Sei sulla pagina 1di 13

Journal of Controlled Release 161 (2012) 680–692

Contents lists available at SciVerse ScienceDirect

Journal of Controlled Release


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

Review

Hydrogels for protein delivery in tissue engineering


Roberta Censi a,⁎, Piera Di Martino a, Tina Vermonden b, Wim E. Hennink b
a
School of Pharmacy, University of Camerino, via S. Agostino 1, 62032, Camerino (MC), Italy
b
Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences,Utrecht University, PO Box 80082, 3508 TB Utrecht, The Netherlands

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

Article history: Tissue defects caused by diseases or trauma present enormous challenges in regenerative medicine. Recently, a
Received 13 January 2012 better understanding of the biological processes underlying tissue repair led to the establishment of new ap-
Accepted 2 March 2012 proaches in tissue engineering which comprise the combination of biodegradable scaffolds and appropriate
Available online 8 March 2012
cells together with specific environmental cues, such as growth or adhesive factors. These factors (in fact pro-
teins) have to be loaded and sustainably released from the scaffolds in time. This review provides an overview
Keywords:
Hydrogels
of the various hydrogel technologies that have been proposed to control the release of bioactive molecules of in-
Growth factors terest for tissue engineering applications.
Protein delivery In particular, after a brief introduction on bioactive protein drugs that have remarkable relevance for tissue
Tissue engineering engineering, this review will discuss their release mechanisms from hydrogels, their encapsulation and immobiliza-
Controlled release tion methods and will overview the main classes of hydrogel forming biomaterials used in vitro and in vivo to release
them.
Finally, an outlook on future directions and a glimpse into the current clinical developments are provided.
© 2012 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 680
2. Proteins in the healing process: an introduction to their delivery approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 681
3. Protein release strategies from hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 681
3.1. Physical entrapment of growth factors into hydrogels: general principles and release mechanisms . . . . . . . . . . . . . . . . . . 681
3.1.1. Synthetic polymers for the physical encapsulation of growth factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
3.1.2. Natural polymers for the physical encapsulation of growth factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
3.2. Covalent binding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 688
3.3. Dual/multiple delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 689
4. Conclusions and outlook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 689
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 690
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 690

1. Introduction proved crucial for achieving effective tissue repair or replacement. There-
fore, modern tissue engineering aims at assisting the re-growth of func-
Controlled drug delivery has seen rapid advances in the last few de- tional tissues by combining cells and engineering materials with
cades with the introduction of novel biomaterials and technologies that signaling biomolecules [1,2]. Biomolecular signals that are mainly
found application in all fields of pharmaceutical and biomedical sciences. growth factors or other cytokines, chemoattractants, adhesion proteins
Particularly, with the advent of protein therapeutics, the need for con- and many others, must be locally delivered in their active form and
trolled delivery systems able to enhance protein's pharmacokinetic and with a sustained release profile. Among the existing technologies, hydro-
pharmacodynamic properties became more urgent. Nowadays, proteins gels – water-swollen, cross-linked polymer networks – have emerged as
are used in the treatment of many diseases but also in the area of tissue particularly promising materials for tissue engineering, as they can act
engineering, where supply of biomolecular cues that mimic the environ- both as scaffolding materials and/or releasing matrices for biologically
ment of natural tissues and promote the communication between cells active and cell modulating substances. Their water content, soft nature
and porous structure mimic biological tissues and make them suitable
⁎ Corresponding author. Tel.: + 39 0737 402215; fax: + 39 0737 402457. to accommodate cells and to encapsulate and release water-soluble com-
E-mail address: roberta.censi@unicam.it (R. Censi). pounds like proteins in a controlled fashion.

0168-3659/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
doi:10.1016/j.jconrel.2012.03.002
R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692 681

After highlighting the rationale for using of delivery technologies to molecules not only enhances tissue regeneration, but also prevents
improve the effectiveness of biomolecular cues, this mini-review briefly unwanted and potentially harmful side-effects at other locations than
describes the main hydrogel systems that have been studied recently as the target. Various strategies have been investigated to achieve con-
releasing matrices for biomolecules involved in healing processes. Strat- trolled protein delivery from hydrogels. They comprise ‘direct’ and ‘in-
egies for encapsulation, (transient) immobilization and controlled re- direct’ delivery approaches. Direct release occurs through physical
lease are also discussed. Finally, hydrogel-based releasing technologies encapsulation, non-covalent binding, covalent immobilization to the
with potential clinical impact in tissue engineering are outlined and delivery system via hydrolytically or enzymatically degradable linkers
an outlook on future directions is provided. and the use of double carriers, where protein loaded micro/nano-
spheres are embedded in hydrogels and the release of the active is
based on a combination of mechanisms (diffusion and or degradation).
2. Proteins in the healing process: an introduction to their
‘Indirect’ approaches rely on gene therapy and cell transplantation.
delivery approaches
Gene therapy is realized through the expression of genetic material
encoding for the desired protein that is delivered in the target tissue,
Cell fate and behavior are highly influenced by a number of factors
while in cell transplantation, specific proteins are secreted by cells
and interactions with the surrounding microenvironment. Signaling
that are encapsulated in a hydrogel [12,13].
molecules create an effective communication between cells and extra-
In the following section the ‘direct’ approaches for protein deliv-
cellular matrix (ECM) and orchestrate the complex cascade of events
ery in tissue engineering will be discussed.
that leads to successful regeneration of damaged tissues [3]. Of all mol-
ecules currently identified as key components of the wound healing
process, growth factors play a pivotal role in the information transfer 3. Protein release strategies from hydrogels
mechanism between cell and ECM. Although the natural dynamics of
the cellular microenvironment are difficult to emulate in an artificial Proteins can be loaded into hydrogels through a manifold of mecha-
setting, it is currently widely accepted that the self-healing capacity of nisms and strategies. They can be physically entrapped in hydrogels or
an organ or tissue can be augmented by the integration of growth fac- adsorbed to the matrix by specific interactions, non-covalent or covalent
tors. In fact, they accelerate the proliferation and differentiation of binding via degradable linkers and subsequently released via diffusion,
recruited or implanted cells and promote the re-growth of tissues and swelling, erosion, degradation or a specific trigger such as pH, tempera-
organs not capable of self-healing otherwise [4–6]. ture, etc. Also, a combination of multiple delivery systems can be used to
Growth factors are large polypeptides that modulate cellular prolif- achieve modular release of multiple proteins. Depending on the release
eration, differentiation, migration, adhesion and gene expression upon mechanism, different release profiles can be obtained. Table 2 overviews
binding to specific receptors on the surface of target cells. Examples of the technologies further described in the following sections.
growth factors and their applications in tissue engineering are listed
in Table 1. Unlike other proteins (i.e. hormones), growth factors act lo- 3.1. Physical entrapment of growth factors into hydrogels: general
cally and possess short diffusion distances through the ECMs, owing to principles and release mechanisms
their short half-lives. Typically, growth factors are enzymatically or
chemically degraded or deactivated in physiological conditions in a Physical encapsulation of growth factors in order to obtain a con-
very limited time frame. To mention, basic fibroblast growth factor trolled release of the active is a frequently applied technique for local
(bFGF) has a half-life of only 3 min after intravenous administration; growth factor delivery in tissue engineering. Its relatively simplicity
[7] similarly, less than 30 min are needed to halve the plasma concen- represents a clear advantage over more sophisticated encapsulation/
tration of vascular endothelial growth factor (VEGF) [8] and platelet- release methods and the majority of hydrogel systems relies on this
derived growth factor (PDGF), isolated from platelets, has a half life of loading process. Loading is done by incubation of the preformed gel
less than 2 min when injected intravenously [9]. with the protein of interest or by adding the protein to the hydrogel
Therefore, the exogenous administration of growth factors for tissue forming monomers/prepolymers. In recent years, attention is shifted
engineering faces important limitations associated to their rapid inacti- to in situ gelling systems which preferably are liquid before adminis-
vation and/or elimination after intravenous delivery, their poor trans- tration and gellify at the site of injection [42]. For these gels, loading
dermal adsorption due to the large size and the hydrolytic and with proteins can easily be established by dissolution of the biother-
proteolytic degradation upon oral administration. Hydrogels are fre- apeutic in the gel forming polymer solution before administration.
quently used to release growth factors in a controlled and effective Typically, the mechanisms that govern the drug release from these
manner and to direct the protein specifically to the wound site hydrogels are controlled by diffusion, swelling, erosion, external
[10,11]. Spatiotemporal control over the delivery of these key signaling stimuli or combinations thereof [43].

Table 1
Typical examples of growth factors and their applications in the field of tissue engineering [11,14].

Growth factor Abbreviation Action Tissue engineering applications

Vascular endothelial growth factor VEGF Migration, proliferation and survival of endothelial cells Blood and lymphatic vessels
Insulin-like growth factor IGF-1 Proliferation, apoptosis inhibition Cartilage, skin, nerve, kidney,
bone, muscle
Hepatocyte growth factor HGF Proliferation, migration, differentiation of mesenchymal stem cells Liver, muscle, bone
Epithelial growth factor EGF Proliferation and differentiation of fibroblast, epithelial, Skin, nerve
mesenchymal and glial cells
Nerve growth factor NGF Proliferation and survival of neural cells Nerve, brain, spine
Bone morphogenetic protein BMP-2/3/7 Differentiation and migration of osteoblasts, renal development Bone, cartilage, kidney
Platelet derived growth factor (2 polypeptide PDGF-AA/BB/AB Embryonic development, proliferation and migration of Bone, skin, muscle, blood vessels
chains A and B forming homodimers endhotelial and smooth muscle cells
AA and BB and heterodimer AB)
Transforming growth factor TGF-α/β Proliferation and differentiation of basal, neural, bone-forming Brain, skin, cartilage, bone
cells, keratinocytes, anti-proliferation epithelial cells
Angiopoietin fibroblast growth factor Ang-1/Ang-2/bFGF Blood vessels maturation Hearth, muscle, blood vessels,
Proliferation endothelial cells bone, skin, nerve
682 R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692

Table 2
Overview of the main release mechanisms and strategies to load and modulate the kinetics of proteins released from hydrogels.

Release mechanism from hydrogels Characteristics Some examples

✓ Fickian diffusion PEG/p(HPMAmlac) [15–17]


✓ Mt/M∞ = k √t PLA-PEG-PLA [18]
✓ Concentration gradient RADA 16 networks [19]
✓ Generally limited to relatively short time spans alginate [20,21]
✓ Protein size b mesh size dex-VS/PEG-4-SH [22]
✓ Geometry determines release rate
✓ Swelling degree controls release rate dextran/P(D,L)LA [23]
✓ Water uptake allows protein to diffuse out oppositely charged dextran microspheres [24]
✓ Swelling can be mediated by degradation

✓ Controlled by physical or chemical erosion PLGA-PEG-PLGA [25,26] PEG/chol-PEG/β-CD [27].


