You are on page 1of 17

Biomaterials 20 (1999) 2287}2303

Bioactive ceramics: the e!ect of surface reactivity


on bone formation and bone cell function
P. Ducheyne*, Q. Qiu
Center for Bioactive Materials and Tissue Engineering, Department of Bioengineering, University of Pennsylvania,
Philadelphia, PA 19104, USA

Abstract

Surface reactivity is one of the common characteristics of bone bioactive ceramics. It contributes to their bone bonding ability and
their enhancing e!ect on bone tissue formation. During implantation, reactions occur at the material}tissue interface that lead to
time-dependent changes in the surface characteristics of the implant material and the tissues at the interface. This review describes
some of the current concepts regarding the surface reactivity of bone bioactive materials and its e!ect on attachment, proliferation,
di!erentiation and mineralization of bone cells. ( 1999 Elsevier Science Ltd. All rights reserved.

Keywords: Bioactivity; Bioactive materials; Tissue engineering

1. Introduction the risk of inducing transmissible diseases [4,5]. There-


fore, considerable attention has been directed towards
Musculoskeletal conditions are among the most fre- the use of synthetic grafts, including hydroxyapatite
quently occurring medical conditions, and they have (HA), tricalcium phosphate (TCP) and bioactive glass
a substantial impact on the health and quality of life of and glass ceramics (BG) [6}10]. However, since it is often
the population. In 1988, musculoskeletal conditions im- desirable to have graft material that will eventually re-
posed a $128 billion burden on the United States eco- sorb, leaving space for new bone formation, the low
nomy [1]. In reconstructive surgery, repair of large bone resorbability of stoichiometric HA is considered to be
defects is a major reparative problem. About 1 230 000 a limiting factor. Furthermore, despite the fact that bone
fractures are treated with osteosynthesis material each growth can occur in porous and dense HA particulates,
year in the USA [1]. Approximately 80% of these require the bone conductive e!ect is limited [11].
adjuvant grafting [1]. In 1988, there were about 400 000 HA, TCP and BG are usually considered bone bioac-
total hip and knee arthroplasties performed and, current- tive ceramics. These are materials which, generally, bond
ly, an increasing number of these procedures are revision to surrounding osseous tissue and enhance bone tissue
surgeries with their concomitant need for bone grafting formation. The chemical, physical and mechanical prop-
[1]. The approaches to these di$cult bone repair prob- erties of these materials have been summarized in pre-
lems include utilization of autografts or allografts [2]. vious reviews [12}14]. Since direct bone bonding to
While the use of autograft material is the preferred tech- bioactive glasses was "rst observed [15], considerable
nique, there are limitations such as donor site morbidity, progress has been made in understanding the basic mech-
limited donor bone supply, anatomical and structural anisms of the formation of bone}biomaterial bond and
problems, and elevated levels of resorption during heal- its e!ect on bone formation. This progress resulted main-
ing [3]. Allografts have the disadvantage of eliciting an ly from two approaches. One focused on studying the
immunological response due to genetic di!erences and bone}biomaterials interface that developed in vivo. The
examination of bonding zone revealed the consistent
presence of an interfacial hydroxyapatite layer [15,16].
* Corresponding author. Department of Bioengineering, Centre for
The other approach used in vitro immersions in
Bioactive Materials and Tissue Enginnering, Univeristy of Pennsyl- simulated physiological #uids or cell containing media
vania, 3320 Smith Walk, Philadelphia, PA 19104, USA. [17,18]. These analyses revealed that reactions occurred

0142-9612/99/$ - see front matter ( 1999 Elsevier Science Ltd. All rights reserved.
PII: S 0 1 4 2 - 9 6 1 2 ( 9 9 ) 0 0 1 8 1 - 7
2288 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

at the implant material surfaces such as dissolution,


precipitation and ion exchange. These reactions were
accompanied by adsorption and incorporation of biolo-
gical molecules [19,20]. The combination of in vivo and
in vitro studies led to a better understanding of surface
reactions of bioactive ceramics in the body and their
e!ects on bone formation and cell function.
In this paper we will primarily review the data from
our own laboratory with the goal to addressing the e!ect
of synthetic graft materials on bone tissue formation and
bone cell function. Since the calcium-phosphate-based
ceramics and glasses are reactive materials, we will "rst
review their physicochemical properties, especially those
leading to surface reactions in vitro and in vivo. In the
Fig. 1. Shear strength as a function of implantation time for controls
second half of the paper, we will review the e!ects of (porous surfaced titanium) and porous specimens with various cal-
physicochemical properties of the bioactive ceramics on cium-phosphate coatings. Note that the greatest e!ect on bone tissue
cellular functions, and we will use this information to ingrowth "xation is associated with coating CAP3, which is the coating
suggest applications of bioactive ceramics in tissue engi- with the highest in vitro dissolution rate (with permission [30]).
neering.

2. Material characteristics a4ect bone tissue formation coatings. In addition, these data show that the intensity
of the e!ect was consistent among the three, early-time
In the 1970s, studies of bioactive glasses and hydroxy- points studied.
apatite focused "rst on the analysis of the bonding inter-
face between these materials and surrounding bone tissue
[15,21]. These studies were followed by data underscor- 3. Mechanisms of bone bioactivity
ing the bene"cial e!ect of a bioactive ceramic lining on
the rate of bonding of porous-coated materials [22]. The ability to bond to bone tissue is a unique property
When a porous stainless-steel "ber network was coated of bioactive ceramics. Analyses of the bone}implant in-
with a slip cast hydroxyapatite (HA) lining, a marked terface revealed that the presence of hydroxyapatite is
increase in bone ingrowth was observed in comparison to one of the key features in the bonding zone. Another
the ingrowth in the same porous metal without HA consistent "nding is that the larger the solubility rate of
lining. This e!ect was pronounced at 2 and 4 weeks, but it the ceramic, the more pronounced is the enhancement
disappeared at 12 weeks [23]. This phenomenon allowed e!ect of bone tissue growth [30}34]. A few examples will
achieving the distinct therapeutic bene"t of faster reha- illustrate this. Among the bioactive ceramics, dense,
bilitation for patients with HA-coated devices. stoichiometric hydroxyapatite has a limited reactivity
The enhancement e!ect on bone tissue ingrowth in vitro [35]. Thus, in in vivo experiments, a lesser e!ect
by calcium-phosphate-based ceramic linings was sub- on bone tissue formation was found [11]. Considering
sequently con"rmed in other experiments [24}29]. How- more reactive calcium-phosphate ceramics, such as bi-
ever, the magnitude of the e!ect varied greatly among phasic calcium-phosphate ceramic consisting of equal
these studies. The variation was likely caused by the amounts of b-tricalcium phosphate and hydroxyapatite,
di!erences in experimental parameters used. Thus, the a decrease in average crystal size and an increase in
hypothesis was formulated that structural and composi- microporosity were observed after implantation in oss-
tional changes of the Ca}P ceramic occur due to process- eous defects in dogs for 6 months [36]. These observa-
ing of the coatings, and that these variations of the tions indicated that dissolution and precipitation
material characteristics a!ect the rate of enhancement of occurred in vivo. At the same time, excellent bone growth
the bioactive ceramic coating. Using an experimental was found in the implantation sites. Yet another illustra-
protocol that extensively controlled factors such as ani- tion is related to machinable bioactive glass}ceramic.
mal model, pore size, pore morphology and properties of X-ray intensity pro"les along the implant}bone interface
the metallic substrate, it was found that there were sta- found a Ca}P rich layer after 16 weeks of implantation
tistically signi"cant variations in bonding strength in the [37].
"rst few weeks after implantation arising from di!erences In a previous review paper we summarized a variety
in ceramic characteristics [30]. Fig. 1 shows that cal- of events that were reported to occur at the bioactive
cium-phosphate coatings signi"cantly enhanced bone tis- ceramic}tissue interface [38]. Fig. 2 schematically shows
sue growth "xation, but that the e!ect di!ered among these phenomena. The list does not imply a ranking in
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2289

is both a solution-mediated as well as surface-controlled


e!ect on cellular activity, organic matrix deposition and
mineralization. The gradual change of the ceramic sur-
face to become a biologically equivalent HA with small
crystal dimensions is a rate determining step in the cas-
cade of events underlying bioactive behavior. All phe-
nomena, collectively, lead to the gradual incorporation of
the bioactive implant into developing bone tissue.
To understand the bone}material bonding mecha-
nisms, the atomic and molecular phenomena occurring
at the material surface and their e!ects on relevant reac-
Fig. 2. Schematic diagram representing the events which take place at tion pathways of cells and tissues must be elucidated.
the interface between bioactive ceramics and the surrounding biological Furthermore, for mechanisms to be uniquely identi"ed, it
environment (with permission [38]). is important to establish the extent of each of these
reactions and the sequence in which they occur. Thus, we
will summarize some of the current insight on these issues
terms of time sequence or importance: (1) dissolution in the following sections.
from the ceramic [15,16,33,35,39}41]; (2) precipitation
from solution onto the ceramic [16,32,36,40,42]; (3) ion
exchange and structural rearrangement at the ceramic} 4. Reaction-related surface modi5cations
tissue interface [11,15,16,32,36,43]; (4) interdi!usion from
the surface boundary layer into the ceramic [44]; (5) 4.1. Dissolution, precipitation and ion exchange reactions
solution-mediated e!ects on cellular activity [11,19,
45}47]; (6) deposition of either the mineral phase (a), or Formation of a biologically equivalent apatitic surface,
the organic phase (b), without integration into the ce- a common characteristic of bioactive materials, can be
ramic surface [32,36,40,48,49]; (7) deposition with integ- reproduced in vitro by immersion experiments using
ration into the ceramic [11,36,40]; (8) chemotaxis to the a simulated physiologic solution that mimics the typical
ceramic surface [11]; (9) cell attachment and prolifer- ion concentrations in body #uids. These in vitro physico-
ation [19,46,50,51]; (10) cell di!erentiation [11]; and (11) chemical analyses are useful to explain the observations
extracellular matrix formation [48,49,51]. on ex vivo specimens [35,42]. Such experiments have
The observation of what transpires at the interface, shown that the materials with high solubility also readily
however, does not represent a mechanistic explanation induce the precipitation of a biologically equivalent
for the e!ect which bioceramics have on bone tissue apatite on their surface. Using this in vitro immersion
formation. We can focus on mechanisms by relying on an methodology, considerable evidence has been obtained
increasing body of evidence which suggests that bone revealing the mechanisms of surface reactions
bonding and bone tissue ingrowth enhancement are the [17,40,42,52,53]. The reactions include dissolution, pre-
result of multiple, parallel and sequential reactions at the cipitation and ion exchange accompanied by absorption
material}tissue interface. The interaction is being de- and incorporation of biological molecules [15}19,31].
scribed in broad terms here, but will be elaborated upon Daculsi et al. used transmission electron microscopy to
in the next sections. These interactions are related to determine the in vivo degradation of biphasic implant
either physicochemical phenomena that occur in the materials which were various mixtures of hydroxyapatite
presence or absence of cells, or are related to reactions and tricalcium phosphate [36]. Implants were inserted in
a!ected by cellular activity. An important aspect of the surgically created periodontal defects. At six months,
overall reaction sequence between these materials and a direct correlation was established between the rate of
tissues is that, in the absence of a biologically equivalent, dissolution of the material and the quantity of newly
calcium de"cient, carbonate containing hydroxyapatite formed microcrystals with Ca/P ratios similar to those of
(c-Ap) surface upon implantation, dissolution, precipita- bone apatite. The transmission electron microscopical
tion and ion exchange reactions lead to a biologically studies carried out by Jarcho et al. [21], and Tracy and
equivalent apatitic surface on the implanted material. Doremus [54], who each used dense, stoichiometric hy-
This reaction does not proceed by itself, but is accom- droxyapatite, showed direct apposition of bone crystals
panied by parallel reactions, such as adsorption and onto the synthetic apatite lattice. Whereas in these
incorporation of biological molecules and attachment of studies solid}solution ion exchange was not addressed,
surrounding cells. Furthermore, cells that have adhered Davies et al. [55] documented in a more recent study
to the ever-reacting material surface interact with the a limited in vivo reactivity of this material.
material and produce some of the surface changes. In the A similar result of limited reactivity of dense, stoi-
inverse direction, i.e. from material to environment, there chiometric hydroxyapatite was also found by Schepers
2290 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

