Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Biomaterials View Article Online

Science
View Journal | View Issue

www.rsc.org/biomaterialsscience Volume 1 | Number 1 | January 2013 | Pages 1–100


Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G
Downloaded on 26 March 2013

ISSN 2047-4830

REVIEW ARTICLE
María Vallet-Regí et al.
A tissue engineering approach based on the use of bioceramics for bone repair
View Article Online

Biomaterials
Science
REVIEW

A tissue engineering approach based on the use of


Cite this: Biomater. Sci., 2013, 1, 40 bioceramics for bone repair
Antonio J. Salinas,a,b Pedro Esbritc,d and María Vallet-Regí*a,b
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

Biomimetics takes advantage of natural strategies for the solution of technological problems, including
the proper design of biomaterials. Living bone exhibits a hierarchical porosity with both giant and nano-
metric pores which must be reproduced for the design of biomaterials for hard tissue repair. Bioactive
and degradable bioceramics are a good alternative for the manufacture of scaffolds. Tissue engineering
approaches to improve bone regeneration include strategies supporting endogenous osteoblast
adhesion, proliferation (osteoconduction), osteoinduction by growth factors, and osteoprogenitors.
Understanding the natural ossification mechanisms and the role of biomolecules involved in this process
is a requirement for the design of bone tissue scaffolds. Mesoporous bioactive ceramics, namely meso-
Received 15th June 2012,
porous silica and templated glasses with nanometric pores to host growth factors, conformed into 3D
Accepted 17th July 2012
Downloaded on 26 March 2013

scaffolds with micrometric porosity by rapid prototyping, are a good option for bone regeneration. In
DOI: 10.1039/c2bm00071g
this regard, biomolecules such as well characterized bone morphogenetic proteins and others under
www.rsc.org/biomaterialsscience current research, such as osteostatin and osteoprogenitors, are promising strategies in bone tissue
engineering applications. Future developments in biomaterials will
a come in both micro- and nano- scales, and molecular and cell
Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia,
Universidad Complutense de Madrid, Madrid, Spain. E-mail: vallet@farm.ucm.es; biology approaches will provide suitable solutions to the demanding
Tel: +34 91 394 1861; Fax: +34 91 394 1786 needs of these compounds.
b
Networking Research Center on Bioengineering, Biomaterials and Nanomedicine
(CIBER-BBN), Madrid, Spain
c
Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria
(IIS)-Fundación Jiménez Díaz, Madrid, Spain
d
Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad
(RETICEF), Instituto de Salud Carlos III, Madrid, Spain

Antonio Salinas graduated in Pedro Esbrit graduated in Chem-


Chemistry at the Universidad istry at Universidad Complu-
Complutense de Madrid (UCM) tense, and received his Ph.D. in
and PhD at the same university Biochemistry from Universidad
in 1992. He is Associate Pro- Autónoma, in Madrid, Spain. He
fessor of Inorganic Chemistry was a post-doc Fulbright fellow
and member of the Biomedical at Barnes and Jewish Hospitals
Research Networking Center on (St. Louis, MO), and visiting
Bioengineering, Biomaterials researcher at Veterans Adminis-
and Nanomedicine, CIBER-BBN. tration Hospital (West Haven,
He was visiting Professor at the CT). He currently holds an
Universities of Florida, Aveiro Associate Chief position as Head
Antonio J. Salinas and Paris. Dr. Salinas has pub- Pedro Esbrit of Bone and Mineral Metabolism
lished more than 100 articles Laboratory at Instituto de Inves-
and books chapters most of them in the field of bioceramics and tigación Sanitaria-Fundación Jiménez Díaz, Madrid. Dr. Esbrit
participated in 22 research projects. He has more than 1,500 has published >100 papers and several books in the field of bone
world-wide citations (ISI Web of Knowledge). Dr. Salinas is Corre- and mineral metabolism, particularly focusing on the actions of
sponding Academician of the Spanish Royal National Academy of PTHrP, in which he is one of the 10-top experts according to
Pharmacy. BiomedExperts.

40 | Biomater. Sci., 2013, 1, 40–51 This journal is © The Royal Society of Chemistry 2013
View Article Online

Biomaterials Science Review

1. State of the art: the process of bone of-the-art biomaterials are being developed as bone tissue
engineering strategies to improve bone repair.
regeneration and repair. How nature works
Bone regeneration after a fracture and the healing of small 1.1 Bone: a model to imitate
bone defects are very effective processes. However, a challenge Nature is an important source of inspiration in the develop-
arises with an increasing number of fractures associated with ment of new implant materials. In this sense, the design of
poor healing through various causes such as osteoporosis, these biomaterials relies on imitating the structure and func-
fractures involving bone sites of poor vascularity or large bone tion of biological systems (see Fig. 1). The biomimetic
tissue loss.1 In this regard, the number of fractures related to approach is based on the fact that after billions of years of
osteoporosis has doubled in the last decade, in part related to evolution, nature has learned which systems are the most
the significant increase in life span in Western societies.2 appropriate in terms of optimal properties with minimal
Osteoporosis occurs frequently in women after menopause resources.9,10 Thus, biomimetics take advantage of these
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

due to the loss of estrogen, but bone loss is also known to natural strategies for the solution of specific problems in all
occur for both men and women with aging. In fact, the branches of materials science and engineering.
increasing number of “non-union” fractures in the aging popu- Bone is formed by a series of biomineralization processes, a
lation is a significant clinical problem. A number of other situ- sequence of physicochemical reactions that give rise to
ations related to osteopenia/osteoporosis, namely diabetes organic–inorganic nanocomposites with exceptional mechan-
mellitus and glucocorticoid treatment, are known to affect ical properties that would be impossible to reach with pure
optimal skeletal healing after trauma or osteotomy.3–8 This materials.10 This tissue is made up of a cellular component
represents a major challenge to our health systems, and deter- and an extracellular matrix consisting of an organic phase,
mines that bone regeneration following maxillo-facial and mainly type 1 collagen, and an inorganic ceramic phase of
orthopaedic surgery is an important clinical issue in regenera- calcium-deficient carbonated hydroxyapatite (CHA) nanocrys-
Downloaded on 26 March 2013

tive medicine. Therefore, therapeutic strategies to improve tals.11 As observed in Fig. 2, the main characteristics of biologi-
bone healing in these circumstances are of utmost impor- cal apatites are: variable composition, nanometric crystal size,
tance. Therapies derived from a better knowledge of the cellu- calcium deficiency, the presence of carbonate groups and
lar and molecular events in bone healing together with state- structural disorder. Another important feature of the apatite
structure is the ability to accommodate different ions in its
three ionic sublattices (calcium, phosphate and hydroxy).11
These apatite nanocrystals grow at ordered mineralization
sites of collagen molecules in bone.
María Vallet-Regí was born in More than 200 bone pieces with different lengths and
Las Palmas, Spain. She studied shapes constitute the human skeleton.12 However, all of them
Chemistry at the Universidad present a hierarchical structure that ranges from the collagen-
Complutense de Madrid (Spain) CHA nanocomposite, the lamellae, lacunae and osteons to the
and received her PhD at the macroscopic material. Another important characteristic of
same university in 1974. She is bone is its porosity. In this sense, bone tissue with less than
full professor of Inorganic Chem- 20% porosity is known as cortical (or compact) bone, whose
istry and head of the research most important function is to provide mechanical support
group Smart Biomaterials in the
Department of Inorganic and
Bioinorganic Chemistry of the
Faculty of Pharmacy at Universi-
María Vallet-Regí dad Complutense de Madrid.
Prof. Vallet-Regí has written
more than 600 articles and several books. She was the most cited
Spanish scientist according to ISI Web of Knowledge, in the field
of Materials Science in these past decades. She is Fellow of Bioma-
terials Science and Engineering at the International College of
Fellows of Biomaterials Science and Engineering (ICF-BSE), Num-
bered Fellow of the Spanish Royal Academy of Engineering and the
Royal National Academy of Pharmacy. She has received the Prix
Franco-Espagnol 2000 from Societé Française de Chimie, the
Spanish Royal Society of Chemistry (RSEQ) award in Inorganic
Chemistry 2008, the Spanish National Research Award in Engin-
eering 2008, FEIQUE Research award 2011 and the RSEQ Fig. 1 Nature is an ideal inspiration to solve technological problems associated
Research Award and Gold Medal 2011. with implant materials.

