You are on page 1of 9

Materials Science & Engineering C 83 (2018) 99–107

Contents lists available at ScienceDirect

Materials Science & Engineering C


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

Review

Mesoporous silica-based bioactive glasses for antibiotic-free antibacterial T


applications

Seray Kayaa, Mark Cresswellb, Aldo R. Boccaccinia,
a
Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
b
Lucideon Ltd., Stoke-on-Trent, ST4 7LQ, UK

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

Keywords: Bioactive glasses (BGs) are being used in several biomedical applications, one of them being as antibacterial
Mesoporous materials. BGs can be produced via melt-quenching technique or sol-gel method. Bactericidal silver-doped sol-
Antibacterial gel derived mesoporous silica-based bioactive glasses were reported for the first time in 2000, having the
Bioactive glass composition 76SiO2-19CaO-2P2O5-3Ag2O (wt%) and a mean pore diameter of 28 nm. This review paper dis-
Sol-gel technique
cusses studies carried out exploring the potential antibacterial applications of drug-free mesoporous silica-based
Antibiotic-free
BGs. Bioactive glasses doped with metallic elements such as silver, copper, zinc, cerium and gallium are the point
of interest of this review, in which SiO2, SiO2-CaO and SiO2-CaO-P2O5 systems are included as the parent glass
compositions. Key findings are that silica-based mesoporous BGs offer a potential alternative to the systemic
delivery of antibiotics for prevention against infections. The composition dependent dissolution rate and the
concentration of the doped elements affect the antibacterial efficacy of BGs. A balance between antibacterial
activity and biocompatibility is required, since a high dose of metallic ion addition can cause cytotoxicity.
Typical applications of mesoporous BGs doped with antibacterial ions include bone tissue regeneration, multi-
functional ceramic coatings for orthopedic devices and orbital implants, scaffolds with enhanced angiogenesis
potential, osteostimulation and antibacterial properties for the treatment of large bone defects as well as in
wound healing.

1. Introduction Various amounts of other oxides are incorporated in silicate, borate


or phosphate BGs to impart particular properties to the material; for
The first bioactive glass (BG) was invented by Prof. Larry Hench at instance, CaO, K2O, Na2O and MgO are useful to adjust the surface
the University of Florida in 1969 [1]. This bioactive glass has a com- reactivity in the biological environment; ZnO, CuO and Ag2O allow the
position of 46.1SiO2-24.4Na2O-26.9CaO-2.6P2O5 (mol%), it was later release of biologically active ions with antibacterial properties [11].
termed 45S5 Bioglass®, and exhibits as a key property the formation of BGs can be produced by two different methods; namely the classic
a bond with bone [1,2]. From a compositional viewpoint, bioactive oxide melting procedure and the sol-gel method. Since sol-gel BGs are
glasses can be basically divided into three groups, depending on the more bioactive and bioresorbable [12–14] than melt-quenched glasses,
representative network former oxide present in the formulation, i.e., in some applications the use of sol-gel BGs is preferred. The sol-gel
SiO2-based (silicate), B2O3-based (borate) and P2O5-based (phosphate) method has advantages of compositional purity and molecular mixing,
systems. The first group comprises a wide range of glass formulations, allowing control of the sample porosity by changing the processing
including 45S5 Bioglass® and other typical compositions such as 1393 conditions and/or by adding suitable templates [15]. The use of tem-
BG (wt%: 53SiO2-6Na2O-12K2O-5MgO-20CaO-4P2O5) [3–5] and S53P4 plates during the sol-gel process generates products with an ordered
BG (53%SiO2-23%Na2O-20%CaO-4%P2O5) [6–8] commercially named mesoporous structure which induces higher bioactivity with respect to
as BonAlive® (BonAlive Biomaterials, Turku, Finland). melt-quenched glasses [16] and makes them suitable for use in drug
Borate glasses are characterized by their higher reactivity in com- delivery applications [17–20].
parison to silica-based glasses, which results in faster bioactive kinetics Bone reconstruction surgeries can be compromised by osteomyelitis
[5,9]. Phosphate glasses are resorbable materials and their dissolution caused by bacteria infection and to treat these, systemic antibiotic ad-
rate can be tuned according to their oxide composition [10]. ministration, surgical debridement, wound drainage and implant


Corresponding author.
E-mail address: aldo.boccaccini@ww.uni-erlangen.de (A.R. Boccaccini).

https://doi.org/10.1016/j.msec.2017.11.003
Received 31 May 2017; Received in revised form 23 August 2017; Accepted 9 November 2017
Available online 10 November 2017
0928-4931/ © 2017 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

removal are applied [21]. These methods are not always effective, so glasses) and MBGs (Mesoporous bioactive glasses). In addition, the
the patients may require subsequent surgeries to combat bacterial in- schematic diagram in Fig. 2 (right) gives information about the biolo-
fections. Introduction of a local drug release system into the implant gical function of some metallic ions that can be added to MBG com-
site can be another approach to treat the problem [22]. This treatment positions to enhance their biological functionalities [37].
offers high drug delivery efficiency, continuous action, reduced toxicity Prevention against bacterial infection is an essential need in or-
and convenience to the patient [21,23]. Mesoporous bioactive glasses thopedic surgery due to the significant medical complications to pa-
(MBGs) are highly attractive for such applications. tients related to infections [38]. Although strict hygienic protocols and
In 2004, for the first time, Yan et al. [20] developed MBGs with the preventive antibiotic prophylaxes have drastically reduced the per-
combination of sol-gel and surfactant templating methods. The ob- centage of postoperative infections, many bacterial species have de-
tained MBG particles were around several tens of micrometers in dia- veloped selective resistance against antibiotics that can cause serious
meter, with highly ordered 5 nm mesopore channels. In comparison infections, which are hard to recover from and usually lead to second
with non-mesoporous bioactive glasses (NBGs), MBGs have enhanced operations.
surface area, higher pore volume, better ability to induce in vitro An alternative to the systemic delivery of antibiotics is the use of
apatite mineralization in simulated body fluid (SBF) and excellent cy- orthopedic devices or prostheses made from synthetic materials with
tocompatibility [24–27]. intrinsic antibacterial properties [39–42]. In this context, silica-based
For the preparation of MBGs, the addition of structure-directing bioactive glasses offer a potential alternative to the systemic delivery of
agents (e.g. Pluronic P123, F127 and cetrimonium bromide, CTAB) is antibiotics for prevention against bacterial infections. The antibacterial
essential to obtain well-ordered pore structures. Under appropriate properties of bioactive glasses have been investigated by Stoor et al.
synthesis conditions, these agents self-organize into micelles. Micelles [43–45] from the clinical point of view. According to their study on
link the hydrolysed silica precursors through the hydrophilic compo- S53P4 BG, in an aqueous environment, ions (Ca2 +, Na+, PO43 −, and
nent and self-assemble to form an ordered mesophase [18,28]. Then, Si4 +) are released from the S53P4 BG which results in a rise in pH and
the mixture reaction system undergoes evaporation-induced self-as- osmotic pressure in its vicinity. These factors potentially influence the
sembly (EISA) process. Once the mixture is dry and the surfactant has viability of oral microorganisms at the dentogingival margin indicating
been removed, a well-ordered mesoporous structure is obtained, having potential for use in dental applications. The antibacterial effects of a
high surface area and high porosity. The basic process to produce me- paste made of a bioactive glass (S53P4) on oral microorganisms were
soporous silica is illustrated in Fig. 1 [28]. examined [43]. It was found that Actinomyces naeslundii bacteria lost its
In 2006 and 2008 MBG powders with 58S and 77S compositions viability within 10 min when exposed to calcium, phosphorus, silicon
having excellent in vitro bioactivity were prepared by hydrothermal and sodium dissolution products. The loss of viability was 60 min for
treatment using P123 [19,29]. By using the same method, in 2008 Li Actinobacillus actinomycetemcomitans, Porphyromonas gingivalis, and
et al. [30] prepared Mg, Zn or Cu containing MBG particles. Later in Streptococcus mutans bacteria. Actinobacillus actinomycetemcomitans has
2010 CaO-SiO2 MBG particles were synthesized for hemostatic appli- been suggested to play a role in juvenile periodontitis [46,47], Por-
cations [31]. phyromonas gingivalis has been associated with destructive periodontal
There is a general trend to upgrade the properties of MBGs with lesions in adults [48–50] and Streptococcus mutans is considered to play
some of the well-known therapeutic ions [11,32–34]. By substituting a major role in caries [51].
small amounts of oxides, the osteogenesis, antibacterial capacity, an- Indeed the antibacterial effect of BGs is used in clinical settings to
giogenesis or cementogenesis [34–36] effects of MBGs can be improved. combat osteomyelitis [52–55], an infection of bone and bone marrow.
Fig. 2 (left) illustrates the year of discovery and the average time Polymethyl methacrylate (PMMA) beads mixed with antibiotics have
required for the in vitro bioactive response of three different silicate been used in the treatment of osteomyelitis [56]. However, after two
glass families, MPGs (melt-prepared glasses), SGGs (sol-gel bioactive weeks of insertion into the bone defect, these beads must be removed by

Fig. 1. Steps required to synthesize mesoporous silica from


a micellar solution, according to Arcos and Vallet-Regi [28].
(Reproduced with permission of Elsevier).