et al. [11] who implanted granules of this material in


various bone tissue sites of the beagle mandible. This
species and site were chosen to compare the in vivo
behavior of several calcium-phosphate ceramics and
glasses in view of their possible or actual use in clinical
dentistry. Dense hydroxyapatite, porous hydroxyapatite
and bioactive glass 45S5 (nominal composition by
weight: 45% SiO , 24.5% Na O, 24.5% CaO, 6% P O )
2 2 2 5
were implanted [11]. Four types of bone defects were
created and "lled at the time of surgery: tooth extrac-
tions, occlusal and buccal cavities in the mandible, and
periodontal defects. The materials were evaluated after 1,
2, 3, 6, 12 and 24 months after implantation. On dense
hydroxyapatite granules, bone tissue conduction was
consistently observed at the early time points. Bone tis-
sue grew from the preexisting bone along the particles
that were closest to the defect wall. However, bone tissue
did not grow further than about 1 mm into the defect
after about 12 weeks. In addition, the presence of multi-
nucleated giant cells was continuously observed from
that time on and the ceramic surface exhibited a moth
eaten appearance resulting from the local resorption of
the material. In the areas where giant cells were present,
there was no new bone tissue formation. Osteoconduc-
tion and giant cell-mediated resorption frequently had
taken place on separate sides of a single particle. These
phenomena are shown in Fig. 3a and b.
This experiment showed beyond doubt that dense,
stoichiometric hydroxyapatite is osteoconductive in vivo.
However, it also revealed that this material only displays
a limited bone bioactivity. This behavior can be ex-
plained by the mechanisms of bioactivity formulated
above. In this instance, it su$ces to focus on physico- Fig. 3. Histological section of dense, stoichiometric hydroxyapatite.
chemical phenomena of dissolution, and dissolution} (a) After three months, bone tissue has grown along the particle sur-
precipitation, without need to address cellular activity face, but only over a distance of about 1 mm. (b) From six months on,
per se. Among all calcium-phosphate ceramic (CPCs) the particle surfaces have a moth eaten appearance caused by multinuc-
studied, dense, stoichiometric hydroxyapatite is the one leated cell resorption. The resorption is not paralleled by a bone
formation process (with permission [11]).
that dissolved the slowest [35]. In addition to its slow
dissolution reaction kinetics, in vitro experiments simula-
ting precipitation reactions in vivo, showed that the pre-
cipitation rate is also amongst the slowest. Furthermore, not unequivocally support this hypothesis. The tissue
the precipitate that forms on its surface is not a biolo- response to a porous carbonated apatite material im-
gically equivalent HA, but is a precursor phase with planted in beagle, mandibular bone was very similar to
greatly lower Ca/P ratio than 1.67 [42]. Thus, even that with dense, stoichiometric hydroxyapatite [11]. Os-
though this material can intrinsically react with sur- teoconduction from the bone defect wall occurred over
rounding bone tissue, its reaction rate is very slow. We a limited distance and bone tissue conduction into the
suggest that competing reactions take over quickly and center of the defects was not observed. In addition,
that, from that time forward, bone tissue formation does a chronic, mild multinucleated cell resorption of the
no longer take place on the hydroxyapatite surface. In material started as early as 3 months after implantation.
clinical dentistry this sequence of events is well known, These observations are shown in Fig. 4a and b [11,56].
since most sites treated with dense, stoichiometric hy- Our in vitro experiments suggest that the characteristic
droxyapatite never fully heal. at the basis of this limited osteoconductivity is the
If a key element for bone bioactive behavior is the considerable induction time for precipitation reactions
development of a carbonated apatite surface, one might on the material to get underway, impeded as they are by
expect an excellent response by using a carbonated apa- protein adsorption to the material surface. Transposing
tite as implant material. However, implantation data do these data to the in vivo situation, protein molecules
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2291

phate concentration in solution prior to the subsequent


drop associated with the calcium-phosphate precipita-
tion [17].
When discussing reaction-induced surface modi"ca-
tions of bioactive ceramics, it is also useful to review that
materials with excellent tissue response to bone generally
undergo a change of their surface properties. In many
instances, a calcium-phosphate layer can be found at the
interface with the tissues. This observation was also made
for other ceramics than the ones discussed so far (sol}gel
processed silica and titania], metallic materials such as
titanium, and various polymers such as polyethylene
oxide/polybutylene terephthalate copolymers and
tyrosine derived polycarbonates [59}67]. Using in vitro
modeling results, it was suggested that this phenomenon
was related to the presence of hydroxyl (Si}OH and
Ti}OH) or carbonyl groups (C}OH) at the material sur-
faces [62]. But, as is the case for stoichiometric
hydroxyapatite, the conditions under which a calcium-
phosphate layer forms in vitro do not necessarily mimic
the in vivo conditions well, nor are the reaction kinetics
such that competing reactions (not modeled in vitro)
dominate the events at the biomaterial surface in vivo. If
the previous suggestion would be correct, the implication
would be that materials do not need to contain calcium
and phosphate in order to bond with bone tissue. How-
ever, given that the in vitro modeling studies did not
address the mechanisms by which Ca and P containing
phases form on hydroxylated surfaces, de"nitive word as
to whether this question can be reduced to a purely
inorganic event, is still needed. In fact, it would appear
that an event without intervening biological components
Fig. 4. Tissue response to porous, carbonated hydroxyapatite. (a) At is a strained concept for, essentially, an organic reaction
24 months, bone does not make contact with the particles. (b) At microcosm. This observation may draw credence from
3 months, a particle lined with multinucleated cells (with permission
an analogous case, i.e. the issue of the reaction
[11]).
sequence at the surface of bioactive glasses. In what
follows we will provide evidence to suggest that this
surface reaction layer does not form as a result of inor-
adsorb onto the implant material upon implantation, ganic reactions "rst, followed by biologically driven
and signi"cantly slow down the formation of a bone events next, as has been proposed through the 1970s and
mineral surface layer [53,57]. As a corollary, bone bioac- 1980s [68]. In fact, serum proteins do have a major e!ect
tive behavior is then not strongly expressed without this on the properties of the surface reaction layer, as will be
carbonated apatitic surface layer. described next.
Among the studies on the partially crystallized Kyoto
glass formulations, which are mixtures of glass and apa- 4.2. Ewect of serum proteins on the surface reactions
tite or wollastonite, two reports are noteworthy. Two of bioactive glass
separate bioactive glass ceramic plates, implanted in soft
tissue sites in close proximity to one another, were found To explore the e!ect of proteins on the formation of
to be "rmly attached to each other after 3 months of surface reaction layers on bioactive glass, we analyzed
implantation [58]. Such a phenomenon can be explained the atomic and molecular changes in bioactive glass 45S5
by assuming that there was a precipitation and sub- surface during immersion in protein-free bu!er solution
sequent growth of crystals eventually bridging the pre- and protein-containing culture medium [69]. When im-
existing gap between the two plates. Although this mersed in serum-free solutions, atomic force microscopy
experiment did not address the surface dissolution, the (AFM) showed that the surface of bioactive glass reacted
same glass ceramic, immersed in a simulated physiolo- non-uniformly. After 5 min of immersion, the glass sur-
gical solution, caused an increase in calcium and phos- face showed a rough texture due to the formation of
2292 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