✓ Surface erosion mostly for physical gels PEG-P(D,L)LA [28]
✓ Constant release rate
✓ Hydrolytical or enzymatic degradation
✓ Protein size > mesh size

✓ Stimuli lead to gel changes and drug release nitrilotriacetic acid/GyrB [29]
✓ Stimuli: temperature, pH, biomolecules, drugs, magnetic field, etc… PEG-VS-MMP [30]
PEG acrylate-MMP [31]

✓ Adsorption of proteins through weak interaction (i.e. electrostatic, hydro- Fibrin-heparin [32,33]
phobic, H-bonding) PEG-IDAA/Ni+ 2Cu+ 2 [34]
✓ Affinity with binding sites: heparin, antibodies, chelating ions, peptides, thiol acrylate PEG [35]
molecular imprinting

✓ Attachment of drugs to matrix via covalent bonds polyacrylamide [36,37]


✓ Cleavable linker PEG-RGD-MMP [38]
✓ Hydrolytical or enzymatic cleavage

✓ Combination of multiple carriers: i.e. protein-loaded micro/nanoparticles PEG-GMA/gelatin [39]


embedded in hydrogels Fibrin-PLGA [40]
✓ Release of multiple proteins through combination of mechanisms and rates HAMC/PLGA [41]

The release mechanism depends on both the characteristics of the on Fickian diffusion, polymer disentanglement and dissolution in water
polymeric network and the protein. When the hydrogel pores are big- [50].
ger than the hydrodynamic radius of the protein, diffusion is the driv- Erosion-controlled systems have increased in number since the
ing force for release, with a diffusion rate depending on the protein development of synthetic biodegradable polymers. In these systems,
size and the water-content of the gel (‘free-volume’) [44]. On the the mobility of the drug in the homogeneous non-degraded polymer
other hand, when the hydrogels pores are smaller than the protein di- matrix is limited and drug release is then governed by the degrada-
ameter, swelling or erosion/degradation (bulk or surface) is needed tion rate of the polymer, porosity increase, and drug diffusion mainly
for release. Generally speaking, the majority of the gel matrices in the surface of the polymer matrix.
reported to date exhibit diffusion controlled release, following Higu- The possibility to tailor the release kinetics of proteins from hydrogels
chi's kinetics, implying that the release is proportional to the square can be achieved through the use of excipients or by changing the cross-
root of time [45]. This release profile was demonstrated particularly linking density of the polymer network. In this particular instance, the
beneficial for the delivery of several growth factors for tissue engi- use of synthetic polymers is extremely advantageous because they offer
neering applications. For example, Brown et al. and Boerckel et al. the opportunity to fine tune their chemical structure to achieve modular
demonstrated that bone regeneration was enhanced when rhBMP-2 release. However, also natural polymer networks can be tailored to some
was released down a concentration gradient [46,47]. The diffusional extent by changing polymer concentration and crosslink density. Also ex-
spatiotemporal growth factor presentation proved effective not only tensive work on the development of hybrid networks in which natural
for bone regeneration, but also for other engineered tissues like polymers are synthetically modified or combined with synthetic poly-
blood vessels [20,21]. mers, has been proposed to combine beneficial properties of both kinds
The Ritger–Peppas equation is often used to fit release data and de- of polymers with respect to mechanical properties, tailorability in terms
termine the underlying release mechanism: M t/M∞ = ktn with M t/M∞ of biodegradability as well as biocompatibility and cytocompatibility.
the fractional drug release at time, t and k a constant incorporating The geometry of the hydrogel-based depot also affects the release
structural and geometric characteristics of the device. If n = 0.5, the re- rate and the duration. Generally, the bigger is the device, the longer the
lease is governed by Fickian diffusion. If n = 1, molecules are released by diffusion distances become and the longer the release consequently
surface erosion, while both mechanisms play a role if n has a value be- lasts. In recent years, patterning techniques utilized in tissue engineering
tween 0.5 and 1 [48,49]. (i.e. stereolithography, bioprinting) have been used to adjust surface area
Swelling-controlled systems depend on water uptake and changes in and consequently modulate release profiles of the biomolecules [51–53].
drug diffusivity within the matrix. Swelling increases polymer flexibility Deviation from the described release behaviors is observed when a
and makes pores bigger, resulting in higher drug mobility with some- specific stimulus – typically temperature, pH, presence of certain mole-
times n values that exceed 1. As a consequence, drug release depends cules – leads to a physical or chemical change in the network structure,
R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692 683

for instance, hydrogels swell or shrink in response to a certain trigger been used to encapsulate and deliver several proteins for intramyocar-
thereby modulating the release of encapsulated drugs/proteins. dial delivery. This peptide-based hydrogel has also been used to deliver
platelet-derived growth factor BB (PDGF-BB) [66], stromal cell-derived
3.1.1. Synthetic polymers for the physical encapsulation of growth factors factor-1 (SDF-1) [67] and insulin-like growth factor I (IGF-I) [68] to de-
Among the various synthetic polymers studied for the delivery of crease myocardial infarct. The observed slow and controlled release of
growth factors in tissue repair, PEG-based networks play a prominent the active proteins has been ascribed to diffusion and to some extent
role. to interaction between protein and polymer. The amphiphilic nature
Burdick et al. investigated the feasibility of a PEG-based hydrogel of the self-assembling polypeptide leads to interactions with the loaded
platform for the replacement and delivery of neurotrophins to the cen- protein, resulting in reduced diffusivity and slower release kinetics.
tral nervous system. For this purpose, triblock copolymers based on Hydrogels that showed degradation mediated release of model
PEG and acrylated poly(lactic acid) (PLA-b-PEG-b-PLA) were synthesized proteins and that potentially can be used to physically encapsulate
and hydrogels formed by photopolymerization. Three neurotrophins, cil- and release growth factors with a zero order release kinetics are for
iary neurotrophic factor (CNTF), brain-derived neurotrophic factor example those formed by stereocomplexation between star PEG copo-
(BDNF) and neurotrophin-3 (NT-3) were encapsulated and released in lymerized with the stereoforms D or L of PLA [28] or those based on
vitro mainly by diffusion in 20 to 80 days depending on polymer concen- PLGA-PEG-PLGA (ReGel) systems [25,26] and inclusion complexes of
tration (10 to 30%) and neurotrophin size. In vitro, the released proteins PEG/cholesterol-PEG/β-cyclodextrin [27].
stimulated the proliferation of a TF-1 cells transfected with the α-subunit Stimuli sensing hydrogels have also been investigated for growth fac-
of the CNTF receptor and stimulated outgrowth of a greater number of tor release in tissue engineering. Recently, Weber et al. proposed a drug-
neuritis from retinal explants than in control culture conditions [18]. sensing hydrogel based on gyrase sub-unit B (GyrB), reversibly cross-
These types of hydrogels, initially described by Hubbell and coworkers linked by coumermycin and able to release VEGF upon addition of an ami-
[54], have been extensively investigated for growth factor delivery nocoumarin antibiotic, novobiocin. The polymer forming the hydrogel is
[55,56] because they offer several benefits, including the possibility to based on polyacrylamide functionalized with nitrilotriacetic acid chelat-
easily modify the network properties by changing the macromer chem- ing a Ni2+ ion to which GyrB can bind through a hexahistidine sequence.
istry and solution concentration. For instance, the degradation and swell- The addition of novobicin causes the cross-links to be partly broken,
ing of the hydrogels are dictated by the number of lactic acid repeat units, resulting in opening of the network structure and release of VEGF [29].
the acrylation efficiency, the molecular weight of the PEG core and the Hubbell et al. synthesized hydrogels comprising multiarm vinyl
concentration of macromer in the prepolymer solution [57]. Also, the sulfone-terminated PEG, a monocysteine containing adhesion pro-
type of degradable unit (e.g., lactic vs caproic acid) alters network degra- tein, and a bis(cysteine) metalloprotein substrate protein (MMP).
dation rates [58]. These hydrogels were studied for tissue engineering purposes and
Similar release and network concepts apply to other PEG/polyesters both cells and vascular endothelial growth factor (VEGF105) were en-
hydrogels, like those based on PEG-poly(lactic-co-glycolic acid) (PLGA)- capsulated in the hydrogel. The release of the encapsulated factor is
PEG triblock copolymers [59]. These materials display lower critical solu- triggered by matrix metalloproteinases (MMPs) [30]. MMPs are are
tion temperature behavior and were processable avoiding the use of high zinc-dependent endopeptidases capable of degrading extracellular
temperatures to dissolve the polymer. It was shown that upon subcuta- matrix proteins and they influence cell behavior such as cell prolifer-
neous injection (rat model) the hydrogels were stable for one month ation, migration (adhesion/dispersion), differentiation, angiogenesis,
[60]. TGF-β1 was loaded into these hydrogels and used as a slow releas- apoptosis, and host defense. MMPs sensing hydrogels were devel-
ing drug reservoir aimed for wound healing purposes. Significant levels oped with the aim to mimic the natural processes involved in tissue
of re-epithelialization, cell proliferation and collagen organization were repair. Other examples of semi-synthetic hydrogels were described
observed [61]. Porcine growth hormone (pGH) and Zn-pGH were sus- by Phelps et al. whose design provides incorporated VEGF, enzyme-
tainably released from PLGA-PEG-PLGA hydrogels for 10–14 days in degradable sites and arginine-glycine-aspartic acid (RGD) cell adhe-
vitro with no initial burst and a good correlation with in vivo data, that sive ligands. Similarly to the system proposed by Hubbell, these ma-
confirmed a weight gain in hypophysectomized rats equivalent to con- trices use PEG as the main building block because it has been shown
ventional daily injections of 5 mg pGH for 14 consecutive days. Approxi- to act as a protein-resistant and cell non-adherent background. The
mately 85% of the glycosylated granulocyte colony stimulating factor network is based on PEG diacrylate which is photocrosslinked in the
loaded in PLGA-PEG-PLGA hydrogels was released over a 12-day period presence of MMP-degradable polypeptide functionalized with two
and similarly to pGH, showed similar efficacy in rats as compared to PEG-acrylate groups, a mono-PEG-acrylate RGD and a mono-PEG-
daily i.v. injections [62]. Censi et al. studied the protein release from acrylate VEGF. Recently, Phelps et al. have demonstrated the benefits
photopolymerized thermosensitive PEG-based networks formed upon of this approach in a mouse hind limb femoral artery ligation model.
gelation of methacrylated poly(hydroxypropyl methacrylamide lac- After 7 days, mice treated with implanted hydrogels showed a 50% in-
tate)-PEG- poly(hydroxypropyl methacrylamide lactate) (p(HPMAm- crease in perfusion to the limbs and 100% increase in perfusion to the
lac)-PEG-p(HPMAm-lac)) triblock copolymers [63]. Proteins are released feet as compared to untreated mice. The matrix alone (not containing
from this network by Fickian diffusion and, similarly to previously de- VEGF) performed as well as soluble VEGF injections, indicating that
scribed systems, network properties and release behavior can be tailored the engineered degradable and adhesive hydrogel has itself a benefi-
by tuning polymer chemistry and concentration [16,17,64]. The gels cial healing or supportive effect. Engineered matrix containing VEGF
showed a good biocompatibility in rats [65] and their potential for carti- performed better than injections of soluble VEGF, acting synergisti-
lage tissue engineering application has been demonstrated [51]. cally as a directive scaffold and a growth factor delivery vehicle [31].
Peptide based hydrogels were also used for protein delivery and re-
lease. A class of ionic self-complementary oligopeptides named RAD 16 3.1.2. Natural polymers for the physical encapsulation of growth factors
and commercialized as PuraMatrix was introduced by Zhang et al. These Hydrogels designed with natural polymers as building blocks display
peptides consist of alternating hydrophilic and hydrophobic amino multiple advantages over synthetic polymer networks with respect to
acids that form β-sheet structures having one polar surface with com- their biocompatibility, biodegradability and good cell adhesion proper-
plementary charged ionic side chains and a non-polar surface with ala- ties. Therefore, extensive work is available on biopolymer-based hydro-
nines. These peptides are able to spontaneously self-assemble into gels for cell and growth factor encapsulation in regenerative medicine
stable macroscopic matrices or nanofibers. The monovalent cations [69–72].
that are needed for the peptide assembly are sequestered from the The main classes of natural polymers studied in hydrogel formula-
physiological environment [19]. The described family of peptides has tions are polysaccharides and proteins/polypeptides.
684 R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692

its C-5 epimer α-L-guluronic acid (G), respectively, covalently linked


in different sequences or blocks. This polymer block consists of consec-
utive G-residues, consecutive M-residues or alternating M and G resi-
dues. Depending on the block composition, alginate has different
conformational preferences and behavior. G-rich blocks of the polymer
are able to bind divalent cations of which Ca2+ is frequently used for
crosslinking alginate to obtain hydrogels. Ca2+ ions bind to the G units
of different polymers chains cooperatively in a so-called egg-box ar-
rangement. The partial oxidation of alginate using sodium periodate
makes the polysaccharide biodegradable [75,76], and these oxidized al-
ginates are therefore particularly appealing for biomedical applications.
An alginate hydrogel patch to deliver stromal cell-derived factor-1
(SDF-1), a naturally occurring chemokine that is rapidly overexpressed
in response to tissue injury, was described by Rabbany et al. Alginate
patches were loaded with either purified recombinant human SDF-1
protein or plasmid expressing SDF-1 and the kinetics of SDF-1 release
were measured both in vitro and in vivo in mice. They demonstrated
Fig. 1. Most commonly used polysaccharides for hydrogel preparation for biomedical that SDF-1 plasmid- and protein-loaded patches were able to release
applications (M = mannuronic acid, G = guluronic acid). therapeutic product over hours to days, with faster release for SDF-1
protein (in vivo K(d) 0.55 days) than SDF-1 plasmid (in vivo K(d)
3.1.2.1. Polysaccharides. Polysaccharides are in general hydrophilic 3.67 days). Prolonged (induction of) SDF-1 release of SDF-1 resulted
polymers and are therefore very suitable for the design of hydrogels. in accelerated healing (9 days) and reduced scarring of acute surgical
The most commonly used polysaccharides in recent hydrogel research wounds in Yorkshire pigs [77].
aimed for protein delivery are alginate, hyaluronic acid, chitosan and Although the biological properties of alginate are well recognized,
dextran (Fig. 1) [73,74]. Representatives of the hydrogel technologies some limitations in tailoring the mechanical properties, the pore size
based on these polysaccharides are briefly discussed below with respect and the release kinetics of proteins from these gels still remain. Many re-
to their use for growth factor delivery. searchers investigated several modified alginates to overcome these lim-
3.1.2.1.1. Alginate. Alginate is a linear polysaccharide composed of itations [78–83]. For example, Ruvinov et al. examined the release of
homopolymeric blocks of 1-4′-linked β-D-mannuronic acid (M) and hepatocyte growth factor (HGF) from modified alginate hydrogels and