100
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

Fig. 2. (Left): Discovery years of three bioactive silicate glass types


showing the average time required for achieving bioactivity, de-
termined by the formation of HA on the surface upon immersion in
SBF. (Right): Typical metallic ions added to these glasses and their
biological functions.
(Modified from ref. [37]).

surgery [57], since beads release about 24% ( ± 11%) of their anti- free BGs which achieve antibacterial effects exclusively by the release
biotic content (mini-beads even up to 93 ± 1.4%) [58]. Besides the of metallic ions. Therefore the aim of this review is to summarize the
antibiotic release issue, unexpected bone loss can occur when these information available in the open literature on the antibacterial prop-
beads are used [59]. erties of mesoporous silica-based bioactive glasses with emphasis on the
Having discovered that S53P4 bioactive glass has antimicrobial specific antibacterial effects of therapeutic ionic dissolution products
properties [4,7,54], this glass took the place of polymer beads in the without any drugs or antibiotics added.
treatment of chronic osteomyelitis [53,54,60]. Polymer beads are ef- The studies reviewed in this article consider thus the antibacterial
fective against osteomyelitis treatment when they are mixed with an- activity of sol-gel derived SiO2-CaO and SiO2-CaO-P2O5 mesoporous
tibiotics; however in this case they have to be removed by surgery due bioactive glasses based on incorporation of antibacterial metallic ions
to their release of antibiotics and bone loss issues [58,59]. Compared into the glass structure. The composition dependent dissolution rate
with polymer beads, bioactive glasses are osteoconductive and bio- and the concentration of the doped elements affect the antibacterial
compatible materials [60]. In addition to their potential use in bone and efficiency of bioactive glasses. Studies have shown that a balance be-
tissue repair applications, BGs provide antibacterial and blood vessel tween antibacterial activity and biocompatibility is required since a
promoting properties [11,45]. high dose of metallic ion addition can lead to cytotoxicity [76,77],
A range of polymer based antibacterial carriers is available which these aspects are also addressed in this review.
could be combined with BGs for achieving improved antibacterial ef-
fects. For example, cationic proteolytic-resistant polymers with poly-
amide backbones have been tested for their antimicrobial activities and 2. Mesoporous silica-based bioactive glasses doped with
their cytotoxicity against primary human dermal fibroblasts [61]. These antibacterial elements
polymers were demonstrated to elicit fast bactericidal activity against
A. baumanii, E. coli, K. pneumonia and P. aeruginosa and were non-toxic 2.1. Silver doped-MBGs
to epithelial cells which made them safe and potentially useable as
broad spectrum ophthalmic antibiotics. Gelatin and antifungal drug- Silver ion has been the most studied metal ion as a biocide (defined
loaded gelatin fiber mats have been produced via electrospinning in the European legislation as a chemical substance or microorganism
method [62]. Due to their cell compatibility and antifungal properties, intended to destroy, deter, render harmless, or exert a controlling effect
electrospun antimicrobial nanofibers can be used in several applica- on any harmful organism by chemical or biological means), as it pre-
tions, as scaffolds in controlled drug delivery, wound dressings, tissue sents a broad and strong antimicrobial behavior [78–80]. Ag has no
engineering, stem cell regeneration and differentiation, and also in food toxicity effect to human cells in suitably low concentrations, and it can
packaging [63–66]. In addition, gentamicin has been incorporated into provide bioactive glasses with extra functionality in addition to their
a layer by layer (LbL) coating with clay interlayer barrier to achieve a inherent osteoconductivity and bone bonding ability [81–85]. As me-
controlled bactericidal effect [67–69]. However, the combination of tallic silver reacts with moisture on the skin surface or with wound
such polymeric substrates (antibiotic carriers) with BGs to exploit po- fluids, silver ions are released that damage bacterial RNA and DNA,
tential synergistic effect of antibiotics and antibacterial ions has not hence inhibiting replication. In the case of silver, inactivation of critical
been investigated. enzymes of the respiratory chain (e.g. succinate dehydrogenase) by
In general terms, three approaches have been used to investigate the metal binding to thiol groups and induction of hydroxyl radicals appear
antibacterial activity of bioactive glasses. The first one is based on to play a major role [86].
bioactive glasses that change the local physiological conditions when Silver is potentially cytotoxic if released in great amounts and thus,
implanted in the body. The second one includes doping the bioactive strict control of the introduced Ag amount is necessary. It is well-known
glasses with antibacterial metallic elements, so that the glass degrades that surface functionalization of glasses can occur by ion-exchange
by releasing ions and induces the bactericidal effect. The last approach treatments while enabling the bulk structure and main properties to
investigates antibiotic added to bioactive glasses [17,34,35,70–72]. remain unaltered. In such a way silver can be introduced by ion ex-
Moreover, mesoporous bioactive glasses, due to their ordered meso- change in the outer layers of glass surfaces, conferring to them anti-
pores which can incorporate biomolecules and drugs, are suitable to bacterial properties while maintaining the intrinsic BG characteristics
achieve the synergistic release of drugs and antibacterial metallic ions [76,77,87].
to increase the effectiveness against bacteria and to impart other bio- A wide range of silver doped silica glasses (Ag-BG) have been de-
logical effects [73–75]. veloped by sol-gel methods [81,82,88–95] to create bioactive glasses
Considering the interest in combatting infections with minimal use exhibiting inhibitory effects on bacterial growth.
of antibiotics [33,38], however, it is important to investigate antibiotic- Among these studies, BGs exhibiting mesoporous structure were
produced via sol-gel methods [81,82,92] and via a structure-directing

101
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

agent assisted sol-gel technique [94,96,97]. mesoporous bioactive glasses have already been proven in many studies
In this context, incorporation of Ag2O into sol-gel produced bioac- [95,101,102]. Due to their controllable dissolution rate, soluble glasses
tive glass compositions (45S5 BG: 46.1SiO2-24.4Na2O-26.9CaO- represent an attractive delivery system for antibacterial copper ions.
2.6P2O5 (mol%)) has been reported to minimize the risk of microbial In one of these relevant investigations, 62.3SiO2–28.9CaO-8.6P2O5
contamination through the leaching of Ag+ ions [81,82]. (wt%) sol-gel produced bioactive glass was doped with 4.7 and 9 wt%
In 2000, [81] the incorporation of 3 wt% Ag2O in 76SiO2-19CaO- CuO to investigate its behavior against E. coli DH5α ampicillin-resistant
5P2O5 (wt%) sol-gel produced mesoporous bioactive glass was shown to and S. mutans bacteria [95]. These Cu-doped glasses had MBC of
exhibit a bacteriostatic effect on Escherichia coli (E. coli) with 0.01% 100–150 mg/ml for E. coli and 5–10 mg/ml for S. mutans bacteria. The
reduced bacterial growth without compromising the glass bioactivity. Cu release in SBF from 4.7 wt% CuO doped MBG was in the range
Sol-gel derived 3 wt% Ag2O incorporated mesoporous silica bioactive 20–60 ppm and for 9 wt% doped ones, it was in the range 20–80 ppm.
glass in the SiO2-CaO-P2O5 system showed good antibacterial property A further study [101] was carried out to prepare structure directing
against E. coli, Pseudomonas aeruginosa (P. aeruginosa) and Staphylo- agent assisted sol-gel derived copper-doped (molar: 0, 1, 2 and 5%)
coccus aureus (S. aureus). The effect was attributed to the leaching of MBG scaffolds. These scaffolds were shown to possess good bio-
silver ions from the glass matrix [82]. It has been confirmed that Ag+ compatibility with no significant cytotoxic effect on human bone
ions in concentrations of 0.05–0.20 mg/ml released from Ag-doped BGs marrow stromal cells (hBMSC) and in addition the incorporation of
inhibited the growth of different bacterial strains (E. coli, P. aeruginosa 5 mol% Cu induced a significant antibacterial effect against E. coli. The
and S. aureus) [82]. released Cu2 + ions from MBG scaffolds significantly inhibited the
In the study of Saravanapavan et al. [88], the introduction of Ag2O viability of Escherichia coli, as shown in Fig. 3 [101].
into sol-gel derived bioactive glass compositions was aimed at mini- A glass system made by sol-gel method with the composition
mising the risk of microbial contamination through the potential anti- 75SiO2-(25-x)CaO-5P2O5-xCuO (x = 0, 2, 5 mol%) was incorporated
microbial activity of the leaching Ag+ ions. It was shown that the into a nanocoating to produce nanocomposite films for wound healing
bioactive glass doped with Ag2O was bacteriostatic and elicited a rapid applications [102]. 5 mol% CuO doped films had better antibacterial
bactericidal reaction. activity than pure and 2 mol% CuO doped films against E. coli, due to
Another study reported that 60SiO2-2Ag2O-34CaO-4P2O5 (mol%) increased Cu ion release into DMEM medium after 7 days (0.0491 ppm
bioactive glass produced by sol-gel method was suitable to coat surgical vs 0.0391 ppm, respectively).
sutures. These sutures were shown to be bioactive, and had anti- For the development of wound dressings, prevention against bac-
microbial and bactericidal properties against Staphylococcus epidermidis terial infections and enhancement of angiogenesis are the key issues
(S. epidermidis) [89,90]. Moreover, 1 mol% Ag2O incorporated sol-gel which have to be taken into account. Wounds heal faster with the
derived 70S30C (70SiO2-30CaO) (mol%) bioactive glass scaffold was transportation of nutrients and removal of waste products from the
produced, which released 0.95 μg/ml of Ag+ ions in 23 h, giving a tissues; therefore new blood vessel formation (angiogenesis) is very
bactericidal effect on E. coli, P. aeruginosa and S. aureus cultures [91]. important [103,104]. When a wound is infected by bacteria, the process
A study comparing the antibacterial activity of a sol-gel derived of healing will take longer [105]; which has to be prevented or treated
silver-doped 64SiO2-26CaO-10P2O5 (mol%) BG with its undoped with appropriate biomaterials like BGs [102]. In terms of these re-
counterpart against E. coli species showed that the silver-free BG had quirements, Cu-BG nanocomposite films are promising biomaterials for
neither bacteriostatic nor bactericidal effects; however 5 mol% Ag2O wound dressing applications also providing antibacterial properties.
doped BG was found to have > 99% killing efficiency towards E. coli Composites of nanofibrillated cellulose (NFC) and Cu doped (2 and
[92]. In related studies, 4 and 8 wt% Ag2O were added to 5 mol%) MBG which have molar ratios of Si/Ca/P = 80/15/5 were
62.3SiO2–28.9CaO-8.6P2O5 (wt%) sol-gel obtained bioactive glass to developed in aerogel form [106]. After successful production of these
investigate and to compare their antibacterial behavior with that of composites, their dissolution behavior, mineralization, cytotoxicity,
undoped BG against E. coli DH5α ampicillin-resistant and Streptococcus angiogenic performance and the antibacterial properties were ex-
mutans (S. mutans) [95]. amined against E. coli.
Both silver loadings demonstrated minimum bactericidal con- Cu-MBGs were shown to have ordered and hexagonally packed
centration (MBC), of 100–150 mg/ml for E. coli and 5–10 mg/ml for S. mesoporous structure, high surface area and large volume of pores.
mutans, whereas the MBC for the undoped BG was > 300 mg/ml. These endow Cu-MBGs with high bioactivity when the material is im-
3 wt% Ag2O was incorporated into structure-directing agent mersed in SBF.
(Pluronic P123) assisted sol-gel derived 73SiO2-13CaO-11P2O5 (wt%) When Cu-MBGs were added to NFC fibrils, they retained the ab-
mesoporous bioactive glass system to investigate the antibacterial sorption capability of NFC aerogels, therefore controlling the moisture
properties of this glass [94]. Ag-MBG showed a very good anti-bacterial around the wounds, which is beneficial to support the wound healing
effect against S. aureus strain, with strong evidence of bactericidal ac- process.
tivity at 0.5 mg/ml of glass concentration. MBC decreased from These aerogels enhanced the gene expression of VEGF A, VEGF C,
2.2 × 109 to 2 × 104 CFU/ml from undoped to 3 wt% Ag2O doped PDGF B and FGF 2 in the culture of 3T3 fibroblasts. 5% Cu doped MBG-
MBG (> 99.9% killing efficiency). NFC composite induced HUVEC sprouting and promoted ECM pro-
58S (60SiO2-36CaO-4P2O5) (mol%) mesoporous BGs produced via duction. Both the NFC-Cu-MBG composites and Cu-MBG (2 and 5 mol%
structure directing agent (P123) assisted sol-gel method was doped with Cu) killed the E. coli strains, the effect of 5 mol% Cu being higher than
1 mol% Ag2O and it was shown that this glass exhibited 100% killing that of 2 mol% Cu MBG.
efficiency against E. coli and S. aureus [98]. In a later study, MBGs with 2% molar percentage of Cu (molar ratio
Table 1 presents a summary of compositions, synthesis methods and Cu/Ca/Si = 2/13/85) and 5% molar percentage of Cu (molar ratio Cu/
results obtained from the studies involving silver containing MBGs Ca/Si = 5/10/85) were prepared by a one-pot ultrasound-assisted sol-
discussed above. gel procedure and the final textural features, the in vitro bioactive re-
sponses and the antibacterial properties of the synthesized MBGs, were
2.2. Copper doped-MBGs tested against E. coli, S. aureus and S. epidermidis [107].
2% mol Cu-MBG had 550 m2 g− 1 surface area, 2.6 nm size of me-
Copper is a naturally occurring element in the human body being sopores, showing in vitro bioactive behavior and a sustained release of
essential to numerous metabolic processes [99,100]. It is, in the right Cu2 + ions into SBF. When 2% mol Cu-MBG was exposed to 1 day of
quantities, non-toxic to human tissues, but is known to have a strong incubation in the three bacteria, approximately 30 to 40% of inhibition
effect on microorganisms. The antibacterial properties of CuO doped of bacteria growth was observed. For 3 days of incubation, E. coli and S.