This journal is © The Royal Society of Chemistry 2013 Biomater. Sci., 2013, 1, 40–51 | 41
View Article Online

Review Biomaterials Science


Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

Fig. 2 Top: Schematic image of CHA nanocrystals reinforcing the triple helix of Fig. 3 Economic impact of different products in US Orthopedic Biomaterials
collagen. Middle: Cancellous bone observed by scanning electron microscopy market in 2009 (Adapted from ref. 15). “Others” category includes the big
and CHA nanocrystals by transmission electron microscopy. Bottom: Main fea- markets for sealants, hemostats and anti-adhesion agents.
tures of biological apatites.

influence of mechanical stimuli that greatly influence bone


(strength and stiffness). On the other hand, cancellous (trabecu- healing in adults.8 At the bone injury site, mesenchymal cells
Downloaded on 26 March 2013

lar or spongy) bone can contain up to 90% of porosity. This type interact with inflammatory cells in an orderly fashion by yet
of bone provides a large surface area which determines the key poorly understood mechanisms.16 Recent evidence also
role of the skeleton in ion homeostasis and haematopoiesis. Fur- suggests that osteoclasts as well as osteoprogenitors might
thermore, pores with different sizes, from one to several hundred have an active role in the early response to injury, as occurs in
micrometers are present in bone; those below 1 μm in diameter the bone remodeling units in the bone surface.17,18
New bone formation is initiated by the condensation of
are responsible for bioactivities involving protein interactions.
This hierarchical porosity must be reproduced in the design of mesenchymal stem cells (MSCs), which then leads to either
new biomaterials for regeneration and repair of hard tissues.13 the formation of a cartilage template in the bone marrow
(endochondral ossification) or direct differentiation into osteo-
There is general agreement on both scaffolds required for
osteoconduction and osteogenic growth factors as key elements blasts at the periostium (intramembranous or appositional ossi-
for applications in bone repair and regeneration.14 The increas- fication). The former ossification mechanism predominates in
most cases of fracture healing, but rigid fixation of the injury
ing importance of the latter factors, mainly bone morphogenic
proteins and other growth factors, is illustrated in Fig. 3 causes primary bone apposition as the major repair mechan-
showing the participation of different products in the US Ortho- ism.8,19 The general pattern of endochondral ossification after
fracture includes several chronological phases (Fig. 4).
pedic Market in 2009.15 These agents represent 20% of this
global market, with an expected growth of 30–35% each year in Immediately following trauma, vascular endothelium integ-
the next five years. This growth is 3-fold higher than that rity is lost, giving rise to disruption of the blood supply and
expected for the US global market for orthopedic biomaterials. hematoma formation. This is accompanied by an inflamma-
tory response, related to the production of pro-inflammatory
cytokines—namely tumour necrosis factor-α, interleukin-1
1.2 Cell and molecular mechanisms underlying the process (IL-1), and IL-6—from aggregated platelets, which have chemo-
of bone healing tactic activity towards lymphocytes, monocytes-macrophages
Bone repair—a key process for resolution of orthopedic and fibroblasts (inflammatory cells), and also endothelial
trauma which causes bone disjunction—is a unique process cells.20 Macrophages can also produce and secrete the afore-
involving the interplay of complex cellular and molecular mentioned cytokines and various growth factors, platelet-
events to generate new bone instead of a fibrous scar which is derived growth factor, fibroblast growth factors, insulin-like
the final outcome in other connective tissues. In contrast to growth factors, the transforming growth factor-β superfamily,
bone remodeling in adults, which requires a relatively long including bone morphogenetic proteins (BMPs) and vascular
period to complete at random sites (called “bone remodeling endotelial growth factor (VEGF), which induce angiogenesis and
units”) throughout the skeleton, bone repair occurs in a com- bone formation from recruited periosteal progenitors, as well as
pressed time frame and in a precise location. The latter basi- extracellular matrix synthesis.21–23
cally recapitulates normal bone formation during Neovascularization of injured tissue is key to successful
embryogenesis, except for some events such as inflammation, bone repair, providing oxygen, facilitating metabolic waste
the limited number of osteoprogenitors available and the key removal, and delivering progenitor cells of haematopoietic

42 | Biomater. Sci., 2013, 1, 40–51 This journal is © The Royal Society of Chemistry 2013
View Article Online

Biomaterials Science Review

The proliferation and differentiation of osteoblasts from


pluripotent MSCs is a complex process under systemic and
local control. Many of the regulatory factors underlying this
control constitute a matter of investigation as putative targets
or therapies to stimulate bone repair.8,31 Of interest, these
factors appear to act through common intracellular signaling
pathways—namely mitogen-activated protein kinase (MAPK)
and Wnt/β-catenin pathways8—to induce proliferation of osteo-
progenitors and activation of Runx2, a key transcription factor
for osteoblast differentiation.32 The fact that MAPK signaling
acts as an important mediator of pro-inflammatory cytokines
determines that its therapeutic manipulation might be
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

restrained to the early phase in bone repair, which involves


inflammation.33 The canonical Wnt/β-catenin pathway has an
important role in bone formation, and recently, its impact on
bone regeneration has also been examined in mice. Thus, sys-
Fig. 4 Phases of endochondral ossification in fracture healing. The well micro-
vascularized periostium is shown, consisting of an outer fibrous layer and the
temic administration of an adenovirus expressing an inhibitor
inner cambium which is rich in osteoprogenitors. Immediately after injury, of this pathway into transgenic mice was found to arrest the
hematoma formation and inflammation take place. Osteoprogenitors are then program of tissue regeneration in the mouse tibia.34 On the
recruited from the periosteum to differentiate into chondrocytes or osteoblastic other hand, it has been suggested that pre-treatment of bone
cells (depending on the oxygen supply). New bone is thus starting to be formed
marrow cells with Wnt/β-catenin pathway activators may
at the borders of the injury site. Simultaneously, a callus mostly made up of
hypertrophic cartilage develops and begins to revascularize. Eventually, osteo-
increase their osteogenic capacity;35 a particularly relevant
Downloaded on 26 March 2013

