the other hand
IPNs with carboxymethyl cellulose (CMC) for the pH dependent show pH responsive behavior due to the presence of carboxylic acid
release of ketoprofen [122]. Pal et al. reported the use of microwave groups on the backbone. The temperature and pH responsive
assisted synthesis for preparing PAAm-g-Alg polymers [123]. Liu properties of PNIPAAm and alginates were combined by a grafting
et al. described the synthesis of PMMA-g-Alg using potassium technique. Amine terminal PNIPAAm (PNIPAAm-NH2) was rst
ditelluratoargenate (III) as initiator [124]. Isklan et al. reported the synthesized and reacted with the carboxyl groups on the alginate
grafting of itaconic acid onto alginates using CAN as well as benzoyl backbone using EDC/NHS coupling. Hydrogel properties of bulk
peroxide initiators [125,126]. Xiao et al. described the synthesis of grafted PNIPAAm-g-Alg were compared to surface grafting of PNI-
polyvinyl alcohol grafted alginates (PVOH-g-Alg) to prepare ferro- PAAm onto Ca-hydrogel surfaces. Temperature dependent behavior
magnetic particles [127]. Vinyl acetate (VAc) was rst grafted to of PNIPAAm-g-Alg hydrogels was evident through a noticeable
create PVAc-g-Alg which was subsequently hydrolyzed to yield decrease in the swelling ratio above 30e35  C. Such behavior was
PVOH-g-Alg. The resulting graft copolymer was physically cross- not observed in the case of non-grafted alginate gels. Fig. 34 shows
linked using Fe (II) ions. Fe (II) was subsequently oxidized to Fe (III), a schematic representing the temperature dependent behavior of
creating ferromagnetic microparticles containing entrapped Fe3O4. PNIPAAm-g-Alg hydrogels. The pH responsive behavior was
Kadokawa et al. described a chemoenzymatic route to prepare evident through higher swelling ratios at increasing pH values due
amylose-grafted-alginates, and disintegrable beads thereof [128].
Amylose can be prepared by a phosphorylase-catalyzed enzymatic
polymerization initiated from a maltooligosaccharide primer. Thus,
an amine functional maltooligosaccharide primer was rst
synthesized and attached to the alginate backbone via aqueous
EDC/NHS coupling. Phosphorylase-catalyzed polymerization was
then performed to propagate amylose chains from the alginate
backbone (Fig. 33). Amylose-g-Alg thus synthesized was used to
form Ca-crosslinked gel beads containing entrapped uorescein (a
model dye). Release of the dye from the beads was studied as they
were exposed to b-amylase, an enzyme that catalyzes amylose
hydrolysis. Attachment of amylose chains impacted Ca-gel forma-
tion and for very long amylose chain lengths, gel formation did not
occur. On b-amylase treatment, the gel beads disintegrated causing
dye release.
Fig. 34. Schematic showing the temperature dependent behavior in PNIPAAm-g-Alg
Lee et al. described the grafting of poly(N-isopropylacrylamide)
hydrogels [129]. Reprinted from Polymer 2002; 43(26); Kim J.H., Lee S.B., Kim S.J., Lee
(PNIPAAm) to the alginate backbone to make temperature/pH Y.M.; Rapid temperature/pH response of porous alginate-g-poly(N-isopropylacrylamide)
responsive hydrogels [129]. PNIPAAm hydrogels are well known for hydrogels; 7549e7558; Copyright 2002, with permission from Elsevier.
3302 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

Fig. 35. Synthesis of PDMAEMA-g-OAlg.

Fig. 36. Schematic showing the effect of pH and ionic strength on PDMAEMA-g-OAlg hydrogels [130]. Reprinted from Int J Pharm 2009; 371(1e2); Gao C., Liu M., Chen S., Jin S.,
Chen J.; Preparation of oxidized sodium alginate-graft-poly((2-dimethylamino) ethyl methacrylate) gel beads and in vitro controlled release behavior of BSA; 16e24; Copyright
2009, with permission from Elsevier.