102
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

Table 1
Summary of reviewed publications on silver containing MBGs used in antibacterial applications.

Reference Composition Synthesis method Antibacterial Results

Bellantone, Coleman, and Hench 76SiO2, 19CaO, 5P2O5, 3Ag2O (wt%) Sol-gel Bacteriostatic effect on E. coli MG1655 with 0.01% reduced
2000 [81] bacterial growth
Bellantone, Williams, and Hench 76SiO2, 19CaO, 5P2O5, 3Ag2O (wt%) Sol-gel MBCs (99.9% killing) for AgBG of 1, 0.5, and 0.5 mg/ml
2002 [82] For E. coli, P. aeruginosa, and S. aureus, respectively
Balamurugan et al. 2008 [92] 64SiO2, 26CaO, 10P2O5, 5Ag2O (mol Sol-gel MBC of Ag-BG: 1 mg/ml for E. coli (> 99% killing efficiency)
%)
Palza et al. 2013 [95] 62.3SiO2, 28.9CaO, 8.6P2O5, 4 and Sol-gel 4Ag2O and 8Ag2O: MBC 100–150 mg/ml for E. coli and 5–10 mg/
8Ag2O (wt%) ml for S. mutans
Gargiulo et al. 2013 [94] 73SiO2, 13CaO, 11P2O5, 3Ag2O (wt Structure directing agent MBC decreased from 2.2 × 109 to 2 × 104 CFU/ml for 3Ag2O
%) assisted sol-gel doped BG (> 99.9% killing efficiency)
H. Zhu et al. 2014 [98] 60SiO2, 36CaO, 4P2O5, 1Ag2O (mol Structure directing agent 100% killing efficiency towards E. coli and S. aureus
%) assisted sol-gel

development than the ones with 7 ZnO (mol%). This is likely due to the
amount of Zn2 + ions released from the 4 mol% ZnO scaffold being
more effective than the amount release from the 7 mol% ZnO scaffolds.
In addition, 4 mol% ZnO scaffolds provided better antibacterial prop-
erties against S. aureus, which can also be related with the more ef-
fective Zn ion release from 4 mol% ZnO doped MBG than the 7 mol%
doped one.
In a recent study [37], the inclusion of 4 ZnO (mol%) substituting
SiO2 in 80SiO2-15CaO-5P2O5 (mol%) BG scaffolds was reported to
drastically increase the amount of dead bacterial cells (S. aureus). Also,
this amount of added ZnO increased the osteoblast development when
zinc containing scaffolds were soaked in extracts of culture medium for
1, 3 and 6 days.
Atkinson et al. [117] prepared three MBGs having composition
70SiO2-(26-x)CaO-4P2O5-xZnO (x = 0, 3 and 5 mol%) by the combined
sol-gel process and polymer templating methods. Their antibacterial
properties were tested against Bacillus subtilis (B. subtilis) and Pseudo-
monas aeruginosa (P. aeruginosa). 3 mol% ZnO doped MBGs had 40%
inhibition for B. subtilis and up to 31–35% inhibition for P. aeruginosa
after 2 h of incubation. 5 ZnO (mol%) doped MBG showed the highest
Fig. 3. Released Cu2 + ions from MBG scaffolds significantly inhibited the viability of antibacterial effect with 91.3% inhibition for B. subtilis after 2 h in-
bacteria according to ref. [101]. ⁎0Cu-MBG group compared to blank control (p < 0.05).
⁎⁎
cubation, as well as 89.4% inhibition for P. aeruginosa. The higher an-
5Cu-MBG group compared to blank control and 0Cu-MBG group (p < 0.01). The Cu-
tibacterial inhibition achieved by the 5 mol% ZnO doped MBGs in
containing scaffolds significantly inhibited bacterial viability compared to scaffolds
without Cu. comparison to the 3 mol% ones can be correlated with higher zinc
(Modified from ref. [101]). concentration determined in the SBF solution after the 14th day of
immersion. 2 ppm of zinc concentration was observed for 5 mol% ZnO
MBGs and 1.8 ppm for the 3 mol% doped ones. However, the 5 mol%
aureus were killed to an extent of 70 to 75% and S. epidermis was nearly
ZnO composition had a lower rate of hydroxyapatite precipitation,
up to 50%.
which is a prominent marker of bioactivity for BGs [118]. The authors
According to the final properties, this Cu-MBG which has both ex-
suggested that the best performing material from this study for implant
cellent bioactivity and antimicrobial property can be considered a
applications was the 3 mol% ZnO added MBG, since it displayed
suitable candidate to prevent infections and to treat bone defects.
medium HA precipitation and antibacterial properties.

2.3. Zinc doped-MBGs 2.4. Cerium doped-MBGs

Zinc has important effects in the development, formation and me- It has been reported that Ce3 + ions reduce enamel demineraliza-
tabolism of bone cells [108–110], in the growth of blood vessels [111] tion, are neuroprotective [119] and promote antibacterial behavior
and as antibacterial agent [112]. It also improves wound healing when added into biomaterials [120].
[113,114]. Zinc provides antibacterial activity by inhibiting glycolysis, Goh et al. [120] added 1, 5 and 10 mol% CeO2 into 50SiO2-45CaO-
transmembrane proton translocation and acid tolerance in bacterial 5P2O5 (mol%) mesoporous silicate glasses via quick alkali mediated sol-
cells [115]. Having high impact on bone formation and growth and gel method. The antibacterial properties were investigated using the
with its antibacterial properties, zinc ions are being increasingly con- quantitative viable count method. It was found that 5 and 10 mol% Ce-
sidered in combination with BGs for bone regeneration applications MBGs exhibited significant antibacterial properties compared to 1 mol
[110]. % Ce and undoped samples. The antibacterial mechanism of cerium
80SiO2-15CaO-5P2O5 (mol%) mesoporous bioactive glass (MBG) (III) ion was reported to be by binding rapidly to E. coli cells, inhibition
scaffolds doped with 4 and 7 ZnO (mol%) were investigated for their of endogenous respiration of cells as well as penetration into the cy-
cytocompatibility and antibacterial properties against S. aureus [116]. 4 toplasm of the cells and interfering with their metabolic functions
ZnO (mol%) substitution was shown to make the MBG a suitable can- [121]. All samples were confirmed to have suitable bioactivity, with
didate for bone regeneration applications [116] since scaffolds with this induced formation of apatite particles upon immersion in simulated
composition led to better human osteoblast-like (HOS) cell body fluid (SBF). These Ce-MBGs were reported to have potential