clast-mediated remodeling of the newly formed bone leads to the restoration of manoeuvre in conditions associated with a low number of
structural bone integrity (modified from the Primer on the Metabolic Bone Dis- osteoprogenitors (e.g., aging and diabetes mellitus).
eases and Disorders of Mineral Metabolism, 7th Edition. www.asbmrprimer. A reduction in the number of osteoprogenitors as well as a
org).
decreased proliferative and differentiation capacity of pre-
osteoblasts to osteoblasts, associated with an overall remodel-
ing capacity decline, correlates with old age.36–38 Alterations in
origin.8,24 Whereas mechanical stability preserves vascular osteoblast function related to aging might be accounted for at
integrity and promotes osteogenesis, instability disrupts micro- least in part by increased oxidative stress and inflammation
vessels producing hypoxic conditions that favor chondrogenesis, status.39,40 Several recent studies have explored the putative
as occurs in the central avascular region of the callus.25 This age-related alterations in the temporal pattern of gene
determines that stabilized fractures have less cartilage and a expression during bone regeneration in rats and humans.18,41
reduced callus volume. VEGF, which is expressed under the In old rats, the machinery of bone healing based on the gene
control of hypoxia-inducible factor (HIF)-1α under low oxygen profile seems to be conserved with age despite the delay in
tension, is a prototype osteogenic–angiogenic factor that can new bone formation to bridge the fracture gap.41 In the callus
expand the osteoprogenitor pool by interaction with the endo- after hip fracture in elder human subjects, the expression of
thelium.26 Osteogenic factors such as BMPs and parathyroid inflammation-related genes is highest at the earlier phase, and
hormone (PTH)-related protein (PTHrP) stimulate the expression it shifts towards bone remodeling genes later on after frac-
of VEGF by osteoblastic cells.27,28 The critical role of this factor ture.18 Interestingly, expression of Sost—the sclerostin-encod-
in bone repair has been well exemplified in mice with con- ing gene, a key modulator of bone remodeling—was rapidly
ditional overexpression of HIF1α in mature osteoblasts (using reduced in the callus, suggesting that this would allow osteo-
the osteocalcin promoter) undergoing distraction osteogenesis blasts to escape from its inhibitory effect to promote bone for-
to stimulate bone regeneration, showing a robust angiogenic mation.18 These observations provide new insights for specific
response closely related to new bone formation.1 The key final interventions targeting excessive inflammation and Sost to
step to correct the bone defect consists of newly formed woven promote bone regeneration in elder subjects.42,43
(or disorganized) bone replacement by lamellar bone through
the activity of osteoclasts and osteoblasts (bone remodeling).
Current evidence also points to the role of osteoclastogenesis
and osteoclast-mediated bone resorption during both early and 2. Bioceramics to promote bone
late stages of bone repair. Osteoprogenitors may activate osteo- regeneration. From the raw materials to
clast-mediated resorption of damaged bone, while osteoclasts scaffolds
themselves can modulate osteoblast activity and thus contribute
to bone formation.19,29 This evidence can explain the reported Synthetic materials used for bone repair can be divided into:
deleterious effects of bone resorption inhibitors, namely ceramics, polymers, metals and composites where at least two
bisphosphonates, on callus formation and remodeling.18,30 classes of materials are combined together.44 Ceramics have a

This journal is © The Royal Society of Chemistry 2013 Biomater. Sci., 2013, 1, 40–51 | 43
View Article Online

Review Biomaterials Science

structure which mimics that of natural bone and thus were


considered attractive materials in this respect.45,46 This cat-
egory of materials includes oxides, phosphates, carbonates,
nitrides, carbides, carbons and glasses. Those designed to
interact with living tissues are called bioceramics,46,47 and
they are the subject of this review.
The so-called bioinert bioceramics elicit a foreign body
reaction which gives rise to a non-adherent fibrous capsule iso-
lated from surrounding bone, which accounts for the aseptic
loss of many implants.48 For this reason, research efforts at
the end of the past century were devoted to biodegradable and
bioactive bioceramics, sometimes denoted as second gener-
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

ation bioceramics. Biodegradable bioceramics are designed to


fulfil specific functions for a given time period, helping in the
self-repair processes of the living organism, and are sub-
sequently resorbed. The critical aspect in the design of these
bioceramics is their relatively fast degradation rate, with the Fig. 5 Main bioceramics classified as a function of its reactivity with living
loss of their mechanical properties, compared to the slower tissues.

new bone formation process. Bioactive bioceramics can react


with physiological fluids, but only at surface level, giving rise
to a sequence of surface reactions producing a strong bond silicon in the structure of bioactive bioceramics appears to
between the material and bone.47 Several calcium phosphates confer them osteoinduction features.49 However, a major draw-
Downloaded on 26 March 2013

such as tricalcium phosphates (alpha or beta polymorphs) or back of calcium phosphates and silica-based bioceramics is
calcium sulphates are good examples of biodegradable their poor mechanical strength, namely brittleness and low
bioceramics. Likewise, well known bioactive ceramics are resistance to fatigue. They exhibit excellent features for filling
hydroxyapatite (HA) and certain compositions of glasses and small bone defects, but their brittleness is a drawback for
glass-ceramics. Bioactive and biodegradable bioceramics are repairing large bone defects. This is especially so in highly
clinically used as bone fillers, bone cements or for coating porous bioceramics and scaffolds with high porosity percen-
metallic implants.47 Fig. 5 shows representative bioceramics of tages and pores larger than 100 μm in diameter. Consequently,
each category. Because the in vivo reactivity of these materials for bone substitution and repair, these bioceramics, are
depends on its surface area, one single material such as HA mainly used as fillers in non-load bearing applications or as
can be classified in different categories depending on its par- coatings of metallic prostheses, but also can be proposed for
ticle size. the manufacture of scaffolds, as will be described in the fol-
Present research in bioceramics is based mainly on biologi- lowing section.
cal criteria since it pursues bone tissue “regeneration” rather Next, the major features of some calcium phosphates
than its mere “replacement”. Thus, current efforts are devoted including synthetic apatites, biphasic mixtures, bone cements
towards the synthesis of bioceramics—so-called third gener- and coatings, and silica-based materials including bioglasses,
ation bioceramics49—which are able to induce a cell response mesoporous ordered silica-based materials and templated
leading to osteogenesis.50 In this regard, research has mainly glasses will be described. Of all the synthetic apatites, HA
focused on porous resorbable or bioactive bioceramics that are [Ca10(PO4)6(OH)2], is the most widely studied for its structural
used to manufacture porous scaffolds able to host cells and and chemical similarities with the inorganic component of
bioactive agents (growth factors, hormones, peptides). The bone.54,55 HA is biocompatible, bioactive and osteoconductive.
signals triggered by these biomolecules should promote bone To mimic biological apatites, synthetic apatites must present
formation. Moreover, advanced bioceramics such as ordered nanometric particle size and contain 4–8 wt% of CO32−
mesoporous silica-based materials or organically modified bio- ions.56,57 HA structure can accept the inclusion of many ions,
ceramics are now under investigation in this respect.51–53 such as Na+, K+, Mg2+, Sr2+, Cl−, F−, HPO42−, etc.57 This can be
exploited in the design of HA with specific surface structure
2.1 Second generation bioceramics and electric charge in order to improve the material behaviour
Most widely used second generation bioceramics are calcium in biological environments. In addition, the presence of
salts—phosphates and sulphates—and silica (SiO2)-based bio- certain elements (i.e., strontium, zinc or silicate) that can be
ceramics which are extensively used in orthopaedics and den- released in the regenerating bone environment might facilitate
tistry because of their biocompatibility and capacity to the process of bone healing. Carbonate and strontium ions
promote osteointegration. Many of these bioceramics are also also facilitate apatite dissolution and implant resorption. Sili-
osteoconductive, meaning their ability to provide the appropri- cate ions increase the mechanical strength of apatite and
ate surface to support osteoblast adhesion and proliferation accelerate its bioactive response. Thus, the synthesis of substi-
and therefore, bone formation.50 Furthermore, the presence of tuted apatites is a current research trend.