to chain expansion from the presence of ionic carboxylate groups groups takes place leading to the formation of polyelectrolyte
on the backbone. complexes with the PDMAEMA cations. This causes a decrease in
Liu et al. reported the grafting of poly((2-dimethylamino)ethyl the equilibrium swelling ratio. Increasing the ionic strength (NaCl
methacrylate) (PDMAEMA) onto oxidized Na-alginate (OAlg) to concentration) initially causes an increase in the swelling ratio due
study the in vitro controlled release behavior of BSA protein from to disruption of the polyelectrolyte complexes, and later decreases
the hydrogels [130]. PDMAEMA is a water soluble polymer with the swelling ratio as the gel network is destroyed by Na-Ca ion
tertiary amine groups containing side chains, and exhibits LCST exchange.
behavior. To use PDMAEMA for biomedical applications, the
synthesis of PDMAEMA-g-OAlg hydrogel beads was described. As 4. Conclusions and prospects
mentioned previously in this paper Na-alginates do not degrade in
aqueous media, but partially oxidized alginates do [64]. OAlg was Advances in the biochemistry of the complex alginate poly-
therefore rst synthesized from Na-alginate by periodate oxidation, saccharides have provided opportunities to control mono-
wherein the vicinal 2-OH and 3-OH groups on the backbone were saccharide sequence that are unique in polysaccharide chemistry
converted to aldehydes. The aldehyde groups were then reacted [131]. This provides insight into the structure-property relation-
with amine terminal PDMAEMA to form a stable Schiffs base in the ships of alginates that can illuminate its important natural func-
form of an imine (Fig. 35). Grafting of PDMAEMA chains to the tions, for example in structural gels in kelp and in protective
alginate chain allowed control over the equilibrium swelling ratio bacterial lms made by P. aeruginosa (which play a critical part in
of hydrogels employing pH and ionic strength of the media. Fig. 36 the progression of cystic brosis [3]). The developing ability to
shows an illustration of the swelling behavior. As the pH of the chemically modify alginate creates signicant potential for tailoring
medium is increased above 3.0, ionization of the carboxylic acid alginate materials for particular applications. The ability of
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3303

alginates to form hydrogels under mild pH and temperature [3] Ramsey DM, Wozniak DJ. Understanding the control of Pseudomonas aeru-
ginosa alginate synthesis and the prospects for management of chronic
conditions, along with the inherent biocompatibility of most algi-
infections in cystic brosis. Mol Microbiol 2005;56:309e22.
nates, makes these materials particularly attractive for biomedical [4] Zimmermann H, Shirley S, Zimmermann U. Alginate-based encapsulation of
applications including drug delivery, tissue engineering, and cells: past, present, and future. Curr Diab Rep 2007;7:314e20.
implantation of protected living cells. More importantly, chemical [5] Ghidoni I, Chlapanidas T, Bucco M, Crovato F, Marazzi M, Vigo D, et al.
Alginate cell encapsulation: new advances in reproduction and cartilage
derivatization is the most preferred method to create next gener- regenerative medicine. Cytotechnology 2008;58:49e56.
ation alginate biomaterials having enhanced or altogether new [6] Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog
properties. A thorough understanding of the chemistry of alginates Polym Sci 2012;37:106e26.
[7] Soonshiong P, Feldman E, Nelson R, Heintz R, Yao Q, Yao ZW, et al. Long-term
is vital to this goal. Preparation of such tailored derivatives however reversal of diabetes by the injection of immunoprotected islets. Proc Natl
has been limited by two main factors; the complete lack of organic Acad Sci USA 1993;90:5843e7.
solubility of alginic acid, and the proclivity for alginic acid to [8] Hay ID, Rehman ZU, Ghafoor A, Rehm BHA. Bacterial biosynthesis of algi-
nates. J Chem Tech Biotechnol 2010;85:752e9.
decompose under acidic, alkaline, and reductive conditions. Even [9] Remminghorst U, Rehm BH. Bacterial alginates: from biosynthesis to appli-
so, the advent of carbonyl activating reagents that work in water, cations. Biotechnol Lett 2006 Nov;28:1701e12.
for example DCC and sulfonated N-hydroxysuccinimide, has [10] Gacesa P. Bacterial alginate biosynthesiserecent progress and future pros-
pects. Microbiology 1998;144:1133e43.
permitted selective modication of the carboxyl groups of alginic [11] Rehm BHA, Valla S. Bacterial alginates: biosynthesis and applications. Appl
acid. This permits not only modication of bulk properties like Microbiol Biotechnol 1997;48:281e8.
solubility, but also attachment of functional substituents like tar- [12] Franklin MJ, Ohman DE. Mutant analysis and cellular localization of the AlgI,
AlgJ, and AlgF proteins required for O acetylation of alginate in Pseudomonas
geting ligands and uorophores. Modication of the hydroxyl
aeruginosa. J Bacteriol 2002;184:3000e7.
groups in aqueous media is more challenging, but has been [13] Franklin MJ, Douthit SA, McClure MA. Evidence that the algI/algJ gene
accomplished under heterogeneous conditions, for example by gel cassette, required for O acetylation of Pseudomonas aeruginosa alginate,
acylation, and using techniques that can work in water, for example evolved by lateral gene transfer. J Bacteriol 2004;186:4759e73.