103
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

applications in the bone regeneration field. promising antibacterial properties [120]. Gallium ions are effective
against bone desorption and for the treatment of osteoporosis and
2.5. Gallium doped-MBGs cancer related hypercalcemias [137,138]. Ga is also effective against
organisms causing tuberculosis and malaria in human beings as well as
The US Food and Drug Administration (FDA) has approved Ga3 + as against Pseudomonas aeruginosa [139,140]. Moreover, besides limiting
an effective agent for treating bone resorption, autoimmune disease, for bacteria attachment, these ions can inhibit bacteria replication by da-
inhibition of biofilm formation, for bone, colon and prostate cancer maging bacteria RNA and DNA, i.e. silver ions, or decreasing bacterial
treatments [122] and also for fighting against both Gram-positive and Fe uptake, i.e. gallium ions. Silver has been used more than the other
Gram-negative bacteria [37,123–128]. antibacterial ions (Cu, Zn, Ce and Ga) as it has a broad and strong
Salinas and Vallet-Regí [37] carried out a study on the production of antimicrobial behavior and has no toxicity effect to human cells in
scaffolds made of mesoporous 3.5 Ga2O3 (mol%) substituted for SiO2 in suitable concentrations, thus providing extra functions to BGs in addi-
80SiO2-15CaO-5P2O5 (mol%) bioactive glasses. Ga3 + ions were hardly tion to osteoconductivity and bone bonding ability.
released from the glass network; therefore it is unlikely that they im-
parted antibacterial property to the glasses. Acknowledgement
A recent study done by Pourshahrestani et al. [126] investigated the
effects of 1–3 Ga2O3 (mol%) addition on the antibacterial properties of The authors would like to acknowledge the European Union's
80SiO2-15CaO-5P2O5 (mol%) ordered mesoporous bioactive glasses, in Horizon 2020 research and innovation programme under the Marie
which they found that 3 Ga2O3 (mol%) addition had the highest anti- Skłodowska-Curie grant agreement No 643050, project “HyMedPoly”.
bacterial rate against S. aureus with 99% after 12 h of incubation. The
highest antibacterial rate can be related with the highest Ga ion con- Funding
centration after the release into Tris–HCl solutions. After 150 h of in-
cubation, solutions of 1, 2 and 3 mol% Ga2O3 doped MBGs had around This work was supported by the European Union's Horizon 2020
0.1, 0.23 and 0.32 ppm of Ga ion concentrations, respectively. research and innovation programme under the Marie Skłodowska Curie
Moreover, Sanchez-Salcedo et al. [127] synthesized mesoporous [grant number 643050] project “HyMedPoly”.
glasses with the composition 80SiO2-15CaO-5P2O5 (mol%), containing
5 mol% of Ga2O3 via evaporation induced self-assembly method. It was References
shown that the Ga3 + concentrations released from MBG into Dulbecco's
Modified Eagle Medium (DMEM) and Todd Hewitt Broth (THB) were in [1] L.L. Hench, R.J. Splinter, W.C. Allen, T.K. Greenlee, Bonding mechanisms at the
the non-cytotoxic levels, also in the effective antibacterial range against interface of ceramic prosthetic materials, J. Biomed. Mater. Res. 5 (1971)
117–141, http://dx.doi.org/10.1002/jbm.820050611.
P. aeruginosa and not far from the effective range against S. aureus. In [2] L.L. Hench, Opening paper 2015 — some comments on bioglass: four eras of
DMEM, the maximum Ga3 + concentration obtained was 2.5 ppm, discovery and development, Biomed. Glass. 1 (2015) 1–11, http://dx.doi.org/10.
which is below the toxicity limit of Ga3 + in blood plasma (14 ppm) 1515/bglass-2015-0001.
[3] A. Hoppe, B. Sarker, R. Detsch, N. Hild, D. Mohn, W.J. Stark, et al., In vitro re-
[38]. In THB the release of Ga3 + concentration was 9.8 ppm, which is activity of Sr-containing bioactive glass (type 1393) nanoparticles, J. Non-Cryst.
140 times higher than the IC90 (drug concentration inhibiting 90% of Solids 387 (2014) 41–46, http://dx.doi.org/10.1016/j.jnoncrysol.2013.12.010.
parasite's activity) of P. aeruginosa and 2 times lower than that of S. [4] D. Zhang, O. Leppäranta, E. Munukka, H. Ylänen, M.K. Viljanen, E. Eerola, et al.,
Antibacterial effects and dissolution behavior of six bioactive glasses, J. Biomed.
aureus [117]. Therefore, this MBG was considered a promising material Mater. Res. A 93 (2010) 475–483, http://dx.doi.org/10.1002/jbm.a.32564.
for bone regeneration applications. [5] M. Brink, T. Turunen, R.P. Happonen, A. Yli-Urpo, Compositional dependence of
bioactivity of glasses in the system Na2O-K2O-MgO-CaO-B2O3-P2O5-SiO2, J.
Biomed. Mater. Res. 37 (1997) 114–121, http://dx.doi.org/10.1002/(SICI)1097-
3. Summary and outlook
4636(199710)37:1<114::AID-JBM14>3.3.CO;2-7.
[6] O. Lepparanta, M. Vaahtio, T. Peltola, D. Zhang, L. Hupa, M. Hupa, et al.,
BGs are being used in bone regeneration applications as fillers in Antibacterial effect of bioactive glasses on clinically important anaerobic bacteria
composites [129], as scaffolds for bone tissue engineering [72,77,130] in vitro, J. Mater. Sci. Mater. Med. 19 (2008) 547–551, http://dx.doi.org/10.
1007/s10856-007-3018-5.
and as coatings of implants [131]. The failure of implants may happen [7] E. Munukka, O. Leppäranta, M. Korkeamäki, M. Vaahtio, T. Peltola, D. Zhang,
due to bacterial colonization or infections [38]. These failures are et al., Bactericidal effects of bioactive glasses on clinically important aerobic
mostly solved by the administration of systemic antibiotics to the pa- bacteria, J. Mater. Sci. Mater. Med. 19 (2008) 27–32, http://dx.doi.org/10.1007/
s10856-007-3143-1.
tient. This solution can lead to allergic reactions, microbial flora de- [8] L. Hupa, Melt-derived bioactive glasses, Bioact. Glas. Mater. Prop. Appl. 2011, pp.
pletion and bacterial resistance. 3–28, , http://dx.doi.org/10.1016/B978-1-84569-768-6.50001-6.
Due to these limitations of antibiotics, the modification of bioma- [9] R.F. Brown, M.N. Rahaman, A.B. Dwilewicz, W. Huang, D.E. Day, Y. Li, et al.,
Effect of borate glass composition on its conversion to hydroxyapatite and on the
terials with alternative antibacterial agents [132] is being intensively proliferation of MC3T3-E1 cells, J. Biomed. Mater. Res. A 88 (2009) 392–400,
investigated. In this perspective, bioactive and soluble glasses are http://dx.doi.org/10.1002/jbm.a.31679.
capable of controlled and sustained release of antimicrobial ions (e.g. [10] E.A. Abou Neel, D.M. Pickup, S.P. Valappil, R.J. Newport, J.C. Knowles, Bioactive
functional materials: a perspective on phosphate-based glasses, J. Mater. Chem. 19
Ag, Cu, Zn, Ce and Ga) and thus have the potential to allow localized (2009) 690–701, http://dx.doi.org/10.1039/b810675d.
antimicrobial treatments which are advantageous compared to systemic [11] A. Hoppe, N.S. Guldal, A.R. Boccaccini, A review of the biological response to ionic
antibiotic delivery systems. Osteogenesis and angiogenesis properties dissolution products from bioactive glasses and glass-ceramics, Biomaterials 32
(2011) 2757–2774, http://dx.doi.org/10.1016/j.biomaterials.2011.01.004.
are also promoted with the release of some of the therapeutic ions, e.g.
[12] M. Cerruti, G. Magnacca, C. Morterra, Characterization of sol–gel bioglasses with
Cu, Zn, Ce and Ga [11,37,102,116,120]. In low concentrations silver the use of simple model systems: a surface-chemistry approach, J. Mater. Chem.
has no toxicity effect to human cells, and it can also give antibacterial, (2003) 1279–1286, http://dx.doi.org/10.1039/b300961k.
osteoconductivity and bone bonding properties to BGs [81–85].Copper [13] R. Li, A.E. Clark, L.L. Hench, An investigation of bioactive glass powders by sol-gel
processing, J. Appl. Biomater. 2 (1991) 231–239, http://dx.doi.org/10.1002/jab.
ion is widely used due to its vascularization/angiogenesis stimulation 770020403.
properties [133]. Zinc is a cofactor in metabolic processes in bone, [14] B. Lei, X. Chen, Y. Wang, N. Zhao, G. Miao, Z. Li, et al., Fabrication of porous
articular tissues and immune system functions [110,134,135]. Bone bioactive glass particles by one step sintering, Mater. Lett. 64 (2010) 2293–2295,
http://dx.doi.org/10.1016/j.matlet.2010.07.066.
formation and mineralization are stimulated by activating aminoacyl- [15] C. Brinker, G. Scherer, Sol-gel science: the physics and chemistry of sol-gel pro-
tRNA synthetase in osteoblastic cells via the zinc doping of BGs [116]. cessing, Adv. Mater. 3 (1990) 912, http://dx.doi.org/10.1186/1471-2105-8-444.
Cerium has been shown to enhance the proliferation, differentiation [16] I. Izquierdo-Barba, D. Arcos, Y. Sakamoto, O. Terasaki, A. Lopez-Noriega,
M. Vallet-Regi, High-performance mesoporous bioceramics mimicking bone mi-
and mineralization of primary osteoblasts [136]. Besides promoting neralization, Chem. Mater. 20 (2008) 3191–3198, http://dx.doi.org/10.1021/
osteogenesis, cerium incorporated materials have demonstrated