Fig. 2. Biophysical properties and vascular endothelial growth factor (VEGF) release from alginate gels. Degradation of gels formed from high molecular weight non-oxidized algi-
nate (triangles), binary molecular weight non-oxidized alginate (white squares) and binary molecular weight partially oxidized alginate (black squares) in phosphate-buffered sa-
line (A). The viscosities of solutions of high (white bar), low (black bar) and binary (grey bar) molecular weight partially oxidized alginate were assessed (B). Release kinetics of
VEGF165 from gels formed from binary molecular weight alginate, either partially oxidized (black squares) or non-oxidized (white squares) (C). Greater endothelial cell prolifer-
ation was observed in cells cultured with VEGF released from binary molecular weight partially oxidized alginate gels (grey), as compared to medium without VEGF (black) and
control VEGF added to medium (white), as determined from the cell number counts from day 0 to day 4 (D). Reproduced from ref. [85].
R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692 685

studied its potential to induce angiogenesis in vivo. They developed a neovascularization. The delivery system, modified with adhesive se-
cross-linkable affinity-binding alginate by introducing sulfoester groups quences and loaded with growth factor and cells, was used to induce
into the uronic acids of alginate. It was shown that alginate-sulfate was the formation of a depot of vascular progenitor cells (outgrowth endo-
capable to bind heparin-binding proteins, like growth factors, with equi- thelial cells (OECs)) in vivo. It was observed that OECs were viable dur-
librium binding constants similar to that of heparin. The affinity binding ing the studied time frame and that the encapsulated VEGF induced the
to alginate-sulfate retarded the release of HGFThe bioconjugate HGF- cells to migrate out of the scaffold. Local and controlled delivery of the
alginate-sulfate was combined with alginate in an aqueous solution morphogen was effective in stimulating the cells to repopulate the dam-
and a hydrogel was formed ex vivo by partial cross-linking in presence aged tissue and participate in regeneration of a vascular network, while
of CaCl2. Upon injection into ischemic myocardial tissue the injectable bolus injections was ineffective in preventing necrotic toe (Fig. 3) [89].
hydrogel completed its cross-linking by sequestration of endogeneous 3.1.2.1.2. Dextran. Dextran consists of α-1,6-linked D-
Ca2+. A cumulative protein release of 75% was observed after 6 h and glucopyranoses with some degree of 1,3-branching. Various methods
in the following 5 days a cumulative release of approximately 85% was to cross-link dextran hydrogels have been developed and its high num-
achieved [84]. This profile is typical of diffusion-based delivery systems ber of available hydroxy groups presents many options for derivatization
that release the drug down a concentration gradient, as described for an- and subsequent physical or chemical crosslinking [90]. For example,
other alginate hydrogel reported in Fig. 2 [85]. A recent method Hennink et al. developed a hydrogel system potentially suitable for the
employed to make alginate networks more stable and tailorable is delivery of proteins relevant for tissue engineering, where a dextran
through the preparation of interpenetrating polymer networks (IPN's) backbone is derivatized with hydroxyethyl methacrylate (HEMA) moie-
[83]. An IPN is defined as a network comprising two or more polymers ties. This polymer in aqueous medium was chemically cross-linked by
that are partially interlaced on a molecular scale but not chemically radical polymerization of the dextran-bound methacrylate groups to
bounded to each other and that can be separated only after chemical yield hydrogel based microspheres [91,92]. These microspheres released
bond breakage [86]. IPNs are designed to combine the advantageous model proteins as well as therapeutic relevant proteins (IL2 [93]; hgH
properties of different polymers. This combination often translates in [94]) in a controlled manner for days to weeks. The same group also de-
good modulation of the mechanical and biological characteristics of the veloped a physically crosslinked hydrogel system by grafting the dextran
final hydrogel, derived from the additive or synergistic effect of the poly- backbone with either D- or L-oligo-lactate chains containing 5 to 15 lactic
mers' properties. Pescosolido et al. developed IPN hydrogels based on acid units. The hydrogel formation was driven by stereocomplexation
Ca2+-alginate and photocross-linked dextran-methacrylate derivatives, and preclinical studies demonstrated the suitability of this hydrogel for
of which the mechanical properties were notably improved by the syner- the controlled delivery of IL-2. The density of oligo-lactates and the
gistic effect of the polymers composing the IPN system. These hydrogels grafting density allowed tailoring of the release and degradation rate.
proved effective in the controlled release of proteins [83]. In another The release of the encapsulated protein was dependent on diffusion
study, it was demonstrated that complete delivery and a better control [23]. Further efforts were addressed by Hennink et al. towards the devel-
over VEGF release kinetics were achieved by cross-linking alginate mi- opment of dextran based delivery platforms for proteins, potentially suit-
croparticles with Zn2+ instead of Ca2+ [87,88]. able for growth factor and cytokine controlled release. Self-assembled
Silva et al. described alginate hydrogels that were covalently modi- macroscopic hydrogels based on oppositely charged dextran micro-
fied with RGD sequences and loaded with growth factor VEGF, and spheres were prepared and their release behavior investigated. It was
that were lyophilized to form microporous scaffolds to induce found that native proteins were released by diffusion/swelling [24].

Fig. 3. Analysis of angiogenesis in ischemic hindlimbs after OEC transplantation. (A) Implantation of blank scaffolds, bolus injection of OECs and VEGF (same quantities as placed in
scaffolds), transplantation of OECs on scaffolds lacking VEGF (alginate scaffold OEC), and transplantation of OECs on scaffolds presenting VEGF121 [alginate scaffold (VEGF) OEC].
Photomicrographs of tissue sections from ischemic hindlimbs of SCID mice at postoperative day 15, immunostained for the mouse endothelial cell marker CD-31 (B), and
human CD-31 (C). Reproduced from ref. [89].
686 R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692

Fig. 4. (A) Reaction of dextran with p-maleimidophenyl isocynate (PMPI) and (B) preparation of dextran-peptide hydrogel through the Michael-type reaction of peptide cross-
linker to Dex-PMPI. Reproduced from ref [95].

More recently, dextran-peptide bioconjugates were synthesized by loaded with multiple growth factors affects the neovascularization
Shoichet et al. In their approach, the polysaccharide was modified of ischemic and wounded tissues. Dextran-allyl isocyanate-
with p-maleimidophenyl isocyanate (PMPI) thereby introducing malei- ethylamine (Dex-AE) derivatives with different degrees of substitu-
mide functionalities in the backbone (Dex-PMPI). A peptide cross- tion were synthesized and mixed with PEG diacrylate at different ra-
linker, derived from collagen and susceptible to gelatinase A digestion, tios and subsequently, hydrogel formation was induced by
was synthesized with bifunctional cysteine termini and used to cross- photopolymerization. It was found that tissue ingrowth was favored
link Dex-PMPI (Fig. 4). It was demonstrated that this type of hydrogel by reducing the degree of substitution of cross-linking groups, which
was cytocompatible and mimicked the degradation and remodeling of resulted in reduced rigidity, increased swelling, increased VEGF re-
the ECM through the activity of cell-secreted enzymes [95]. lease rate, and rapid hydrogel disintegration. Furthermore, the re-
Michael addition cross-linking reaction was used to induce network lease of multiple angiogenic GFs increased the size and number of
formation between dextran vinyl sulfone conjugates (dex-VS) and tet- newly formed functional vessels [96]. In another study, hybrid
rafunctional mercapto poly(ethylene glycol) (PEG-4-SH). The release hydrogels based on combinations of glycidyl methacrylated dextran
of several proteins including bFGF from hydrogels of different polymer and gelatin, processed into different physical structures (micro-
concentrations was studied. Also these networks showed diffusional spheres, cylinders) have been synthesized and used to deliver
release and a certain extent of tailorability of the bFGF [22]. growth factors, including BMP-2and IGF-1 [97].
In a recent study, Sun and coworkers demonstrated that the ma- 3.1.2.1.3. Hyaluronan. Hyaluronan or hyaluronic Acid (HA) is a
nipulation of the cross-link density of dextran based hydrogels linear glycosaminoglycan composed of repeating disaccharide units
R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692 687

of D-glucuronic acid and N-acetylglucosamine [98,99]. This naturally and high pH's. Chitosan has mucoadhesive properties originating from
occurring polysaccharide is negatively charged under physiological its cationic nature [92]. To make this polymer suitable for hydrogel for-
conditions has molecular weights up to 10 7 Da. It is found mainly in mation, water-soluble and cross-linkable derivatives were synthesized.
the extracellular matrix (ECM) and in the synovial fluids of joints One of these approaches was realized by grafting 4-azidobenzoic acid to
where it acts as a lubricant by which it reduces the friction of bones the available free amine groups of lactose-modified chitosan. In the
due to its unique viscoelastic properties [100]. presence of UV light the azido groups are converted into nitrenes that
Because of its biocompatible, attractive physical properties and are highly reactive species with the free amine groups on chitosan
possibilities for further chemical modifications, HA is an extensively yielding networks [115]. VEGF and FGF-2 were loaded into photo
studied polysaccharide for the development of delivery systems for cross-linkable modified chitosan hydrogels and their release and effica-
tissue engineering applications [101–107]. However, unmodified cy in inducing neovascularization in ischemia models were studied
hyaluronic acid in aqueous solution displays poor mechanical proper- [116–118]. The interactions between the nitrene groups and the
ties. Therefore, to be suitable for protein delivery, HA-gels need phys- amine groups of the proteins were identified as the reason for the in-
ical and/or chemical stabilization. Usually, the carboxylic groups of complete release from these types of hydrogel [119,120]. Yeo et al.
hyaluronic acid are derivatized to introduce functionalities that reported that 80% of initially loaded VEGF was retained in the network
make the polymer suitable for cross-linking [108–113]. after cross-linking [118]. In situ gelling double networks of oxidized
In situ photopolymerization was applied by Park et al. to crosslink dextrans and thiolated chitosan (semi-interpenetrating networks)
thermosensitive hyaluronic acid/Pluronic composite hydrogels that re- were recently proposed for healing purposes by Chen et al. These net-
leased human growth hormone with kinetics correlated with the mass works showed faster gelation kinetics as compared to auto-gelling thio-
erosion [105]. Subsequently, Patterson et al. synthesized glycidyl methac- lated chitosan hydrogels [121]. Recently, comprehensive and complete
rylate modified hyaluronan hydrogels with different crosslink densities reviews on chitosan hydrogels were published concerning their drug/
and thus of different degradation rates and used them to prepare photo- protein delivery aspects [122–125].
polymerized hydrogels as BMP-2 and/or VEGF releasing matrices for bone
regeneration. They demonstrated that all matrices displayed diffusion- 3.1.2.2. Proteins. Collagen, fibrin and gelatin are naturally occurring, en-
like release behavior of entrapped proteins and their degradation and re- zymatically degradable proteins that have been extensively exploited as
lease rates modulated the formation of mature bone, specifically affecting main component in matrices for drug delivery and tissue engineering
the organization of the collagen matrix. Additionally, the co-delivery of an purposes. Many of the hydrogel systems based on gelatin and collagen
angiogenic molecule (VEGF) in conjunction with an osteoinductive mol- are cross-linked using harsh conditions that exploit gluteraldehyde or
ecule (BMP-2) increased the extent of formed mineralized tissue [102]. water-soluble carbodiimides and that can be detrimental for the encap-
Crosslinked, PEG diacrylate/thiolated hyaluronan hydrogels were sulated proteins [126]. However, also milder preparation conditions
studied for the delivery of multiple growth factors (VEGF and/or were investigated. For instance, non-covalently crosslinked fibrillar col-
Ang-1) in both presence or absence of heparin, a molecule that forms lagen can be used to create hydrogels by entanglements of collagen fi-
complexes, stabilizes the structure and modulates the release of bers [127]. The mesh sizes of these entangled collagen fibers are quite
growth factors, according to the dissociation constant of the complex large and therefore diffusion of only very large proteins can be con-
growth factor/heparin. Greater neovascularization was observed trolled. Nevertheless, the protein release rate may still be lower than
when the hydrogels were loaded with both growth factors [106,113]. expected based on diffusion coefficients due to weak interactions be-
Similar studies were conducted by the same group both in vitro and tween collagen and loaded proteins [127].
in vivo using in situ gelled thiolated gelatin/thiolated hyaluronic acid/ Gelatin has a long history in clinical use with examples of gelatin
PEG diacrylate based networks with or without disulfide bridged hep- based systems with a variety of physical geometries. Gelatin derived
arin as part of the network and releasing combinations of growth fac- from collagen can be positively or negatively charged depending whether
tors like VEGF, Ang-1, PDGF and KGF. It was found that release collagen depolymerization is carried out at acidic or basic conditions. The
followed first order kinetics, with gelatin slightly increasing the charge of gelatin network can be utilized to complex oppositely charged
growth factor release rate, and heparin, slowing down the kinetics, be- proteins serving as additional feature to control release rate [128]. Gelatin
cause of its complexation with the growth factors. The extent of revas- based hydrogels have been used for the delivery of FGF-2 [129], HGF and
cularization and blood vessel maturation was found greater in gels VEGF [130]. Nowadays, gelatins can be produced in yeast cells recombi-
containing both heparin and gelatin than in those containing only gel- nantly leading to slightly different polymer properties. The advantage is
atin or only heparin [104,114]. that properties such as molecular weight, amino acid sequences and iso-
Other cross-linked hyaluronic acid networks developed for tissue en- electric points can be tailored precisely. Degradation times, swelling
gineering purposes are for example those studied by Varghese et al. based properties and ultimately also drug release kinetics are affected by the de-
on aldehyde-HA/hydrazide-HA for the burst-free release of rhBMP-2 sign of recombinant gelatin. Only a few recombinant gelatin hydrogels
[107] or those developed by Leach et al. composed of photocross-linked have been investigated for their protein delivery properties [131,132]. Fi-
glycidyl HA or photocross-linked glycidyl HA/acrylate 4-arms PEG [108]. brin is a fibrous, non-globular protein derived from fibrinogen, a protein
Other HA cross-linkable hydrogels for protein delivery that have been of the blood coagulation cascade. Fibrinogen, a water soluble protein, is
proposed recently are based on tyramine-HA [109], on disulfide bridged converted into fibrin that polymerizes to yield a gel due to the action of
thiolated-HA [111], divinyl sulfone-HA/divinyl sulfone PEG [110] and adi- thrombin. Fibrin networks are gradually resorbed in vivo by the secretion
pic acid dihydrazide-HA/methacrylate-HA [112]. of fibrinolytic enzymes and as such has been widely used for formulating
Since HA is negatively charged at physiological pH, the protein re- depot systems for proangiogenic growth factor delivery. Commercial fi-
lease rate will be affected by the charge of the protein. Generally brin glues are also used in the clinic for in situ application.
speaking, cationic proteins are slower released than anionic or neu- In an early clinical trial, VEGF was injected in an in situ forming fi-
tral proteins. The release of proteins is not only influenced by charge brin matrix into the right popliteal region of a man suffering from
interactions with the polymer matrix, but also by the enzymatic deg- claudication. Although substantial growth of newly formed vessels
radation of HA-based gels in the presence of hyaluronidase, which is was observed, a high initial burst occurred within the first 24 h. To
present in biological tissues [109,112]. overcome this issue, VEGF was later covalently bound to the matrix
3.1.2.1.4. Chitosan. Chitosan is a copolymer of glucosamine and N- and released by enzymatic cleavage [133,134]. Similarly, FGF-2 was
acetylglucosamine and is derived from the natural polymer chitin electrostatically immobilized in fibrin hydrogels using heparin
(=poly N-acetylglucosamine) by (partial) deacetylation. The polymer [135]. These strategies showed in animal model enhanced neovascu-
is positively charged at low pH's and uncharged and insoluble at neutral larization and reduced fibrosis and inflammation.
688 R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692