[14] Franklin MJ, Ohman DE. Identication of algF in the alginate biosynthetic
by acetal formation. The discovery of organic solvents for the tet- gene cluster of Pseudomonas aeruginosa which is required for alginate
rabutylammonium salts of alginic acid (TBAF in polar aprotic acetylation. J Bacteriol 1993;175:5057e65.
solvents) greatly expands the potential for alginate modication, [15] Gimmestad M, Sletta H, Ertesvag H, Bakkevig K, Jain S, Suh SJ, et al. The
Pseudomonas uorescens AlgG protein, but not its mannuronan C-5-
encompassing a much wider variety of reagents. By analogy with epimerase activity, is needed for alginate polymer formation. J Bacteriol
cellulose [132], these new solvent systems should also enable 2003;185:3515e23.
enhanced exploration of chemoselectivity (e.g., OH vs. COOH [16] Chitnis CE, Ohman DE. Cloning of Pseudomonas aeruginosa algG, which
controls alginate structure. J Bacteriol 1990;172:2894e900.
reaction) and regioselectivity (M vs. G, 2-OH vs. 3-OH within each [17] Franklin MJ, Chitnis CE, Gacesa P, Sonesson A, White DC, Ohman DE. Pseu-
monosaccharide), because of the more selective reagents and domonas aeruginosa AlgG is a polymer level alginate C5-mannuronan
milder conditions that are now possible. Such regio- and chemo- epimerase. J Bacteriol 1994;176:1821e30.
[18] Zimmermann U, Klck G, Federlin K, Hannig K, Kowalski M, Bretzel RG, et al.
selectivity should enable combined biochemical and chemical Production of mitogen-contamination free alginates with variable ratios of
routes to alginate derivatives with exquisite control over both mannuronic acid to guluronic acid by free ow electrophoresis. Electro-
monosaccharide sequence and substitution position, to a degree phoresis 1992;13:269e74.
[19] Klck G, Frank H, Houben R, Zekorn T, Horcher A, Siebers U, et al. Production
unmatched in polysaccharide chemistry to date (the only compa-
of puried alginates suitable for use in immunoisolated transplantation. Appl
rable examples involve de novo synthesis of polysaccharides by Microbiol Biotechnol 1994;40:638e43.
laborious, standard carbohydrate chemistry involving multiple [20] Haug A, Larsen B, Smidsrd O. A study of the constitution of alginic acid by
protection, deprotection and glycosylation steps [60]). In addition partial hydrolysis. Acta Chem Scand 1966;20:183e90.
[21] Haug A, Larsen B, Smidsrd O. Studies on the sequence of uronic acid resi-
to the anticipation of progress against the important challenges of dues in alginic acid. Acta Chem Scand 1967;21:691e704.
overcoming regiocontrol and chemoselectivity issues in alginate [22] Painter TJ, Smidsrd O, Haug A. A computer study of the changes in
derivative synthesis, we can anticipate that researchers in the near composition-distribution occurring during random depolymerisation of
a binary linear heteropolysaccharide. Acta Chem Scand 1968;22:1637e48.
future will be addressing analytical issues. The ability to use tech- [23] Larsen B, Smidsrd O, Painter TJ, Haug A. Calculation of the nearest-
niques like multidimensional NMR experiments, partial and full neighbour frequencies in fragments of alginate from the yields of free
polysaccharide hydrolysis, combined LC/MS and GC/MS, and other monomers after partial hydrolysis. Acta Chem Scand 1970;24:726e8.
[24] Smidsrod O, Whittington SG. Monte Carlo investigation of chemical inho-
spectroscopic and chromatographic techniques to give a more mogeneity in polymers. Macromolecules 1969;2:42e4.
complete picture of substitution position and sequence will be [25] Penman A, Sanderson GR. A method for the determination of uronic acid
essential accompaniment to the anticipated synthetic advances. In sequence in alginates. Carbohydr Res 1972;25:273e82.
[26] Grasdalen H, Larsen B, Smidsrd O. Pmr study of the composition and
this way the great promise of alginates as biomaterials for sequence of uronate residues in alginates. Carbohydr Res 1979;68:23e31.
biomedical and other applications can be fully realized. [27] Grasdalen H. High-Field, H-1-Nmr Spectroscopy of alginate - sequential
structure and linkage conformations. Carbohydr Res 1983;118:255e60.