particles on the surface. With increasing immersion time, [71]. As will be described below, bioactive glass granules
the particles increased in size and number. Unlike the can be internally hollowed out leading to shells of cal-
reaction on the substrates immersed in serum-free solu- cium-phosphate in which osteogenesis can be observed
tion, a uniform layer of globules, presumably proteins, [11]. Bioactive glass granules were immersed under inte-
covered the substrates immersed in serum-containing gral (no solution exchange during the experiment, there-
solutions. As the immersion time increased, the density of by simulating stagnant #uid conditions) or di!erential
globules increased and the layer thickened. Rutherford conditions (conditions simulating continuous #uid #ow
backscattering spectroscopy (RBS) of the outermost sur- in vivo) in tris bu!ered solution complemented with
face layers revealed that substrates formed Si-rich surface either plasma electrolyte (TE) or with electrolyte and
layers in the "rst hour of immersion in the absence or 10% serum (TES-10). Only when the solution was con-
presence of serum proteins. However, the RBS spectra tinuously replenished, thereby avoiding Si saturation in
also showed that the formation of the calcium phosphate solution, and only when the solution contained serum
layer was di!erent in serum-containing solutions relative proteins, was full Si dissolution from the core of the
to the serum-free solutions. FTIR analysis showed that, granules observed [71]. This result was ascribed to the
in the serum-free solution, crystalline hydroxyapatite was di!erent structure and composition of the surface reac-
formed by transformation from the initially formed tion layer that formed in the presence of serum. In
amorphous calcium phosphate. In contrast, immersion in the serum-free solution, the carbonated hydroxyapatite
serum-containing solutions only produced an amor- surface protects the glass from further corrosion. In
phous calcium phosphate. contrast, the reaction layer formed in serum-containing
Prior to this experiment, Radin et al. already demon- solutions does not o!er su$cient protection from con-
strated, using one-parametric variations in solution com- tinued corrosion. This surface reaction layer is slightly
position, that the surface reactions on bioactive glass porous and comprises proteins, silica and amorphous
45S5 were exquisitely dependent upon the modeling con- calcium phosphate [71]. In the presence of serum pro-
ditions [70]. The solutions used were tris bu!er, tris teins, the apatite formation is extensively delayed, a "nd-
bu!er complemented with plasma electrolyte and/or ing which is consistent with other data on the e!ect of
serum, and serum. After a short immersion time (3 h) proteins on nucleation and crystal growth of apatite
a crystalline, carbonated hydroxyapatite (c-HA) layer [57,72}74].
formed only in tris. Reaction surfaces of di!erent struc- In the present section, data were summarized docu-
ture, morphology and composition were observed in all menting that in solutions more closely approaching the
other solutions after either short- and long-term immer- physiological state, the maturation of amorphous cal-
sions. They comprised two layers with the layer in con- cium-phosphate to crystalline hydroxyapatite does not
tact with the bulk consisting mainly of Si; the outer layer take place readily. The proteinaceous layer which ad-
was composed of Si, Ca and P, was amorphous and had sorbs onto the glass interferes with the solid to liquid
a Ca/P ratio of about 1. Serum proteins adsorbed on the interaction of the amorphous calcium-phosphate layer.
BG surfaces at the early stages of the solution-mediated Whereas amorphous calcium phosphate can form exclus-
BG reactions. Formation of a crystalline carbonated HA ively as the result of physicochemical phenomena in the
layer was delayed up to three or more days in solution solid glass phase, it is suggested here that adsorbed
with plasma ions. And again, in the presence of serum, serum proteins impede the nucleation and growth
only amorphous surfaces composed of Si, Ca and P were reactions by which it would transform to carbonated
observed for any time up to seven days of immersion apatite. The reactions in the glass are not blocked, how-
[70]. ever, as Ca and P di!usion leads to a continuously
Given the repeated observation of the absence of thickening of the Ca}P rich zone under the adsorbed
crystalline hydroxyapatite when proteins co-adsorb, it protein layer. The adsorption of the protein layer may
is unlikely that, in vivo, adsorption of biological also be critical in terms of providing attachment sites for
molecules will take place subsequent to the transforma- bone cells such as osteoblasts and their progenitors. The
tion of an amorphous calcium-phosphate rich layer to e!ect of adsorbed molecules is the subject of the next
carbonated apatite, as was previously suggested [68]. sections.
The present data suggest that serum proteins adsorb in
tandem with the occurrence of solution-mediated reac-
tions leading to formation of a silica-gel. Amorphous 5. Stimulation of cellular function0e4ect of porosity
calcium-phosphate phases accumulate in this Si-rich
matrix. 5.1. Marrow-derived osteoprogenitor cells
A signi"cantly di!erent behavior between immersion in porous matrices
in serum protein-free and serum protein-containing solu-
tions was also observed in experiments intended to In vivo studies have clearly documented that bioactive
model silicon dissolution from bioactive glass particles ceramics a!ect cellular function. A series of experiments
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2293

using porous hydroxyapatite and bone marrow cells


showed that the osteoprogenitor nature of cells from
bone marrow was activated more readily in heterotopic
sites when they were cultured with porous, carbonated
hydroxyapatite than when implanted by themselves
[45,75}77]. With these studies, Caplan and his associates
were "rst to describe the idea of culturing cells capable of
expressing the osteoblastic phenotype with the intent to
synthesize arti"cial bone grafts. These authors focused
primarily on issues related to stem cell preparation.
When cultured on appropriate templates, the cell cul-
tures produced extracellular matrix. Investigators from
this laboratory also used periosteal-derived cells.
Nakahara et al. [78] showed that when the cells were
Fig. 5. Bone tissue formation in excavated bioactive glass particles of
combined with porous calcium-phosphate ceramics and narrow size range. Bone tissue is stained red. Also note the channels
implanted in a subcutaneous site in athymic mice, bone connecting the interior of the particle with the surrounding milieu (with
tissue was formed. permission [116]).
In these studies using marrow stroma-derived osteo-
progenitor cells, it is important to realize that it is pri-
marily the intrinsic capacity of the osteoprogenitor cells excavated bioactive glass particles was independently
that produces the upregulation to cells of the osteoblast veri"ed by Gatti et al. [80]. Bone biopsies taken from
lineage. Okumura et al. [79] demonstrated that stem cell sinus lift treatment sites at the time of implant insertion
di!erentiation to cells expressing the osteoblastic pheno- con"rmed the excavation process and bone tissue forma-
type also occurred in porous titanium. However, the tion within the excavated particles in a clinical setting
pattern of bone tissue formation was di!erent. Whereas [81].
in porous hydroxyapatite it started at the ceramic sur- The excavation of the particles itself is related to the
face, it started in the middle of the pore and was much well-documented surface reactions on bioactive glass, as
slower in porous titanium. These data suggested that well as to an extensive control of the glass granule size.
even though the intrinsic capability of pluripotential cells Interfacial reactions on bioactive glass lead to the forma-
led to di!erentiation along the osteoblast pathway, the tion of two surface layers: an inner silica gel layer and an
osteogenic potential of these cells was also stimulated by outer calcium-phosphate rich layer [82]. In the dog man-
the bioactive material surface. dible, the thickness of a reaction layer on bioactive glass
45S5 was found to be about 150}170 lm at 3 months
5.2. Bioactive glass granules of narrow size range [83]. Thus, with a particle size that is about double the
reaction layer thickness, the particles fully react. These
Another example of stimulation of cellular function is reactions produce phase transformations, which in turn
the phenomenon of bone tissue formation with bioactive may provoke dimensional variations and cause cracking.
glass granules of narrow size range. In the aforemen- Macrophages move into the particles through these
tioned beagle study with porous HA, bioactive glass cracks and assist with the removal of the interior silica gel
particles with a well-controlled particle size were also by phagocytosis [11,84]. What remains is a shell of
implanted. Bioactive glass granules of narrow size range calcium-phosphate, which was previously formed as
(300}355 lm, as determined by sieving) elicited bone tis- a biological reaction product. Osteoprogenitor cells mi-
sue formation throughout 5 mm defects in the beagle grate into the protective space of these in situ formed
mandible as soon as 1 month after implantation [11]. hollow chambers and undergo di!erentiation to osteo-
The critical phenomenon taking place "rst was the blasts. This di!erentiation does not occur outside these
formation of internal pouches in each of the particles. biologically formed calcium-phosphate chambers. The
This was followed by di!erentiation of osteoprogenitor proposed mechanism for the excavation process also
cells to osteoblasts actively laying down bone tissue in allows to explain why large particles do not react
these pouches. Subsequently, bone tissue grew out of the throughout and, as such, do not cause the di!erentiation
excavations, surrounded the particles and connected of precursor cells. For smaller particles, it was shown that
with bone tissue formed around the neighboring par- they elicited an in#ammatory response in maxillofacial
ticles. As a result, defects were quickly repaired in about applications [85,86]. Considering that in particle prep-
3}6 months, as each particle served as a nucleus for arations with a large variation of particle size, there is
repair [11]. Fig. 5 shows bone tissue formation in an always a much larger number of smaller particles than
excavated particle. This "nding of upregulation of os- larger ones, a restricted size range is important to achiev-
teoprogenitor cells to bone tissue-forming osteoblasts in ing this unique phenomenon of promotion of cell
2294 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

di!erentiation throughout the treated defect. The re- drawn into the developing surface reaction layers, di!er-
stricted size range is also important to allow optimum ences in reaction kinetics can also lead to di!erences in
vascularity development. When a wide size range of par- factor concentrations at the glass surface. Second, di!er-
ticles is installed, the space between the larger particles is ences in #uid #ow can produce di!erences in shear stress
more readily "lled up, thereby obstructing the tissue between inside and outside.
repair [10]. We used "nite element analysis to model numerically
compositional variations of the extracellular #uids, spe-
5.3. Bone tissue induction in porous calcium phosphates ci"cally the Na`, Ca`2, and HPO~2 concentrations. We
4
also modeled the #uid #ow around and within porous
Ripamonti [87] and his coworkers observed a bone bioactive glass [93]. For the range of interstitial #uid
induction phenomenon in porous hydroxyapatite. This velocities examined, the analysis revealed several signi"-
"nding was unusual in that the porous specimens were cant "ndings. First, the creeping #ow of interstitial #uid
not preloaded with cells. In addition, the implantation around the bioactive glass granules was dominated by
sites were soft tissue sites, unlike in the Schepers et al. viscous forces [low Reynolds number #ow), producing
study [11] where di!erentiation of progenitors to osteo- shear stresses at the granule}#uid interface. Second, the
blasts caused by a synthetic material were observed in #uid inside the granules remained stagnant, resulting in
a bony site. The "nding of induction by Ripamonti et al., zero shear stress at the granule}#uid interface. Di!usion
however, did not occur until after longer implantation was the dominant mass transport mechanism within the
durations. In addition, it was not consistently observed in porous granules. Third, the governing mass transport
all animal species. Porous hydroxyapatite obtained after mechanism around the granules depended on the #ow
hydrothermal conversion of the calcium carbonate exo- "eld. At low velocities, di!usion dominated the transport
skeleton of coral was implanted in the rectus abdominis of chemical species. At intermediate velocities, di!usion
of adult rabbits, dogs, and baboons. Minimal amounts of and convection governed the mass transport, while at
bone formed in specimens harvested from rabbits and high velocities, convection was the controlling transport
dogs. In contrast, 10.4 and 25.9% of the pore space was mechanism [93]. These numerical analyses documented
"lled in specimens retrieved from baboons after 3 and variations in #ow and stress patterns, reaction rates and
6 months, respectively [87,88]. Other workers have solute concentrations between the interior and exterior of
con"rmed the phenomenon of bone induction in porous the granules. As an outcome of these analyses, the hy-
bioactive ceramics implanted in soft tissue sites [89}91]. pothesis was formulated that variations in material sur-
face composition, medium composition and shear stress
5.4. The ewect of protected spaces on bone formation caused the observed di!erences in cellular function. Spe-
ci"cally, any of the cellular activities leading up to tissue
As early as in 1965, Urist observed that &2new bone formation, including attachment, proliferation, di!erenti-
was deposited in pockets or excavation chambers "lled ation and extracellular matrix formation, could be im-
with proliferating osteoprogenitor cells. The process be- plied. This broad hypothesis was the basis for a number
gan within a few weeks and was complete in a few of subsequent experiments.
months. How much new bone could be attributed to the
osteoconduction of cells growing in from the walls of the
host bone, and how much could be accounted for by 6. E4ects of surface reactions on cellular function
osteogenic induction, was not apparent from experiments
on normal, healthy bones2' [92]. Clearly, pores, and 6.1. Surface modixed bioactive glass stimulates
pockets which resemble pores, have an e!ect beyond the the osteoblast phenotype in vitro
e!ect produced by growth factors and other signaling
molecules. In every study summarized in this section the In an e!ort to reproduce the e!ect of BG in that it
protective space in excavations or inside pores was essen- stimulated the osteoblast phenotype in excavated glass
tial to stimulate di!erentiation of precursors to osteo- pouches [11], we cultured neonatal rat calvaria osteo-
blasts. blasts on porous BG templates. Brighton and coworkers
Addressing the phenomenon of di!erentiation in the documented that these cells expressed the osteoblast
excavated bioactive glass granules, we hypothesized that phenotype, but their osteoblastic markers were not ex-
the e!ect could be due to two main sets of reasons [93]. pressed till after 17}21 days [94}97]. In our experiments,
First, the bioactive glass is a reactive material and, as the osteoblast phenotype was already expressed after
a result, solid to solution interaction in the limited space 7 days. Fig. 6 is a scanning electron micrograph of the
of the pouch will be modi"ed from the reactions without cross section of porous BG showing the formation of
transport limitations outside the particles. Although we extracellular matrix after 7 days in cell culture. The cells
speci"cally focused on the ionic species of the glass, given also exhibited a high alkaline phosphatase activity and
that growth factors and signaling molecules can be synthesized collagen type I and osteocalcin [49].
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2295