attached to fibrin on one site and to heparin on the other extremity.


Growth factors of different affinity for heparin (bFGF >bNGF, BDNF,
neutrophin-3) [32,33] were loaded and released with slower kinetics
as compared to fibrin hydrogels. In a similar approach adopted by Kiick
et al. four arms thiolated PEGs were reacted with monomaleimide-
functionalized low molecular weight heparin and cross-linked in aque-
ous medium after addition of heparin-binding peptide modified PEGs.
This delivery system as well other similar hydrogels have been exten-
sively used for the controlled delivery of many growth factors for differ-
ent applications [145–148].
Heparinization was applied also to collagen [149,150], alginate
[151], hyaluronan [152] and pluronic [153] using different chemis-
tries and cross-link methods.
Polymers were complexed with growth factors binding heparin also
by electrostatic interactions. Sulfonation of hyaluronan and uronic acid
monomers of alginate allowed non-covalent interaction with heparin-
Fig. 5. Gelation mechanism and protein encapsulation methods for PEGylated fibrin
binding proteins [80,84]. Although heparin bound hydrogels are defi-
gels. Reproduced from ref. [136].
nitely the most studied affinity systems to date, some challenges still re-
main such as unstable protein-heparin bonding in vivo, large amounts
Recently, Drinnan et al. reported on an elegant approach on the of heparin necessary to achieve sufficient drug loading and heparin-
multimodal release of different proteins from the same hydrogel. induced thrombocytopenia [154]. Affinity hydrogel systems based on
They developed an injectable PEGylated fibrin gel designed for the re- the mechanism of metal-ion-chelation and binding of histidine tagged
lease of PDGF-BB and TGF-β1 with distinct kinetics (Fig. 5). Growth proteins have been proposed as an effective alternative to the
factors were loaded into PEGylated fibrin gels via 3 mechanisms: en- heparin-containing systems. To mention, Metters et al. developed
trapment, conjugation through a homobifunctional amine reactive PEG-co-methacrylated iminoadiacetic acid displaying affinity binding
PEG linker, and physical adsorption on the fibrin matrix. PDGF-BB for histidine tagged proteins through divalent metal ions such as nickel
was entrapped during thrombin-mediated crosslinking leading to its and copper. The authors also elaborated a mathematical model to pre-
diffusion-controlled release over 2 days. TGF-β1 was both conjugated dict the release rate of proteins from the hydrogels [34].
through the PEG linker and bound to the matrix via physical adsorp- Phage display is a technique that has been exploited to identify pep-
tion, delaying the release rate of TGF-β1 up to 10 days. Further, the tides with affinities for growth factors that could be coupled to the gel-
release rate was highly correlated to gel degradation rate, indicating ling polymer [155].
that TGF-β1 release was degradation-controlled [136]. As an example Anseth et al. developed an affinity peptide-
Fibrin hydrogels were also used to fabricate circuits for the con- functionalized PEG hydrogel with the ability to sequester monocyte che-
trolled release of chondroitinase ABC (a bacterial enzyme capable of motactic protein 1 (MCP-1), a chemokine that induces the chemotaxis of
digesting the glycosaminoglycan (GAG) side chains from chondroitin monocytes, dendritic cells, and memory T-cells. Affinity peptides were
sulphate proteoglycans) in spinal cord lesion treatment. Sustained de- immobilized in PEG hydrogels via a thiol-acrylate photopolymerization.
livery of the protein in vivo for about 3 weeks was demonstrated as The release of encapsulated recombinant MCP-1 from PEG hydrogels
compared to bolus injection. Furthermore, after 3 weeks, six times was tailored by altering the spacer distance between the affinity peptide
more bioactive chondroitinase ABC and 37% lower levels of inhibitory and the crosslinking site [35]. Antibodies were also exploited for the spe-
glycosaminoglycans (GAG) were found in the spinal cord when hydro- cific binding of proteins onto scaffolds [156].
gels were used vs intraspinal injection of enzyme solution [137]. Molecular imprinting is a technique used to synthesize biomimetic
3.1.2.2.1. Affinity-based drug release. Affinity based drug delivery sys- polymer networks with template-shaped cavities that have affinity for
tems utilize physical interactions between the therapeutic drug and the specific molecules of interest (the templates). The nature of the interac-
delivery system to manipulate drug loading and control release kinetics. tion can vary from non-covalent bonds to metal coordination and even
The easiest method to modulate release kinetics of loaded proteins is covalent bonds. This technique has been applied for drug and protein
physical adsorption, involving the formation of ionic complexes by elec- delivery and recognition. However, although some success has been
trostatic interactions between charged polymers oppositely charged pro- achieved in the molecular imprinting of small molecule drugs, huge
teins [138–140]. Many charged polymers like hyaluronan, chitosan, challenges still exist with imprinting of proteins [42,77,125,157].
alginate, acidic gelatin can be potentially used to retard drug release [89].
Heparin sulfate is a naturally occurring, highly sulfated anionic
glycosaminoglycan found in the ECM that is a natural matrix responsi- 3.2. Covalent binding
ble for immobilizing and releasing various proteins that influence the
natural processes of adhesion, migration, proliferation, differentiation. Proteins, mostly exploiting their reactive amine and thiol groups,
This glycosaminoglycan possesses a specific binding domain for many can be covalently bound to polymer matrices via functional groups
growth factors that interact with heparin via non-covalent bonding like hydroxy, amine, carboxyl groups that, if not naturally present in
[32,141]. Besides enhancing matrix ability to retain the encapsulated the structure of the polymer, have to be introduced by functionalization
protein, it has been shown that affinity binding with heparin also stabi- reactions, blending or co-polymerization. The release of the protein is
lizes the drug, preserving its structure and functionality during handling mediated either via hydrolysis or reduction reactions or (cell-mediated)
of the material [142]. Heparin is generally incorporated within the de- enzymatic cleavage. This type of mechanism leads to on-demand re-
livery system through many strategies as addressed in recent state of lease of loaded proteins, mimicking the enzymatic activity naturally
art reviews of affinity-based drug delivery [11,143,144]. Therefore, occurring in ‘healthy’ ECM. However, stability and maintenance of
this paper deals only with some general aspects of this topic. biological activity of the protein may represent an issue.
One approach to incorporate heparin in a hydrogel structure is to co- Verheyen et al. recently reported on an efficient strategy to intro-
valently link the heparin moiety to the polymer. Many examples of duce methacrylamide groups on the lysine residues of a model protein
growth factor delivery from heparin modified polymer hydrogels have (lysozyme) for immobilization and triggered release from a polyacryl-
been reported. Hubbell used a bi-domain peptide that was covalently amide or dextran hydrogel network. They designed a novel spacer
R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692 689

unit containing a disulfide bond, such that the release of the protein can (MC) were designed by Shoichet et al. for independent delivery of one
be triggered by reduction [36,37]. or more therapeutic proteins, from 1 to 28 days, for the ultimate appli-
PEG hydrogels containing pendant RGD peptides can be formed cation of spinal cord injury repair. To achieve a diversity of release
using amine-functionalized MMP ligands as a cross-linker. Close prox- profiles, they exploited the combination of fast diffusion-
imity of the gel to cells was mediated by RGD peptide and BMP-2 controlled release of dissolved solutes from the HAMC itself and
bound to the matrix was delivered to the site of the bone defect [38]. slow drug release from PLGA particles dispersed within the gel
Other popular systems that rely on covalent cross-linking are those con- [41]. Methacrylated hyaluronan networks encapsulating alginate
taining peptide linkers susceptible to the enzymatic activity [158–160]. microparticles enhanced mesenchymal stem cell chondrogenesis
following delivery of TGF-β3 in vitro and in vivo [164].
3.3. Dual/multiple delivery systems

