Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Materials Science and Engineering C 61 (2016) 965–978

Contents lists available at ScienceDirect

Materials Science and Engineering C

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

Review

Antibacterial titanium surfaces for medical implants


S. Ferraris ⁎, S. Spriano
Department of Applied Science and Technology, Institute of Materials Physics and Engineering, Politecnico di Torino, C.so Duca degli Abruzzi 24, Torino 10129, Italy

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

Article history: Bacterial contamination is a critical problem in different fields (ranging from everyday life to space missions, and
Received 4 June 2015 from medicine to biosensing). Specifically, in the case of medical implants, foreign materials are preferential sites
Received in revised form 16 November 2015 for bacterial adhesion and microbial contamination, which can lead to the development of prosthetic infections.
Accepted 28 December 2015
These problems can in turn lead to the necessity of a prolonged antibiotic therapy (which can last for years) and
Available online 31 December 2015
eventually to the removal of the device, with a consequent significant increase in the hospitalization times and
Keywords:
costs, together with a stressful, painful and critical situation for the patient. Commercially pure titanium and
Antibacterial its alloys are the most commonly used materials for permanent implants in contact with bone, and the preven-
Titanium tion of infections on their surface is therefore a crucial challenge for orthopaedic and dental surgeons. The prob-
Silver lem of the bacterial contamination of medical implants is briefly described in the first part of the present review.
Copper Then the most important inorganic antibacterial agents (Ag, Cu and Zn) are described, and this is followed by a
Zinc review of the reported attempts of their introduction onto the surface of Ti-based substrates.
© 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 966
2. Bacterial contamination of medical implants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 966
3. Antibacterial agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 967
4. Surface modifications that can be introduced to obtain antibacterial metallic surfaces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 967
4.1. The introduction of silver onto titanium surfaces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 968
4.1.1. Ion implantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 968
4.1.2. Electrochemical techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
4.1.3. In situ synthesis of silver nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
4.1.4. Ion exchange . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
4.1.5. Sol–gel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
4.1.6. Sputtering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 970
4.1.7. Plasma spray . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 970
4.1.8. Alloys . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
4.2. Copper introduction onto titanium surfaces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
4.2.1. Ion implantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
4.2.2. Ti–Cu alloys . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
4.2.3. Sputtering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
4.2.4. Sol–gel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 973
4.2.5. Chemical vapour deposition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 973
4.3. Zinc introduction onto titanium surfaces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 973
4.4. Other strategies adopted to impart antibacterial properties to titanium surfaces . . . . . . . . . . . . . . . . . . . . . . . . . . . . 974
5. Conclusions and future outlooks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 975
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 976

⁎ Corresponding author.
E-mail address: sara.ferraris@polito.it (S. Ferraris).

http://dx.doi.org/10.1016/j.msec.2015.12.062
0928-4931/© 2016 Elsevier B.V. All rights reserved.
966 S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978