[28] Pawar SN, Edgar KJ. Chemical modication of alginates in organic solvent
Acknowledgements systems. Biomacromolecules 2011;12:4095e103.
[29] Sikorski P, Mo F, Skjk-Brk G, Stokke BT. Evidence for egg-box-compatible
We gratefully acknowledge support for SNP from USDA NIFA interactions in calcium-alginate gels from ber X-ray diffraction. Bio-
macromolecules 2007;8:2098e103.
(USDA NIFA) grant number 2010-34489-20784, and from the [30] Donati I, Holtan S, Mrch YA, Borgogna M, Dentini M, Skjk-Brk G. New
Sustainable Engineered Materials Institute at Virginia Tech hypothesis on the role of alternating sequences in calcium-alginate gels.
(Sustainable Engineered Materials Institute at Virginia Tech), the Biomacromolecules 2005;6:1031e40.
[31] Morch YA, Donati I, Strand BL, Skjk-Brk G. Effect of Ca2, Ba2, and Sr2 on
Institute for Critical Technology and Applied Science for facility
alginate microbeads. Biomacromolecules 2006;7:1471e80.
support, and the Macromolecules and Interfaces Institute for [32] Skjk-Brk G, Grasdalen H, Smidsrd O. Inhomogeneous polysaccharide
educational support. ionic gels. Carbohydr Polym 1989;10:31e54.
[33] Draget KI, Skjk-Brk G, Smidsrd O. Alginic acid gels: the effect of alginate
chemical composition and molecular weight. Carbohydr Polym 1994;25:
References 31e8.
[34] Timell TE. The acid hydrolysis of glycosides: I. General conditions and the
[1] Draget KI. Alginates. In: Phillips GO, Williams PA, editors. Handbook of effect of the nature of the aglycone. Can J Chem 1964;42:1456.
hydrocolloids; 2009. p. 379e95. [35] Smidsrd O, Haug A, Larsen B. The inuence of pH on the rate of hydrolysis of
[2] Boateng JS, Matthews KH, Stevens HNE, Eccleston GM. Wound healing acidic polysaccharides. Acta Chem Scand 1966;20:1026e34.
dressings and drug delivery systems: a review. J Pharm Sci 2008;97: [36] Tsujino I, Saito T. A new unsaturated uronide isolated from alginase hydro-
2892e923. lysate. Nature 1961;192:970e1.
3304 S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305

[37] Preiss J, Ashwell G. Alginic acid metabolism in bacteria. J Biol Chem 1962; [71] Babak VG, Skotnikova EA, Lukina IG, Pelletier S, Hubert P, Dellacherie E.
237:309e16. Hydrophobically associating alginate derivatives: surface tension properties
[38] Haug AL, Larsen B, Smidsrd O. The degradation of alginates at different pH of their mixed aqueous solutions with oppositely charged surfactants.
values. Acta Chem Scand 1963;17:1466e8. J Colloid Interface Sci 2000;225:505e10.
[39] Haug AL, Larsen B, Smidsrd O. Alkaline degradation of alginate. Acta Chem [72] Pelletier S, Hubert P, Lapicque F, Payan E, Dellacherie E. Amphiphilic deriv-
Scand 1967;21:2859e70. atives of sodium alginate and hyaluronate: synthesis and physico-chemical
[40] Haug A, Larsen B. Phenolic compounds in brown algae: I. The presence of properties of aqueous dilute solutions. Carbohyd Polym 2000;43:343e9.
reducing compounds in Ascophyllum nodosum (L.) Le Jol. Acta Chem Scand [73] Leonard M, Rastello de Boisseson M, Hubert P, Dellacherie E. Production of
1958;12:650. microspheres based on hydrophobically associating alginate derivatives by
[41] Smidsrd O, Haug A, Larsen B. The inuence of reducing substances on the dispersion/gelation in aqueous sodium chloride solutions. J Biomed Mater
rate of degradation of alginates. Acta Chem Scand 1963;17:1473e4. Res A 2004;68A:335e42.
[42] Smidsrd O, Haug A, Larsen B. Degradation of alginate in the presence of [74] Galant C, Kjoniksen AL, Nguyen GTM, Knudsen KD, Nystrom B. Altering
reducing compounds. Acta Chem Scand 1963;17:2628e37. associations in aqueous solutions of a hydrophobically modied alginate in
[43] Leo WJ, Mcloughlin AJ, Malone DM. Effects of sterilization treatments on the presence of beta-cyclodextrin monomers. J Phys Chem B 2006;110:
some properties of alginate solutions and gels. Biotechnol Prog 1990;6: 190e5.