44 | Biomater. Sci., 2013, 1, 40–51 This journal is © The Royal Society of Chemistry 2013
View Article Online

Biomaterials Science Review

The most popular biphasic mixtures of calcium phosphates


are based on HA and β-tricalcium phosphate, β-Ca3(PO4)2,
(β-TCP) mixtures that evolve to CHA under physiological con-
ditions.58 The dissolution of resorbable β-TCP acts as a source
for new bone by the increase of Ca2+ and PO43− ion concen-
tration in the local environment, simultaneously creating extra
porosity that favours a suitable in vivo behaviour of the
material. This biphasic material can be injected, used as a
coating or as bulk in bone replacement. Other biphasic mix-
tures are under investigation including calcium phosphates,
bioactive glasses, and calcium sulphates, etc.
Bone cements based on calcium salts, mainly phosphates,
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

carbonates or sulphates, present excellent biocompatibility


and bone-repair properties.59,60 Most of the injectable calcium
phosphate-based cements evolve to calcium phosphate apatite
during the setting reaction. These cements harden in situ and
can be slowly resorbed. During this gradual process, the bone
grows and replaces the cement. Research is under way to
Fig. 6 Different uses of calcium phosphate bioceramics.
shorten the curing time and improve the mechanical
toughness.
Coatings of calcium phosphates over metallic substrates engineering as 3D scaffolds in association with osteogenic
have been used to overcome the main limitation of calcium agents, as will be detailed later. These bioactive matrices
Downloaded on 26 March 2013

phosphates as bioceramics: their poor mechanical properties. present osteoconduction and osteoinduction.
Common substrates are titanium alloys, Ti6Al4V and commer- In recent years, silica-based ordered mesoporous biocera-
cially pure Ti. Calcium phosphate coatings are usually pro- mics attracted interest as templates for drug delivery appli-
duced by plasma spray because of the rapid deposition rate at cations.68 These mesoporous materials have the same
a relatively low cost.61,62 However, this method presents several chemical composition, namely silica, and are denoted by
drawbacks: (i) the presence of highly soluble amorphous different acronyms, e.g., MCM-41, SBA-15, MCM-48, among
calcium phosphate in the coatings produce resorption pro- others. Mesoporous silica-based bioceramics exhibit a high
blems; (ii) the poor adherence of the bioceramic-substrate; (iii) surface area (ca. 1000 m2 g−1), high pore volume (ca. 1 cm3
the instability of HA at high temperatures; and (iv) the phase g−1), narrow pore size distribution in the mesopore region
transition of titanium at high temperatures. Nowadays, (2–50 nm), and a pore channel system homogeneously orga-
alternative techniques to obtain calcium phosphate coatings nized in 2D or 3D arrangements. The surface of these
are under investigation including physical vapor deposition, materials covered with silanol groups should govern
chemical vapor deposition, magnetron sputtering, electrophor- their interaction with the host tissue, although the kinetics of
etic deposition, pulsed laser deposition and dip coating. Some the nanoapatite layer formation is slow.69 Therefore, in the last
of these techniques permit higher control of the coating thick- few years a great interest was devoted towards the so-called
ness and crystallinity or can take place at low temperatures, an templated glasses,70 which exhibit analogous textural proper-
interesting advantage compared with plasma spray. In Fig. 6, ties to mesoporous materials but a greater bioactivity response
different uses of calcium phosphate bioceramics are that made them very attractive in bone regeneration
presented. technologies.
For silica-containing bioceramics, bioglasses were the first
synthetic materials for which bioactivity was described.63 They 2.2 3D bioceramic scaffolds for bone tissue engineering
contain silica as the network former together with other met- In bone tissue engineering, synthetic scaffolds should fulfil
allic oxides as modifiers. Among them, Bioglass® 45S5 (45% various requirements—they must provide mechanical stability
SiO2 24.5% NaO2, 24.5% CaO and 6% P2O5), obtained by tra- as well as an appropriate environment for new bone formation;
ditional “quenching-of-a-melt” method, is the best character- e.g., they must favour cell attachment as well as cell growth
ized and used bioceramic of this group.63 and differentiation.71–75 Bioceramics can be manufactured as
In the 1990s, the synthesis of porous bioactive bioglasses by scaffolds in this regard. The in vivo behaviour of such scaffolds
the sol–gel method began.64–67 This technique requires lower depends on both the intrinsic properties of the bioceramic
temperatures, which facilitates the possible incorporation of and specific features resulting from their fabrication
biologically active molecules or living cells into the bioglasses. method.76–78
These bioglasses exhibited high surface areas and porosity as One advantage of bioceramic scaffolds lies in the fact that
well as an abundance of surface silanol (Si–OH) groups, which they usually have hierarchical porosity with highly intercon-
greatly improves their bioactive response. Nowadays, the most nected pores analogous to bone. Pores can be classified in
promising application of bioactive bioglasses is in bone tissue three categories depending on the role they play. Those with

This journal is © The Royal Society of Chemistry 2013 Biomater. Sci., 2013, 1, 40–51 | 45
View Article Online

Review Biomaterials Science

diameters in the range 150 to 400 μm and larger are needed


for cell colonization and vascularisation.79 Another group of
pores under 20 μm is necessary for capillary ingrowths and
cell-matrix interactions. In the case of scaffolds made of meso-
porous ordered silica-based bioceramics or templated glasses,
pores of a few nanometers are present, which allow the
inclusion of osteogenic factors to decorate the scaffold. This
confers a very important added value to the resulting scaffold:
the capacity of hosting drugs for locally treating different bone
morbidities and pathologies, such as bone infection, osteo-
porosis or cancer. The scaffold processing method should thus
preserve the original mesoporosity combined with macropor-
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

osity. Fig. 7 shows the different pore sizes required for several
physiological functions of bone tissue.
Several conformation methods are available to obtain
scaffolds at room temperature, a manoeuvre that permits the
Fig. 8 Bone regeneration and local drug delivery by mesoporous-based 3D
inclusion of different biomolecules in the material. An appro- scaffolds.
priate and useful method to produce scaffolds as bone
implants is rapid prototyping (RP) 3D printing,80,81 which
makes possible the design of the correct scaffold structure
the loading solution (including pH) are all parameters that
based on tomography information of the bone tissue to repair.
highly affect the adsorption capacity of the bioceramics
Then, with a computer-assisted design, suitable scaffolds with
scaffolds.83 However, this capacity can be improved by pre-
Downloaded on 26 March 2013

the appropriate shape and porosity can be obtained for


vious chemical functionalization of scaffold mesopores with
different clinical requirements. Generally, biodegradable or
different organic groups tailored to interact with a particular
bioactive bioceramics are used for this purpose; a bioceramic
factor of interest.53 This approach allows fine control over the
paste is prepared to load in the injector and to mould the
drug load and a predictable release pattern.83,84
scaffold with a previously designed macroporosity. A combi-
The efficacy of local administration of osteoinductive
nation of different methods, such as sol–gel chemistry, double
agents, e.g., peptides and growth factors, may be improved by
polymer templating and RP, has lead to 3D mesoporous bio-
their covalent grafting to the scaffolds, which determines a
glass-based scaffolds exhibiting hierarchical pore networks,
sustained exposure in the bone microenvironment.85,86 These
with giant (30–1000 μm), macro- (10–30 μm) and meso- (5 nm)
osteoinductive agents would act as attracting signals for bone
porosity.
cells.
An attractive strategy in bone tissue engineering has been
the incorporation of osteogenic factors with the aim to
improve the osteoinductive properties of the scaffolds (Fig. 8).
Incorporation of these factors relies on conventional impreg- 3. Symbiosis between nature and materials
nation techniques using manufactured scaffolds.82 The type of
pore and exposed (inner and outer) surface, and the quality of A traditional approach in tissue engineering strategies involves
the implantation of different types of osteogenic cells seeded
onto suitable templates, including bioceramics. Cells of the
osteoblastic lineage with different differentiation status can be
used to drive bone tissue regeneration.87,88 In addition,
different sources of osteoprogenitors have been tested for their
capacity to regenerate bone (reviewed in ref. 31 and 89). MSCs
—defined as adherent multipotent cells capable of differentiat-
ing into osteogenic cells90—have distinctive and interesting
features such as immunosuppressive, cell protective, and
angiogenic activities.91 Current data support the notion that
MSCs are indeed pericytes residing on blood vessels; in this
context, an insult causing vasculature disruption would make
these cells available to help regenerate the resulting damaged
tissue.92,93 MSCs are relatively abundant in the bone marrow
from which they can be easily isolated and expanded in cul-
tures for use in different bone tissue engineering schemes.
Fig. 7 Porosity features involved in the design of scaffolds for bone tissue The major drawback of MSCs from various origins, however,
engineering. lies in their limited quantity and/or differentiation