1. Introduction organized and co-operative community, as in a city, protecting them-


selves from external adversity [17,19].
Bacterial contamination is a serious problem that can affect numer- Biofilm formation and development can be divided into a sequence
ous applications, from home and everyday life, to food handling and of events [17,18,19,20]:
storage, medical implants, clinical settings, biosensors and even to
space missions [1–8]. 1. A first reversible attachment of bacteria to the solid surface, mainly
The case of medical implants is particularly critical since an infection due to cellular Brownian motion and microbe-surface interactions:
can develop in a patient who has a compromised health and immune electrostatic, gravitational or Van der Waals forces, and the hydro-
system. Moreover, implant materials are preferential sites for bacterial phobic degree of the bacteria and of the matching biomaterial. In par-
adhesion. The development of a prosthetic infection actually involves ticular the roughness and surface topography have a great influence
about 1% of all total joint replacements, but this percentage dramatically at this stage.
increases in the case of revision surgery [9–11]. For example in the den- 2. Bacteria accumulation and irreversible attachment, which are medi-
tal field, peri-implantitis has been observed for up to 14.4% of implants ated by specific molecular and cellular interactions (adhesion pro-
in the first 5 years, but a higher incidence has been documented for lon- teins, proteinaceous appendages and EPS production)
ger follow-ups and after a first septic failure [12]. This complication 3. Biofilm maturation, the formation of bacterial microcolonies and the
leads to the necessity of a prolonged antibiotic therapy (which can last entrapment of planktonic cells in the EPS
for years) and eventually to the removal of the implant. The conse- 4. The detachment of some bacteria for the colonization of new surfaces
quence of these post-operative complications, and of the lack of effec-
tive treatments, is a significant increase in hospitalization times and As an example dental plaque is a biofilm typical of natural tooth and
costs, together with a stressful, painful and critical situation for the pa- dental implants [12]. When it colonizes the dental implant, generally
tient. In addition it should be recalled that a significant increase in anti- through the transmucosal abutment, and lead to peri-implantitis devel-
biotic resistant bacterial strains has been observed, and this has led to opment, it constitutes one of the major causes of implant failure [12]. In
the necessity of innovative prevention and treatment strategies [11]. particular a dynamic biofilm composition has been reported for dental
A brief description of the bacterial infection of medical implants is implant surfaces [21]: the first bacteria that approach the surface
reported in the first part of the present paper. Then a review of the are generally Streptococci and Actinomyces species, whose adhesion is
scientific papers concerning the surface modification of titanium affected to a great extent by the surface characteristics. Later, during bio-
substrates intended for medical implants is reported with a specific film maturation, other bacterial strains, such as Fusobacterium nucleatum
focus on the introduction of inorganic antibacterial agents (Ag, Cu, and Prevotella intermedia (firstly) and Porphyromonas gingivalis,
Zn). To the best of the authors' knowledge, these specific topics have Capnocytophaga gingivalis and Actinomyces israelii (later), appear, but
not been reviewed in the recent literature. In fact, the most recent re- they are influenced less by material surface properties. Furthermore it
views deal with antibiotic biomaterials for dental applications [12] has been observed that implant surface properties (roughness, hydrophi-
and antibacterial coatings for titanium implants [13,14]. However, in re- licity, chemical composition and even sterilization method) affect the
cent years, research has been focused on the investigation of alternative early colonization of the material but not biofilm maturation directly
antibacterial agents to antibiotics, as well as on surface functionalization [21]. On the other hand the type and amount of early surface contamina-
and modification, instead of coatings. This is due to the numerous draw- tion significantly influence the consequent plaque development on im-
backs related to the systemic use of antibiotics (development of antibi- plant surface [21].
otic resistant bacterial strains) as well as to delamination and Specifically the main pathogens involved in orthopaedic and
bioresorption related to the coatings on implants [11,15]. cardiovascular prosthetic infection are Staphylococcus aureus and Staph-
The aim of the present review is to collect and compare the recent ylococcus epidermidis; the former is the main colonizer of metallic im-
researches on surface modification of titanium surfaces in order to con- plants, while the latter is the main colonizer of polymeric surfaces [18,
fer antibacterial properties by the introduction of inorganic antibacterial 6].
agents, with a specific focus on metal ions (Ag, Cu, Zn). Since bacteria are 0.5–2 μm-sized cells, they can be modelled as
colloidal particles that interact with solid surfaces [16]. However, this
2. Bacterial contamination of medical implants simplification seems to be too approximate. The main problems of this
simplification are related to the heterogeneous structure of the mi-
Bacterial contamination of solid synthetic surfaces, which is a phe- crobes and to their above cited complex features, which mediate bacte-
nomenon that has been widely documented, depends on the specific ria–surface interactions (filamentous proteinaceous appendages and
characteristics of both the involved microbes and of the materials. produced exopolymeric substances). At the same time, the surface char-
As far as bacteria are concerned, their filamentous proteinaceous ap- acteristics of solid surfaces exposed to a biological environment change,
pendages (e.g. Pili, flagella) can support adhesion to solid surfaces; due to protein absorption and can favour bacterial adhesion if compared
these appendages are, on a nanoscale, about 10 nm in diameter and to bare inorganic ones.
100 nm in length. Their dimension is comparable with some features As far as the features of the surfaces are concerned, their chemical
typical of innovative surfaces (nano-roughness and pattern produced composition, charge, wettability and roughness are the main factors
on synthetic surfaces in order to promote cellular adhesion and differ- that can affect the interaction with bacteria [20,21]. For example
entiation) intended for medical implants and must be taken in consider- roughness, in the tenth of micron range, has been reported to dramati-
ation in the design of that substrates in order to avoid bacterial cally increase bacterial adhesion [22,23], while lower Ra values than
adhesion. Bacteria produce exopolymeric substances (mainly polysac- 0.2 μm, as well as surface nano-textures, do not improve biofilm forma-
charides and other macromolecules) which contribute to biofilm forma- tion [24,25,21]. Moreover it has been reported that rough and/or
tion [16] and consequent permanent contamination of implanted hydrophobic surfaces lead to higher bacterial colonization [21]. As an
surfaces. A biofilm can be defined as a community of different micro- example calcium phosphate coatings and roughened dental implants
organisms that are protected in a self-produced polysaccharidic matrix have shown increased bacterial contamination and peri-implantitis
(the EPS described above), and which are irreversibly attached to a sur- development [12].
face [17,18]. A biofilm protects organized bacteria from both fluid shear Moreover, protein absorption from biological fluid leads to changes
stress and the action of systemic pharmacological therapies. Moreover, in the original surface characteristics of the biomaterials, and conse-
it acts as a storage facility for nutrients and substances [17]. Moreover quently influences bacterial adhesion. For example it has been observed
bacteria live in the polymeric protective structure of the biofilm, as an that a greater surface absorption of plasma proteins enhances bacterial
S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978 967