Biomedical hydrogels that can deliver multiple growth factors in a 4. Conclusions and outlook
multimodal mode and provide desirable pore structure and porosity
to potentially encapsulate cells, have considerable potential as future The importance of growth and adhesive factors and their synergistic
therapeutic tools in tissue engineering. The use of growth factors interplay during healthy tissue development is nowadays a well-
loaded microspheres embedded in the hydrogel structure is a com- established concept. However, their administration in the form of bolus
mon approach to multimodal protein delivery. injection to promote tissue regeneration and repair has shown to be
Microparticles of acidic and basic gelatin were used as carriers for the often ineffective and potentially harmful due to the short duration of ac-
individual delivery of two different growth factors and embedded in a hy- tion of the encapsulated protein drug. Such a delivery approach of
drogel matrix, composed of glycidyl methacrylated dextran (Dex-GMA)/ growth factors has been applied in a number of clinical trials of unsatis-
gelatin. This hybrid system allowed the independent release of BMP-2 factory outcome. While some phase I clinical trials (angiogenic gene
or IGF-1(Fig. 6) that facilitated cell attachment, proliferation, metabolism, therapy and human growth factor FGF-I infusion for coronary artery dis-
and osteoblastic differentiation of cells in a synergistic manner [39,161]. ease) [165,166] have reported promising results, phase II clinical trials,
Fibrin hydrogels were complexed with heparin-functionalized PLGA recruiting a larger number of patients, have not shown the expected
nanoparticles for the release of VEGF for revascularization purposes [40]. benefits to patients. To mention, VIVA (Vascular endothelial growth fac-
A similar approach was adopted by Burdick et al., who formulated tor in Ischemia for Vascular Angiogenesis) trial conducted on 178 pa-
PLGA microspheres into PLA-PEG-PLA hydrogels for the delivery of mul- tients by intravenous and intracoronary infusion of VEGF [167] and
tiple neutrophins with individual release rate [18]. Similarly, Biondi FIRST trial comprising intracoronary infusions of FGF-2 on 337 patients
et al. used PLGA microspheres in collagen gels and collagen-HA semi- [168], did not result in effective commercially available formulations
interpenetrating networks. The protein release kinetics and delivery for the treatment of cardiovascular diseases. On the other hand, positive
onset strongly depended on the complex interplay between protein outcomes resulted from clinical trials on growth factors administered
transport through the PLGA matrix and in the collagen-based release through the use of delivery systems that controlled to some extent the
media, and water sequestration within the scaffold [162,163]. Physical protein release rate. In this way the problems associated with the appli-
hydrogel blends composed of hyaluronan (HA) and methyl cellulose cations of bolus injections are overcome. For example, after successful
clinical trials, two growth factors BMP-2 (clinical trial BESTT) [169,170]
and BMP-7 (clinical trial OP-1 Putty) [171,172] immobilized in collagen
sponge and matrix, respectively, are currently commercially available
for the treatment of bone fractures and defects. Also in the field of cardio-
vascular diseases, encapsulation of FGF-2 into alginate microcapsules led
to some positive results in clinical trial phases (clinical trial Polymer)
[173,174]. Finally, Regranex® Gel is the only growth factor delivery sys-
tem of PDGF that obtained full FDA approval for clinical use.
Generally speaking, formulation of proteins in controlled delivery
systems allow to sustain the release and lower the doses of potent
growth factors, that, if administered by infusion, need supraphysiolo-
gical concentrations, leading to severe side-effects and do not assure
therapeutic efficacy over a sufficient time-span, because of rapid deg-
radation/elimination. Moreover, the design of smart delivery systems
may offer the possibility to deliver multiple growth factors with inde-
pendent release rates and to load or attract cells for the regeneration
of damaged tissues. This review summarizes the efforts that have
been addressed on facilitating the delivery of single or multiple
growth factors with appropriate control over temporal and spatial
presentation of these biomolecular cues. Current available technolo-
gies that have demonstrated potential in the modulation of release
profiles, according to the specific therapeutic needs, including tradi-
tional diffusion/swelling/degradation mediated, on-demand, affinity
and covalent binding based delivery are presented.
However, some challenges still exist. For example, highly inconve-
nient encapsulation methods often involving post-loading techniques
and low encapsulation efficiency, the stability of the protein, the safety
and biocompatibility of the delivery devices and the difficulties to trans-
late in vitro results to in vivo situation. Further efforts in the design or
Fig. 6. Cumulative protein(s) release of BMP-2 and/or IGF-1 from scaffolds containing
optimization of materials with minimal tissue response, sufficient stiff-
BMP-2/IGF-1 combination (BMP-2 and IGF-1) (C); or from scaffolds containing a mix- ness, appropriate degradation and release profiles are needed in order
ture of microparticles loaded with BMP-2 or IGF-1 (F). Reproduced from ref. [39]. to make the translational step between academia and clinics successful.
690 R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692

Acknowledgement [28] C. Hiemstra, Z. Zhong, S.R. Van Tomme, M.J. van Steenbergen, J.J.L. Jacobs, W.D.
Otter, W.E. Hennink, J. Feijen, In vitro and in vivo protein delivery from in situ
forming poly(ethylene glycol)-poly(lactide) hydrogels, J. Control. Release 119
This project was financially supported by L'Òreal – Unesco, “For (2007) 320–327.
Women in Science 2011”. [29] M. Ehrbar, R. Schoenmakers, E.H. Christen, M. Fussenegger, W. Weber, Drug-
sensing hydrogels for the inducible release of biopharmaceuticals, Nat. Mater.
7 (2008) 800–804.
[30] M. Lutolf, G. Raeber, A. Zisch, N. Tirelli, J. Hubbell, Cell-responsive synthetic
References hydrogels, Adv. Mater. 15 (2003) 888–892.
[31] E.A. Phelps, N. Landázuri, P.M. Thulé, W.R. Taylor, A.J. García, Bioartificial matrices
[1] E. Lavik, R. Langer, Tissue engineering: current state and perspectives, Appl. for therapeutic vascularization, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 3323–3328.
Microbiol. Biotechnol. 65 (2004) 1–8. [32] S.E. Sakiyama-Elbert, J.A. Hubbell, Development of fibrin derivatives for controlled
[2] A.G. Mikos, S.W. Herring, P. Ochareon, J. Elisseeff, H.H. Lu, R. Kandel, F.J. Schoen, release of heparin-binding growth factors, J. Control. Release 65 (2000) 389–402.
M. Toner, D. Mooney, A. Atala, M.E.V. Dyke, D. Kaplan, G. Vunjak-Novakovic, En- [33] S.E. Sakiyama-Elbert, J.A. Hubbell, Controlled release of nerve growth factor
gineering Complex Tissues, Tissue Eng. 12 (2006) 3307–3339. from a heparin-containing fibrin-based cell ingrowth matrix, J. Control. Release
[3] E. Bell, Tissue Engineering in Perspective, in: R.P. Lanza (Ed.), Principles of Tissue 69 (2000) 149–158.
Engineering, edn 2, Academic Press, San Diego, 2000. [34] C.-C. Lin, A.T. Metters, Metal-chelating affinity hydrogels for sustained protein
[4] S. Kobsa, W.M. Saltzman, Bioengineering approaches to controlled protein deliv- release, J. Biomed. Mater. Res. A 83A (2007) 954–964.
ery, Pediatr. Res. 63 (2008) 513–519. [35] C.-C. Lin, P.D. Boyer, A.A. Aimetti, K.S. Anseth, Regulating MCP-1 diffusion in affinity
[5] S.T. Andreadis, D.J. Geer, Biomimetic approaches to protein and gene delivery for hydrogels for enhancing immuno-isolation, J. Control. Release 142 (2010) 384–391.
tissue regeneration, Trends Biotechnol. 24 (2006) 331–337. [36] E. Verheyen, L. Delain-Bioton, S. Van Der Wal, N. El Morabit, A. Barendregt, W.E.
[6] R. Vasita, D.S. Katti, Growth factor-delivery systems for tissue engineering: a Hennink, C.F. Van Nostrum, Conjugation of methacrylamide groups to a model
materials perspective, Expert Rev. Med. Devices 3 (2005) 29–47. protein via a reducible linker for immobilization and subsequent triggered re-
[7] E.R. Edelman, M.A. Nugent, M.J. Karnovsky, Perivascular and intravenous admin- lease from hydrogels, Macromol. Biosci. 10 (2010) 1517–1526.
istration of basic fibroblast growth factor: vascular and solid organ deposition, [37] E. Verheyen, L. Delain-Bioton, S. der Wal, N. El Morabit, W.E. Hennink, C.F. van