46 | Biomater. Sci., 2013, 1, 40–51 This journal is © The Royal Society of Chemistry 2013
View Article Online

Biomaterials Science Review

capacity;94,95 these hurdles are presently a subject of intense


investigation through different approaches.96,97 In addition,
elucidating the true role of the host contribution to bone
tissue regeneration is paramount to optimize tissue engineer-
ing approaches. Of interest in this line, MSCs and more com-
mitted cells of the osteoblast lineage appear to have a different
capability to recruit host endothelial cells and, therefore, to
increase vascularization.93,97
Osteopromotion can be achieved by using factors which
enhance the healing process. Thus, a common strategy to
promote bone healing relies on the use of osteogenic factors
which are either directly implanted as recombinant proteins or
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

embedded in a suitable carrier such as bioceramics previous


to implantation.98,99 Osteoinduction properties are conferred
Fig. 9 A comparison between bioactivities of osteostatin and BMP-2.
to these biomaterials by growth factors which can induce
differentiation of MSCs along the bone repair process.31
BMP-2 and BMP-7 in particular, have been widely used in
animal models and also in randomized clinical trials for were highly osteoconductive and osteoinductive by promoting
healing fractures and stimulating spine fusion. BMP-2 has infiltration of proliferating osteogenic precursor cells and con-
proven essential for MSCs recruitment and expansion; it is the nective tissue formation (Fig. 10). In addition, these biocera-
only United States Food and Drug Administration (FDA)- mics induced revascularization of the defect, related to an
approved molecular therapy for use in the management of increased immunostaining for VEGF in the healing bone
fracture repair and spine non unions.100 In fact, BMP-2 rep- tissue.104 More recently, these biomaterials were also found to
Downloaded on 26 March 2013

resented 18% of the US orthopaedic biomaterials market in improve the early stage of bone healing in the same bone frac-
2009 (See Fig. 3). However, some concerns have recently been ture model in osteoporotic rabbits.108 Thus, these osteostatin-
raised due to the poor outcomes of BMP-2 administration in loaded bioceramics appear to be an attractive approach for
some cases.101,102 bone tissue engineering applications even in the setting of
Recently, our group has started to explore the advantage of osteopenia.
the small peptide osteostatin, consisting of the 107–111
domain (Thr-Arg-Ser-Ala-Trp) of PTHrP, to promote bone
healing. Fig. 9 shows the comparison of some biological 4. Future perspectives
characteristics of osteostatin and BMP-2. As can be seen, local
application of osteostatin is expected to bring significant Second generation bioceramics including those bioactive and
advantages with respect to BMP-2. We have recently demon- resorbable bioceramics are being used today as implants for
strated that osteostatin loading onto silica-based ceramics con- bone regeneration and repair in Orthopedics and Dental appli-
ferred osteogenic features to these materials, including cations. Third generation bioceramics, those driving bone
stimulation of osteoblastic cell growth and differen- regeneration in various bone tissue engineering schemes, are
tiation.103,104 Moreover, osteostatin has recently been proven to now under intense research in the hope of significant develop-
increase the osteogenic efficacy of fibroblast growth factor-2 as ments in the near future.
coated onto Si-doped hydroxyapatite by enhancing its angio- A representative picture of the current research interests in
genic potential in vitro.105 These data are consistent with the biomaterials can be obtained from the program of the Nine
recently described anabolic features of the native peptide, World Biomaterials Congress that took place in Chengdu
PTHrP107–139, independent of its concomitant inhibitory effect (China) from the 1st to the 5th of June 2012. The central topic
on bone resorption.106,107 Attractive properties of osteostatin of the Congress was: “Innovative Biomaterials and Crossing
lie in the fact that it can affect bone cells at <nM concen- Frontiers in Biomaterials and Regenerative Medicine”. Over
trations and its structure is not likely to be degraded by pro- 3,000 communications were classified in 87 Symposia that
teolysis. This prompted us to further investigate whether were divided into 167 scientific sessions. The main topic was
osteostatin-coated bioceramics might be able to improve “scaffolds”, which was included in 12 sessions, followed by
osteointegration and accelerate bone repair after a cavitary “cell-materials interactions”, which were discussed in 8 ses-
defect in the femoral epiphysis of healthy rabbits. This in vivo sions. In addition, the following topics were present in 6 ses-
model, in which trabecular bone damage predominates, has sions: “nanomaterials, gene delivery and hydrogels”. Other 5
been proven to be suitable for testing biomaterials. Histologi- sessions were devoted to “tissue inducing biomaterials, bio-
cal analysis revealed the absence of significant inflammation mimetics, and molecular imaging”, while four more sessions
or bone resorption within the time of study (4 and 8 weeks) dealt with “bioinorganics and bioceramics, surface modifications,
after implantation.104 At 8 weeks, microcomputerized tomogra- hybrid materials and polymer design”. These 12 topics rep-
phy (μCT) analysis showed that osteostatin-loaded biomaterials resented almost 50% of all scientific sessions of the Congress.

This journal is © The Royal Society of Chemistry 2013 Biomater. Sci., 2013, 1, 40–51 | 47
View Article Online

Review Biomaterials Science

Acknowledgements
Funding for the authors came from grants by Comisión Inter-
ministerial de Ciencia y Tecnología (CICYT, Spain) (MAT2008-
736 and CSO2010-11384-E), Comunidad Autónoma de Madrid
(CAM; S2009/MAT-1472), Instituto de Salud Carlos III
(RETICEF RD06/0013/1002) and Fundación de Investigación
Médica Mutua Madrileña.
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

References
1 C. Wann, S. R. Gilbert, Y. Wang, X. Cao, X. Shen,
G. Ramaswamy, K. A. Jacobsen, Z. S. Alaq, A. W. Eberhard,
Fig. 10 3D reconstruction microtomography images of mesoporous silica bio-
ceramics with SBA 15 structure, unloaded and loaded with osteostatin before
L. C. Gerstenfeld, T. A. Einhorn, L. Deng and
being implanted for 8 weeks in a cavitary defect model performed in the rabbit T. L. Clemens, Proc. Natl. Acad. Sci. U. S. A., 2008, 105,
femur. 686.
2 The bone and joint decade. www.boneandjointdecade.org.
2005.
3 R. Gruber, H. Koch, B. A. Doll, F. Tegtmeier, T. A. Einhorn
Downloaded on 26 March 2013