adhesion [26]. Furthermore absorption of fibronectin can favour micro- bind to the bacterial cell membrane and are also able to penetrate the
bial attachment, while albumin absorption can inhibit it [20,6]. cell. The antibacterial activity of silver nanoparticles mainly depends
In particular increased bacterial adhesion has been documented for on their dimension and shape [31,36]. On the other hand the toxicity
inert metallic surfaces (e.g. steel), compared to more reactive ones of these nanoparticles is currently under investigation; it should be
(e.g. titanium), this phenomenon can be explained by the lower tissue underlined that few systematic studies have focused on Ag-
integration of inert substrates [6]. As widely reported in the literature, nanoparticles, despite their widespread application [37,38].
after implantation, a sort of “race for the surface” starts between cells The antibacterial activity of silver ions [39] and nanoparticles [40,
and bacteria: the consequence is a lower bacterial contamination of sur- 41], and their mechanism of action, have been demonstrated in vitro
faces that are easily colonized by cells [6,27]. on gram-positive (S. aureus) and gram-negative (E. coli) strains. Numer-
A slightly higher bacterial contamination (Escherichia coli) has been ous studies report values of the Minimum Inhibitory concentration
observed for Ti6Al4V, compared to the T6Al7Nb alloy [28]. Experimental (MIC) of silver ions against bacteria (including S. aureus and E. coli)
data comparing bacterial contamination cp Ti and Ti 6Al4V alloy are not and few ones consider also copper and zinc ones [42,43,44,45]. A signif-
extensively reported in literature, a study related to surface modifica- icant variability in the values can be registered among the various stud-
tion of grade 2 and grade 5 [29] Ti did not evidence important difference ies; however a difference of two orders of magnitude in the MIC of Ag+
between the bacterial contamination of the two substrates. compared to the Cu2+ and Zn2+ ones is generally reported. A compari-
Finally a certain ability to reduce bacterial colonization has been re- son between MIC values of Ag+, Cu2+ and Zn2+ and the corresponding
ported for several pure metals, and a rank of “antibacterial” metal has Lethal Doses 50 (LD50) values (for L929 mouse fibroblasts cells) [46] is
been proposed: gold, titanium, cobalt, vanadium, aluminium, chromium summarized in Table 1.
and iron [21], even though the effective antibacterial behaviour of these Moreover the antibacterial activity of metallic or oxide nanoparticles
surfaces has not yet been univocally demonstrated. Moreover, a distinc- (e.g. Ag, Ag/Au coated iron oxides, ZnO, CuO, TiO2, Al2O3, and Ga)
tion should be made between the anti-adhesive, bacteriostatic, bacteri- against various bacterial strains has been demonstrated [47,48].
cidal and cytotoxic effects of these elements. Finally Minimum Inhibitory Concentrations of metallic and oxide
nanoparticles against different bacterial strains, including S. aureus
3. Antibacterial agents and E. coli have been reported in the literature [43,49,50]. MIC values
comprised in the range 3–180 μg/ml have been reported for Ag nano-
Bacterial infections are usually treated and prevented by the local particles with 3–95 nm diameters [43,49,50]. As far as copper nanopar-
and systemic application of antibiotics. The main problems related to ticles (9–95 nm) are concerned values in the range 20–2500 μg/ml have
antibiotic therapies are the increasing development of antibiotic resis- been reported [49,50] for the MIC. Slightly lower values can be found for
tant bacterial strains (e.g. Meticilin Resistant Staphylococcus aureus — copper oxides nanoparticles (22–95 nm), in particular 100–5000 μg/ml
MRSA) and their narrow spectrum of activity, which results in the diffi- for CuO nanoparticles and 500–5000 μg/ml for Cu2O ones [50]. Finally
culty of treating infections caused by several bacteria. The development MIC in the range 2055–5000 μg/ml have been reported for ZnO nano-
of antibiotic resistant bacterial strains has recently been defined as particles (22–95 nm) [50].
“global threat” to human health that needs to be addressed [30]. These data underline the significantly higher antibacterial action of
In this context, an increasing interest in alternative antibacterial silver, compared to copper and zinc, which can support the higher num-
agents has been witnessed. In particular inorganic antibacterial agents ber of publication concerning silver enriched surfaces compared to Cu
can represent a challenging strategy to fight bacterial infections, with- and Zn enriched ones, as discussed in the following.
out the use of antibiotics.
It must be evidenced that most of the reported studies were con- 4. Surface modifications that can be introduced to obtain antibacte-
ducted using planktonic cells and they do not investigate biofilm forma- rial metallic surfaces
tion and growth: this is a not negligible limit when clinical implications
have to be deducted from lab data. This is in agreement with the usual Various techniques have been explored in the scientific literature
procedure of preliminary works: a general standard procedure for the and in industrial and clinical applications in order to obtain titanium
evaluation of antibacterial activity of biomaterials has not been defined surfaces enriched with antibacterial agents. In particular three main cat-
yet. egories of surface modifications can be considered for inorganic antibac-
Metal ions, salts and nanoparticles are part of one of the most impor- terial agents:
tant classes of inorganic antibacterial agents. Among these agents, silver
has long been known and used as an antibacterial agent [31]. Moreover 1) surface enrichment of the inorganic antibacterial agents without the
an effective antibacterial effect has also been pointed out for other metal introduction of a layer
ions, such as copper and zinc. 2) growth of a surface oxide layer (TiO2) and its doping with the anti-
The mechanisms of action of metals against bacteria have been re- bacterial agent (in the same passage or in a second step)
cently reviewed [32]. 3) deposition of a coating (TiO2 or foreign one, e.g. hydroxyapatite —
For example silver ions can effectively bind to thiol groups, a charac- HAp) and its doping with the antibacterial agent (in the same pas-
teristic of many proteins, and play a structural and functional role in the sage or in a second step)
bacterial cells, thus causing the death of microorganism. Ag ions can
alter the function and structure of proteins in the bacterial cell wall, These strategies and the main techniques used for their application
and lead to its consequent rupture. Moreover, they can bind and alter are briefly schematized in Fig. 1. The literature references for the report-
several enzymes, which are crucial for cellular respiration and metabo- ed strategies are cited and compared in the following paragraphs.
lism. Finally, they can interfere with DNA through cell division and rep-
lication [33]. The multiple actions of silver ions explain the reduced
Table 1
resistance development, compared to antibiotics, and also their broad
Minimum Inhibitory Concentrations (MIC), against various bacterial strains (including
spectrum of activity, which includes many bacteria, as well as fungi S. aureus and E. coli) and Lethal Doses 50 (LD50) for L929 mouse fibroblast cells.
and yeasts [31,33]. An analogous mechanism of action has been de-
MIC [μg/ml] LD50 [mmol/l]
scribed for other metal ions (e.g. Cu, Zn) [34,35].
Increasing interest in silver nanoparticles, as antibacterial agents, Ag+ 0.03–8 [42,43,45] 3.5 ∗ 10−3 [46]
has been shown [36,37,38]. Ag-nanoparticles have a large surface area, Cu2+ 256–448 [44,45] 2.3 ∗ 10−1 [46]
Zn2+ 768 [45] 3.6 ∗ 10−3 [46]
which allows a high silver ion release. Moreover, they can directly
968 S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978

Fig. 1. Strategies and main techniques for the introduction of inorganic antibacterial agents onto titanium surfaces.