51e3. [75] Burckbuchler V, Kjoniksen AL, Galant C, Lund R, Amiel C, Knudsen KD, et al.
[44] Chamberlain NH, Cunningham GE, Speakman JB. Alginic acid diacetate. Rheological and structural characterization of the interactions between
Nature 1946;158:553. cyclodextrin compounds and hydrophobically modied alginate. Bio-
[45] Wassermann A. Alginic acid-acetate. J Chem Soc; 1948:197. macromolecules 2006;7:1871e8.
[46] Schweiger RG. Acetylation of alginic acid. I. Preparation and viscosities of [76] Augst AD, Kong HJ, Mooney DJ. Alginate hydrogels as biomaterials. Macromol
algin acetates. J Org Chem 1962;27:1786e9. Biosci 2006;6:623e33.
[47] Schweiger RG. Acetylation of alginic acid. II. Reaction of algin acetates with [77] Shapiro L, Cohen S. Novel alginate sponges for cell culture and trans-
calcium and other divalent ions. J Org Chem 1962;27:1789e91. plantation. Biomaterials 1997;18:583e90.
[48] Grasdalen H, Larsen B, Smidsrd O. C-13-Nmr studies of alginate. Carbohydr [78] Donati I, Vetere A, Gamini A, Coslovi A, Campa C, Paoletti S. Galactose-
Res 1977;56:C11e5. substituted alginate 1: Preliminary characterization and study of gelling
[49] Grasdalen H, Larsen B, Smidsrd O. C-13-Nmr studies of monomeric properties. Biomacromolecules 2003;4:624e31.
composition and sequence in alginate. Carbohydr Res 1981;89:179e91. [79] Donati I, Coslovi A, Gamini A, Vetere A, Campa C, Paoletti S. Galactose-
[50] Skjk-Brk G, Paoletti S, Gianferrara T. Selective acetylation of mannuronic substituted alginate 2: conformational aspects. Biomacromolecules 2004;5:
acid residues in calcium alginate gels. Carbohydr Res 1989;185:119e29. 186e96.
[51] Skjk-Brk G, Zanetti F, Paoletti S. Effect of acetylation on some solution and [80] Donati I, Draget KI, Borgogna M, Paoletti S, Skjk-Brk G. Tailor-made
gelling properties of alginates. Carbohydr Res 1989;185:131e8. alginate bearing galactose moieties on mannuronic residues: selective
[52] Khler S, Heinze T. New solvents for cellulose: dimethyl sulfoxide/ammo- modication achieved by a chemoenzymatic strategy. Biomacromolecules
nium uorides. Macromol Biosci 2007;7:307e14. 2005;6:88e98.
[53] Sun H, DiMagno SG. Anhydrous tetrabutylammonium uoride. J Am Chem [81] Yang J, Goto M, Ise H, Cho C-S, Akaike T. Galactosylated alginate as a scaffold
Soc 2005;127:2050e1. for hepatocytes entrapment. Biomaterials 2002;23:471e9.
[54] Coleman RJ, Lawrie G, Lambert LK, Whittaker M, Jack KS, Grndahl L. [82] Irwin MA. The liver: biology and pathobiology. 5th ed.; 2009.
Phosphorylation of alginate: synthesis, characterization, and evaluation of [83] Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels as synthetic
in vitro mineralization capacity. Biomacromolecules 2011;12:889e97. extracellular matrix materials. Biomaterials 1999;20:45e53.
[55] Alban S, Schauerte A, Franz G. Anticoagulant sulfated polysaccharides: part I. [84] Rowley JA, Mooney DJ. Alginate type and RGD density control myoblast
Synthesis and structureeactivity relationships of new pullulan sulfates. phenotype. J Biomed Mater Res 2002;60:217e23.
Carbohydr Polym 2002;47:267e76. [85] Draget KI, Smidsrd O, Skjk-Brk G. Alginates from algae. In: Steinbchel A,
[56] Linhardt RJ, Claude S. Hudson Award address in carbohydrate chemistry. Rhee SK, editors. Polysaccharides and polyamides in the food industry:
Heparin: structure and activity. J Med Chem 2003;46:2551e64. properties, production, and patents; 2005. p. 1e30.
[57] Ronghua H, Yumin D, Jianhong Y. Preparation and in vitro anticoagulant [86] Thu B, Bruheim P, Espevik T, Smidsrd O, SoonShiong P, Skjk-Brk G.
activities of alginate sulfate and its quaterized derivatives. Carbohydr Polym Alginate polycation microcapsules 2. Some functional properties. Biomate-
2003;52:19e24. rials 1996;17:1069e79.