104
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

cm800172x. doping amount of ZnO in MgO-ZnO solid solution, Int. J. Inorg. Mater. 2 (2000)
[17] C. Wu, J. Chang, Y. Xiao, Mesoporous bioactive glasses as drug delivery and bone 451–454, http://dx.doi.org/10.1016/S1466-6049(00)00045-3.
tissue regeneration platforms, Ther. Deliv. 2 (2011) 1189–1198, http://dx.doi. [43] P. Stoor, E. Söderling, J.I. Salonen, Antibacterial effects of a bioactive glass paste
org/10.4155/tde.11.84. on oral microorganisms, Acta Odontol. Scand. 56 (1998) 161–165, http://dx.doi.
[18] D. Arcos, A. López-Noriega, E. Ruiz-Hernández, O. Terasaki, M. Vallet-Regí, org/10.1080/000163598422901.
Ordered mesoporous microspheres for bone grafting and drug delivery, Chem. [44] P. Stoor, E. Söderling, R. Grénman, Bioactive glass S53P4 in repair of septal per-
Mater. 21 (2009) 1000–1009, http://dx.doi.org/10.1021/cm801649z. forations and its interactions with the respiratory infection-associated micro-
[19] W. Xia, J. Chang, Well-ordered mesoporous bioactive glasses (MBG): a promising organisms Heamophilus influenzae and Streptococcus pneumoniae, J. Biomed. Mater.
bioactive drug delivery system, J. Control. Release 110 (2006) 522–530, http:// Res. 58 (2001) 113–120, http://dx.doi.org/10.1002/1097-4636(2001)
dx.doi.org/10.1016/j.jconrel.2005.11.002. 58:1<113::AID-JBM170>3.0.CO;2-V.
[20] X. Yan, C. Yu, X. Zhou, J. Tang, D. Zhao, Highly ordered mesoporous bioactive [45] P. Stoor, E. Söderling, R. Grenman, Interactions between the bioactive glass S53P4
glasses with superior in vitro bone-forming bioactivities, Angew. Chem. Int. Ed. 43 and the atrophic rhinitis-associated microorganism Klebsiella ozaenae, J. Biomed.
(2004) 5980–5984, http://dx.doi.org/10.1002/anie.200460598. Mater. Res. 48 (1999) 869–874, http://dx.doi.org/10.1002/(SICI)1097-
[21] L. Zhao, X. Yan, X. Zhou, L. Zhou, H. Wang, J. Tang, et al., Mesoporous bioactive 4636(1999)48:6<869::AID-JBM16>3.0.CO;2-6.
glasses for controlled drug release, Microporous Mesoporous Mater. 109 (2008) [46] M.G. Newman, S.S. Socransky, E.D. Savitt, D.A. Propas, A. Crawford, Studies of the
210–215, http://dx.doi.org/10.1016/j.micromeso.2007.04.041. microbiology of periodontosis, J. Periodontol. 47 (1976) 373–379, http://dx.doi.
[22] V. Mouriño, A.R. Boccaccini, Bone tissue engineering therapeutics: controlled drug org/10.1902/jop.1976.47.7.373.
delivery in three-dimensional scaffolds, J. R. Soc. Interface 7 (2010) 209–227, [47] J. Slots, The predominant cultivable organisms in juvenile periodontitis, Scand. J.
http://dx.doi.org/10.1098/rsif.2009.0379. Dent. Res. 84 (1976) 1–10.
[23] Y. Zhu, Y. Zhang, C. Wu, Y. Fang, J. Yang, S. Wang, The effect of zirconium in- [48] J. Slots, The predominant cultivable microflora of advanced periodontitis, Scand.
corporation on the physiochemical and biological properties of mesoporous J. Dent. Res. 4 (1977) 114–121, http://dx.doi.org/10.1111/j.1600-0722.1977.
bioactive glasses scaffolds, Microporous Mesoporous Mater. 143 (2011) 311–319, tb00541.x.
http://dx.doi.org/10.1016/j.micromeso.2011.03.007. [49] C.A. Spiegel, S.E. Hayduk, G.E. Minah, G.N. Krywolap, Black-pigmented
[24] X. Yan, X. Huang, C. Yu, H. Deng, Y. Wang, Z. Zhang, et al., The in-vitro bioactivity Bacteroides from clinically characterized periodontal sites, J. Periodontal Res. 14
of mesoporous bioactive glasses, Biomaterials 27 (2006) 3396–3403, http://dx. (1979) 376–382, http://dx.doi.org/10.1111/j.1600-0765.1979.tb00234.x.
doi.org/10.1016/j.biomaterials.2006.01.043. [50] A.C.R. Tanner, C. Haffer, G.T. Bratthall, R.A. Visconti, S.S. Socransky, A study of
[25] E. Leonova, I. Izquierdo-Barba, D. Arcos, A. López-Noriega, N. Hedin, M. Vallet- the bacteria associated with advancing periodontitis in man, J. Clin. Periodontol. 6
Regí, et al., Multinuclear solid-state NMR studies of ordered mesoporous bioactive (1979) 278–307, http://dx.doi.org/10.1111/j.1600-051X.1979.tb01931.x.
glasses, J. Phys. Chem. C 112 (2008) 5552–5562, http://dx.doi.org/10.1021/ [51] J.M. Tanzer, J. Livingston, A.M. Thompson, The microbiology of primary dental
jp7107973. caries in humans, J. Dent. Educ. 65 (2001) 1028–1037.
[26] A. García, M. Cicuéndez, I. Izquierdo-Barba, D. Arcos, M. Vallet-Regí, Essential [52] L. Drago, D. Romano, E. De Vecchi, C. Vassena, N. Logoluso, R. Mattina, et al.,
role of calcium phosphate heterogeneities in 2D-hexagonal and 3D-cubic Bioactive glass BAG-S53P4 for the adjunctive treatment of chronic osteomyelitis of
SiO2 − CaO − P2O5 mesoporous bioactive glasses, Chem. Mater. 21 (2009) the long bones: an in vitro and prospective clinical study, BMC Infect. Dis. 13
5474–5484, http://dx.doi.org/10.1021/cm9022776. (2013) 584, http://dx.doi.org/10.1186/1471-2334-13-584.
[27] M. Alcaide, P. Portoles, A. Lopez-Noriega, D. Arcos, M. Vallet-Regi, M.T. Portoles, [53] J. McAndrew, C. Efrimescu, E. Sheehan, D. Niall, Through the looking glass;
Interaction of an ordered mesoporous bioactive glass with osteoblasts, fibroblasts bioactive glass S53P4 (BonAlive®) in the treatment of chronic osteomyelitis, Ir. J.
and lymphocytes, demonstrating its biocompatibility as a potential bone graft Med. Sci. 182 (2013) 509–511, http://dx.doi.org/10.1007/s11845-012-0895-5.
material, Acta Biomater. 6 (2010) 892–899, http://dx.doi.org/10.1016/j.actbio. [54] C.L. Romano, N. Logoluso, E. Meani, D. Romano, E. De Vecchi, C. Vassena, et al., A
2009.09.008. comparative study of the use of bioactive glass S53P4 and antibiotic-loaded cal-
[28] D. Arcos, M. Vallet-Regi, Sol-gel silica-based biomaterials and bone tissue re- cium-based bone substitutes in the treatment of chronic osteomyelitis: a retro-
generation, Acta Biomater. 6 (2010) 2874–2888, http://dx.doi.org/10.1016/j. spective comparative study, Bone Jt J 96 (B) (2014) 845–850, http://dx.doi.org/
actbio.2010.02.012. 10.1302/0301-620X.96B6.33014.
[29] W. Xia, J. Chang, Preparation, in vitro bioactivity and drug release property of [55] N.A.P. Van Gestel, J. Geurts, D.J.W. Hulsen, B. Van Rietbergen, S. Hofmann,
well-ordered mesoporous 58S bioactive glass, J. Non-Cryst. Solids 354 (2008) J.J. Arts, Clinical applications of S53P4 bioactive glass in bone healing and os-
1338–1341, http://dx.doi.org/10.1016/j.jnoncrysol.2006.10.084. teomyelitic treatment: a literature review, Biomed. Res. Int. 2015 (2015), http://
[30] X. Li, X. Wang, D. He, J. Shi, Synthesis and characterization of mesoporous dx.doi.org/10.1155/2015/684826.
CaO–MO–SiO2–P2O5 (M = Mg, Zn, Cu) bioactive glasses/composites, J. Mater. [56] J.H. Calhoun, M.M. Manring, M. Shirtliff, Osteomyelitis of the long bones, Semin.
Chem. 18 (2008) 4103, http://dx.doi.org/10.1039/b805114c. Plast. Surg. 23 (2009) 59–72, http://dx.doi.org/10.1055/s-0029-1214158.
[31] X. Wu, J. Wei, X. Lu, Y. Lv, F. Chen, Y. Zhang, et al., Chemical characteristics and [57] J. Geurts, J.J. Chris Arts, G.H.I.M. Walenkamp, Bone graft substitutes in active or
hemostatic performances of ordered mesoporous calcium-doped silica xerogels, suspected infection. Contra-indicated or not? Injury 42 (2011), http://dx.doi.org/
Biomed. Mater. 5 (2010) 35006, http://dx.doi.org/10.1088/1748-6041/5/3/ 10.1016/j.injury.2011.06.189.
035006. [58] G. Walenkamp, Small PMMA beads improve gentamicin release, Acta Orthop.
[32] A. Hoppe, A.R. Boccaccini, Biological impact of bioactive glasses and their dis- Scand. 60 (1989) 668–669, http://dx.doi.org/10.3109/17453678909149599.
solution products, Front. Oral Biol. 17 (2015) 22–32, http://dx.doi.org/10.1159/ [59] T.F. Calton, T.K. Fehring, W.L. Griffin, Bone loss associated with the use of spacer
000381690. blocks in infected total knee arthroplasty, Clin. Orthop. Relat. Res. (1997)
[33] V. Mourino, J.P. Cattalini, A.R. Boccaccini, Metallic ions as therapeutic agents in 148–154.
tissue engineering scaffolds: an overview of their biological applications and [60] N.C. Lindfors, Bioactive glass S53P4 as a bone graft substitute in the treatment of
strategies for new developments, J. R. Soc. Interface 9 (2012) 401–419, http://dx. osteomyelitis, Bioact. Glas. Mater. Prop. Appl. 2011, pp. 209–216, , http://dx.doi.
doi.org/10.1098/rsif.2011.0611. org/10.1016/B978-1-84569-768-6.50009-0.
[34] C. Wu, J. Chang, Mesoporous bioactive glasses: structure characteristics, drug/ [61] R. Lakshminarayanan, V.A. Barathi, M. Venkatesh, N.K. Verma, S. Liu, X.J. Loh,
growth factor delivery and bone regeneration application, Interface Focus 2 R.W. Beuerman, Membrane selectivity of cationic polyamides and rational design
(2012) 292–306, http://dx.doi.org/10.1098/rsfs.2011.0121. of proteolyticresistant antimicrobial peptides, Invest. Ophthalmol. Vis. Sci. 57
[35] C. Wu, J. Chang, Multifunctional mesoporous bioactive glasses for effective de- (2016) 333.
livery of therapeutic ions and drug/growth factors, J. Control. Release 193 (2014) [62] R. Lakshminarayanan, R. Sridhar, X.J. Loh, M. Nandhakumar, V.A. Barathi,
282–295, http://dx.doi.org/10.1016/j.jconrel.2014.04.026. M. Kalaipriya, et al., Interaction of gelatin with polyenes modulates antifungal
[36] C. Wu, W. Fan, Y. Zhu, M. Gelinsky, J. Chang, G. Cuniberti, et al., Multifunctional activity and biocompatibility of electrospun fiber mats, Int. J. Nanomedicine 9
magnetic mesoporous bioactive glass scaffolds with a hierarchical pore structure, (2014) 2439–2458, http://dx.doi.org/10.2147/IJN.S58487.
Acta Biomater. 7 (2011) 3563–3572, http://dx.doi.org/10.1016/j.actbio.2011.06. [63] M. Ignatova, I. Rashkov, N. Manolova, М. Ignatova, I. Rashkov, N. Manolova,
028. Drug-loaded electrospun materials in wound-dressing applications and in local
[37] A.J. Salinas, M. Vallet-Regí, Glasses in bone regeneration: a multiscale issue, J. cancer treatment, Expert Opin. Drug Deliv. 10 (2013) 469–483, http://dx.doi.org/
Non-Cryst. Solids 432 (2016) 9–14, http://dx.doi.org/10.1016/j.jnoncrysol.2015. 10.1517/17425247.2013.758103.
03.025. [64] S. Puttipipatkhachorn, J. Nunthanid, K. Yamamoto, G.E. Peck, Drug physical state
[38] D. Campoccia, L. Montanaro, C.R. Arciola, The significance of infection related to and drug-polymer interaction on drug release from chitosan matrix films, J.
orthopedic devices and issues of antibiotic resistance, Biomaterials 27 (2006) Control. Release 75 (2001) 143–153, http://dx.doi.org/10.1016/S0168-3659(01)
2331–2339, http://dx.doi.org/10.1016/j.biomaterials.2005.11.044. 00389-3.
[39] A.M. Mulligan, M. Wilson, J.C. Knowles, The effect of increasing copper content in [65] D. Kai, G. Jin, M.P. Prabhakaran, S. Ramakrishna, Electrospun synthetic and
phosphate-based glasses on biofilms of Streptococcus sanguis, Biomaterials 24 natural nanofibers for regenerative medicine and stem cells, Biotechnol. J. 8
(2003) 1797–1807, http://dx.doi.org/10.1016/S0142-9612(02)00577-X. (2013) 59–72, http://dx.doi.org/10.1002/biot.201200249.
[40] T.N. Kim, Q.L. Feng, J.O. Kim, J. Wu, H. Wang, G.C. Chen, et al., Antimicrobial [66] C. Dhand, M. Venkatesh, V.A. Barathi, S. Harini, S. Bairagi, E. Goh Tze Leng, et al.,
effects of metal ions (Ag+, Cu2 +, Zn2 +) in hydroxyapatite, J. Mater. Sci. Mater. Bio-inspired crosslinking and matrix-drug interactions for advanced wound dres-
Med. 9 (1998) 129–134, http://dx.doi.org/10.1023/A:1008811501734. sings with long-term antimicrobial activity, Biomaterials 138 (2017) 153–168,
[41] J.C. Wataha, P.E. Lockwood, A. Schedle, Effect of silver, copper, mercury, and http://dx.doi.org/10.1016/j.biomaterials.2017.05.043.
nickel ions on cellular proliferation during extended, low-dose exposures, J. [67] J. Min, R.D. Braatz, P.T. Hammond, Tunable staged release of therapeutics from
Biomed. Mater. Res. 52 (2000) 360–364, http://dx.doi.org/10.1002/1097- layer-by-layer coatings with clay interlayer barrier, Biomaterials 35 (2014)
4636(200011)52:2<360::AID-JBM16>3.0.CO;2-B. 2507–2517, http://dx.doi.org/10.1016/j.biomaterials.2013.12.009.
[42] O. Yamamoto, J. Sawai, T. Sasamoto, Change in antibacterial characteristics with [68] X. Zhu, Loh X. Jun, Layer-by-layer assemblies for antibacterial applications,