Fig. 7. Alkaline phosphatase (AP) activity of 7-day cell cultures on


porous bioactive glass conditioned by three procedures. &BG-0' is the
glass surface onto which serum proteins were adsorbed for 1 h; &BG-48'
Fig. 6. SEM micrograph of the fractured surface of the porous modi"ed
is the glass surface transformed to a crystalline hydroxyapatite, but not
bioactive glass template seeded with neonatal rat calvaria osteoblasts
subjected to a protein adsorption treatment; &BG 2-step' is the glass
for 7 days. Note the uniform formation of extracellular matrix and
treated sequentially such that it was transformed to a crystalline hy-
bone-like tissue throughout the porous disk (with permission [49]).
droxyapatite onto which serum proteins adsorbed. Note that cells
grown on &BG 2-step' showed high AP activity, and in contrast, the
other two conditions not (with permission [18]).
In this study we also documented that the expression
of the osteoblastic phenotype critically depended on the
preparation of the glass surface. Prior to our experi- single-step immersions [101]. Furthermore, the DNA
ments, a limited e!ect of the BG substrates in cell culture content did not indicate a loss of cellular viability on
was reported [98,99]. A con#uent monolayer was not those surfaces that were either calcium phosphate with-
observed until 12 days of culture using fetal rat calvaria out adsorbed proteins, or the glass surface with adsorbed
osteoblasts. Additionally, only a &weak' expression of the proteins only.
osteocalcin was observed at this time point. A better
cellular response in vitro was achieved by these authors 6.2. Serum protein adsorption
by adding various oxides which rendered the glass sur-
face less reactive. However, the oxides selected were The data summarized in the previous section evoked
among those that produced an adverse e!ect on the rate the hypothesis that concentration of serum proteins at
of mineralized bone formation in vivo, as reported by the glass surface transformed to calcium-phosphate, ser-
Gross et al. [100]. We overcame the issue of limited ved to enhance the expression of the osteoblast pheno-
reactivity by treating the glass surface prior to cell seed- type. Thus, we measured the amount of protein that
ing. The bioactive glass was transformed into a calcium absorbed from the tissue culture medium. A greater
phosphate surface by immersing it in serum-free, physio- quantity of serum proteins absorbed onto the calcium-
logical solution in a "rst step (step 1), followed by immer- phosphate-coated glass than onto the untreated BG.
sion in serum-containing media (step 2). Fig. 7 shows that However, a yet larger amount absorbed onto
with this 2-step surface preparation, the cells grown on stoichiometric HA, which was used concurrently [20].
the modi"ed glass surface maintained critical features of This was surprising, as in vivo results clearly documented
the osteoblast phenotype, such as a high alkaline phos- a more intense bioactive e!ect for BG than HA [11,102].
phatase activity, among others [101]. This phenotype Thus, we reasoned that the e!ect of the serum protein
was not expressed if the cells were seeded on BG tem- absorption treatment was related to the species that
plates conditioned with only one of the steps (immersion absorbed and found, in fact, signi"cantly di!erent
in either simulated physiological saline to produce a cal- absorption pro"les for untreated BG, calcium-phos-
cium-phosphate surface without pre-adsorbed proteins, phate-coated BG and stoichiometric HA [20]. Fig. 8
or immersion in tissue culture medium to produce a glass shows the protein absorption spectra on each of these
surface with adsorbed proteins). The cells exhibited a low three materials. Whereas on HA the spectrum of ab-
alkaline phosphatase activity. It is also noteworthy that sorbed proteins re#ected the composition of the medium,
equivalent amounts of protein were produced by cells there was a preferential absorption pattern to each of the
inoculated on the templates regardless of the speci"c two types of BG specimens. Especially, the broad band of
conditioning treatment, 2-step or either one of the proteins at molecular weights in excess of 200 kDa which
2296 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

(1) BG immersed in the electrolyte containing, serum free,


simulated physiological solution (BG coated with a Ca}P
surface, but without pre-absorbed proteins), (2) BG im-
mersed in tissue culture medium (BG with a layer of
proteins), (3) BG treated consecutively in the electrolyte
solution and the tissue culture medium (BG coated with
a Ca}P surface onto which "bronectin was extensively
adsorbed), (4) HA immersed in tissue culture medium
(HA extensively coated with a broad spectrum of pro-
teins). The results shown in Fig. 9 reveal a much greater
cellular attachment to the Ca}P and protein-coated BG
than to any other surface. These data demonstrate that
the preferential absorption of "bronectin is a "rst impor-
tant observation to explain BG's e!ect on bone cell
function, and as a corollary, bone tissue formation [20].

6.3. The ewect of xbronectin on osteoblast cell


attachment to bioactive glass

Having found that "bronectin preferentially absorbed


onto calcium-phosphate-coated BG, it is possible that
the e!ect on bone cell function of adsorption of the ligand
was due exclusively to the quantity of "bronectin ab-
sorbed, and was not dependent upon the e$cacy of the
ligand's attachment function. Thus, we also addressed the
question whether, for the same concentration of adsor-
bed "bronectin onto various material surfaces, there
would be di!erences in cell attachment. We quantita-
tively determined cellular attachment to bioactive glass
as a function of bioactive surface reaction stage and
Fig. 8. (a) Densitometry of di!erent serum protein bands present in the quantity of adsorbed "bronectin. It was found that for
polyacrylamide gel of the protein fractions that are present in the tissue
culture medium (TCM), adsorbed onto synthetic hydroxyapatite (HA),
untreated BG (BG-0) and BG of which the surface was transformed to
crystalline calcium phosphate (BG 2-step). The intensity of the protein
bands was visualized using Image Quant software and the Molecular
Dynamics Personal Densitometer. Noteworthy is that the crystalline
calcium phosphate layer on BG (BG 2-step) extensively adsorbed
proteins in a molecular weight range 210}250 KDa; this protein was
minimally adsorbed on BG-0. Furthermore, while &BG 2-step' appeared
to be very selective, HA adsorbed a wide range of proteins. (b) Western
blot analysis of serum proteins adsorbed on BG 2-step (lane 1), BG-0
(lane 2), HA (lane 3), tissue culture plastic dish (lane 5). Bovine serum
"bronectin and TCM are present in (lane 4) and (lane 6), respectively.
BG 2-step selectively adsorbed higher amounts of "bronectin than HA
or BG-0. The intensity of "bronectin bands decreased in the order BG
2-step'BG-0'HA (with permission [20]).

absorbed onto calcium-phosphate-coated BG was note-


worthy. Western blot analysis showed that this band
Fig. 9. DNA content of MC3T3-E1 cells attached on BG with di!erent
arose from extensive, preferential absorption of "bronec- surface modi"cations and HA ceramic. &BG-0' is the glass surface onto
tin onto the calcium-phosphate-coated BG [20]. By vir- which serum proteins were adsorbed for 1 h; &BG-48' is the glass surface
tue of "bronectin being an ubiquitous molecule, which, transformed to a crystalline hydroxyapatite, but not subjected to a pro-
among other functions, also serves as an attachment tein adsorption treatment; &BG 2-step' is the glass treated sequentially
molecule between substrate and cell membrane of an- such that it was transformed to a crystalline hydroxyapatite onto which
serum proteins adsorbed. Note that a signi"cantly larger number of
chorage-dependent cells, an experiment was formulated cells adhered to the glass surface that had been transformed to a cal-
to assess cellular attachment. Four di!erent materials or cium-phosphate layer prior to adsorbing serum proteins and seeding
material surfaces were prepared prior to cell seeding: the cells (with permission [20]).
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2297