4.1. The introduction of silver onto titanium surfaces substrates [53,54]. A change in the zeta potential, through a less
negative value (at about −40 mV at pH 7) than the control (at about
Extensive literature is available on this topic, but the comparison be- −100 mV at pH 7), as well as an increase in hydrophobicity and surface
tween the various research work results quite difficult because of the nano-hardness have been observed after these treatments [53,54]. Al-
different test methodologies employed (release media and times, bacte- most no Ag release (b10 ppb in 60 days) has been observed for these
rial strains, cellular type and culture conditions). Depending on the surfaces, but significant antibacterial behaviour (95–100% reduction)
employed technique, silver has been introduced as both ions and as against E. coli and S. aureus [53,54] and good tolerability, by the osteo-
metal nanoparticles. All the reported materials have resulted in antibac- blasts, have been documented [53]. Considering the negligible Ag+ re-
terial behaviour, but the mechanisms and times of action are different, lease observed for these surfaces electrostatic interactions and
according to whether ions and/or nanoparticles are involved: a system- microgalvanic phenomena have been suggested in order to explain
atic comparison of the various actions of ions and nanoparticles will be the antibacterial action, however, the mechanism of this action still re-
of particular interest in future works, since it has not yet been fully in- quires further investigation.
vestigated. Biocompatibility, with respect to osteoblasts, depends on Titanium oxide layers, doped with silver, have also been obtained by
the amount of silver released. Moreover it should be considered that means of ion beam assisted deposition (IBAD), performed in an oxygen
dental and orthopaedic applications suffer from similar problems atmosphere, on titanium substrates, polished [55] or plasma spray coat-
concerning bone-implant contact, but they differ significantly ed [56], using silver and titanium targets [55]. Metallic silver has been
concerning the required time of action (much longer than 1 month, introduced into the oxide layer (constituted by TiO, Ti2O3 and TiO2)
for dental implants), as well as soft tissue-implant interaction (the during the IBAD process. An increase in surface tension and energy
gum is in contact with the collar of dental implants). has been recorded for higher Ag concentrations. The realization of dif-
Either a higher hydrophobicity or hydrophilicity has been obtained, ferent sizes of Ag nanoparticles (4–25 nm), by means of the IBAD tech-
after silver addition, because the various modifications proposed pro- nique [56], has pointed out better antibacterial behaviour against E. coli
duced surfaces with different roughness and surface composition of and S. aureus for particles larger than 5 nm, together with a good toler-
the final material. Roughness magnitude and topography, as well as sur- ability by the osteoblasts. Considering the negligible silver release ob-
face chemical composition are the main factors affecting surface served for these surfaces (b10 ppb Ag cm2 after 70 days in water,
wettability. analogous to the one cited before for PIII process [53,54]) the antibacte-
rial activity (in the dark) has been attributed to an electron storage
capability of silver nanoparticles [56].
4.1.1. Ion implantation Moreover ion beam assisted deposition has been employed to obtain
Ion implantation has been employed for the introduction of silver Ag-doped hydroxyapatite, which is used as a coating on titanium sub-
ions onto the surface of steel, c.p. titanium and Ti6Al7Nb and Ti6Al4V al- strates, with increasing crystallinity from the top (surface) to the bot-
loys [51,52]. Depending on the ion dose (up to 2 ∗ 1017 ions/cm2) an in- tom (substrate interface) of the coating [57]. Silver nanoparticles of
creasing silver content can be introduced on the metallic surfaces [51, about 5–30 nm were dispersed through the coating. The less crystalline
52]. Antibacterial metals have been obtained against S. aureus [51,52] portion of the coating can be rapidly dissolved in solution, while the
for ion implantation in the range 1 ∗ 1016 ions/cm2–2 ∗ 1017 ions/cm2 more crystalline part leads to stability of the surface layer and acts like
[51,52]. Biocompatibility for osteoblasts cells has been verified for Ag a silver reservoir. Accordingly, the release of silver into water it initially
implanted (b1 ∗ 1017 ions/cm2) Ti6Al4V surfaces [52]. Finally an resulted in a high value (from 0.4 to 0.8 ppm in 24 h and from 0.78 to
improvement/maintenance in wear/corrosion resistance has been 1.70 in 172 h, depending on the silver content introduced) and it then
observed for Ag ion implanted metals [51]. it decreased (for up to 15 days). The introduction of 3 wt.% of silver
The Plasma Immersion Ion Implantation (PIII) process induces the allowed a surface to be obtained that was antibacterial against
precipitation of silver nanoparticles (5–8 nm in diameter) onto titanium S. aureus and biocompatible for osteoblast cells.
S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978 969