105
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

Biomater. Sci. 3 (2015) 1505–1518, http://dx.doi.org/10.1039/C5BM00307E. Biotechnol. Bioprocess Eng. 17 (2012) 746–754, http://dx.doi.org/10.1007/
[69] P. Gentile, M.E. Frongia, M. Cardellach, C.A. Miller, G.P. Stafford, G.J. Leggett, s12257-012-0046-x.
et al., Functionalised nanoscale coatings using layer-by-layer assembly for im- [94] N. Gargiulo, A.M. Cusano, F. Causa, D. Caputo, P.A. Netti, Silver-containing me-
parting antibacterial properties to polylactide-co-glycolide surfaces, Acta soporous bioactive glass with improved antibacterial properties, J. Mater. Sci.
Biomater. 21 (2015) 35–43, http://dx.doi.org/10.1016/j.actbio.2015.04.009. Mater. Med. 24 (2013) 2129–2135, http://dx.doi.org/10.1007/s10856-013-
[70] M.N. Rahaman, D.E. Day, B. Sonny Bal, Q. Fu, S.B. Jung, L.F. Bonewald, et al., 4968-4.
Bioactive glass in tissue engineering, Acta Biomater. 7 (2011) 2355–2373, http:// [95] H. Palza, B. Escobar, J. Bejarano, D. Bravo, M. Diaz-Dosque, J. Perez, Designing
dx.doi.org/10.1016/j.actbio.2011.03.016. antimicrobial bioactive glass materials with embedded metal ions synthesized by
[71] V. Mouriño, A.R. Boccaccini, Bone tissue engineering therapeutics: controlled drug the sol-gel method, Mater. Sci. Eng. C 33 (2013) 3795–3801, http://dx.doi.org/10.
delivery in three-dimensional scaffolds, J. R. Soc. Interface 7 (2010) 209–227, 1016/j.msec.2013.05.012.
http://dx.doi.org/10.1098/rsif.2009.0379. [96] F. Baino, S. Fiorilli, R. Mortera, B. Onida, E. Saino, L. Visai, et al., Mesoporous
[72] J. Hum, A.R. Boccaccini, Bioactive glasses as carriers for bioactive molecules and bioactive glass as a multifunctional system for bone regeneration and controlled
therapeutic drugs: a review, J. Mater. Sci. Mater. Med. 23 (2012) 2317–2333, drug release, J. Appl. Biomater. Biomech. 10 (2012) 12–21, http://dx.doi.org/10.
http://dx.doi.org/10.1007/s10856-012-4580-z. 5301/JABB.2012.9036.
[73] J. Ye, J. He, C. Wang, K. Yao, Z. Gou, Copper-containing mesoporous bioactive [97] R. Phetnin, S.T. Rattanachan, Preparation and antibacterial property on silver
glass coatings on orbital implants for improving drug delivery capacity and anti- incorporated mesoporous bioactive glass microspheres, J. Sol-Gel Sci. Technol. 75
bacterial activity, Biotechnol. Lett. 36 (2014) 961–968, http://dx.doi.org/10. (2015) 279–290, http://dx.doi.org/10.1007/s10971-015-3697-1.
1007/s10529-014-1465-x. [98] H. Zhu, C. Hu, F. Zhang, X. Feng, J. Li, T. Liu, et al., Preparation and antibacterial
[74] C. Wu, Y. Zhou, W. Fan, P. Han, J. Chang, J. Yuen, et al., Hypoxia-mimicking property of silver-containing mesoporous 58S bioactive glass, Mater. Sci. Eng. C 42
mesoporous bioactive glass scaffolds with controllable cobalt ion release for bone (2014) 22–30, http://dx.doi.org/10.1016/j.msec.2014.05.004.
tissue engineering, Biomaterials 33 (2012) 2076–2085, http://dx.doi.org/10. [99] M.L. Turski, D.J. Thiele, New roles for copper metabolism in cell proliferation,
1016/j.biomaterials.2011.11.042. signaling, and disease, J. Biol. Chem. 284 (2009) 717–721, http://dx.doi.org/10.
[75] J.-H. Lee, A. El-Fiqi, N. Mandakhbayar, H.-H. Lee, H.-W. Kim, Drug/ion co-de- 1074/jbc.R800055200.
livery multi-functional nanocarrier to regenerate infected tissue defect, [100] J.F. Collins, J.R. Prohaska, M.D. Knutson, Metabolic crossroads of iron and copper,
Biomaterials 142 (2017) 62–76, http://dx.doi.org/10.1016/j.biomaterials.2017. Nutr. Rev. 68 (2010) 133–147, http://dx.doi.org/10.1111/j.1753-4887.2010.
07.014. 00271.x.
[76] S. Di Nunzio, C. Vitale Brovarone, S. Spriano, D. Milanese, E. Verné, V. Bergo, [101] C. Wu, Y. Zhou, M. Xu, P. Han, L. Chen, J. Chang, et al., Copper-containing me-
et al., Silver containing bioactive glasses prepared by molten salt ion-exchange, J. soporous bioactive glass scaffolds with multifunctional properties of angiogenesis
Eur. Ceram. Soc. 24 (2004) 2935–2942, http://dx.doi.org/10.1016/j. capacity, osteostimulation and antibacterial activity, Biomaterials 34 (2013)
jeurceramsoc.2003.11.010. 422–433, http://dx.doi.org/10.1016/j.biomaterials.2012.09.066.
[77] P.J. Newby, R. El-Gendy, J. Kirkham, X.B. Yang, I.D. Thompson, A.R. Boccaccini, [102] J. Li, D. Zhai, F. Lv, Q. Yu, H. Ma, J. Yin, et al., Preparation of copper-containing
Ag-doped 45S5 Bioglass®-based bone scaffolds by molten salt ion exchange: pro- bioactive glass/eggshell membrane nanocomposites for improving angiogenesis,
cessing and characterisation, J. Mater. Sci. Mater. Med. 22 (2011) 557–569, antibacterial activity and wound healing, Acta Biomater. 36 (2016) 254–266,
http://dx.doi.org/10.1007/s10856-011-4240-8. http://dx.doi.org/10.1016/j.actbio.2016.03.011.
[78] A.B.G. Lansdown, Silver in health care: antimicrobial effects and safety in use, [103] A.A. Gorustovich, J.A. Roether, A.R. Boccaccini, Effect of bioactive glasses on
Curr. Probl. Dermatol. 33 (2006) 17–34, http://dx.doi.org/10.1159/000093928. angiogenesis: a review of in vitro and in vivo evidences, Tissue Eng. B Rev. 16
[79] W.K. Jung, H.C. Koo, K.W. Kim, S. Shin, S.H. Kim, Y.H. Park, Antibacterial activity (2010) 199–207, http://dx.doi.org/10.1089/ten.teb.2009.0416.
and mechanism of action of the silver ion in Staphylococcus aureus and Escherichia [104] A. Aguirre, A. González, M. Navarro, Ó. Castaño, J.A. Planell, E. Engel, Control of
coli, Appl. Environ. Microbiol. 74 (2008) 2171–2178, http://dx.doi.org/10.1128/ microenvironmental cues with a smart biomaterial composite promotes en-
AEM.02001-07. dothelial progenitor cell angiogenesis, Eur. Cell. Mater. 24 (2012) 90–106 (doi:-
[80] S. Pal, Y.K. Tak, J.M. Song, Does the antibacterial activity of silver nanoparticles vol024a07 [pii]).
depend on the shape of the nanoparticle? A study of the Gram-negative bacterium [105] J. Grzybowski, M.K. Janiak, E. Oidak, K. Lasocki, J. Wrembel-Wargocka, A. Cheda,
Escherichia coli, J. Biol. Chem. 290 (2015) 1712–1720, http://dx.doi.org/10.1128/ et al., New cytokine dressings. II. Stimulation of oxidative burst in leucocytes in