the same amounts of adsorbed "bronectin, the attach-


ment strength to treated bioactive glass was signi"cantly
greater than for untreated glass, control glass or stoichio-
metric hydroxyapatite [103]. The treated BG surfaces
were either amorphous calcium phosphate (obtained by
immersion for one day in serum free, simulated physiolo-
gical solution: BG1d) or a crystalline calcium phosphate
(obtained by immersion in the same solution for 7 days:
BG7d).
The osteoblast cell attachment experiments were per-
formed on substrates that were incubated in "bronectin
solutions for 30 min, then in 1% bovine serum albumin
solution for 30 min to block non-speci"c adhesion. Cells
were seeded onto the disks (34 000 cells/cm2) and allowed
to attach for 15 min. The disks were spun in a rotating
disk device and the fraction of adherent cells at a given
stress level were counted and normalized to the number
of cells not experiencing any stress (cells at the center of
the disk). These results were plotted as a function of shear
stress and "tted to a logistic equation to estimate the
critical shear stress (q ) for 50% detachment (Fig. 10). The
# Fig. 10. Cell detachment pro"les for substrates coated with equal
q was used as a measure of attachment strength. The amounts of "bronectin using a 0.1 lg/ml "bronectin solution
# (10 dyn/cm2"1 N/m2). The specimens used included non-reactive
adherent fraction decreased sigmoidally as the shear
stress increased. s increased as the "bronectin concen- borosilicate glass as control glass (CG); stoichiometric hydroxyapatite
# (sHA); untreated bioactive glass (BG0); and bioactive glass of which the
tration increased. For "bronectin-treated substrates, surface was transformed to an amorphous calcium phosphate (BG1d)
s was 50% higher for treated bioactive glasses (BG1d,
# or crystalline carbonated hydroxyapatite (BG7d) by a 1 or 7 day
BG7d) than for untreated glass (BG0), a control non- immersion treatment respectively. Cell adhesion was signi"cantly high-
bioactive glass and HA. er on BG of which the surface had been transformed to calcium
The data clearly documented that the surface treat- phosphate (BG1d, BG7d), than on unreacted bioactive glass (BG0),
control glass (CG) or stoichiometric hydroxyapatite (sHA) (with per-
ment of bioactive glass resulted in enhanced "bronectin- mission [103]).
mediated cell attachment. Since the surface treatment did
not a!ect the amount of adsorbed "bronectin, these data
suggested that the observed increase in attachment di!erentiation. On HA, proliferation is slow to start in
strength resulted from di!erences in "bronectin confor- comparison to the proliferative activity on the calcium-
mation. Previous studies documenting "bronectin con- phosphate-coated BG. In addition, it continues through-
formational changes resulting from adsorption onto out the two week duration of the experiment without
material surfaces can be invoked to explain our own slowing down, all while di!erentiation activity is not
results [104]. Thus, an enhanced biological response activated. Thus, the activity of this cell line was stimu-
resulted from bioactive glass surface modi"cations which lated towards extracellular matrix formation on BG, but
mimicked the changes in vivo [20]. not on HA [105].
It is known that the ligand "bronectin connects to the
cell membrane via integrin receptors, which are also 6.4. Synergism between the calcium-phosphate
activated by this interaction. The activation of integrins surface on bioactive glass and growth factors
then triggers cytoplasmic reactions, and thereby, stimu-
lates the intracellular signaling pathway. The evidence As the previous sections documented, concentration of
that the attachment function of "bronectin was better "bronectin at the surface of bioactive glass transformed
expressed when it was absorbed to calcium-phosphate- to calcium-phosphate-serves to enhance osteoblast func-
coated BG suggests the possibility that this molecule may tion. These data raise a tempting idea, namely that other
also activate the integrins' intracellular signaling much biologically active molecules present in solution may
more readily than when adsorbed to other material surfa- also adsorb onto bioactive calcium-phosphates, and
ces. This implies that the cellular functions such as prolif- especially the calcium-phosphate layer grown on bio-
eration and di!erentiation will be stimulated. The data active glass. Such adsorbed molecules, together with
shown in Fig. 11a and b support this hypothesis an optimally bioactive calcium-phosphate would then
[105]. On treated BG, MC3T3-E1 cell proliferation is extensively a!ect cellular function. This idea of con-
extensive in the "rst few days after cell seeding. After centrated adsorption is the inverse of using cal-
about 7 days, proliferation slows down at the expense of cium-phosphate ceramics and glasses as a vehicle for
2298 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

medium. Second, since we synthesized the glass by


a sol}gel technique, we incorporated BMP-2 throughout
the glass matrix. The xerogel matrix allows di!usion of
the molecule through its porous network and, as such,
the molecule is released in a controlled, continuous way
[108]. Control conditions were the rat marrow stromal
cells by themselves and treated glass disks with cells, but
without addition of BMP-2 to the medium.
Fig. 12 summarizes the alkaline phosphatase activity
of the cells cultured under these various conditions [111].
It is evident that the glass treated to simulate in vivo
conditions leading to a calcium-phosphate surface stimu-
lated the di!erentiation of the osteoprogenitor cells. In
fact, the alkaline phosphatase activity of these cells was
higher than that of the cells seeded on the tissue culture
plastic dishes. Under the conditions of the experiment,
the alkaline phosphatase activity of cells grown on the
glass was also higher than that of cells cultured under the
same conditions, but in medium with BMP-2 added. All
data taken together suggested that the e!ect of the glass
by itself followed from the adsorption of growth factors
that are naturally present in the medium. This could be
readily deduced from the results of the three conditions
in which the sol}gel glass was combined with BMP-2.
When the molecule was added to the medium, it
adsorbed onto the Ca}P surface and a!ected cells in
a more extensive way than under any other condition at
the early, i.e. 6 day, time point. In the other cases the
molecule had to di!use to the surface, and therefore it can
be assumed that less was available to stimulate plu-
ripotential precursor cells to the di!erentiated function at

Fig. 11. DNA content (a) and alkaline phosphatase activity (b) of
MC3T3-E1 cells grown on bioactive glass transformed into crystalline
hydroxyapatite (BG-180H) or synthetic hydroxyapatite (HA). Both ma-
terials were subjected to an immersion in tissue culture medium prior to
cell seeding in order to pre-adsorb proteins. Note that proliferation
starts much sooner on BG-180H than on HA. Around day 7 it slows
down to express the di!erentiated function. In contrast on HA prolifer-
ation continued throughout the experiment in a similar manner as in
control conditions (no substrates*not shown here) (with permission
[105]).

controlled release of incorporated biologically active


molecules, such as, e.g. for the release of growth factors
[106}110]. Thus, in analogy to the observation of en- Fig. 12. Average normalized alkaline phosphatase (AP) activity of rat
hanced function of "bronectin, we addressed the question marrow stromal cells in controls, in which cells were cultured on tissue
culture plastic without and with BMP added to culture medium (&C'
whether bone morphogenetic protein adsorbed into the and &C-BT' respectively), and in experimental groups. Cells were grown
calcium-phosphate surface on BG would a!ect di!erenti- on room-temperature processed silica sol}gel materials with a cal-
ation of osteoblast precursor cells more extensively than cium-phosphate surface; BMP was added to the culture medium (SG-
when added to the culture medium by itself [111]. In this BT), or the BMP was incorporated in the calcium-phosphate surface
experiment we also included two other conditions for (SG-BP), or the BMP was incorporated throughout the glass specimen
(SG-BI). All showed an elevated AP activity over the corresponding
combining BMP-2 (BMP-2, Genetics Institute, Cam- control groups. Cells grown on the glass without added factor (SG) also
bridge, MA) with bioactive glass. First, cells were seeded showed an increased AP activity when compared with cells on tissue
on the surface treated glass and BMP-2 was added to the culture plastic (C) (with permission [111]).
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2299

this "rst time point. At the later time point, i.e. 10 days, this synthesis of bone tissue. Especially when made in porous,
disadvantage of di!usion through the solid disappeared resorbable form, and conditioned to develop a bone-like
and all conditions wherein the glass and BMP-2 were surface prior to being seeded with pluripotential cells
combined, stimulated the expression of the osteoblast capable of expressing the osteoblastic phenotype, these
phenotype in a similar, major way (assays of other pheno- templates lead to expeditious and abundant in vitro
typic markers were performed to con"rm this) [111]. synthesis of extracellular matrix with most important
Another interesting "nding is the synergistic e!ect be- characteristics of bone tissue [49]. In addition, when
tween the calcium-phosphate surface reaction layer on growth factors with established bene"cial e!ect on bone
the glass, and the growth factor used. In fact, there was tissue formation would be incorporated in the templates
a sevenfold increase in expression of the di!erentiated and be released in a time and dose controlled fashion, it is
function by osteoprogenitor cells in comparison to cells possible to further optimize in vitro bone tissue engineer-
to which BMP-2 was added without the glass [111]. ing concepts.
Summing up, the data suggest that concentration of Surface modi"ed bioactive glass templates are resorb-
osteoinductive proteins which are naturally present in able and are gradually replaced by bone tissue. This
bone tissue repair sites or the addition of a recombinant follows from an implantation experiment in which the
form of these growth factors lead to a biologically very typical femoral bone defects encountered in revision hip
potent state of the molecules. The results also suggest arthroplasties were simulated [113]. In this study, a uni-
that the calcium-phosphate surface layers formed on this cortical window defect measuring 1.1]4.5 mm (width
Si-containing, biologically reactive glass material, play over length) was created bilaterally in the femoral dia-
an important role in stimulating osteoblastic di!erenti- physis of adult, male syngeneic rats. Porous, surface
ation of osteoprogenitor cells from the bone marrow. modi"ed bioactive glass ceramics (pSMC) sca!olds were
prepared from 45S5 bioactive glass granules. Two tis-
sue-engineered constructs were synthesized, namely the
7. Applications in tissue engineering sca!old with bone marrow stromal cells seeded at the
time of surgery (&primary'), or these sca!olds with culture
7.1. Bioactive scawolds for bone tissue engineering expanded cells producing extracellular bone-like matrix
(&hybrid'). Defects were treated randomly with pSMC,
The use of in vitro synthesized bone tissue with mar- primary, hybrid, or left untreated (&sham') to compare
row cells obtained from the patient is an appealing idea healing rates at 2, 4, and 12 weeks.
to avoid the profound limitations of biological and syn- At 2 weeks, the long bones treated with the hybrid and
thetic bone grafts. Advantages of in vitro synthesized primary constructs had 40% more bone in the defect
bone constructs include that large segments can be pro- than bones with the pSMC. Signi"cant increases of bone
duced when allografts would appear to be unavoidable tissue formation in the defect occurred over time for all
because of large defect dimensions, and that cells and groups, resulting in approximately 40% bone in the
tissue cultured outside the body could aid in the repair of defect for all treatment groups by 12 weeks. By 12 weeks,
defects at sites with limited growth potential. the surface modi"ed bioactive glass sca!old was fully
The main components of tissue-engineered bone are replaced by in situ formed calcium-phosphate. The ratio
signaling molecules, responding cells and biocompatible of transformed sca!old to defect size decreased signi"-
or bioactive matrix materials [112]. It is expected that cantly for all groups over time. The ultimate torque to
ideal matrix materials stimulate osteoprogenitor cell failure and sti!ness were a!ected by treatment and by
function and the expression of the osteoblastic pheno- time. By 2 weeks, defects treated with hybrid had the
type. They should also integrate with tissues, gradually highest sti!ness and this property was not signi"cantly
be degraded and fully replaced by bone tissue. di!erent from the one of intact bone. Between 2 and
The premise of the bone tissue engineering program in 4 weeks, there were signi"cant increases in sti!ness for
our laboratory is that for bone cell function and extracel- primary and sham, resulting in no signi"cant di!erence
lular matrix formation to be optimized, the critical ma- among these groups and intact bone by 4 weeks. By 12
terial properties that stimulate signaling molecule and weeks, all treatments had comparable sti!ness and
bone cell function must be identi"ed. It is in this context torque to intact bone.
that the data related to the e!ect of bioactive ceramics and Porous, surface modi"ed bioactive ceramic integrated
glasses on cell function are very useful. This is especially well with bone tissue in the healing of a skeletal defect site
true, given that in an arti"cial or in vitro system, osteo- and resorbed in concert with bone formation, which
progenitor cells, as well as bone cells themselves can rap- allows for improvement of the long bone's structural
idly change their phenotype. Hence, the substrate needs to integrity over time. The osteogenic activity of both tis-
promote the expression of the bone cell phenotype. sue-engineered constructs, osteoprogenitor cells seeded
Surface modi"ed bioactive glass templates combine onto sca!olds at the time of surgery, or cells expanded to
several requirements of the ideal template for in vitro form in vitro synthesized bone}sca!old, was similar.
2300 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

Bone formation and the return of normal torsional prop- approach to repairing and regenerating diseased inter-
erties were enhanced for the tissue-engineered constructs vertebral discs.
as compared to the sca!old alone.