No doubt that they will be major topics in the field of Bio- and J. O. Hollinger, Exp. Gerontol., 2006, 41, 1080.
materials in the next years. But the question arises as to how 4 P. Peichl, L. A. Holzer, R. Maier and G. Holzer, J. Bone
biomaterials research will evolve? Interesting advances in this Joint Surg. Am., 2011, 93, 1583.
regard were presented in specific sessions such as “growth 5 E. C. Gaetti-Jardim, J. F. Santiago-Junior, M. C. Goiato,
factor delivery” or “bone tissue engineering”. E. P. Pellizer, O. Magro-Filho and E. G. Jardim Jr, J. Cranio-
Regarding the growth factor delivery topic, various comp- fac. Surg., 2011, 22, 1111.
lementary approaches are now envisioned to promote bone 6 L. Fernández de Castro, D. Lozano, S. Dapía, S. Portal-
healing. As a promising approach, the off-label use of PTH1–34 Núñez, J. R. Caeiro, E. Gómez-Barrena and P. Esbrit,
as intermittent subcutaneous injection—an approved anabolic Tissue Eng. Part A, 2010, 16, 1157.
therapy in osteoporotic patients109—has recently been pro- 7 A. Gandhi, H. A. Beam, J. P. O’Connor, J. R. Parsons and
posed to promote bone regeneration.110 This type of adminis- S. S. Lin, Bone, 2005, 37, 482.
tration of PTH1–34 increased the rate of callus formation and 8 F. Deschaseaux, L. Sensébé and D. Heymann, Trends Mol.
its mechanical properties.110-112 The use of a degradable Med., 2009, 15, 417.
matrix with plasmid coding for PTH and BMP-4 as implants 9 Y. Bar-Cohen, in Biomimetics Nature-Based Innovation, ed.
has also proven to be efficient to fill a femoral gap in rats.113 A Y. Bar-Cohen, Taylor and Francis (CRC Press), Boca Raton,
pulsatile release system, consisting of PTH-loaded alginate FL, 2011. p. 14.
disks and several polyanhydride isolation layers alternately 10 L. T. Kuhn, D. J. Fink and A. H. Heuer, in Biomimetic
stacked within the device, has also been developed.114 This Materials Chemistry, ed. S. Mann, Wiley-VCH, United
biocompatible system, as well as a matrix-bound engineered Kingdom, 1996, p. 41.
active fragment of human PTH1–34, whose development is 11 M. Vallet-Regí and D. Arcos, Biomimetic Nanoceramics in
associated with matrix remodeling,115 hold promise for the Clinical Use: From Materials to Applications, Royal Society
controlled delivery of PTH locally into a bone injury site. of Chemistry, Cambridge, 2008.
Attempts have also been made to manipulate the controlled 12 D. G. Steele and C. A. Bramblett, The Anatomy and Biology
release of various osteotropic agents with variable kinetics of the Human Skeleton, Texas A&M University Press, 1988,
with the aim of achieving an optimal bone healing p. 10.
response.116 13 W. Frieb and J. Werner, in Handbook of Porous Solids, ed.
The use of superparamagnetic mesoporous silica particles F. Shüth, S. Kienneth, W. Sing and W. Kamps, Wiley-VCH,
as part of “nanogate” systems with a potential “on/off” factor Weinheim, Germany, 2000, p. 2923.
release is also under investigation.117 14 M. Schieker, H. Seitz, I. Drosse, S. Seitz and W. Mutschler,
A consistent notion throughout all the plenary lectures in Eur. J. Trauma, 2006, 32, 114.
the aforementioned Congress was that biomaterials science is 15 P. Driscoll 2009, http://mediligence.com/blog/2009/01/30/
moving into the nanometric arena. Combined with smart orthopedic-biomaterials-market-growth-strongest-in-us/
molecular and cell biology strategies, they will provide a solu- 16 L. Claes, S. Recknagel and A. Ignatius, Nat. Rev. Rheuma-
tion to many of the current orthopedic demands. tol., 2012, 8, 133.

48 | Biomater. Sci., 2013, 1, 40–51 This journal is © The Royal Society of Chemistry 2013
View Article Online

Biomaterials Science Review

17 H. Schell, J. Lienau, D. R. Epari, P. Seebeck, C. Exner, 37 M. E. Martínez, M. T. del Campo, S. Medina, M. Sánchez,
S. Muchow, H. Bragulla, N. P. Haas and G. N. Duda, Bone, M. J. Sánchez-Cabezudo, P. Esbrit, P. Martínez, I. Moreno,
2006, 38, 547. A. Rodrigo, M. V. Garcés and L. Munuera, Calcif. Tissue
18 J. Caetano-Lopes, A. Lopes, A. Rodrigues, D. Fernandes, Int., 1999, 64, 280.
I. P. Perpétuo, T. Monjardino, R. Lucas, J. Monteiro, 38 P. Martínez, I. Moreno, F. De Miguel, V. Vila, P. Esbrit and
Y. T. Konttinen, H. Canhão and J. E. Fonseca, PLoS One, M. E. Martínez, Bone, 2001, 29, 35.
2011, 6, e16947. 39 M. Almeida, L. Han, M. Martín-Millán, C. A. O’Brien and
19 F. Tortelli, T. Tasso, F. Loiacono and R. Cancedda, Bio- S. C. Manolagas, J. Biol. Chem., 2007, 282, 27298.
materials, 2010, 31, 242. 40 S. Salvioli, M. Capri, S. Valensin, P. Tieri, D. Monti,
20 T. Kon, T. J. Cho, T. Aizawa, M. Yamazaki, N. Nooh, E. Ottaviani and C. Franceschi, Curr. Pharm. Des., 2006,
D. Graves, L. C. Gerstenfeld and T. A. Einhorn, J. Bone 12, 3161.
Miner. Res., 2001, 16, 1004. 41 R. A. Meyer Jr, B. R. Desai, D. E. Heiner, J. Fiechtl,
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

21 H. C. Pape, R. Marcucio, C. Humphrey, C. Colnot, S. Porter and M. H. Meyer, J. Orthop. Res., 2006, 24, 1933.
M. Knobe and E. J. Harvey, J. Orthop. Trauma, 2010, 24, 42 E. C. Wahl, J. Aronson, L. Liu, J. L. Fowlkes,
522. K. M. Thrailkill, R. C. Bunn, R. A. Skinner, M. J. Miller,
22 C. M. Champagne, J. Takebe, S. Offenbacher and G. E. Cockrell, L. M. Clark, Y. Ou, C. M. Isales,
L. F. Cooper, Bone, 2002, 30, 26. T. M. Badger, M. J. Ronis, J. Sims and C. K. Lumpkin Jr,
23 W. Lehmann, C. M. Edgar, K. Wang, T. J. Cho, J. Bone Miner. Res., 2010, 25, 114.
G. L. Barnes, S. Kakar, D. T. Graves, J. M. Rueger, 43 M. S. Ominsky, C. Li, X. Li, H. L. Tan, E. Lee, M. Barrero,
L. C. Gerstenfeld and T. A. Einhorn, Bone, 2005, 36, 300. F. J. Asuncion, D. Dwyer, C. Y. Han, F. Vlasseros,
24 S. Portal-Núñez, D. Lozano and P. Esbrit, Histol. Histo- R. Samadfam, J. Jolette, S. Y. Smith, M. Stolina,
pathol., 2012, 27, 559. D. L. Lacey, W. S. Simonet, C. Paszty, G. Li and H. Z. Ke,
Downloaded on 26 March 2013