AEM.02218-06. vitro and reduction of viable bacteria within an infected wound, Int. J. Pharm. 184
[81] M. Bellantone, N.J. Coleman, L.L. Hench, Bacteriostatic action of a novel four- (1999) 179–187, http://dx.doi.org/10.1016/S0378-5173(99)00064-2.
component bioactive glass, J. Biomed. Mater. Res. 51 (2000) 484–490, http://dx. [106] X. Wang, F. Cheng, J. Liu, J.-H. Smått, D. Gepperth, M. Lastusaari, et al.,
doi.org/10.1002/1097-4636(20000905)51:3<484::AID-JBM24>3.0.CO;2-4. Biocomposites of copper-containing mesoporous bioactive glass and nanofi-
[82] M. Bellantone, H.D. Williams, L.L. Hench, Broad-spectrum bactericidal activity of brillated cellulose: biocompatibility and angiogenic promotion in chronic wound
Ag2O-doped bioactive glass, Antimicrob. Agents Chemother. 46 (2002) healing application, Acta Biomater. 46 (2016) 286–298, http://dx.doi.org/10.
1940–1945, http://dx.doi.org/10.1128/AAC.46.6.1940-1945.2002. 1016/j.actbio.2016.09.021.
[83] T.J. Berger, J.A. Spadaro, S.E. Chapin, R.O. Becker, Electrically generated silver [107] A. Bari, N. Bloise, S. Fiorilli, G. Novajra, M. Vallet-Regí, G. Bruni, et al., Copper-
ions: quantitative effects on bacterial and mammalian cells, Antimicrob. Agents containing mesoporous bioactive glass nanoparticles as multifunctional agent for
Chemother. 9 (1976) 357–358, http://dx.doi.org/10.1128/AAC.9.2.357. bone regeneration, Acta Biomater. (2017) 1–12, http://dx.doi.org/10.1016/j.
[84] M.A. Hollinger, Toxicological aspects of topical silver pharmaceuticals, Crit. Rev. actbio.2017.04.012.
Toxicol. 26 (1996) 255–260, http://dx.doi.org/10.3109/10408449609012524. [108] L. Fong, K. Tan, C. Tran, J. Cool, M.A. Scherer, R. Elovaris, et al., Interaction of
[85] J.L. Clement, P.S. Jarrett, Antibacterial silver, Metal-Based Drugs 1 (1994) dietary zinc and intracellular binding protein metallothionein in postnatal bone
467–482, http://dx.doi.org/10.1155/MBD.1994.467. growth, Bone 44 (2009) 1151–1162, http://dx.doi.org/10.1016/j.bone.2009.02.
[86] O. Gordon, T.V. Slenters, P.S. Brunetto, A.E. Villaruz, D.E. Sturdevant, M. Otto, 011.
et al., Silver coordination polymers for prevention of implant infection: thiol in- [109] A. Grandjean-Laquerriere, P. Laquerriere, E. Jallot, J.M. Nedelec, M. Guenounou,
teraction, impact on respiratory chain enzymes, and hydroxyl radical induction, D. Laurent-Maquin, et al., Influence of the zinc concentration of sol-gel derived
Antimicrob. Agents Chemother. 54 (2010) 4208–4218, http://dx.doi.org/10. zinc substituted hydroxyapatite on cytokine production by human monocytes in
1128/AAC.01830-09. vitro, Biomaterials 27 (2006) 3195–3200, http://dx.doi.org/10.1016/j.
[87] E. Vernè, S. Di Nunzio, M. Bosetti, P. Appendino, C. Vitale Brovarone, G. Maina, biomaterials.2006.01.024.
et al., Surface characterization of silver-doped bioactive glass, Biomaterials 26 [110] P. Balasubramanian, L.A. Strobel, U. Kneser, A.R. Boccaccini, Zinc-containing
(2005) 5111–5119, http://dx.doi.org/10.1016/j.biomaterials.2005.01.038. bioactive glasses for bone regeneration, dental and orthopedic applications,
[88] P. Saravanapavan, J.E. Gough, J.R. Jones, L.L. Hench, Antimicrobial macroporous Biomed. Glass. 1 (2015) 51–69, http://dx.doi.org/10.1515/bglass-2015-0006.
gel-glasses: dissolution and cytotoxicity, Annu. Meet. Int. Soc. Ceram. Med. [111] R. Pasqualini, C.F. Barbas, W. Arap, Vessel maneuvers: zinc fingers promote an-
254–256 2004, pp. 1087–1090, , http://dx.doi.org/10.4028/www.scientific.net/ giogenesis, Nat. Med. 8 (2002) 1353–1354, http://dx.doi.org/10.1038/nm1202-
KEM.254-256.1087. 1353.
[89] J.J. Blaker, S.N. Nazhat, A.R. Boccaccini, Development and characterisation of [112] C. Lang, C. Murgia, M. Leong, L.-W. Tan, G. Perozzi, D. Knight, et al., Anti-in-
silver-doped bioactive glass-coated sutures for tissue engineering and wound flammatory effects of zinc and alterations in zinc transporter mRNA in mouse
healing applications, Biomaterials 25 (2004) 1319–1329, http://dx.doi.org/10. models of allergic inflammation, Am. J. Phys. Lung Cell. Mol. Phys. 292 (2007)
1016/j.biomaterials.2003.08.007. L577–84, http://dx.doi.org/10.1152/ajplung.00280.2006.
[90] J. Pratten, S.N. Nazhat, J.J. Blaker, A.R. Boccaccini, In vitro attachment of [113] Y.H. Cho, S.J. Lee, J.Y. Lee, S.W. Kim, C.B. Lee, W.Y. Lee, et al., Antibacterial
Staphylococcus epidermidis to surgical sutures with and without Ag-containing effect of intraprostatic zinc injection in a rat model of chronic bacterial prostatitis,
bioactive glass coating, J. Biomater. Appl. 19 (2004) 47–57, http://dx.doi.org/10. Int. J. Antimicrob. Agents 19 (2002) 576–582, http://dx.doi.org/10.1016/S0924-
1177/0885328204043200. 8579(02)00115-2.
[91] J.R. Jones, L.M. Ehrenfried, P. Saravanapavan, L.L. Hench, Controlling ion release [114] A.B.G. Lansdown, U. Mirastschijski, N. Stubbs, E. Scanlon, M.S. Agren, Zinc in
from bioactive glass foam scaffolds with antibacterial properties, J. Mater. Sci. wound healing: theoretical, experimental, and clinical aspects, Wound Repair
Mater. Med. 17 (2006) 989–996, http://dx.doi.org/10.1007/s10856-006-0434-x. Regen. 15 (2007) 2–16, http://dx.doi.org/10.1111/j.1524-475X.2006.00179.x.
[92] A. Balamurugan, G. Balossier, D. Laurent-Maquin, S. Pina, A.H.S. Rebelo, J. Faure, [115] T.N. Phan, T. Buckner, J. Sheng, J.D. Baldeck, R.E. Marquis, Physiologic actions of
et al., An in vitro biological and anti-bacterial study on a sol–gel derived silver- zinc related to inhibition of acid and alkali production by oral streptococci in
incorporated bioglass system, Dent. Mater. 24 (2008) 1343–1351, http://dx.doi. suspensions and biofilms, Oral Microbiol. Immunol. 19 (2004) 31–38, http://dx.
org/10.1016/j.dental.2008.02.015. doi.org/10.1046/j.0902-0055.2003.00109.x.
[93] N. Nezafati, F. Moztarzadeh, S. Hesaraki, Surface reactivity and in vitro biological [116] S. Sanchez-Salcedo, S. Shruti, A.J. Salinas, G. Malavasi, L. Menabue, M. Vallet-
evaluation of sol gel derived silver/calcium silicophosphate bioactive glass, Regi, In vitro antibacterial capacity and cytocompatibility of SiO2-CaO-P2O5 meso-