4.1.2. Electrochemical techniques rapid ion release and consequent Ag accumulation in the culture medi-
A titanium oxide enriched in silver has been obtained by means of um induced a limited cytotoxic effect on the osteoblasts. The realization
plasma electrolytic oxidation (PEO) on Ti6Al7Nb, in an electrolyte of smaller nanotubes, the modulation of the oxidation reaction in water,
based on calcium acetate/calcium glycerophosphate, containing com- the deposition of a hydroxyapatite, polymeric or even “customized”
mercial Ag nanoparticles (7–25 nm) [58,59]. Silver nanoparticles were layer (able to favour ion release only in the presence of bacterial toxins)
introduced onto the surface of the oxide, into its pore walls and embed- have been proposed as possible solutions for the modulation of silver re-
ded throughout the entire coating thickness. An increase in surface lease and the reduction of the cytotoxic effect [65].
roughness and wettability was detected, after the surface modification The presence of a nanotube layer (with or without silver doping) has
process [59]. The silver enriched surface showed the ability to complete- induced a significant improvement in substrate wettability (the contact
ly kill methicillin-resistant S. aureus after a culture of 24 h [59]. angle of titanium surfaces was 42.9° for water and 23.2° for
Moreover titanium oxide layers, enriched in silver, have been obtained diiodomethane and it drops to 0° after nanotubes deposition) [67].
on Ti6Al4V alloy by means of anodic spark deposition, in an aqueous elec- The antibacterial activity of these typologies of coatings has been
trolyte containing silver nanoparticles [60]. The silver was detected as demonstrated against S. aureus [65,66,67] and E. coli [67], and has
small particles (mainly metallic), distributed within a 100 nm thick sur- been maintained for up to 30 days for TiO2 nanotubes containing Ag
face layer. The Ag ion release from the treated surface, in PBS, presented nanoparticles at the highest concentration [65]. An increase in the anti-
a maximum at 24 h (1.2 ng ∗ g−1 ∗ mm−2) and then remained constant bacterial activity with the nanotube diameter has been observed, with
for 15 days (0.4 ng ∗ g−1 ∗ mm−2). The silver containing surfaces present- an optimum value for 100 nm [67].
ed effective antibacterial behaviour (complete killing after 12 h incuba- Furthermore silver nanoparticles, stabilized by citrate (6 nm obtain-
tion, superior than pure silver) against gentamicin-resistant S. epidermidis. ed by means of a NaBH4 reduction of AgNO3), have been deposited,
Plasma Electrolytic Oxidation (PEO), in an electrolyte containing Ca, through absorption, onto a polished titanium substrate [68]. The parti-
P and Ag-nanoparticles, has also been performed on a plasma spray (PS) cles aggregated in multi-layer islands (100–300 nm). The surface cover-
titanium coating on a Ti6Al4V alloy [61] in order to obtain a surface layer age depended on the absorption time. A better adhesion to the substrate
with interconnected micro- and nano-porosity and antibacterial activi- was observed for the aggregated particles than for the isolated ones. Ef-
ty. A TiO2 microporous (0.07–5 μm pore) layer, rich in Ca, P and silver fective antibacterial behaviour (both reduction of bacterial adhesion
nanoparticles, was obtained. The coating follows the starting surface and bactericidal effect) against Pseudomonas aeruginosa was
macro-roughness of the PS coating. The Ca/P ratio was similar to that demonstrated.
of the hydroxyapatite one and the Ag nanoparticles were dispersed Finally alginate/chitosan polyelectrolyte multilayers, modified by
both on the surface and in the pores of the coating. dopamine, have been obtained on commercially pure titanium, using
Composite nanoparticles of TiO2 (23 nm) and Ag (4 nm) have been the layer-by-layer technique. These layers were enriched with silver
obtained by means of nucleophilic reaction, and were then deposited nanoparticles, using the reduction ability of dopamine on AgNO3 [69].
on titanium substrates by electrophoresis in an aqueous solution [62]. The thus obtained coatings were biocompatible for fibroblasts and anti-
The optimization of the deposition parameters allowed homogeneous bacterial against E. coli and S. aureus (inhibition halo of 2.85 ± 0.24 mm
surfaces to be obtained, with a limited number of cracks. The mechani- and 2.13 ± 0.15 respectively).
cal properties of the surfaces were acceptable and comparable with
those reported in literature (HV ≈ 210 kg/mm2, E ≈ 29 GPa and 4.1.4. Ion exchange
Kic ≈ 0.86 MPA m1/2). The surfaces presenting TiO2 and TiO2-Ag nano- Silver has been introduced onto titanium surfaces by means of an
particles were able to induce apatite precipitation, after 1 week of ion-exchange process, after the formation of a sodium titanate layer in
soaking in a simulated body fluid. However, a reduction in bioactivity a NaOH solution, through the exchange between Na+ and Ag+ ions
was observed when the silver content was increased. A decrease in [70,71]. Silver acetate [70] and nitrate [71] were considered as sources
the availability of the Ti–OH groups, because of the formation of Ti–O– for the Ag+ ions. The surface introduction of silver, both in ionic and me-
Ag bonds, has been suggested as an explanation of this phenomenon. tallic (nanoparticles) form, has also been recorded [70]. A consistent Ag
Finally a Plasma Electrolytic Process (a combination of plasma elec- release was observed in a physiological saline solution (300–510 ppb),
trolytic oxidation and electrophoretic deposition) has been applied to in a PBS (320–440 ppb) and in a foetal bovine serum (64,000–
c. p Ti substrates, in order to obtain composite coatings (about 83 μm 82,000 ppb), after 24 h. The significantly higher value in the serum
thick), and made of titanium oxide and silver enriched hydroxyapatite can be attributed to the high affinity between the Ag ions and the pro-
(Ag-HAp) [63]. The silver enriched hydroxyapatite was prepared by teins [70]. A faster silver release from the titanate, due to the presence
adding 2.5 wt.% of silver to a HAp microwave synthesis, in order to ob- of silver ions, and a slower one from the metallic nanoparticles, have
tain antibacterial behaviour, without altering the bioactivity and bio- been suggested for these surfaces [70]. Effective antibacterial behaviour,
compatibility of the HAp. Ag-HAp particles (60–70 nm) were added to against S. aureus [71] and Meticillin Resistant Staphylococcus aureus
the electrolyte for the Plasma Electrolytic Process. The obtained coatings (MRSA) [70], was observed. Bioactive behaviour (apatite forming ability
improved the corrosion resistance of the surface, both at a physiological in simulated body fluid) was observed for all the treated surfaces, before
(7.4) and an acidic pH (4.5, simulation of osteoclasts resorption). The and after Ag-introduction [71].
coatings were also bioactive and presented antibacterial behaviour Moreover the introduction of silver ions onto biocompatible [72] and
(inhibition zone) against E. coli. bioactive [73] glass/glass-ceramic coatings, deposited on titanium sub-
strates, has been considered. The possibility of tailoring the silver con-
4.1.3. In situ synthesis of silver nanoparticles tent, and consequently the antibacterial activity, by varying the ion
Silver nanoparticles have been obtained on different TiO2 substrates, exchange condition, has been verified. Silver ion release in SBF up to
through a UV reduction in AgNO3 solutions [64,65,66,67]. The amount of 4 μg/mm2 has been obtained. Ag-doped coatings are antibacterial
deposited nanoparticles depends on the concentration of the silver ni- against stock and clinically isolated S. aureus strains (2–5 mm inhibition
trate solution [65,66], while their size can be modulated by varying halos and 95–100% reduction of bacterial adhesion), bioactive (promote
the irradiation time [66]. hydroxyapatite precipitation in simulated body fluid) and biocompati-
For example the TiO2 nanotubes (130 nm diameter and 7 μm ble (for osteoblasts cells) [72,73].
length), enriched with silver nanoparticles (10–20 nm), showed a sig-
nificant Ag release in PBS: it is maximum in the first days (0.15– 4.1.5. Sol–gel
0.45 ppm after 24 h depending on the silver content) and then slowed The sol–gel technique has been applied to prepare TiO2–Ag compos-
down over 14 days (a half bacterial reduction after 14 days) [65]. The ite nanoparticles [74], powders [75,76] or coatings; spin-coating has
970 S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978