25 J. D. Boerckel, B. A. Uhrig, N. J. Willett, N. Huebsch and J. Bone Miner. Res., 2011, 26, 1012.
R. E. Guldberg, Proc. Natl. Acad. Sci. U. S. A., 2011, 108, 44 A. S. Hoffman, in Biomaterials Science, ed. B. D. Ratner,
E674. A. S. Hoffman, F. J. Schoen and J. E. Lemons, Elsevier Aca-
26 M. Grellier, N. Ferreira-Tojais, C. Bourget, R. Bareille, demic Press, Amsterdam, 2004. p. 67.
F. Guillemot and J. Amédée, J. Cell. Biochem., 2009, 106, 45 M. Vallet-Regí and E. Ruiz-Hernández, Adv. Mater., 2011,
390. 23, 5177.
27 S. Akeel, A. El-Awady, K. Hussein, M. El-Refaey, 46 M. Vallet-Regi and A. J. Salinas, in Bone Repair Biomater-
M. Elsalanty, M. Sharawy and M. Al-Shabrawey, Arch. Oral ials, ed. J. A. Planell, S. M. Best, D. Lacroix and A. Merolli,
Biol., 2012, 57, 445. CRC Press Boca Raton, 2009, p. 194.
28 P. Esbrit, M. V. Alvarez-Arroyo, F. de Miguel, O. Martín, 47 L. L. Hench, J. Am. Ceram. Soc., 1998, 81, 1705.
M. E. Martínez and C. Caramelo, J. Am. Soc. Nephrol., 48 D. G. Castner and B. D. Ratner, Surf. Sci., 2002, 500, 28.
2000, 11, 1085. 49 L. L. Hench and J. M. Polak, Science, 2002, 295, 1014–
29 L. Pederson, M. Ruan, J. J. Westendorf, S. Khosla and 1017.
M. J. Oursler, Proc. Natl. Acad. Sci. U. S. A., 2008, 105, 50 T. Albrektsson and C. Johansson, Eur. Spine J., 2001, 10
20764. (Suppl 2), S96.
30 E. Shimizu, J. Tamasi and N. C. Partridge, J. Dent. Res., 51 A. J. Salinas and M. Vallet-Regí, Z. Anorg. Allg. Chem.,
2012, 91, 268. 2007, 1762.
31 K. Arvidson, B. M. Abdallah, L. A. Applegate, N. Baldini, 52 M. Vallet-Regí, I. Izquierdo-Barba and M. Colilla, Philos.
E. Cenni, E. Gómez-Barrena, D. Granchi, M. Kassem, Transact. A Math. Phys. Eng. Sci., 2012, 370, 1400.
Y. T. Konttinen, K. Mustafa, D. P. Pioletti, T. Sillat and 53 M. Vallet-Regí, M. Colilla and B. González, Chem. Soc.
A. Finne-Wistrand, J. Cell. Mol. Med., 2011, 15, 718. Rev., 2011, 40, 596.
32 B. L. Vaes, P. Ducy, A. M. Sijbers, J. M. Hendriks, E. P. van 54 J. Juhasz and S. Best, J. Mater. Sci., 2012, 47, 610.
Someren, N. G. de Jong, E. R. van den Heuvel, W. Olijve, 55 J. C. Elliott, Structure and Chemistry of the Apatites
E. J. van Zoelen and K. J. Dechering, Bone, 2006, 39, 724. and Other Calcium Orthophosphates, Elsevier, London,
33 F. H. Zhou, B. K. Foster, X. F. Zhou, A. J. Cowin and 1994.
C. J. Xian, J. Bone Miner. Res., 2006, 21, 1075. 56 J. C. Elliott, J. Appl. Crystallogr., 1980, 13, 618.
34 J-B. Kim, P. Leucht, K. Lam, C. Luppen, D. T. Berge, 57 R. Z. LeGeros, Calcium phosphates in oral biology and
R. Nusse and J. A. Helms, J. Bone Miner. Res., 2007, 22, medicine, in Monographs in Oral Science, ed. H. M. Myers,
1913. S. Karger, Basel, 1991.
35 U. Krause, S. Harris, A. Green, J. Ylostalo, S. Zeitouni, 58 J. M. Bouler, R. Z. LeGeros and G. Daculsi, J. Biomed.
N. Lee and C. A. Gregory, Proc. Natl. Acad. Sci. U. S. A., Mater. Res., 2000, 51, 680.
2010, 107, 4147. 59 L. C. Chow, J. Ceram. Soc. Jpn., 1991, 99, 954.
36 P. J. Roholl, E. Blauw, C. Zurcher, J. A. Dormans and 60 M. P. Ginebra, F. C. M. Driessens and J. A. Planell, Bioma-
H. M. Theuns, J. Bone Miner. Res., 1994, 9, 355. terials, 2004, 25, 3453.

This journal is © The Royal Society of Chemistry 2013 Biomater. Sci., 2013, 1, 40–51 | 49
View Article Online

Review Biomaterials Science

61 L. M. Sun, C. C. Berndt, K. A. Gross and A. Kucuk, 89 S. Ohba, F. Yano and U-i. Cheng, BoneKEy, 2009, 6,
J. Biomed. Mater. Res., 2001, 58, 570. 405.
62 L. L. Yan, Y. Leng and L. T. Weng, Biomaterials, 2003, 24, 90 L. Da Silva Meirelles, A. I. Caplan and N. B. Nardi, Stem
2585. Cells, 2008, 26, 2287.
63 L. L. Hench, R. J. Splinter, W. C. Allen and T. K. Greenlee, 91 A. I. Caplan and J. E. Dennis, J. Cell. Biochem., 2006, 98,
J. Biomed. Mater. Res., 1971, 2, 117. 1076.
64 R. Li, A. E. Clark and L. L. Hench, J. Appl. Biomater., 1991, 92 P. Bianco, P. G. Robey and P. J. Simmons, Cell. Stem Cell,
2, 231. 2008, 2, 313.
65 M. M. Pereira, A. E. Clark and L. L. Hench, J. Biomed. 93 J. M. Sorrell, M. A. Baber and A. I. Caplan, Tissue Eng. Part
Mater. Res., 1994, 28, 693. A, 2009, 15, 1751.
66 A. J. Salinas, M. Vallet-Regí and I. Izquierdo-Barba, J. Sol- 94 S. Shi, S. Gronthos, S. Chen, A. Reddi, C. M. Counter,
Gel Sci. Technol., 2001, 21, 13. P. G. Robey and C. Y. Wang, Nat. Biotechnol., 2002, 20,
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

67 M. Vallet-Regí, C. V. Ragel and A. J. Salinas, Eur. J. Inorg. 587.


Chem., 2003, 1029. 95 L. Duplomb, M. Dagouassat, P. Jourdon and D. Heymann,
68 M. Vallet-Regí, A. Rámila, R. P. del Real and J. Pérez-Par- Stem Cells, 2007, 25, 544.
iente, Chem. Mater., 2001, 13, 308. 96 Y. Chen, V. Bloemen, S. Impens, M. Mohecen, F. P. Luyten
69 M. Vallet-Regí, M. L. Ruiz-González, I. Izquierdo-Barba and J. Schrooten, Tissue Eng. Part C Methods, 2011, 17,
and J. M. González-Calbet, J. Mater. Chem., 2006, 16, 26. 1211.
70 A. López-Noriega, D. Arcos, I. Izquierdo-Barba, 97 A. I. Caplan, Tissue Eng. Part B., 2009, 15, 195.
Y. Sakamoto, O. Terasaki and M. Vallet-Regí, Chem. 98 H. Autefage, F. Briand-Mesange, S. Cazalbou, C. Drouet,
Mater., 2006, 18, 3137. D. Fourmy, S. Goncalves, J. P. Salles, C. Combes,
71 J. R. Jones and L. L. Hench, Curr. Opin. Solid State Mat. P. Swider and C. Rey, J. Biomed. Mater. Res., Part B.,
Downloaded on 26 March 2013

Sci., 2003, 7, 301. 2009, 91B, 706.