7.2. Intervertebral disc tissue engineering 8. Conclusions

Bone tissue engineering using surface modi"ed bioac- Within excavated bioactive glass granules, osteo-
tive glass sca!olds relies on the e!ect of the calcium- progenitor cells di!erentiate and lay down bone tissue. In
phosphate that forms at the glass surface on bone cell vitro experiments, summarized in this paper, have re-
function. When osteoblasts were cultured on glass, the vealed some of the critical events for bioactive glass to
apatite layer was related to the formation of calci"ed stimulate the expression of the osteoblast phenotype. We
extracellular matrix [105]. It is of value to note, though, suggest that the events that were modeled in vitro are
that the calcium-phosphate layer, which can be formed in also likely to take place in vivo, although other phe-
various ways on the glass surface, can also perform other nomena, which have yet to be analyzed, may also con-
functions. Especially, the e!ect on non-mineralizing cells tribute to the observed di!erentiation in the excavated
is interesting to explore. glass particles.
We focused on studying the e!ect on nucleus pulposus Whereas tissue engineering has found its way into the
cells by virtue of the signi"cance of the underlying clinical clinical practice of restoring skin tissue in severely burnt
problem that was thereby addressed. Spinal fusion is patients and patients with chronic decubitus wounds,
frequently used as a treatment for degenerative disc dis- methods to regenerate tissues of the musculoskeletal sys-
ease, especially in cases where instability is a concern. tem are still in the preclinical phase. Numerous are the
Fusion provides load bearing and passive restraint to the statements emphasizing the importance of carrier mate-
motion segment. However, joint motion, which is the rials for optimal delivery of cells and biological signaling.
primary function of the disc, is eliminated. As a result, In this context, resorbable, bioactive glass is an excellent
other motion segments become overloaded, leading to material for the stimulation and delivery of cells and
accelerated degeneration. Other undesirable long-term molecules to achieve repair of bone tissue.
e!ects associated with spinal fusions have also been re- The studies on the e!ect of reactivity of bioactive
ported [114]. Thus, we pursued tissue engineering prin- materials on bone cell function have shown their unique
ciples for treating the intervertebral disc and regenerating role in bone bonding and bone formation processes.
its nucleus pulposus (NP) tissue. Investigations into the mechanisms of bone}bioactive
When studying cultures with nucleus pulposus cells, ceramic interaction provide guidance for the design of
we observed the formation of an apatite layer on the future biomaterials that enhance cell attachment, stimu-
bioactive glass surface with and without cells by day 21 late bone growth and ultimately integrate with the newly
[115]. However, the layer formed in the cultures with developed tissue bed. However, not every question re-
nucleus pulposus cells di!ered from the one formed in garding the successful use of bioactive ceramics as tissue
cultures with osteoblasts. Speci"cally, the layer was thin- engineering sca!old materials has yet been answered. It
ner and only poorly crystallized. Furthermore, the min- would appear that it is most pressing to elucidate the
eral was restricted to a layer of globules on the glass composition and structure of the mineral phase that
surface. With nucleus pulposus cells the function of this exquisitely a!ects the function of various biological mol-
layer as an attachment substratum is probably the preva- ecules, and the relationship between this inorganic phase
lent function. Accordingly, this would imply that the and the spatial structure of the adsorbing proteins.
layer binds "bronectin and other serum molecules, which
then mediate the initial attachment of the nucleus pul-
posus cells and facilitate their subsequent proliferation. Acknowledgements
In fact, FTIR analysis revealed carbonyl, ester and amide
group bands in the spectrum. Nucleus pulposus cells Support over the years from various agencies is grate-
were found to rapidly attach to the surface-treated glass fully acknowledged. Part of the work was supported
substrate, colonizing it within 12 h. They proliferated by the NIH (AR-40194, DE-10639, DE-13051), NSF
forming a lawn of cells and matrix by day 21. DNA (BCS-9202314, BCS-9309053), NASA (NAG9-817,
analysis showed an increase in cell number with time. NAG8-1483) and VA (1189-RA).
Among other markers, these cells expressed CD-44, a cell
surface glycoprotein that binds hyaluronic acid, which in
References
turn can bind to "bronectin [115]. All of these observa-
tions point to the qualities of surface modi"ed bioactive [1] Praemer A, Furner S, Rice DP. Musculoskeletal conditions in
glass as a carrier for nucleus pulposus cells in vitro, and the United States. Park Ridge: Am Acad Orthop Surg 1992.
suggest that glass-cell hybrids provide an exciting novel p. 1}85.
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2301

[2] Damien CJ, Parsons JR. Bone graft and bone graft substitutes: [20] El-Ghannam A, Ducheyne P, Shapiro IM. E!ect of serum pro-
a review of current technology and applications. J Appl Bio- tein adsorption on osteoblast adhesion to bioactive glass and
mater 1992;2:187}208. hydroxyapatite. J Orthop Res 1999;17:340}5.
[3] Glowacki J, Mulliken JB. Demineralized bone implants. Clin [21] Jarcho M, Kay JF, Gumaer KI, Doremus RN, Drobeck HP.
Plast Surg 1985;12(2):233}41. Tissue, cellular and subcellular events at a bone}ceramic hy-
[4] Buck BE, Malinin TI, Brown MD. Bone transplantation and droxylapatite interface. J Bioeng 1977;1:79}92.
human immunode"ciency virus. An estimate of risk of acquired [22] Ducheyne P, Hench L. Comparison of the skeletal "xation of
immunode"ciency syndrome (AIDS). Clin Orthop 1989; porous and bioreactive materials. Trans 1st Mtg Europ Soc
240:129}36. Biomater 1977:2PS}P0.
[5] Binderman I, Fin N. Bone substitutes-organic, inorganic, and [23] Ducheyne P, Hench LL, Kagan A, Martens M, Bursens A,
polymeric: cell material interactions. In: Yamamuro T, Hench Mulier JC. E!ect of hydroxyapatite impregnation on skeletal
LL, Wilson J, editors. CRC handbook of bioactive biomaterials. bonding of porous coated implants. J Biomed Mater Res 1980;
Boca Raton: CRC Press, 1990. p. 45}61. 14:225}37.
[6] Passuti N, Daculsi G, Martin S. Macroporous calcium-phos- [24] Berry JL, Geiger JM, Moran JM, Skraba JS, Greenwald AS. Use
phate ceramics for long bone surgery in human and dogs*clini- of tricalcium phosphate or electrical stimulation to enhance the
cal and histological studies. In: Heimke G, Soltesz U, Lee A, bone-porous implant interface. J Biomed Mater Res 1986;
editors. Clinical implant materials. Amsterdam: Elsevier, 1990. 20:65}77.
p. 255}8. [25] Eschenroeder Jr HC, McLaughlin RE, Reger SI. Enhanced sta-
[7] LeGeros RZ. Calcium phosphate materials in restorative den- bilization of porous-coated metal implants with tricalcium phos-
tistry: a review. Adv Dent Res 1988;2(1):164}80. phate granules. Clin Orthop Rel Res 1987;216:234}46.
[8] Ricci JL, Blumenthal NC, Spivak JM, Alexander H. Evaluation [26] Rivero DP, Fox J, Skipor AK, Urban RM, Galante JO. Calcium
of a low-temperature calcium-phosphate particulate implant phosphate-coated porous titanium implants for enhanced skel-
material: physical}chemical properties and in vivo bone re- etal "xation. J Biomed Mater Res 1988;22:191}201.
sponse. J Oral and Maxillofacial Surg 1992;50:969}78. [27] Mayor MD, Collier JB, Hanes CK. Enhanced early "xation of
[9] de Groot K, Tencer A, Waite P, Nichols J, Kay J. Signi"cance of porous-coated implants using tricalcium phosphate. Trans 32nd
the porosity and physical chemistry of calcium phosphate cer- ORS 1986. p. 340 (Abstract).
amics. Dental and other head and neck uses. In: Ducheyne P, [28] Cook SD, Thomas KA, Kay JF, Jarcho M. Hydroxyapatite-
Lemons J, editors. Bioceramics: materials characteristics versus coated porous titanium for use as an orthopedic biologic attach-
in vivo behavior. New York: NY: Academic Science, 1988. ment system. Clin Orthop Rel Res 1988;230:303}12.
p. 272}7. [29] Oonishi H, Yamamoto M, Ishimaru H, Tsuji E, Kushitani S,
[10] Schepers EJG, Ducheyne P, Barbier L, Schepers S. Bioactive Aono M, Ukon Y. The e!ect of hydroxyapatite coating on bone
glass particles of narrow size range: a new material for the repair growth into porous titanium alloy implants. J Bone Jt Surg
of bone defects. Implant Dent 1993;2:151}6. 1989;71:213}6.
[11] Schepers E, Declercq M, Ducheyne P, Kempeneers R. Bioactive [30] Ducheyne P, Beight J, Cuckler J, Evans B, Radin S. E!ect
glass particulate material as a "ller for bone lesions. J Oral of calcium-phosphate coating characteristics on early post-
Rehab 1991;18:439}52. operative bone tissue ingrowth. Biomaterials 1990;11:
[12] de Groot K. Ceramics of calcium phosphates: preparation and 531}40.
properties. In: de Groot K, editor. Bioceramics of calcium-phos- [31] Ducheyne P, Cuckler JM. Bioactive ceramic prosthetic coatings
phate. Boca Raton: CRC Press, 1983. p. 99}114. [review]. Clin Orthop Rel Res 1992;276:102}14.
[13] Van Raemdonck W, Ducheyne P, De Meester P. Calcium- [32] de Bruijn JD, Bovell YP, van Blitterswijk CA. Structural ar-
phosphate ceramics. In: Ducheyne P, Hastings GW, editors. rangements at the interface between plasma sprayed calcium-
Metal and ceramic biomaterials, vol. II. Strength and surface. phosphates and bone. Biomaterials 1994;15:543}50.
Boca Raton: CRC Press, 1984. p. 143}66. [33] de Bruijn JD, Flach TS, Leenders H, van de Brink J, van
[14] Ducheyne P, Lemons JE, editors. In: Bioceramics: materials Blitterswijk CA. Degradation and interface characteristics
characteristics versus in vivo behavior. New York: NY of plasma sprayed hydroxyapatite coatings with di!erent
Acadamic Science, 1988. crystallinities. In: Yamamuro T, Kokubo T, Nakamura T, edi-
[15] Hench L, Splinter R, Greenlee T, Allen W. Bonding mechanisms tors. Bioceramics, vol. 5. Kyoto: Kobunshi Kankokai, 1992.
at the interface of ceramic prosthetic materials. J Biomed Eng p. 291}8.
1971;2:117}41. [34] Maxian SH, Zawadski JP, Dunn MG. In vitro evaluation of
[16] Neo M, Nakaruma T, Yamamuro T, Ohtsuki C, Kokubo T. amorphous calcium-phosphate and poorly crystallized hy-
Transmission microscopic study of apatite formation on bioac- droxyapatite coating on titanium implants. J Biomed Mater Res
tive ceramics in vivo. In: Ducheyne P, Kokubo T, van Blitter- 1993;27:111}7.
swijk CA, editors. Bone-bonding biomaterials. Leiderdorp, [35] Ducheyne P, Radin S, King L. The e!ect of calcium-phosphate
The Netherlands: Reed Healthcare Communications, 1993. ceramic composition and structure on in vitro behavior.
p. 111}20. I. Dissolution. J Biomed Mater Res 1993;27:25}34.
[17] Kokubo T, Kushitani H, Sakka S, Kitsugi T, Yamamuro T. [36] Daculsi G, LeGeros RZ, Nery E, Lynch K, Kerebel B. Trans-
Solutions able to reproduce in vivo surface-structure changes in formation of biphasic calcium phosphate ceramics in vivo: ultra-
bioactive glass}ceramic A-W. J Biomed Mater Res 1990; structural and physicochemical characterization. J Biomed
24:721}34. Mater Res 1989;23:883}94.
[18] El-Ghannam A, Ducheyne P, Shapiro IM. Formation of surface [37] Holand W, Vogel W, Naumann K, Gummel J. Interface reac-
reaction products on bioactive glass and their e!ects on the tions between machinable bioactive glass}ceramics and bone.
expression of the osteoblastic phenotype and the deposition of J Biomed Mater Res 1985;19:303}12.
mineralized extracellular matrix. Biomaterials 1997; 18:295}303. [38] Ducheyne P, Bianco P, Radin S, Schepers E. Bioactive materials:
[19] Hyakuna K, Yamamuro T, Kotoura Y, Kakutani Y, Kitsugi T, mechanisms and bioengineering considerations. In: Ducheyne
Takagi H, Oka M, Kokubo T. The in#uence of calcium-phos- P, Kokubo T, van Blitterswijk CA, editors. Bone-bioactive bio-
phate ceramics and glass}ceramics on cultured cells and their materials. Leiderdorp, The Netherlands: Reed Healthcare Com-
surrounding media. J Biomed Mater Res 1989; 23:1049}66. munications, 1993. p. 1}12.
2302 P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303