[58] Freeman I, Kedem A, Cohen S. The effect of sulfation of alginate hydrogels on [87] Grasselli M, Diaz LE, Cascone O. Beaded matrices from cross-linked alginate
the specic binding and controlled release of heparin-binding proteins. for afnity and ion exchange chromatography of proteins. Biotechnol Tech
Biomaterials 2008;29:3260e8. 1993;7:707e12.
[59] Fan L, Jiang L, Xu Y, Zhou Y, Shen Y, Xie W, et al. Synthesis and anticoagulant [88] Chang C, Zhang L, Zhou J, Zhang L, Kennedy JF. Structure and properties of
activity of sodium alginate sulfates. Carbohydr Polym 2011;83:1797e803. hydrogels prepared from cellulose in NaOH/urea aqueous solutions. Carbo-
[60] Gama CI, Tully SE, Sotogaku N, Clark PM, Rawat M, Vaidehi N, et al. Sulfation hydr Polym 2010;82:122e7.
patterns of glycosaminoglycans encode molecular recognition and activity. [89] Zhou J, Chang C, Zhang R, Zhang L. Hydrogels prepared from unsubstituted
Nat Chem Biol 2006;2:467e73. cellulose in NaOH/urea aqueous solution. Macromol Biosci 2007;7:804e9.
[61] Rogers CJ, Clark PM, Tully SE, Abrol R, Garcia KC, Goddard WA, et al. Eluci- [90] Kartha KPR, Srivastava H. Reaction of epichlorohydrin with carbohydrate
dating glycosaminoglycan-protein-protein interactions using carbohydrate polymers. II. Starch reaction mechanism and physicochemical properties of
microarray and computational approaches. Proc Natl Acad Sci USA 2011;108: modied starch. Starch/Strke 1985;37:297e306.
9747e52. [91] Moe ST, Skjk-Brk G, Smidsrd O. Covalently cross-linked sodium alginate
[62] Carr M-C, Delestre C, Hubert P, Dellacherie E. Covalent coupling of a short beads. Food Hydrocolloid 1991;5:119e23.
polyether on sodium alginate: synthesis and characterization of the resulting [92] Moe ST, Skjk-Brk G, Elgsaeter A, Smidsrd O. Swelling of covalently
amphiphilic derivative. Carbohyd Polym 1991;16:367e79. crosslinked alginate gels: inuence of ionic solutes and nonpolar solvents.
[63] Scott JE, Tigwell MJ, Phelps CF, Nieduszynski IA. Mechanism of scission of Macromolecules 1993;26:3589e97.
alginate chains by periodate. Carbohydr Res 1976;47:105e17. [93] Yeom CK, Lee K-H. Characterization of sodium alginate membrane cross-
[64] Bouhadir KH, Lee KY, Alsberg E, Damm KL, Anderson KW, Mooney DJ. linked with glutaraldehyde in pervaporation separation. J Appl Polym Sci
Degradation of partially oxidized alginate and its potential application for 1998;67:209e19.
tissue engineering. Biotechnol Prog 2001;17:945e50. [94] Yeom CK, Jegal JG, Lee KH. Characterization of relaxation phenomena and
[65] Marc Duval J, Delestre C, Carr M-C, Hubert P, Dellacherie E. Synthesis and permeation behaviors in sodium alginate membrane during pervaporation
characterization of some covalent dextran-polyoxyethyleneglycol deriva- separation of ethanol-water mixture. J Appl Polym Sci 1996;62:1561e76.
tives. Carbohydr Polym 1991;15:233e42. [95] Patil MB, Veerapur RS, Bhat SD, Madhusoodana CD, Aminabhavi TM. Hybrid
[66] Sinquin A, Hubert P, Dellacherie E. Amphiphilic derivatives of alginate: composite membranes of sodium alginate for pervaporation dehydration of
evidence for intra- and intermolecular hydrophobic associations in aqueous 1,4-dioxane and tetrahydrofuran. Desalination Water Treat 2009;3:11e20.
solution. Langmuir 1993;9:3334e7. [96] Adoor SG, Manjeshwar LS, Bhat SD, Aminabhavi TM. Aluminum-rich zeolite
[67] Maestro A, Gonzalez C, Gutierrez JM. Rheological behavior of hydrophobi- beta incorporated sodium alginate mixed matrix membranes for pervapo-
cally modied hydroxyethyl cellulose solutions: a linear viscoelastic model. ration dehydration and esterication of ethanol and acetic acid. J Memb Sci
J Rheol 2002;46:127e43. 2008;318:233e46.