106
S. Kaya et al. Materials Science & Engineering C 83 (2018) 99–107

macroporous glass scaffolds enriched with ZnO, J. Mater. Chem. B 2 (2014) [129] G. Chouzouri, M. Xanthos, In vitro bioactivity and degradation of poly-
4836–4847, http://dx.doi.org/10.1039/c4tb00403e. caprolactone composites containing silicate fillers, Acta Biomater. 3 (2007)
[117] I. Atkinson, E.M. Anghel, L. Predoana, O.C. Mocioiu, L. Jecu, I. Raut, et al., 745–756, http://dx.doi.org/10.1016/j.actbio.2007.01.005.
Influence of ZnO addition on the structural, in vitro behavior and antimicrobial [130] Q. Fu, E. Saiz, M.N. Rahaman, A.P. Tomsia, Bioactive glass scaffolds for bone
activity of sol–gel derived CaO–P2O5–SiO2 bioactive glasses, Ceram. Int. 42 (2016) tissue engineering: state of the art and future perspectives, Mater. Sci. Eng. C
3033–3045, http://dx.doi.org/10.1016/j.ceramint.2015.10.090. Mater. Biol. Appl. 31 (2012) 1245–1256, http://dx.doi.org/10.1016/j.msec.2011.
[118] T. Kokubo, H. Takadama, How useful is SBF in predicting in vivo bone bioactivity? 04.022.Bioactive.
Biomaterials 27 (15) (2006) 2907, http://dx.doi.org/10.1016/j.biomaterials. [131] M. Mehdipour, A. Afshar, M. Mohebali, Electrophoretic deposition of bioactive
2006.01.017. glass coating on 316L stainless steel and electrochemical behavior study, Appl.
[119] C. Leonelli, G. Lusvardi, G. Malavasi, L. Menabue, M. Tonelli, Synthesis and Surf. Sci. 258 (2012) 9832–9839, http://dx.doi.org/10.1016/j.apsusc.2012.06.
characterization of cerium-doped glasses and in vitro evaluation of bioactivity, J. 038.
Non-Cryst. Solids 316 (2003) 198–216, http://dx.doi.org/10.1016/S0022- [132] T.N. Kim, Q.L. Feng, J.O. Kim, J. Wu, H. Wang, G.C. Chen, et al., Antimicrobial
3093(02)01628-9. effects of metal ions (Ag+, Cu2 +, Zn2 +) in hydroxyapatite, J. Mater. Sci. Mater.
[120] Y.F. Goh, A.Z. Alshemary, M. Akram, M.R. Abdul Kadir, R. Hussain, In-vitro Med. 9 (1998) 129–134, http://dx.doi.org/10.1023/A:1008811501734.
characterization of antibacterial bioactive glass containing ceria, Ceram. Int. 40 [133] N. Kong, K. Lin, H. Li, J. Chang, Synergy effects of copper and silicon ions on
(2014) 729–737, http://dx.doi.org/10.1016/j.ceramint.2013.06.062. stimulation of vascularization by copper-doped calcium silicate, J. Mater. Chem. B
[121] J.M. Sobek, D.E. Talburt, Effects of the rare earth cerium on Escherichia coli, Am 2 (2014) 1100, http://dx.doi.org/10.1039/c3tb21529f.
Soc Microbiol 95 (1968) 47–51. [134] P.D. Saltman, L.G. Strause, The role of trace minerals in osteoporosis, J. Am. Coll.
[122] P. Collery, B. Keppler, C. Madoulet, B. Desoize, Gallium in cancer treatment, Crit. Nutr. 12 (1993) 384–389, http://dx.doi.org/10.1080/07315724.1993.10718327.
Rev. Oncol. Hematol. 42 (2002) 283–296, http://dx.doi.org/10.1016/S1040- [135] M. Yazar, S. Sarban, A. Kocyigit, U.E. Isikan, Synovial fluid and plasma selenium,
8428(01)00225-6. copper, zinc, and iron concentrations in patients with rheumatoid arthritis and
[123] E. Zeimaran, S. Pourshahrestani, B. Pingguan-Murphy, N.A. Kadri, H.A. Rothan, osteoarthritis, Biol. Trace Elem. Res. 106 (2005) 123–132, http://dx.doi.org/10.
R. Yusof, et al., Fabrication and characterization of poly(octanediol citrate)/gal- 1385/BTER:106:2:123.
lium-containing bioglass microcomposite scaffolds, J. Mater. Sci. 50 (2015) [136] G. Zhou, G. Gu, Y. Li, Q. Zhang, W. Wang, S. Wang, et al., Effects of cerium oxide
2189–2201, http://dx.doi.org/10.1007/s10853-014-8782-2. nanoparticles on the proliferation, differentiation, and mineralization function of
[124] L.R. Bernstein, Mechanisms of therapeutic activity for gallium, Pharmacol. Rev. 50 primary osteoblasts in vitro, Biol. Trace Elem. Res. 153 (2013) 411–418, http://
(1998) 665–682. dx.doi.org/10.1007/s12011-013-9655-2.
[125] Y. Kaneko, M. Thoendel, O. Olakanmi, B.E. Britigan, P.K. Singh, The transition [137] R.P. Warrell, R.S. Bockman, C.J. Coonley, M. Isaacs, H. Staszewski, Gallium nitrate
metal gallium disrupts Pseudomonas aeruginosa iron metabolism and has anti- inhibits calcium resorption from bone and is effective treatment for cancer-related
microbial and antibiofilm activity, J. Clin. Invest. 117 (2007) 877–888, http://dx. hypercalcemia, J. Clin. Invest. 73 (1984) 1487–1490, http://dx.doi.org/10.1172/
doi.org/10.1172/JCI30783. JCI111353.
[126] S. Pourshahrestani, E. Zeimaran, N. Adib Kadri, N. Gargiulo, S. Samuel, [138] P. Melnikov, A.R. Teixeira, A. Malzac, B. Coelho M de, Gallium-containing hy-
S.V. Naveen, et al., Gallium-containing mesoporous bioactive glass with potent droxyapatite for potential use in orthopedics, Mater. Chem. Phys. 117 (2009)
hemostatic activity and antibacterial efficacy, J. Mater. Chem. B 4 (2016) 71–86, 86–90, http://dx.doi.org/10.1016/j.matchemphys.2009.05.046.
http://dx.doi.org/10.1039/C5TB02062J. [139] D.M. Pickup, R.M. Moss, D. Qiu, R.J. Newport, S.P. Valappil, J.C. Knowles, et al.,
[127] S. Sanchez-salcedo, A. Salinas, M. Vallet-Regi, Development of mesoporous Structural characterization by X-ray methods of novel antimicrobial gallium-
bioactive glasses able to release antibacterial Ga3 + ions, Conf. Abstr. 10th World doped phosphate-based glasses, J. Chem. Phys. 130 (2009), http://dx.doi.org/10.
Biomater. Congr., Front. Bioeng. Biotechnol. 2016, pp. 5–7, , http://dx.doi.org/10. 1063/1.3076057.
3389/conf.FBIOE.2016.01.01476. [140] R.J. Martens, N.A. Miller, N.D. Cohen, J.R. Harrington, L.R. Bernstein,
[128] F. Minandri, C. Bonchi, E. Frangipani, F. Imperi, P. Visca, Promises and failures of Chemoprophylactic antimicrobial activity of gallium maltolate against in-
gallium as an antibacterial agent, Future Microbiol. 9 (2014) 379–397, http://dx. tracellular Rhodococcus equi, J. Equine. Vet. Sci. 27 (2007) 341–345, http://dx.doi.
doi.org/10.2217/fmb.14.3. org/10.1016/j.jevs.2007.06.007.

107

You might also like