been used on silicon [77] and titanium [78] substrates, while dip- have been obtained on a Ti layer, sputtered onto silicon, by means of
coating has been used on glass [79]. In most of the works [77,74,79, the sputtering deposition of an Ag film, a thermal treatment (700 °C)
75], titanium isopropoxide and silver nitrate were used as precursors and the application of a uniform electric field [87]. The capping of Ag
of TiO2 and Ag, respectively. Tetrabutyl titanate [78] and titanium sul- structures within a TiO2 layer was performed by means of sol–gel dip
phate [76] have also been considered as alternative sources for TiO2 syn- coating in [87], in order to modulate the silver release; the largest quan-
thesis. In the sol–gel process, silver is introduced in the ionic form tity of Ag ions was detected in the solution in the first 8 days (at about
(AgNO3) and converted to metallic nanoparticles during the calcination 550 nM/ml for both Ag nanorods and capped Ag nanorods) and the
process; Ag ions are larger than Ti ones, and, as a result, they cannot quantity then reached a plateau only for the capped structures. Effective
enter the Ti crystal lattice and are reduced to metallic nanoparticles in antibacterial activity of these coatings was verified against E. coli (com-
the thermal treatment [77,79]. They are also effective in inhibiting plete killing of bacteria, 105 CFU/ml, in 1.75–2 h).
TiO2 grain growth [79]. Some authors have witnessed the formation of Moreover the sputtering technique has been employed for the
silver oxides after air annealing [74] or UV irradiation [77]. A noticeable deposition of thin films (at about 600 nm) of hydroxyapatite (HAp)
effect of silver on the crystallization of titanium oxide has also been and Ag-doped hydroxyapatite (Ag-Hap, 0.5 and 1.5 wt.% Ag) on titani-
observed, and a stabilization of rutile, after air annealing, and of anatase, um substrates [88], using pure HAp sintered tiles and silver strips as
after N2 annealing, has been observed [74]. Ion release tests in water targets. An optimal adhesion was obtained on plasma etched titanium
have shown that the main contribution occurs in the first hour (0% damage by tape test), while a certain dissolution of the coating
(0.4 ppm) and continues for up to 1 day (0.26 ppm), with an almost was observed in PBS (Ag reduction to 0.15–0.25 wt.% after 4 weeks in
complete release after 7 days (0.005 ppm) [77,76]. The antibacterial PBS). The Ag containing coatings reduced the adhesion of both
activity of sol–gel TiO2–Ag composites was verified against E. coli (2.5– S. epidermidis (Gram-positive) and P. aeruginosa (Gram-negative) with
7 mm inhibition halo, 100% viability reduction) [77,78]. An improve- a greater effect on the latter, but the effect decreased after 48 h, proba-
ment in TiO2 photocatalytic activity, due to the addition of silver, was bly due to partial coating delamination in the culture medium.
also evidenced [77,64,79]. Finally ZrO2 and ZrO2 doped with silver or copper have been depos-
The sol–gel technique has also been employed for the coating of ited on titanium substrates, by means of twin-gun reactive magnetron
macroporous 3D titanium scaffolds with Ag-doped hydroxyapatite sputtering [89]. Cu and Ag entered the coating in both an oxide and me-
[80]. Titanium scaffolds were acid etched and NaOH treated. A 30 μm tallic form; when silver was deposited, silver nanoparticles appeared on
thick hydroxyapatite coating was obtained starting from a slurry con- the surface. The sputtered zirconia was in monoclinic form and it main-
taining calcium nitrate tetrahydrate, phosphoiric pentoxide, ethyl alco- tained this structure after copper addition, while it became tetragonal
hol and silver nitrate. The antibacterial effect against E. coli and after silver addition. The presence of silver nanoparticles increased sur-
albusStaphylococcus increased with the silver content in the apatite face hydrophobicity (at about 95° compared to the 80° of ZrO2): this
(N95% for 0.8 and 1.6 wt.%), while an opposite trend was registered phenomenon was related to the antibacterial activity of the surface.
for biocompatibility, with respect to osteoblast cells. The introduction Both the Ag and Cu doped ZrO2 coatings demonstrated antibacterial ac-
of 0.8 wt.% of silver into the hydroxyapatite resulted in the best tivity against S. aureus and Actinobacillus actinomycetemcomitans, with a