[68] Sinquin A, Hubert P, Dellacherie E. Intermolecular associations in hydro- [97] Teli SB, Gokavi GS, Sairam M, Aminabhavi TM. Highly water selective sili-
phobically modied derivatives of propyleneglycol alginate. Polymer 1994; cotungstic acid (H4SiW12O40) incorporated novel sodium alginate hybrid
35:3557e60. composite membranes for pervaporation dehydration of acetic acid. Sep
[69] Sinquin A, Houzelle MC, Hubert P, Choplin L, Viriot ML, Dellacherie E. Purif Technol 2007;54:178e86.
Amphiphilic derivatives of propylene glycol alginate: a revisit of their phys- [98] Patil MB, Veerapur RS, Patil SA, Madhusoodana CD, Aminabhavi TM. Prepa-
icochemical behavior in dilute aqueous solution. Langmuir 1996;12:3779e82. ration and characterization of lled matrix membranes of sodium alginate
[70] Sinquin A, Hubert P, Marchal P, Choplin L, Dellacherie E. Rheological prop- incorporated with aluminum-containing mesoporous silica for pervapora-
erties of semi-dilute aqueous solutions of hydrophobically modied tion dehydration of alcohols. Sep Purif Technol 2007;54:34e43.
propylene glycol alginate derivatives. Colloids Surf A Physicochem Eng Asp [99] Bhat SD, Mallikarjuna NN, Aminabhavi TM. Microporous alumino-phosphate
1996;112:193e200. (AlPO4-5) molecular sieve-loaded novel sodium alginate composite
S.N. Pawar, K.J. Edgar / Biomaterials 33 (2012) 3279e3305 3305

membranes for pervaporation dehydration of aqueous-organic mixtures [117] Kurkuri MD, Kumbar SG, Aminabhavi TM. Synthesis and characterization of
near their azeotropic compositions. J Memb Sci 2006;282:473e83. polyacrylamide-grafted sodium alginate copolymeric membranes and their
[100] Kim Y-J, Yoon K-J, Ko S-W. Preparation and properties of alginate superab- use in pervaporation separation of water and tetrahydrofuran mixtures.
sorbent lament bers crosslinked with glutaraldehyde. J Appl Polym Sci J Appl Polym Sci 2002;86:272e81.
2000;78:1797e804. [118] Yang W, Zhang L, Wu L, Li J, Wang J, Jiang H, et al. Synthesis and charac-
[101] Kim JH, Jegal J, Kim JH, Lee KH, Lee Y. Enantioselective permeation of alpha- terization of MMAeNaAlg/hydroxyapatite composite and the interface
amino acid optical isomers through crosslinked sodium alginate membranes. analyse with molecular dynamics. Carbohydr Polym 2009;77:331e7.
J Appl Polym Sci 2003;89:3046e51. [119] Mollah MZI, Khan MA, Hoque MA, Aziz A. Studies of physico-mechanical
[102] Riyajan S-A, Sakdapipanich J. Development of a controlled release neem properties of photo-cured sodium alginate with silane monomer. Carbo-
capsule with a sodium alginate matrix, crosslinked by glutaraldehyde and hydr Polym 2008;72:349e55.
coated with natural rubber. Polym Bull 2009;63:609e22. [120] Kulkarni RV, Setty CM, Sa B. Polyacrylamide-g-alginate-based electrically
[103] Kulkarni AR, Soppimath KS, Aminabhavi TM, Dave AM, Mehta MH. Glutar- responsive hydrogel for drug delivery application: synthesis, character-
aldehyde crosslinked sodium alginate beads containing liquid pesticide for ization, and formulation development. J Appl Polym Sci 2010;115:
soil application. J Control Release 2000;63:97e105. 1180e8.
[104] Chan AW, Whitney RA, Neufeld RJ. Kinetic controlled synthesis of pH- [121] Yang S, Liu G, Cheng Y, Zheng Y. Electroresponsive behavior of sodium
responsive network alginate. Biomacromolecules 2008;9:2536e45. alginate-g-poly (acrylic acid) hydrogel under DC electric eld. J Macromol Sci
[105] Chan AW, Whitney RA, Neufeld RJ. Semisynthesis of a controlled stimuli- Part A 2009;46:1078e82.
responsive alginate hydrogel. Biomacromolecules 2009;10:609e16. [122] Kulkarni RV, Sa B. Novel pH-sensitive interpenetrating network hydrogel
[106] Leone G, Torricelli P, Chiumiento A, Facchini A, Barbucci R. Amidic alginate beads of carboxymethylcellulose e (polyacrylamide-grafted-alginate) for
hydrogel for nucleus pulposus replacement. J Biomed Mater Res-A 2008; controlled release of ketoprofen: preparation and characterization. Curr
84A:391e401. Drug Deliv 2008;5:256e64.