Proc. Natl. Acad. Sci. U. S. A. 15 (1993) 1513–1517. Nostrum, Protein macromonomers for covalent immobilization and subsequent
[8] S.M. Eppler, D.L. Combs, T.D. Henry, J.J. Lopez, S.G. Ellis, J.H. Yi, B.H. Annex, E.R. triggered release from hydrogels, J. Control. Release 148 (2010) e18–e19.
McCluskey, T.F. Zioncheck, A target-mediated model to describe the pharmaco- [38] M.P. Lutolf, F.E. Weber, H.G. Schmoekel, J.C. Schense, T. Kohler, R. Muller, J.A.
kinetics and hemodynamic effects of recombinant human vascular endothelial Hubbell, Repair of bone defects using synthetic mimetics of collagenous extra-
growth factor in humans, Clin. Pharmacol. Ther. 72 (2002) 20–32. cellular matrices, Nat. Biotechnol. 21 (2003) 513–518.
[9] S.J. Lee, Cytokine delivery and tissue engineering, Yonsei Med. J. 41 (2000) 704–719. [39] F.M. Chen, R. Chen, X.J. Wang, H.H. Sun, Z.F. Wu, In vitro cellular responses to
[10] V. Balasubramanian, O. Onaca, R. Enea, D.W. Hughes, C.G. Palivan, Protein deliv- scaffolds containing two microencapulated growth factors, Biomaterials 30
ery: from conventional drug delivery carriers to polymeric nanoreactors, Expert (2009) 5215–5224.
Opin. Drug Deliv. 7 (2010) 63–78. [40] Y.-I. Chung, S.-K. Kim, Y.-K. Lee, S.-J. Park, K.-O. Cho, S.H. Yuk, G. Tae, Y.H. Kim,
[11] P. Tayalia, D.J. Mooney, Controlled growth factor delivery for tissue engineering, Efficient revascularization by VEGF administration via heparin-functionalized
Adv. Mater. 21 (2009) 3269–3285. nanoparticle–fibrin complex, J. Control. Release 143 (2010) 282–289.
[12] U. Nöth, L. Rackwitz, A.F. Steinert, R.S. Tuan, Cell delivery therapeutics for mus- [41] M.D. Baumann, C.E. Kang, J.C. Stanwick, Y. Wang, H. Kim, Y. Lapitsky, M.S.
culoskeletal regeneration, Adv. Drug Deliv. Rev. 62 (2010) 765–783. Shoichet, An injectable drug delivery platform for sustained combination thera-
[13] D.L. Coutu, A.-M. Yousefi, J. Galipeau, Three-dimensional porous scaffolds at the py, J. Control. Release 138 (2009) 205–213.
crossroads of tissue engineering and cell-based gene therapy, J. Cell. Biochem. [42] S.R. Van Tomme, G. Storm, W.E. Hennink, In situ gelling hydrogels for pharma-
108 (2009) 537–546. ceutical and biomedical applications, Int. J. Pharm. 355 (2008) 1–18.
[14] K. Ladewig, Drug delivery in soft tissue engineering, Expert Opin. Drug Deliv. [43] C.-C. Lin, A.T. Metters, Hydrogels in controlled release formulations: network de-
8 (2011) 1175–1188. sign and mathematical modeling, Adv. Drug Deliv. Rev. 58 (2006) 1379–1408.
[15] R. Censi, P.J. Fieten, P. Di Martino, W.E. Hennink, T. Vermonden, In situ forming [44] B. Amsden, Solute diffusion within hydrogels. Mechanisms and models, Macro-
hydrogels by tandem thermal gelling and Michael addition reaction between molecules 31 (1998) 8382–8395.
thermosensitive triblock copolymers and thiolated hyaluronan, Macromolecules [45] R.W. Korsmeyer, N.A. Peppas, Macromolecular and modeling aspects of
43 (2010) 5771–5778. swelling-controlled systems, in: T.J. Roseman, S.Z. Mansdorf (Eds.), Controlled
[16] R. Censi, T. Vermonden, H. Deschout, K. Braeckmans, P. Di Martino, S.C. De Release Delivery Systems, Marcel Dekker, 1983, pp. 77–90.
Smedt, C.F. Van Nostrum, W.E. Hennink, Photopolymerized thermosensitive [46] J.D. Boerckel, Y.M. Kolambkar, K.M. Dupont, B.A. Uhrig, E.A. Phelps, H.Y. Stevens,
poly(HPMAlactate)-PEG-based hydrogels: effect of network design on mechan- A.J. García, R.E. Guldberg, Effects of protein dose and delivery system on BMP-
ical properties, degradation, and release behavior, Biomacromolecules 11 mediated bone regeneration, Biomaterials 32 (2011) 5241–5251.
(2010) 2143–2151. [47] K.V. Brown, B. Li, T. Guda, D.S. Perrien, S. Guelcher, J.C. Wenke, Improving bone
[17] R. Censi, T. Vermonden, M.J. van Steenbergen, H. Deschout, K. Braeckmans, S.C. formation in a rat femur segmental defect by controlling BMP-2 release, Tissue
De Smedt, C.F. van Nostrum, P. di Martino, W.E. Hennink, Photopolymerized Eng. Part A 17 (2011) 1735–1746.
thermosensitive hydrogels for tailorable diffusion-controlled protein delivery, [48] P.L. Ritger, N.A. Peppas, A simple equation for description of solute release II. Fickian
J. Control. Release 140 (2009) 230–236. and anomalous release from swellable devices, J. Control. Release 5 (1987) 37–42.
[18] J.A. Burdick, M. Ward, E. Liang, M.J. Young, R. Langer, Stimulation of neurite out- [49] L. Serra, J. Doménech, N.A. Peppas, Drug transport mechanisms and release ki-
growth by neurotrophins delivered from degradable hydrogels, Biomaterials 27 netics from molecularly designed poly(acrylic acid-g-ethylene glycol) hydro-
(2006) 452–459. gels, Biomaterials 27 (2006) 5440–5451.
[19] S. Zhang, T.C. Holmes, C.M. DiPersio, R.O. Hynes, X. Su, A. Rich, Self-complemen- [50] C.S. Brazel, N.A. Peppas, Modeling of drug release from swellable polymers, Eur.
tary oligopeptide matrices support mammalian cell attachment, Biomaterials 16 J. Pharm. Biopharm. 49 (2000) 47–58.
(1995) 1385–1393. [51] W.S.R. Censi, J. Malda, G. di Dato, P.E. Burgisser, W.J.A. Dhert, C.F. van Nostrum, P.
[20] E.A. Silva, D.J. Mooney, Effects of VEGF temporal and spatial presentation on an- di Martino, T. Vermonden, W.E. Hennink, Printable photopolymerizable thermo-
giogenesis, Biomaterials 31 (2010) 1235–1241. sensitive p(HPMA-lactate)-PEG hydrogel for tissue engineering, Adv. Funct.
[21] R.R. Chen, E.A. Silva, W.W. Yuen, A.A. Brock, C. Fischbach, A.S. Lin, R.E. Guldberg, Mater. 21 (2011) 1833–1842.
D.J. Mooney, Integrated approach to designing growth factor delivery systems, [52] L. Pescosolido, W. Schuurman, J. Malda, P. Matricardi, F. Alhaique, T. Coviello, P.R.
FASEB J. 21 (2007) 3896–3903. Van Weeren, W.J.A. Dhert, W.E. Hennink, T. Vermonden, Hyaluronic acid and
[22] C. Hiemstra, Z. Zhong, M.J. van Steenbergen, W.E. Hennink, J. Feijen, Release of dextran-based semi-IPN hydrogels as biomaterials for bioprinting, Biomacromo-
model proteins and basic fibroblast growth factor from in situ forming degrad- lecules 12 (2011) 1831–1838.
able dextran hydrogels, J. Control. Release 122 (2007) 71–78. [53] T.M. Seck, F.P.W. Melchels, J. Feijen, D.W. Grijpma, Designed biodegradable
[23] G.W. Bos, J.J.L. Jacobs, J.W. Koten, S. Van Tomme, T. Veldhuis, C.F. van Nostrum, W. Den hydrogel structures prepared by stereolithography using poly(ethylene
Otter, W.E. Hennink, In situ crosslinked biodegradable hydrogels loaded with IL-2 are glycol)/poly(d, l-lactide)-based resins, J. Control. Release 148 (2010) 34–41.
effective tools for local IL-2 therapy, Eur. J. Pharm. Sci. 21 (2004) 561–567. [54] A.S. Sawhney, C.P. Pathak, J.A. Hubbell, Bioerodible hydrogels based on photopo-
[24] S.R. Van Tomme, B.G. De Geest, K. Braeckmans, S.C. De Smedt, F. Siepmann, J. lymerized poly(ethylene glycol)-co-poly(α-hydroxy acid) diacrylate macro-
Siepmann, C.F. Van Nostrum, W.E. Hennink, Mobility of model proteins in mers, Macromolecules 26 (1993) 581–587.
hydrogels composed of oppositely charged dextran microspheres studied by [55] J.A. Burdick, M.N. Mason, A.D. Hinman, K. Thorne, K.S. Anseth, Delivery of
protein release and fluorescence recovery after photobleaching, J. Control. Re- osteoinductive growth factors from degradable PEG hydrogels influences osteo-
lease 110 (2005) 67–78. blast differentiation and mineralization, J. Control. Release 83 (2002) 53–63.
[25] Y.J. Kim, S. Choi, J.J. Koh, M. Lee, K.S. Ko, S.W. Kim, Controlled release of insulin from [56] J. Elisseeff, W. McIntosh, K. Fu, T. Blunk, R. Langer, Controlled-release of IGF-I and
injectable biodegradable triblock copolymer, Pharm. Res. 18 (2001) 548–550. TGF-β1 in a photopolymerizing hydrogel for cartilage tissue engineering, J.
[26] S. Choi, M. Baudys, S. Kim, Control of blood glucose by novel GLP-1 delivery Orthop. Res. 19 (2001) 1098–1104.
using biodegradable triblock copolymer of PLGA-PEG-PLGA in type 2 diabetic [57] A.T. Metters, K.S. Anseth, C.N. Bowman, A statistical kinetic model for the bulk
rats, Pharm. Res. 21 (2004) 827–831. degradation of PLA-b-PEG-b-PLA hydrogel networks: incorporating network
[27] F.D. Van Manakker, K. Braeckmans, N.E. Morabit, S.C. De Smedt, C.F. Van Nostrum, non-idealities, J. Phys. Chem. B 105 (2001) 8069–8076.
W.E. Hennink, Protein-release behavior of self-assembled PEG-ß-cyclodextrin/- [58] K.A. Davis, J.A. Burdick, K.S. Anseth, Photoinitiated crosslinked degradable copolymer
PEG-cholesterol hydrogels, Adv. Funct. Mater. 19 (2009) 2992–3001. networks for tissue engineering applications, Biomaterials 24 (2003) 2485–2495.
R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692 691