compromise. more pronounced effect for silver (at about 70% bacterial reduction for
Finally in vivo tests, on a rabbit model, in infected surgical site by Cu doped ZrO2 and 97% reduction for Ag doped one on both strains). A
local introduction of bacteria (S. aureus), have been performed for tita- moderate reduction of bacterial adhesion has been evidenced also for
nium bone screws coated, or not, with a sol–gel coating of TiO2–Ag- ZrO2 coatings compared to bare titanium.
nanoparticles. It was observed that the coating resists the implantation
friction and appears unaltered after the implant in bone. The results re- 4.1.7. Plasma spray
vealed the effective ability of coated screws to reduce biofilm formation Ag–TiO2 coatings have been obtained on titanium substrates by
(no biofilm was observed on coated screws in contrast to uncoated means of air plasma spraying [90]. Metallic silver and silver oxide
ones), with no accumulation of silver in the tissues (cornea, kidneys, (due to the oxidation developed in the air deposition process) were ho-
liver or brain analysed 28 days after implantation) [81]. mogeneously distributed through the surface layer. The introduction of
silver did not alter the wettability or the biocompatibility of the TiO2
4.1.6. Sputtering coating. The Ag release into water was at a maximum after 24 h (at
The sputtering technique has been employed to deposit a TiO2 coat- about 0.11 μg/(ml∗cm2)) and then slowly continued for 28 days. An ef-
ing, containing silver nanoparticles, using Ag–TiO2 composite targets fective antibacterial action against E. coli (100% reduction after 24 h)
[82,83], or Ag and Ti targets, and a reactive atmosphere [84,85]. Silver was observed for these coatings.
was present in metallic form [82,83] in the sputtered layers. The intro- Furthermore plasma spray titanium coatings, sprayed onto Ti6Al4V
duction of silver affected the crystalline structure of the film (some ru- substrates, have been treated in NaOH and in a calcification solution
tile peaks were observed for 2.5 and 20% Ag). The hydrophylicity and containing Ca, PO4, Na and Ag, in order to obtain a silver enriched hy-
photocatalytic activity of the TiO2 films were significantly enhanced by droxyapatite layer on the surface (silver content from 1.50 to
the introduction of silver (up to 5 vol.%), because of the better separa- 5.15 wt.%) [91]. The Ag was released in a simulated body fluid and
tion between the electrons and holes, the increase in oxygen anion rad- showed a maximum at 24 h (at about 0.4 ppm for the highest silver con-
icals (O−2 ) and the reactive surface centres represented by Ti
3+
[82,83]. centration). The antibacterial activity of the coatings was verified
Silver release tests in artificial saliva and in PBS evidenced an Ag amount against E. coli, P. aeruginosa and S. aureus (bacterial reduction higher
higher than the threshold value necessary to obtain antibacterial behav- than 95% was registered for all coatings against the tested bacteria, ex-
iour (0.1 ppb); with a maximum being reached at 24 h [84,85]. An effec- cept for the lowest Ag concentration against S. aureus that induced at
tive antibacterial action against S. aureus was verified, together with about 63% reduction).
biocompatibility for fibroblast cells [84,85]. Finally different coatings (Hydroxyapatite (HAp), Ag2O, SrO or
The deposition of TaN–Ag coatings by means of sputtering has also Ag2O–SrO doped hydroxyapatite) have been deposited on commercial-
been considered on titanium substrates, using Ta and Ag targets and ly pure titanium by means of the plasma spray technique [92] and com-
an Ar or N2 flux [86]. Silver (up to 21.4 at.%) has been introduced in pared; a good adhesion was obtained (16 MPa required for HAp). A
the TaN coating in the form of well distributed Ag nanoparticles with di- similar Ag release rate in PBS was observed for the Ag– and Ag–Sr-
ameter comprised between 15 and 53 nm. An effective antibacterial ac- doped HAp coatings after 7 days of soaking (at about 0.8 ppm at 24 h
tion has been demonstrated against S. aureus (N 90% reduction) together and 2 ppm after 7 days), indicating that Sr does not affect silver release.
with biocompatibility for fibroblasts cells [86]. Finally, Ag nanorods While some signs of cytotoxicity were noted for Ag-HAp, good
S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978 971