[39] Klein CP, Driessen AA, de Groot K, van den Hoo! A. Biode- [58] Kitsugi T, Yamamuro T, Nakamura T, Kokubo T, Takagi M,
gradation behavior of various calcium phosphate materials in Shibuya T, Takeuchi H, Ono M. Bonding behavior between two
bone tissue. J Biomed Mater Res 1983;17:769}84. bioactive ceramics in vivo. J Biomed Mater Res 1987;21:1109}23.
[40] LeGeros RZ, Daculsi G, Orly I, Gregoire M, Heughebaert M, [59] Li P, Bakker D, van Blitterswijk CA. A bone-bonding polymer
Gineste M, Kijkowska R. Formation of carbonate apatite on (Polyactive(R) 80/20) induces hydroxycarbonate apatite forma-
calcium phosphate materials: dissolution/precipitation pro- tion in vitro. J Biomed Mater Res 1997;34:79}86.
cesses. In: Ducheyne P, Kokubo T, van Blitterswijk CA, editors. [60] Levene H, Phuvanartnuruks V, Abramson S, James K, Kohn J.
Bone-bonding biomaterials. Netherlands: Reed Healthcare Chelation of calcium ions by some tyrosine-derived polymers
Communications, 1993. p. 201}12. may be related to improved bone biocompatibility. Trans 25th
[41] Patka P, den Hollander W, Klein CPAT, de Groot K. Behavior Mtg SFB 1999. p. 13 (Abstract).
of ceramics, hydroxyapatite, and tricalcium phosphate in bone. [61] Li P, Ducheyne P. Quasi-biological apatite "lm induced on tita-
Trans Mtg SFB 1989;15:103 (Abstract). nium in a simulated body #uid. J Biomed Mater Res 1998;41:341}8.
[42] Radin SR, Ducheyne P. The e!ect of calcium phosphate ceramic [62] Li P, Ohtsuki C, Kokubo T, Nakanishi K, Soga N, de Groot K.
composition and structure on in vitro behavior. II. Precipita- The role of hydrated silica, titania, and alumina in inducing
tion. J Biomed Mater Res 1993;27:35}45. apatite on implants. J Biomed Mater Res 1994;28:7}15.
[43] Andersson OH, Karlsson KH, Kangasniemi K. Calcium-phos- [63] Cho S-B, Nakanishi K, Kokubo T, Soga N, Ohtsuki C,
phate formation at the surface of bioactive glass in vivo. J Non- Nakamura T. Apatite formation on silica gel in simulated body
Cryst Solids 1990;119:290}6. #uid: its depedence on structures of silica gels prepared in
[44] Ducheyne P, Kim CS, Pollack SR. The e!ect of phase di!erences di!erent media. J Biomed Mater Res 1996;33:145}51.
on the time-dependent variation of the zeta potential of hy- [64] Li P, Nakanishi K, Kokubo T, de Groot K. Induction and
droxyapatite. J Biomed Mater Res 1992;26:147}68. morphology of hydroxyapatite, precipitated from metastable
[45] Ohgushi H, Goldberg VM, Caplan AI. Heterotopic osteogenesis simulated body #uids on sol}gel prepared silica. Biomaterials
in porous ceramics induced by marrow cells. J Orthop Res 1993;14:963}8.
1989;7:568}78. [65] Li P, de Groot K. Calcium phosphate formation within sol}gel
[46] Gregoire M, Orly I, Manankau J. The in#uence of calcium- prepared titania in vitro and in vivo. J Biomed Mater Res
phosphate biomaterials on human bone cell activites. An in vitro 1993;27:1495}500.
approach. J Biomed Mater Res 1990;24:165}77. [66] Pereira MM, Clark AE, Hench LL. Calcium-phosphate forma-
[47] Vrouwenvelder WC, Groot CG, de Groot K. Histological and tion on sol}gel-derived bioactive glasses in vitro. J Biomed
biochemical evaluation of osteoblasts cultured on bioactive Mater Res 1994;28:693}8.
glass, hydroxylapatite, titanium alloy, and stainless steel. J Bio- [67] Li R, Clark AE, Hench LL. An investigation of bioactive
med Mater Res 1993;27:465}75. glass powders by sol}gel processing. J Appl Biomater 1991;
[48] de Bruijn JD, Davies JE, Klein CPAT, de Groot K, van Blitter- 2:231}9.
swijk CA. Biological responses to calcium-phosphate ceramics. [68] Hench LL. Bioceramics: from concept to clinic. J Am Ceram Soc
In: Ducheyne P, Kokubo T, van Blitterswijk CA, editors. Bone- 1991;74(7):1487}510.
bonding biomaterials. Leiderdorp, The Netherlands: Reed [69] E!ah-Kaufman EAB, Ducheyne P, Radin S, Bonnell DA, Com-
Health-care Communications, 1993. p. 57}72. posto RJ. Immersion-induced changes of bioactive glass surfaces
[49] El-Ghannam A, Ducheyne P, Shapiro IM. Bioactive material in the presence or absence of serum proteins. J Biomed Mater
template for in vitro synthesis of bone. J Biomed Mater Res Res 1999, submitted for publication.
1995;29:359}70. [70] Radin S, Ducheyne P, Rothman B, Conti A. The e!ect of in vitro
[50] Keeting PE, Oursler MJ, Weigand KE, Bonde SK, Spelsberg modeling conditions on the surface reactions on bioactive glass.
TC, Riggs BL. Zeolite A increases proliferation, di!erentiation, J Biomed Mater Res 1997;37:363}75.
and transforming growth factor beta production in normal adult [71] Radin S, Ducheyne P, Falaize S, Hammond A. Transformation
human osteoblast-like cells in vitro. J Bone Miner Res of bioactive glass granules into Ca}P shells in vitro. J Biomed
1992;7:1281}9. Mater Res 1999, in press.
[51] Matsuda T, Davies JE. The in vitro response of osteoblasts to [72] Boutin PB, Testerman MC, Wheeler DL, Miller GJ, Schultz GS,
bioactive glass. Biomaterials 1987;8:275}84. Strates BS, Petty RW. Osteogenic e!ects of transforming
[52] Kokubo T, Ito S, Haung ZT, Hayashi T, Sakka S, Kitsugi T, growth-factor-b (TGF-b) with Hydroxyapatite. J Bone Miner
Yamamuro T. Ca}P-rich layer formed on high strength bioac- Res 1993;8:S237.
tive glass}ceramic A-W. J Biomed Mater Res 1990;24:331}43. [73] Johnsson MS-A, Paschalis E, Nancollas GH. Kinetics of min-
[53] Radin S, Ducheyne P. The e!ect of bioactive ceramic composi- eralization, demineralization, and transformation of calcium-
tion and structure on in vitro behavior: III porous versus dense phosphates at mineral and protein surfaces. In: Davies JE,
ceramics. J Biomed Mater Res 1994;28:1303}9. editor. Bone}biomaterial interface. Toronto: University of
[54] Tracy BM, Doremus RH. Direct electron microscopy studies of Toronto Press, 1991. p. 62}75.
the bone}hydroxylapatite interface. J Biomed Mater Res [74] Martin RI, Brown PW. Formation of hydroxyapatite in serum.
1984;18:719}26. Mater Med 1994;5:96}102.
[55] Davies JE, Pilliar RM, Smith DC, Chernecky R. Bone interfaces [75] Ohgushi H, Okumura M, Tamai S, Shors EC, Caplan AI. Mar-
with retrieved alumina and hydroxyapatite ceramics. In: Bon- row cell induced osteogenesis in porous hydroxyapatite and
"eld W, Towner KE, Hastings G, editors. Bioceramics, vol. 4. tricalcium phosphate: a comparative histomorphometric study
Oxford: Butterworth, 1991. p. 199}204. of ectopic bone formation. J Biomed Mater Res 1990;24:1563}70.
[56] Schepers E, Ducheyne P. The application of bioactive glass [76] Goshima J, Goldberg VM, Caplan AI. The origin of bone
particles of narrow size range as a "ller materials for bone formed in composite grafts of porous calcium-phosphate ce-
lesions: a 24 month animal experiment. In: Christiansen D, ramic loaded with marrow cells. Clin Orthop 1991;269:274}83.
Ducheyne P, editors. Bioceramics, vol. 6. Oxford: Butterworth- [77] Dennis JE, Haynesworth SE, Young RG, Caplan AI. Osteogen-
Heinemann, 1993. p. 401}4. esis in marrow-derived mesenchymal cell porous ceramic com-
[57] Radin S, Ducheyne P. The e!ect of serum proteins on solution posites transplanted subcutaneously: e!ect of "bronectin and
induced surface transformations of bioactive ceramics. J Biomed laminin on cell retention and rate of osteogenic expression. Cell
Mater Res 1996;30:273}80. Transplant 1992;1:23}32.
P. Ducheyne, Q. Qiu / Biomaterials 20 (1999) 2287}2303 2303