[59] B. Jeong, Y.H. Bae, S.W. Kim, Thermoreversible gelation of PEG-PLGA-PEG tri- [91] W.N.E. van Dijk-Wolthuis, J.A.M. Hoogeboom, M.J. van Steenbergen, S.K.Y. Tsang,
block copolymer aqueous solutions, Macromolecules 32 (1999) 7064–7069. W.E. Hennink, Degradation and release behavior of dextran-based hydrogels,
[60] B. Jeong, Y.H. Bae, S.W. Kim, In situ gelation of PEG-PLGA-PEG triblock copolymer Macromolecules 30 (1997) 4639–4645.
aqueous solutions and degradation thereof, J. Biomed. Mater. Res. 50 (2000) 171–177. [92] R.J.H. Stenekes, O. Franssen, E.M.G. van Bommel, D.J.A. Crommelin, W.E. Hennink, The
[61] P.Y. Lee, Z. Li, L. Huang, Thermosensitive hydrogel as a Tgf-β1 gene delivery ve- preparation of dextran microspheres in an all-aqueous system: effect of the formula-
hicle enhances diabetic wound healing, Pharm. Res. 20 (2003) 1995–2000. tion parameters on particle characteristics, Pharm. Res. 15 (1998) 557–561.
[62] G.M. Zentner, R. Rathi, C. Shih, J.C. McRea, M.H. Seo, H. Oh, B.G. Rhee, J. Mestecky, Z. [93] C.J. De Groot, J.A. Cadée, J.-W. Koten, W.E. Hennink, W. Den Otter, Therapeutic efficacy
Moldoveanu, M. Morgan, S. Weitman, Biodegradable block copolymers for delivery of IL-2-loaded hydrogels in a mouse tumor model, Int. J. Cancer 98 (2002) 134–140.
of proteins and water-insoluble drugs, J. Control. Release 72 (2001) 203–215. [94] K. Vlugt-Wensink, R. de Vrueh, M. Gresnigt, C. Hoogerbrugge, S. van Buul-Offers,
[63] T. Vermonden, N.E. Fedorovich, D. van Geemen, J. Alblas, C.F. van Nostrum, W.J.A. L. de Leede, L. Sterkman, D. Crommelin, W. Hennink, R. Verrijk, Preclinical and
Dhert, W.E. Hennink, Photopolymerized thermosensitive hydrogels: synthesis, clinical in vitro in vivo correlation of an hgh dextran microsphere formulation,
degradation, and cytocompatibility, Biomacromolecules 9 (2008) 919–926. Pharm. Res. 24 (2007) 2239–2248.
[64] T. Vermonden, S.S. Jena, D. Barriet, R. Censi, J. van der Gucht, W.E. Hennink, R.A. [95] S.G. Lévesque, M.S. Shoichet, Synthesis of enzyme-degradable, peptide-cross-linked
Siegel, Macromolecular diffusion in self-assembling biodegradable thermosensi- dextran hydrogels, Bioconjug. Chem. 18 (2007) 874–885.
tive hydrogels, Macromolecules 43 (2009) 782–789. [96] G. Sun, Y.-I. Shen, S. Kusuma, K. Fox-Talbot, C.J. Steenbergen, S. Gerecht, Func-
[65] R. Censi, S. van Putten, T. Vermonden, P. di Martino, C.F. van Nostrum, M.C. tional neovascularization of biodegradable dextran hydrogels with multiple an-
Harmsen, R.A. Bank, W.E. Hennink, The tissue response to Photopolymerized PEG- giogenic growth factors, Biomaterials 32 (2011) 95–106.
p(HPMAm-lactate)-based Hydrogels, J. Biomed. Mater. Res. A 97 A (2010) 219–229. [97] F.-M. Chen, Y.-M. Zhao, H.-H. Sun, T. Jin, Q.-T. Wang, W. Zhou, Z.-F. Wu, Y. Jin,
[66] P.C.H. Hsieh, M.E. Davis, J. Gannon, C. MacGillivray, R.T. Lee, Controlled delivery Novel glycidyl methacrylated dextran (Dex-GMA)/gelatin hydrogel scaffolds
of PDGF-BB for myocardial protection using injectable self-assembling peptide containing microspheres loaded with bone morphogenetic proteins: formula-
nanofibers, J. Clin. Invest. 116 (2006) 237–248. tion and characteristics, J. Control. Release 118 (2007) 65–77.
[67] V.F.M. Segers, T. Tokunou, L.J. Higgins, C. MacGillivray, J. Gannon, R.T. Lee, Local [98] M. Morra, Engineering of biomaterials surfaces by hyaluronan, Biomacromole-
delivery of protease-resistant stromal cell derived factor-1 for Stem cell recruit- cules 6 (2005) 1205–1223.
ment after myocardial infarction, Circulation 116 (2007) 1683–1692. [99] M. Mori, M. Yamaguchi, S. Sumitomo, Y. Takai, Hyaluronan-based biomaterials
[68] M.E. Davis, P.C.H. Hsieh, T. Takahashi, Q. Song, S. Zhang, R.D. Kamm, A.J. in tissue engineering, Acta Histochem. Cytochem. 37 (2004) 1–5.
Grodzinsky, P. Anversa, R.T. Lee, Local myocardial insulin-like growth factor 1 [100] G. Kogan, L. Šoltés, R. Stern, P. Gemeiner, Hyaluronic acid: a natural biopolymer with a
(IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy broad range of biomedical and industrial applications, Biotechnol. Lett. 29 (2007)
for myocardial infarction, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 8155–8160. 17–25.
[69] B. Balakrishnan, R. Banerjee, Biopolymer-Based Hydrogels for Cartilage Tissue [101] D.D. Allison, K.J. Grande-Allen, Hyaluronan: a powerful tissue engineering tool,
Engineering, Chem. Rev. 111 (2011) 4453–4474. Tissue Eng. 12 (2006) 2131–2140.
[70] N. Hunt, L. Grover, Cell encapsulation using biopolymer gels for regenerative [102] J. Patterson, R. Siew, S.W. Herring, A.S.P. Lin, R. Guldberg, P.S. Stayton, Hyaluro-
medicine, Biotechnol. Lett. 32 (2010) 733–742. nic acid hydrogels with controlled degradation properties for oriented bone re-
[71] S. Huang, X. Fu, Naturally derived materials-based cell and drug delivery sys- generation, Biomaterials 31 (2010) 6772–6781.
tems in skin regeneration, J. Control. Release 142 (2010) 149–159. [103] I.L. Kim, R.L. Mauck, J.A. Burdick, Hydrogel design for cartilage tissue engineering: a
[72] S. Van Vlierberghe, P. Dubruel, E. Schacht, Biopolymer-based hydrogels as scaf- case study with hyaluronic acid, Biomaterials 32 (2011) 8771–8782.
folds for tissue engineering applications: a review, Biomacromolecules (2011), [104] R.A. Peattie, D.B. Pike, B. Yu, S. Cai, X.Z. Shu, G.D. Prestwich, M.A. Firpo, R.J. Fisher,
doi:10.1021/bm200083n. Effect of gelatin on heparin regulation of cytokine release from hyaluronan-
[73] M. Rinaudo, Main properties and current applications of some polysaccharides based hydrogels, Drug Deliv. 15 (2008) 389–397.
as biomaterials, Polym. Int. 57 (2008) 397–430. [105] M.R. Kim, T.G. Park, Temperature-responsive and degradable hyaluronic
[74] T. Coviello, P. Matricardi, C. Marianecci, F. Alhaique, Polysaccharide hydrogels for acid/pluronic composite hydrogels for controlled release of human growth
modified release formulations, J. Control. Release 119 (2007) 5–24. hormone, J. Control. Release 80 (2002) 69–77.
[75] L. Pescosolido, T. Piro, T. Vermonden, T. Coviello, F. Alhaique, W.E. Hennink, P. [106] C.M. Riley, P.W. Fuegy, M.A. Firpo, X. Zheng Shu, G.D. Prestwich, R.A. Peattie,
Matricardi, Biodegradable IPNs based on oxidized alginate and dextran-HEMA Stimulation of in vivo angiogenesis using dual growth factor-loaded crosslinked
for controlled release of proteins, Carbohydr. Polym. 86 (2011) 208–213. glycosaminoglycan hydrogels, Biomaterials 27 (2006) 5935–5943.
[76] K.H. Bouhadir, K.Y. Lee, E. Alsberg, K.L. Damm, K.W. Anderson, D.J. Mooney, Deg- [107] E. Martínez-Sanz, D.A. Ossipov, J. Hilborn, S. Larsson, K.B. Jonsson, O.P. Varghese,
radation of partially oxidized alginate and its potential application for tissue en- Bone reservoir: injectable hyaluronic acid hydrogel for minimal invasive bone
gineering, Biotechnol. Progr. 17 (2001) 945–950. augmentation, J. Control. Release 152 (2011) 232–240.
[77] S.Y. Rabbany, J. Pastore, M. Yamamoto, T. Miller, S. Rafii, R. Aras, M. Penn, Contin- [108] J.B. Leach, C.E. Schmidt, Characterization of protein release from photocrosslink-
uous delivery of stromal cell-derived factor-1 from alginate scaffolds accelerates able hyaluronic acid-polyethylene glycol hydrogel tissue engineering scaffolds,
wound healing, Cell Transplant. 19 (2010) 399–408. Biomaterials 26 (2005) 125–135.
[78] M. Leonard, M.R. De Boisseson, P. Hubert, F. Dalençon, E. Dellacherie, Hydropho- [109] F. Lee, J.E. Chung, M. Kurisawa, An injectable hyaluronic acid-tyramine hydrogel
bically modified alginate hydrogels as protein carriers with specific controlled system for protein delivery, J. Control. Release 134 (2009) 186–193.
release properties, J. Control. Release 98 (2004) 395–405. [110] S.K. Hahn, S. Jelacic, R.V. Maier, P.S. Stayton, A.S. Hoffman, Anti-inflammatory drug de-
[79] A.W. Chan, R.J. Neufeld, Tuneable semi-synthetic network alginate for absorp- livery from hyaluronic acid hydrogels, J. Biomater. Sci. Polym. Ed. 15 (2004)
tive encapsulation and controlled release of protein therapeutics, Biomaterials 1111–1119.
31 (2010) 9040–9047. [111] S.K. Hahn, J.S. Kim, T. Shimobouji, Injectable hyaluronic acid microhydrogels for
[80] I. Freeman, A. Kedem, S. Cohen, The effect of sulfation of alginate hydrogels on controlled release formulation of erythropoietin, J. Biomed. Mater. Res. A 80
the specific binding and controlled release of heparin-binding proteins, Bioma- (2007) 916–924.
terials 29 (2008) 3260–3268. [112] S.K. Hahn, E.J. Oh, H. Miyamoto, T. Shimobouji, Sustained release formulation of
[81] M. George, T.E. Abraham, pH sensitive alginate-guar gum hydrogel for the con- erythropoietin using hyaluronic acid hydrogels crosslinked by Michael addition,
trolled delivery of protein drugs, Int. J. Pharm. 335 (2007) 123–129. Int. J. Pharm. 322 (2006) 44–51.
[82] H.-F. Liang, M.-H. Hong, R.-M. Ho, C.-K. Chung, Y.-H. Lin, C.-H. Chen, H.-W. Sung, Novel [113] S. Cai, Y. Liu, Z.S. Xiao, G.D. Prestwich, Injectable glycosaminoglycan hydrogels for con-
method using a temperature-sensitive polymer (methylcellulose) to thermally gel trolled release of human basic fibroblast growth factor, Biomaterials 26 (2005)
aqueous alginate as a ph-sensitive hydrogel, Biomacromolecules 5 (2004) 6054–6067.
1917–1925. [114] R. Elia, P.W. Fuegy, A. VanDelden, M.A. Firpo, G.D. Prestwich, R.A. Peattie, Stim-
[83] L. Pescosolido, S. Miatto, C. Di Meo, C. Cencetti, T. Coviello, F. Alhaique, P. ulation of in vivo angiogenesis by in situ crosslinked, dual growth factor-
Matricardi, Injectable and in situ gelling hydrogels for modified protein release, loaded, glycosaminoglycan hydrogels, Biomaterials 31 (2010) 4630–4638.
Eur. Biophys. J. 39 (2010) 903–909. [115] K. Ono, Y. Saito, H. Yura, K. Ishikawa, A. Kurita, T. Akaike, M. Ishihara, Photocrosslink-
[84] E. Ruvinov, J. Leor, S. Cohen, The effects of controlled HGF delivery from an able chitosan as a biological adhesive, J. Biomed. Mater. Res. 49 (2000) 289–295.
affinity-binding alginate biomaterial on angiogenesis and blood perfusion in a [116] K. Obara, M. Ishihara, T. Ishizuka, M. Fujita, Y. Ozeki, T. Maehara, Y. Saito, H. Yura,
hindlimb ischemia model, Biomaterials 31 (2010) 4573–4582. T. Matsui, H. Hattori, M. Kikuchi, A. Kurita, Photocrosslinkable chitosan hydrogel
[85] E.A. Silva, D.J. Mooney, Spatiotemporal control of vascular endothelial growth factor containing fibroblast growth factor-2 stimulates wound healing in healing-
delivery from injectable hydrogels enhances angiogenesis, J. Thromb. Haemost. 5 impaired db/db mice, Biomaterials 24 (2003) 3437–3444.
(2007) 590–598. [117] M. Fujita, M. Ishihara, Y. Morimoto, M. Simizu, Y. Saito, H. Yura, T. Matsui, B.
[86] A.D. Jenkins, P. Kratochvìl, R.F.T. Stepto, U.W Suter, Glossary of basic terms in Takase, H. Hattori, Y. Kanatani, M. Kikuchi, T. Maehara, Efficacy of photocros-
polymer science, Pure Appl. Chem. 68 (1996) 2287. slinkable chitosan hydrogel containing fibroblast growth factor-2 in a rabbit
[87] S.M. Jay, B.R. Shepherd, J.W. Andrejecsk, T.R. Kyriakides, J.S. Pober, W.M. model of chronic myocardial infarction, J. Surg. Res. 126 (2005) 27–33.
Saltzman, Dual delivery of VEGF and MCP-1 to support endothelial cell trans- [118] Y. Yeo, W. Geng, T. Ito, D.S. Kohane, J.A. Burdick, M. Radisic, Photocrosslinkable hydro-
plantation for therapeutic vascularization, Biomaterials 31 (2010) 3054–3062. gel for myocyte cell culture and injection, J. Biomed. Mater. Res. B 81B (2007)
[88] S.M. Jay, W.M. Saltzman, Controlled delivery of VEGF via modulation of alginate 312–322.
microparticle ionic crosslinking, J. Control. Release 134 (2009) 26–34. [119] K. Masuoka, M. Ishihara, T. Asazuma, H. Hattori, T. Matsui, B. Takase, Y. Kanatani,
[89] E.A. Silva, E.-S. Kim, H.J. Kong, D.J. Mooney, Material-based deployment en- M. Fujita, Y. Saito, H. Yura, K. Fujikawa, K. Nemoto, The interaction of chitosan
hances efficacy of endothelial progenitor cells, Proc. Natl. Acad. Sci. U. S. A. 105 with fibroblast growth factor-2 and its protection from inactivation, Biomate-
(2008) 14347–14352. rials 26 (2005) 3277–3284.
[90] S.R. Van Tomme, W.E. Hennink, Biodegradable dextran hydrogels for protein de- [120] K. Obara, M. Ishihara, M. Fujita, Y. Kanatani, H. Hattori, T. Matsui, B. Takase, Y.
livery applications, Expert Rev. Med. Devices 4 (2007) 147–164. Ozeki, S. Nakamura, T. Ishizuka, S. Tominaga, S. Hiroi, T. Kawai, T. Maehara,
692 R. Censi et al. / Journal of Controlled Release 161 (2012) 680–692