72 J. R. Jones, J. Eur. Ceram. Soc., 2009, 29, 1275. 99 M. Manzano and M. Vallet-Regí, Prog. Solid State Chem.,
73 J. R. Porter, T. T. Ruckh and K. C. Popat, Biotechnol. Prog., 2012, 40, 17.
2009, 25, 1539. 100 S. Govender, C. Csimma, H. K. Genant, A. Valentin-Opran,
74 F. O’Brien, Mater. Today, 2011, 14, 88. Y. Amit, R. Arbel, H. Aro, D. Atar, M. Bishay, M. G. Börner,
75 B. M. Willie, A. Petersen, K. Schmidt-Bleek, A. Cipitria, P. Chiron, P. Choong, J. Cinats, B. Courtenay, R. Feibel,
M. Mehta, P. Strube, J. Lienau, B. Wildemann, P. Fratzl B. Geulette, C. Gravel, N. Haas, M. Raschke,
and G. Duda, Soft Matter, 2010, 6, 4976. E. Hammacher, D. van der Velde, S. Oevre, L. Nordsletten,
76 Q. Fu, E. Saiz, M. N. Rahaman and A. P. Tomsia, Mater. A. Patel, A. Pohl, W. Rennie, P. Reynders, P. M. Rommens,
Sci. Eng., C., 2011, 31, 1245. J. Rondia, W. C. Rossouw, P. J. Daneel, S. Ruff, A. Rüter,
77 A. J. Wagoner Johnson and B. A. Herschler, Acta Biomater., S. Santavirta, T. A. Schildhauer, C. Gekle, R. Schnettler,
2011, 7, 16. D. Segal, H. Seiler, R. B. Snowdowne, J. Stapert,
78 D. M. Yunos, O. Bretcanu and A. R. Bocaccini, J. Mater. G. Taglang, R. Verdonk, L. Vogels, A. Weckbach,
Sci., 2008, 43, 4433. A. Wentzensen and T. Wisniewski, J. Bone Joint Surg. Am.,
79 O. Gauthier, J. M. Bouler, E. Aguado, P. Pilet and 2002, 84, 2123.
G. Daculsi, Biomaterials, 1998, 19, 133. 101 L. B. Shields, G. H. Raque, S. D. Glassman,
80 S. F. Yang, K. F. Leong, Z. H. Du and C. K. Chua, Tissue M. Campbell, T. Vitaz, J. Harping and C. B. Shields,
Eng., 2002, 8, 1. Spine, 2006, 31, 542.
81 A. García, I. Izquierdo-Barba, M. Colilla, C. López de 102 H. T. Aro, S. Govender, A. D. Patel, P. Hernigou, A. P. de
Laorden and M. Vallet-Regí, Acta Biomater., 2011, 7, 1265. Gregorio, G. I. Popescu, J. D. Goleen, J. Christensen and
82 W. Habraken, J. G. C. Wolke and J. A. Jansen, Adv. Drug. A. Valentin, J. Bone Joint Surg. Am., 2011, 93, 801.
Deliv. Rev., 2007, 59, 234. 103 D. Lozano, M. Manzano, J. C. Doadrio, A. J. Salinas,
83 M. Vallet-Regí, F. Balas and D. Arcos, Angew. Chem., Int. M. Vallet-Regí, E. Gómez-Barrena and P. Esbrit, Acta Bio-
Ed., 2007, 46, 7548. mater., 2010, 6, 797.
84 M. Vallet-Regí, Chem.–Eur. J., 2006, 12, 1. 104 C. G. Trejo, D. Lozano, M. Manzano, J. C. Doadrio,
85 M. Manzano, D. Lozano, D. Arcos, S. Portal-Núñez, A. J. Salinas, S. Dapía, E. Gómez-Barrena, M. Vallet-Regí,
C. L. Orden, P. Esbrit and M. Vallet-Regí, Acta Biomater., N. García-Honduvilla, J. Buján and P. Esbrit, Biomaterials,
2011, 7, 3555. 2010, 31, 8564.
86 A. Baeza, I. Izquierdo-Barba and M. Vallet-Regí, Acta Bio- 105 D. Lozano, M. J. Feito, S. Portal-Núñez, R. M. Lozano,
mater., 2010, 6, 743. M. C. Matesanz, M. C. Serrano, M. Vallet-Regí,
87 M. I. Santos, R. E. Unger, R. A. Sousa, R. L. Reis and M. T. Portolés and P. Esbrit, Acta Biomater., 2012, 8, 2770.
C. J. Kirkpatrick, Biomaterials, 2009, 15, 2373. 106 D. Lozano, L. Fernández-de-Castro, S. Portal-Núñez,
88 A. Arthur, A. Zannettino and S. Gronthos, J. Cell. Physiol., A. López-Herradón, S. Dapía, E. Gómez-Barrena and
2009, 218, 237. P. Esbrit, Br. J. Pharmacol., 2011, 162, 1424.

50 | Biomater. Sci., 2013, 1, 40–51 This journal is © The Royal Society of Chemistry 2013
View Article Online

Biomaterials Science Review

107 L. F. de Castro, D. Lozano, S. Portal-Núñez, M. Maycas, 112 T. T. Andreassen, C. Fledelius, C. Ejersted and H. Oxlund,
M. De la Fuente, J. R. Caeiro and P. Esbrit, J. Cell Physiol., Acta Orthop. Scand., 2001, 72, 304.
2012, 227, 1752. 113 K. Kamo, N. Miyakoshi, Y. Kasukawa, K. Nozaka, H. Sasaki
108 D. Lozano, C. G. Trejo, E. Gómez-Barrena, M. Manzano, and Y. Shimada, J. Bone Miner. Metab., 2010, 28, 634.
J. C. Doadrio, A. J. Salinas, M. Vallet-Regí, N. García-Hon- 114 J. Fang, Y-Y. Zhu, E. Smiley, J. Bonadio, J. P. Rouleau,
duvilla, P. Esbrit and J. Buján, Acta Biomater., 2012, 8, S. A. Goldstein, L. K. McCauley, B. L. Davidson and
2317. B. J. Roessler, Proc. Natl. Acad. Sci. U. S. A., 1996, 93, 5753.
109 R. L. Jilka, Bone, 2007, 40, 1434. 115 I. Arrigí, S. Mark, M. Alvisi, B. von Rechenberg,
110 M. Takahata, H. A. Awad, R. J. O’Keefe, S. V. Bukata and J. A. Hubbell and J. C. Schense, Biomaterials, 2009, 30,
E. M. Schwarz, Cell Tissue Res., 2012, 347, 545. 1763.
111 G. L. Barnes, S. Kakar, S. Vora, E. F. Morgan, 116 J. H. Jeon and D. A. Puleo, Biomaterials, 2008, 29, 3591.
L. C. Gerstenfeld and T. A. Einhorn, J. Bone Joint Surg. 117 E. Ruiz-Hernández, A. Baeza and M. Vallet-Regí, ACS
Published on 01 October 2012 on http://pubs.rsc.org | doi:10.1039/C2BM00071G

Am., 2008, 90(Suppl 1), 120. Nano., 2011, 5, 1259.


Downloaded on 26 March 2013

This journal is © The Royal Society of Chemistry 2013 Biomater. Sci., 2013, 1, 40–51 | 51

You might also like