[107] Xu JB, Bartley JP, Johnson RA. Preparation and characterization of alginate [123] Sen G, Singh RP, Pal S. Microwave-initiated synthesis of polyacrylamide
hydrogel membranes crosslinked using a water-soluble carbodiimide. J Appl grafted sodium alginate: synthesis and characterization. J Appl Polym Sci
Polym Sci 2003;90:747e53. 2010;115:63e71.
[108] Bouhadir KH, Hausman DS, Mooney DJ. Synthesis of cross-linked poly(- [124] Liu Y, Yang L, Li J, Shi Z. Grafting of methyl methacrylate onto sodium algi-
aldehyde guluronate) hydrogels. Polymer 1999;40:3575e84. nate initiated by potassium ditelluratoargentate(III). J Appl Polym Sci 2005;
[109] Shah SB, Patel CP, Trivedi HC. Ceric-induced grafting of acrylate monomers 97:1688e94.
onto sodium alginate. Carbohydr Polym 1995;26:61e7. _
[125] Isklan N, Kursun F, Inal M. Graft copolymerization of itaconic acid onto
[110] Shah SB, Patel CP, Trivedi HC. Fentons reagent-initiated graft copolymeri- sodium alginate using ceric ammonium nitrate as initiator. J Appl Polym Sci
zation of acrylonitrile onto sodium alginate. J Appl Polym Sci 1994;51: 2009;114:40e8.
1421e6. _
[126] Isklan N, Kursun F, Inal M. Graft copolymerization of itaconic acid onto
[111] Shah SB, Patel CP, Trivedi HC. Kinetics and reaction mechanism of Fentons- sodium alginate using benzoyl peroxide. Carbohydr Polym 2010;79:665e72.
reagent-initiated graft copolymerization of acrylonitrile onto sodium algi- [127] Ma P, Xiao C, Li L, Shi H, Zhu M. Facile preparation of ferromagnetic alginate-
nate. J Appl Polym Sci 1994;52:857e60. g-poly(vinyl alcohol) microparticles. Eur Polym J 2008;44:3886e9.
[112] Tripathy T, Pandey SR, Karmakar NC, Bhagat RP, Singh RP. Novel occulating [128] Omagari Y, Kaneko Y, Kadokawa J-I. Chemoenzymatic synthesis of amylose-
agent based on sodium alginate and acrylamide. Eur Polym J 1999;35:2057e72. grafted alginate and its formation of enzymatic disintegratable beads. Car-
[113] Tripathy T, Singh RP. Characterization of polyacrylamide-grafted sodium bohydr Polym 2010;82:394e400.
alginate: a novel polymeric occulant. J Appl Polym Sci 2001;81:3296e308. [129] Kim JH, Lee SB, Kim SJ, Lee YM. Rapid temperature/pH response of porous
[114] Biswal DR, Singh RP. The occulation and rheological characteristics of alginate-g-poly(N-isopropylacrylamide) hydrogels. Polymer 2002;43:
hydrolyzed and unhydrolyzed grafted sodium alginate in aqueous solutions. 7549e58.
J Appl Polym Sci 2004;94:1480e8. [130] Gao C, Liu M, Chen S, Jin S, Chen J. Preparation of oxidized sodium alginate-
[115] Mandal S, Basu SK, Sa B. Ca2 ion cross-linked interpenetrating network graft-poly((2-dimethylamino) ethyl methacrylate) gel beads and in vitro
matrix tablets of polyacrylamide-grafted-sodium alginate and sodium algi- controlled release behavior of BSA. Int J Pharm 2009;371:16e24.
nate for sustained release of diltiazem hydrochloride. Carbohydr Polym [131] Morch YA, Donati I, Strand BL, Skjk-Brk G. Molecular engineering as an
2010;82:867e73. approach to design new functional properties of alginate. Bio-
[116] Toti US, Aminabhavi TM. Synthesis and characterization of polyacrylamide macromolecules 2007;8:2809e14.
grafted sodium alginate membranes for pervaporation separation of water [132] Fox SC, Li B, Xu D, Edgar KJ. Regioselective esterication and etherication of
plus isopropanol mixtures. J Appl Polym Sci 2004;92:2030e7. cellulose - a review. Biomacromolecules 2011;12:1956e72.

Potrebbero piacerti anche