Acceleration of wound healing in healing-impaired db/db mice with a photo- [152] Y. Liu, S. Cai, X.Z. Shu, J. Shelby, G.D. Prestwich, Release of basic fibroblast growth
crosslinkable chitosan hydrogel containing fibroblast growth factor-2, Wound factor from a crosslinked glycosaminoglycan hydrogel promotes wound healing,
Repair Regen. 13 (2005) 390–397. Wound Repair Regen. 15 (2007) 245–251.
[121] H. Zhang, A. Qadeer, W. Chen, In situ gelable interpenetrating double network [153] J.J. Yoon, H.J. Chung, T.G. Park, Photo-crosslinkable and biodegradable pluroni-
hydrogel formulated from binary components: thiolated chitosan and oxidized c/heparin hydrogels for local and sustained delivery of angiogenic growth factor,
dextran, Biomacromolecules 12 (2011) 1428–1437. J. Biomed. Mater. Res. A 83A (2007) 597–605.
[122] N. Bhattarai, J. Gunn, M.Q. Zhang, Chitosan-based hydrogels for controlled, local- [154] L.B. Menajovsky, Heparin-induced thrombocytopenia: clinical manifestations
ized drug delivery, Adv. Drug Deliv. Rev. 62 (2010) 83–99. and management strategies, Am. J. Med. 118 (2005) 21S–30S.
[123] M. Ishihara, K. Obara, S. Nakamura, M. Fujita, K. Masuoka, Y. Kanatani, B. Takase, [155] S.M. Willerth, P.J. Johnson, D.J. Maxwell, S.R. Parsons, M.E. Doukas, S.E.
H. Hattori, Y. Morimoto, M. Ishihara, T. Maehara, M. Kikuchi, Chitosan hydrogel Sakiyama-Elbert, Rationally designed peptides for controlled release of nerve
as a drug delivery carrier to control angiogenesis, J. Artif. Organs 9 (2006) 8–16. growth factor from fibrin matrices, J. Biomed. Mater. Res. A 80A (2007) 13–23.
[124] M. Amidi, E. Mastrobattista, W. Jiskoot, W.E. Hennink, Chitosan-based delivery sys- [156] Y. Zhao, J. Zhang, X. Wang, B. Chen, Z. Xiao, C. Shi, Z. Wei, X. Hou, Q. Wang, J. Dai,
tems for protein therapeutics and antigens, Adv. Drug Deliv. Rev. 62 (2010) 59–82. The osteogenic effect of bone morphogenetic protein-2 on the collagen scaffold
[125] N.W. Turner, C.W. Jeans, K.R. Brain, C.J. Allender, V. Hlady, D.W. Britt, From 3D to conjugated with antibodies, J. Control. Release 141 (2010) 30–37.
2D: a review of the molecular imprinting of proteins, Biotechnol. Progr. 22 [157] E. Verheyen, J.P. Schillemans, M. Van Wijk, M.A. Demeniex, W.E. Hennink, C.F.
(2006) 1474–1489. Van Nostrum, Challenges for the effective molecular imprinting of proteins, Bio-
[126] S.H. De Paoli Lacerda, B. Ingber, N. Rosenzweig, Structure-release rate correlation in materials 32 (2011) 3008–3020.
collagen gels containing fluorescent drug analog, Biomaterials 26 (2005) 7165–7172. [158] A.H. Zisch, M.P. Lutolf, M. Ehrbar, G.P. Raeber, S.C. Rizzi, N. Davies, H. Schmökel,
[127] D.G. Wallace, J. Rosenblatt, Collagen gel systems for sustained delivery and tis- D. Bezuidenhout, V. Djonov, P. Zilla, J.A. Hubbell, Cell-demanded release of VEGF
sue engineering, Adv. Drug Deliv. Rev. 55 (2003) 1631–1649. from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue
[128] S. Young, M. Wong, Y. Tabata, A.G. Mikos, Gelatin as a delivery vehicle for the con- growth, FASEB J. 17 (2003) 2260–2262.
trolled release of bioactive molecules, J. Control. Release 109 (2005) 256–274. [159] P.D. Thornton, G. McConnell, R.V. Ulijn, Enzyme responsive polymer hydrogel
[129] Y. Tabata, A. Nagano, Y. Ikada, Biodegradation of hydrogel carrier incorporating beads, Chem. Commun. (2005) 5913–5915.
fibroblast growth factor, Tissue Eng. 5 (1999) 127–138. [160] M. Ehrbar, S.C. Rizzi, R. Hlushchuk, V. Djonov, A.H. Zisch, J.A. Hubbell, F.E. Weber,
[130] Z. Patel, H. Ueda, M. Yamamoto, Y. Tabata, A. Mikos, In vitro and in vivo release of M.P. Lutolf, Enzymatic formation of modular cell-instructive fibrin analogs for
vascular endothelial growth factor from gelatin microparticles and biodegrad- tissue engineering, Biomaterials 28 (2007) 3856–3866.
able composite scaffolds, Pharm. Res. 25 (2008) 2370–2378. [161] F.M. Chen, Y.M. Zhao, R. Zhang, T. Jin, H.H. Sun, Z.F. Wu, Y. Jin, Periodontal regen-
[131] M. Sutter, J. Siepmann, W.E. Hennink, W. Jiskoot, Recombinant gelatin hydrogels eration using novel glycidyl methacrylated dextran (Dex-GMA)/gelatin scaf-
for the sustained release of proteins, J. Control. Release 119 (2007) 301–312. folds containing microspheres loaded with bone morphogenetic proteins, J.
[132] H. Teles, T. Vermonden, G. Eggink, W.E. Hennink, F.A. de Wolf, Hydrogels of Control. Release 121 (2007) 81–90.
collagen-inspired telechelic triblock copolymers for the sustained release of pro- [162] M. Biondi, L. Indolfi, F. Ungaro, F. Quaglia, M. La Rotonda, P. Netti, Bioactivated
teins, J. Control. Release 147 (2010) 298–303. collagen-based scaffolds embedding protein-releasing biodegradable microspheres:
[133] A.H. Zisch, U. Schenk, J.C. Schense, S.E. Sakiyama-Elbert, J.A. Hubbell, Covalently tuning of protein release kinetics, J. Mater. Sci. Mater. Med. 20 (2009) 2117–2128.
conjugated VEGF–fibrin matrices for endothelialization, J. Control. Release 72 [163] F. Ungaro, M. Biondi, I. d'Angelo, L. Indolfi, F. Quaglia, P.A. Netti, M.I. La Rotonda,
(2001) 101–113. Microsphere-integrated collagen scaffolds for tissue engineering: effect of mi-
[134] M. Ehrbar, S.M. Zeisberger, G.P. Raeber, J.A. Hubbell, C. Schnell, A.H. Zisch, The role crosphere formulation and scaffold properties on protein release kinetics, J. Con-
of actively released fibrin-conjugated VEGF for VEGF receptor 2 gene activation and trol. Release 113 (2006) 128–136.
the enhancement of angiogenesis, Biomaterials 29 (2008) 1720–1729. [164] L. Bian, D.Y. Zhai, E. Tous, R. Rai, R.L. Mauck, J.A. Burdick, Enhanced MSC chondrogen-
[135] H.S. Yang, S.H. Bhang, J.W. Hwang, D.-I. Kim, B.-S. Kim, Delivery of basic fibro- esis following delivery of TGF-β3 from alginate microspheres within hyaluronic acid
blast growth factor using heparin-conjugated fibrin for therapeutic angiogene- hydrogels in vitro and in vivo, Biomaterials 32 (2011) 6425–6434.
sis, Tissue Eng. Part A 16 (2010) 2113–2119. [165] Todd K. Rosengart, Leonard Y. Lee, Shailen R. Patel, Paul D. Kligfield, Peter M.
[136] C.T. Drinnan, G. Zhang, M.A. Alexander, A.S. Pulido, L.J. Suggs, Multimodal release of Okin, Neil R. Hackett, O. Wayne Isom, Ronald G. Crystal, Six-Month Assessment
transforming growth factor-β1 and the BB isoform of platelet derived growth factor of a Phase I Trial of Angiogenic Gene Therapy for the Treatment of Coronary Ar-
from PEGylated fibrin gels, J. Control. Release 147 (2010) 180–186. tery Disease Using Direct Intramyocardial Administration of an Adenovirus Vec-
[137] A.J.T. Hyatt, D. Wang, J.C. Kwok, J.W. Fawcett, K.R. Martin, Controlled release of tor Expressing the VEGF121 cDNA, Ann. Surg. 230 (1999) 466.
chondroitinase ABC from fibrin gel reduces the level of inhibitory glycosamino- [166] B. Schumacher, P. Pecher, B.U. von Specht, T. Stegmann, Induction of neoangio-
glycan chains in lesioned spinal cord, J. Control. Release 147 (2010) 24–29. genesis in ischemic myocardium by human growth factors: first clinical results
[138] J.P. Schillemans, E. Verheyen, A. Barendregt, W.E. Hennink, C.F. Van Nostrum, of a new treatment of coronary heart disease, Circulation 97 (1998) 645–650.
Anionic and cationic dextran hydrogels for post-loading and release of proteins, [167] T.D. Henry, B.H. Annex, G.R. McKendall, M.A. Azrin, J.J. Lopez, F.J. Giordano, P.K.
J. Control. Release 150 (2011) 266–271. Shah, J.T. Willerson, R.L. Benza, D.S. Berman, C.M. Gibson, A. Bajamonde, A.C.
[139] J.P. Schillemans, W.E. Hennink, C.F. van Nostrum, The effect of network charge Rundle, J. Fine, E.R. McCluskey, f.t.V. Investigators, The VIVA Trial, Circulation
on the immobilization and release of proteins from chemically crosslinked dex- 107 (2003) 1359–1365.
tran hydrogels, Eur. J. Pharm. Biopharm. 76 (2010) 329–335. [168] M. Simons, J.A. Ware, Therapeutic angiogenesis in cardiovascular disease, Nat.
[140] J.P. Schillemans, W.E. Hennink, C.F. van Nostrum, Charged dextran hydrogels for Rev. Drug Discov. 2 (2003) 863–871.
post-loading and release of proteins, J. Control. Release 148 (2010) e82–e83. [169] H.T. Aro, S. Govender, A.D. Patel, P. Hernigou, A.P. De Gregorio, G.I. Popescu, J.D.
[141] D.J. Maxwell, B.C. Hicks, S. Parsons, S.E. Sakiyama-Elbert, Development of rationally Golden, J. Christensen, A. Valentin, Recombinant human bone morphogenetic
designed affinity-based drug delivery systems, Acta Biomater. 1 (2005) 101–113. protein-2: a randomized trial in open tibial fractures treated with reamed nail
[142] E.R. Edelman, E. Mathiowitz, R. Langer, M. Klagsbrun, Controlled and modulated fixation, J. Bone Joint Surg. Am. 93 (2011) 801–808.
release of basic fibroblast growth factor, Biomaterials 12 (1991) 619–626. [170] S. Govender, C. Csimma, H.K. Genant, A. Valentin-Opran, Y. Amit, R. Arbel, H. Aro,
[143] N.X. Wang, H.A. Von Recum, Affinity-based drug delivery, Macromol. Biosci. 11 D. Atar, M. Bishay, M.G. Börner, P. Chiron, P. Choong, J. Cinats, B. Courtenay, R.
(2011) 321–332. Feibel, B. Geulette, C. Gravel, N. Haas, M. Raschke, E. Hammacher, D. Van der
[144] K. Lee, E.A. Silva, D.J. Mooney, Growth factor delivery-based tissue engineering: Velde, P. Hardy, M. Holt, C. Josten, R.L. Ketterl, B. Lindeque, G. Lob, H.
general approaches and a review of recent developments, J. R. Soc. Interface Mathevon, G. McCoy, D. Marsh, R. Miller, E. Munting, S. Oevre, L. Nordsletten,
8 (2011) 153–170. A. Patel, A. Pohl, W. Rennie, P. Reynders, P.M. Rommens, J. Rondia, W.C.
[145] M.D. Wood, G.H. Borschel, S.E. Sakiyama-Elbert, Controlled release of glial- Rossouw, P.J. Daneel, S. Ruff, A. Rüter, S. Santavirtal, T.A. Schidhauer, C. Gekle,
derived neurotrophic factor from fibrin matrices containing an affinity-based R. Schnettler, D. Segal, H. Seiler, R.B. Snowdowne, J. Stapert, G. Taglang, R.
delivery system, J. Biomed. Mater. Res. A 89A (2009) 909–918. Verdonik, L. Vogels, A. Weckbach, A. Wentzensen, T. Wisriewsk, Recombinant
[146] N. Yamaguchi, B.-S. Chae, L. Zhang, K.L. Kiick, E.M. Furst, Rheological characteri- human bone morphogenetic protein-2 for treatment of open tibial fractures a
zation of polysaccharide−poly(ethylene glycol) star copolymer hydrogels, Bio- prospective, controlled, randomized study of four hundred and fifty patients, J.
macromolecules 6 (2005) 1931–1940. Bone Joint Surg. Am. 84 (2002) 2123–2134.
[147] T. Nie, A. Baldwin, N. Yamaguchi, K.L. Kiick, Production of heparin-functionalized [171] A.R. Vaccaro, J.P. Lawrence, T. Patel, L.D. Katz, D.G. Anderson, J.S. Fischgrund, J.
hydrogels for the development of responsive and controlled growth factor de- Krop, M.G. Fehlings, D. Wong, The safety and efficacy of OP-1 (rhBMP-7) as a re-
livery systems, J. Control. Release 122 (2007) 287–296. placement for iliac crest autograft in posterolateral lumbar arthrodesis: a long-
[148] N. Yamaguchi, L. Zhang, B.-S. Chae, C.S. Palla, E.M. Furst, K.L. Kiick, Growth factor term (>4 years) pivotal study, Spine 33 (2008) 2850–2862.
mediated assembly of cell receptor-responsive hydrogels, J. Am. Chem. Soc. 129 [172] A.R. Vaccaro, P.G. Whang, T. Patel, F.M. Phillips, D.G. Anderson, T.J. Albert, A.S.
(2007) 3040–3041. Hilibrand, R.S. Brower, M.F. Kurd, A. Appannagari, M. Patel, J.S. Fischgrund, The
[149] M.J.B. Wissink, R. Beernink, J.S. Pieper, A.A. Poot, G.H.M. Engbers, T. Beugeling, safety and efficacy of OP-1 (rhBMP-7) as a replacement for iliac crest autograft
W.G. van Aken, J. Feijen, Binding and release of basic fibroblast growth factor for posterolateral lumbar arthrodesis: minimum 4-year follow-up of a pilot
from heparinized collagen matrices, Biomaterials 22 (2001) 2291–2299. study, Spine J. 8 (2008) 457–465.
[150] M.A. Princz, H. Sheardown, Heparin-modified dendrimer cross-linked collagen [173] R.J. Laham, F.W. Sellke, E.R. Edelman, J.D. Pearlman, J.A. Ware, D.L. Brown, J.P.
matrices for the delivery of basic fibroblast growth factor (FGF-2), J. Biomater. Gold, M. Simons, Local perivascular delivery of basic fibroblast growth factor
Sci. Polym. Ed. 19 (2008) 1201–1218. in patients undergoing coronary bypass surgery: results of a phase I random-
[151] M. Ohta, Y. Suzuki, H. Chou, N. Ishikawa, S. Suzuki, M. Tanihara, Y. Suzuki, Y. ized, double-blind, placebo-controlled trial, Circulation 100 (1999) 1865–1871.
Mizushima, M. Dezawa, C. Ide, Novel heparin/alginate gel combined with basic [174] M. Ruel, R.J. Laham, J.A. Parker, M.J. Post, J.A. Ware, M. Simons, F.W. Sellke, Long-
fibroblast growth factor promotes nerve regeneration in rat sciatic nerve, J. term effects of surgical angiogenic therapy with fibroblast growth factor 2 pro-
Biomed. Mater. Res. A 71A (2004) 661–668. tein, J. Thorac. Cardiovasc. Surg. 124 (2002) 28–34.

Potrebbero piacerti anche