[78] Nakahara H, Goldberg VM, Caplan AI. Culture-expanded per- [99] Vrouwenvelder WC, Groot CG, de Groot K. Better histology
iosteal-derived cells exhibit osteochondrogenic potential in por- and biochemistry for osteoblasts cultured on titanium-doped
ous calcium-phosphate ceramics in vivo. Clin Orthop Rel Res bioactive glass: bioglass 45S5 compared with iron-, titanium-,
1992:291}8. #uorine- and boron-containing bioactive glasses. Biomaterials
[79] Okumura J, Ohgushi H, Ducheyne P. Porous titanium felt as 1994;15:97}106.
a carrier of osteogenic cells. In: Ducheyne P, Christiansen D, [100] Gross U, Brandes J, Strunz V, Bab I, Sela J. The ultrastructure of
editors. Bioceramics, vol. 6. Oxford: Butterworths-Heinemann, the interface between a glass}ceramic and bone. J Biomed Mater
1993. p. 305. Res 1981;15:291}305.
[80] Gatti AM, Ducheyne P, Piattelli A, Schepers E, Trisi L, Chiarini [101] El-Ghannam A, Ducheyne P, Shapiro I. Bioactive glass tem-
L, Monari E. Glass corrosion layers on bioactive glass granules plates for the synthesis of bone-like tissue in vitro. In: Mikos
of uniform size a!ect cellular function. In: Ducheyne P, Chris- AG, Murphy RM, Bernstein H, Peppas NA, editors. Bio-
tiansen D, editors. Bioceramics, vol. 6. Oxford: Butterworth- materials for cell and drug delivery. Pittsburgh: Materials Re-
Heinemann, 1993. p. 395}400. search Society, 1994. p. 257}62.
[81] Furusawa T, Mizunuma K. Osteoconductive properties and [102] Hench LL. Bioactive ceramics. In: Ducheyne P, Lemons JE,
e$cacy of resorbable bioactive glass as a bone-grafting material. editors. Bioceramics: material characteristics versus in vivo be-
Implant Dent 1997;6:93}101. havior. New York: The New York Academy of Sciences, 1988.
[82] Hench LL, Ethridge EC. Biomaterials: an interfacial approach. p. 54}71.
New York: Plenum Press, 1982. [103] Garcia AJ, Ducheyne P, Boettiger D. E!ect of surface reaction
[83] Schepers E, Ducheyne P, Declercq M. Interfacial analysis of stage on "bronection-mediated adhesion of osteoblast-like cells
"ber-reinforced bioactive glass dental root implants. J Biomed to bioactive glass. J Biomed Mater Res 1998;40:48}56.
Mater Res 1989;23:735}52. [104] Grinnell F, Feld MK. Adsorption characteristics of plasma
[84] Schepers EJ, Ducheyne P. Bioactive glass particles of narrow "bronectin in relationship to biological activity. J Biomed Mater
size range for the treatment of oral bone defects: a 1}24 month Res 1981;15:363}81.
experiment with several materials and particle sizes and size [105] El-Ghannam A, Ducheyne P, Shapiro IM. Porous bioactive
ranges. J Oral Rehab 1997;24(3):171}81. glass and hydroxyapatite ceramic a!ect bone cell function in vitro
[85] Low SB, Fetner AE, Clark AE, Hench LL, Wilson-Hench J. along di!erent time lines. J Biomed Mater Res 1997;36:167}80.
Periodontal osseous defect repair. US Patent 1989, Patent [106] Chesmel K, Beight J, Rothman R, Tuan R. TGF-beta enhances
d 4815046. osseointegration in vivo. In: Ducheyne P, Christiansen D, edi-
[86] Wilson J, Low SB. Bioactive ceramics for periodontal treatment: tors. Bioceramics, vol. 6. Oxford: Butterworth-Heinemann Ltd,
comparative studies in the patus monkey. J Appl Biomater 1993. p. 21}6.
1992;3:123}9. [107] Beck LS, Deguzman L, Lee WP, Zioncheck TF, Osaka G,
[87] Ripamonti U. The morphogenesis of bone in replicas of porous Nguyen T, Ongpipattanakul B, Gorrel J, Aufdemorte TB,
hydroxyapatite obtained from conversion of calcium carbonate Plouhar PL. TGF-b1 bound to tricalcium phosphate persists at
exoskeletons of coral. J Bone Jt Surg 1991;73A:692}703. segmental radial defects and induces bone formation. Trans 41st
[88] Ripamonti U. Osteoinduction in porous hydroxyapatite im- Ann Meet ORS 1995;20:593 (Abstract).
planted in heterotopic sites of di!erent animal models. Bio- [108] Santos EM, Radin S, Ducheyne P. Sol}gel derived carrier for
materials 1996;17:31}5. the controlled release of proteins. Biomaterials 1999;
[89] Klein C, de Groot K, Chen W, Li Y, Zhang X. Osseous sub- 20:1695}700.
stance formation induced in porous calcium-phosphate ceram- [109] Nicoll SB, Radin S, Santos EM, Tuan RS, Ducheyne P. In vitro
ics in soft tissues. Biomaterials 1994;15:31}4. release kinetics of biologically active transforming growth factor
[90] de Bruijn JD, Dalmeijer R, de Groot K. Osteoinduction by b1 from a novel xero-gel carrier. Biomaterials 1997;18:853}9.
microstructured calcium-phosphates. Trans 25th Mtg SFB [110] Downes S, DiSilvio L, Klein CPAT, Kayser MV. Growth hor-
1999;22:235 (Abstract). mone loaded bioactive ceramics. J Mater Sci: Mater Med
[91] Toth JM, Lynch KL, Hackbarth DA. Ceramic-induced osteogen- 1991;2:176}80.
esis following subcutaneous implantation of calcium-phosphates. [111] Santos EM, Radin S, Shenker BJ, Shapiro IM, Ducheyne P.
In: Ducheyne P, Christiansen D, editors. Bioceramics, vol. 6. Si}Ca}P xerogels and bone morphogenetic protein act synergis-
Philadelphia: Butterworth-Heinemann, 1993. p. 9}14. tically on rat stromal marrow cell di!erentiation in vitro. J Bio-
[92] Urist MR. Bone: formation by autoinduction. Science 1965; med Mater Res 1998;41:87}94.
150:893}9. [112] Ripamonti U, Reddi AH. Tissue engineering, morphogenesis,
[93] GarcmH a AJ, Ducheyne P. Numerical analysis of extracellular #uid and regeneration of the periodontal tissues by bone mor-
#ow and chemical species transport around and within porous phogenetic proteins. Crit Rev Oral Biol Med 1997;8:154}63.
bioactive glass. J Biomed Mater Res 1994;28:947}60. [113] Livingston T, Ducheyne P, Garino J. Mechanical and histologi-
[94] Brighton CT, Scha!er JL, Shapiro DB, Tang JS, Clark CC. cal evaluation of a tissue engineered bioactive sca!old in vivo.
Proliferation and macromolecular synthesis by rat calvaria bone In: Ohgushi H, Hastings GW, Yoshikawa T, editors. Bio-
cells grown in various oxygen tension. J Orthop Res 1991;9:847}54. ceramics, vol. 12. Singapore: World Scienti"c Publishing Co.,
[95] Brighton CT, McCluskey WP. Response of cultured bone cells 1999. p. 245}8.
to a capacitively coupled electric "eld: inhibition of cAMP [114] Lehmann TR, Spratt KF, Tozzi JE, Weinstein JN, Reinarz SJ,
response to parathyroid hormone. J Orthop Res 1987;6:567}71. el-Khoury GY, Colby H. Long-term follow-up of lower lumbar
[96] Scha!er JL, Brighton CT, Clark CC, Brinly FJ. In vitro growth fusion patients. Spine 1987;12(2):97}104.
of rat calvaria bone cells at various oxygen tensions. Trans [115] Gan JC, Ducheyne P, Vresilovic E, Shapiro IM. Bioactive glass
Orthop Res Soc 1987;12:176. serves as a substrate for maintenance of phenotype of nucleus
[97] Spindler KP, Shapiro DB, Gross SB, Brighton CT, Clark CC. pulposus cells of the intervertebral disk. J Biomed Mater Res
The e!ect of ascorbic acid on the metabolism of rat calvaria 1999, submitted for publication.
bone cells in vitro. J Orthop Res 1989;7:696}701. [116] Schepers E, Pinruethai P. A comparative study of bioactive glass
[98] Vrouwenvelder WC, Groot CG, de Groot K. Behaviour of fetal and hydroxyapatite particles in periodontal bone lesions. In:
rat osteoblasts cultured in vitro on bioactive glass and nonreac- Ducheyne P, Christiansen D, editors. Bioceramics, vol. 6. Ox-
tive glasses. Biomaterials 1992;13(6):382. ford: Butterworth-Heinemann, 1993. p. 113}6.

You might also like