biocompatibity for osteoblasts was instead observed for Ag–Sr-HAp. It that are used to introduce copper onto titanium substrates, in metallic
seems that Sr can reduce and balance the toxic effect of Ag on cells. or oxide form.
Both coatings were antibacterial against P. aeruginosa (almost complete
reduction after 24 h). 4.2.1. Ion implantation
As mentioned before, ion implantation has been employed to
4.1.8. Alloys introduce different antibacterial ions (Ag, Cu) onto metallic substrates
Ti–Ag (Ti–1Ag, Ti–2Ag and Ti–4Ag) [93] and TiNiAg (Ti 49.3, Ni 47.3 [51,97]. As far as copper is concerned, an increased antibacterial activity
and Ag 1.4 at.%) [94] alloys have been developed in order to obtain bulk against S. aureus was observed, as the copper content on the surface in-
antibacterial metals for biomedical applications. creased (0.5–4 1017 ions/cm2, concentrations comparable with the ones
Sand-blasting, by means of large grits, and acid etching of the Ti–Ag previously described for silver). Cu ion implantation induced a slight
alloys resulted in a micro-rough surface, thus much more Ag was ex- improvement in the wear resistance of the metallic substrate. On the
posed, and the alloy was able to release Ag ions in PBS (at about other hand, and contrary to what has been observed for silver implanted
1000 ppb after 24 h and 4000 ppb in 7 days for the highest silver con- surfaces, described in the previous paragraph, a reduction in corrosion
centration) and the amount of released silver was consistent with the resistance was recorded for Cu-enriched samples.
Ag content in the alloy. The antibacterial behaviour of the thus treated
alloy was verified against Streptococcus aureus (N 95% reduction for all 4.2.2. Ti–Cu alloys
the tested concentrations), while biocompatibility was verified on the Ti–Cu alloys with different copper contents (1% and 5%, [96] and
osteoblast cells of mice for all the Ag concentrations [93]. 10 wt.% [98]) have been prepared. Ti2Cu and Cu rich phases have been
Looking at the reported data, the majority of the modified surfaces detected on the 10% Ti–Cu alloy [98]. A significant increase in the
present an antibacterial behaviour due to a silver release higher than micro-hardness, yield strength and compressive strength was observed,
the MIC of Ag+ ions for the specified bacterial strain. However, an anti- as well as a reduction in ductility, for the Ti–Cu alloy, compared to bare
bacterial action has been evidenced for AgNPs obtained on Ti substrate Ti [98]. Unlike what was reported for Cu ion implanted surfaces [97], an
by PIII and IBAD processes, and has been attributed to electrostatic in- increase in the corrosion resistance was observed [98]. No inhibition
teractions, microgalvanic phenomena and electron storage capability halo was observed for the 10% Ti–Cu alloy, but a significant reduction
of AgNPs [53,54,56]. Considering that silver ions release is usually max- in bacteria adhered onto the surface was observed for both the
imum in the first day and no author reported ion release longer than 10 wt.% Ti–Cu alloy [98] and for the 1% and 5% ones [96]. In [98], a low
1 month, the opportunity to obtain silver enriched surfaces able to dem- Cu release (0.05 mg/l after 72 h in a physiological solution) indicated
onstrate an antibacterial action without ion release seems interesting that bacteria can only be killed when they are directly in contact with
and needs further investigations. the surface. In [96], it was shown that antibacterial behaviour against
Another key point, in the development of innovative antibacterial S. aureus and E. coli increased as the copper content in the alloy in-
metallic surfaces for bone contact application, is to guarantee good creased, as well as the cytotoxic effect on fibroblast cells; a 1% Cu con-
bone integration. As reported, there is a sort of “race for the surface” tent was shown to be the most promising one to limit bacterial
upon implantation [6,27] between cells and bacteria. Fast and physio- infection and guarantee tissue integration. No cellular tests were report-
logical bone integration can consequently reduce the risk of bacterial ed in [98].
colonization. Taking into account these considerations, a surface able
to induce optimal bone integration and, at the same time, exert an anti-
4.2.3. Sputtering
bacterial action is extremely promising for clinical applications. Among
Magnetron sputtering has been employed for the deposition of a
the reported research articles, a bioactive behaviour (material ability to
copper containing film on Ti6Al4V [99] and steel [100] substrates. In
bond to bone by the formation of an apatite layer) can only be evi-
the former case, different techniques were considered (direct current
denced for these materials: Ag-doped hydroxyapatite [57,63,80,88,91,
magnetron sputtering, dual magnetron sputtering and dual high
92], Ag-doped bioactive glass coatings [72,73], Ag enriched NaOH treat-
power impulse magnetron sputtering), and different chemical composi-
ed Titanium [70,71] and few TiO2 layers [61,62]. Moreover, in [62] a re-
tions were obtained for the deposited films [99]. Generally, these films
duction in the bioactivity has been evidenced after silver addition, due
were constituted by metallic Ti and Cu, but their oxides were also de-
to the reduction of the free Ti–OH groups. In this context, the authors re-
tected. Cu release was on occasion verified in DMEM (up to 6 mmol/l
cently published the characterization of an innovative titanium surface
in 24 h), depending on the chemical characteristics of the sample; an in-
(obtained by means of a controlled oxidation in Ag-enriched hydrogen
crease in antibacterial behaviour (up to the complete reduction of
peroxide) constituted by titanium oxide embedded with metallic silver
planktonic bacteria and biofilm), as well as in cytotoxicity, was evi-
nanoparticles [95]. This surface is bioactive against S. aureus, biocompat-
denced as the amount of released copper increased. In the latter case,
ible for osteoblasts cells and bioactive (it induces hydroxyapatite pre-
TiN and Cu were deposited [100]. A reduction in the friction coefficient
cipitation after 15 day in SBF). No reduction of the bioactive behaviour
was observed for the coated samples. Unlike what has been reported for
has been registered upon silver addition, in this case.
ion implanted copper [51,97], and according the results reported for Ti–
A summary of the strategies utilized in the scientific literature to in-
Cu alloys [98], an improvement in corrosion resistance was observed for
troduce silver onto titanium surfaces is reported in Table 2. The main
these materials [100], probably because of the deposition of a TiN layer
features (Ag release, antibacterial activity and biocompatibility) of the
on the top of the surface. Antibacterial activity was demonstrated
modified surfaces are also reported.
against E. coli (80–90% bacterial reduction after 24 h, depending on
the coating characteristics).
4.2. Copper introduction onto titanium surfaces A plasma surface alloying technique has also been employed to
obtain a Cu–Ni layer on commercially pure titanium [101]. Composi-
Although an extensive amount of literature exists on the enrichment tional analyses evidenced a higher surface enrichment in Ni than in
of titanium surfaces with silver, very few research papers have consid- Cu, despite of the known cytotoxicity of Ni no biocompatibility stud-
ered the application of copper on titanium, as an inorganic antibacterial ies have been reported in [101]. The 10 μm coating conferred anti-
agent. This is unexpected because lower cytotoxicity is reported for cop- bacterial properties (almost complete reduction of bacterial
per than for silver [96], so copper is a very interesting antibacterial viability by direct counting of the colonies on the sample surface)
agent. The research works have reported various techniques (ion im- against E. coli and S. aureus. Antibacterial activity has been attributed
plantation, alloys, sputtering, sol–gel and chemical vapour deposition) to copper release, but no measurements are reported in [101]. An
972
Table 2
Techniques for the preparation of silver enriched titanium substrates and the related properties.

Technique Chemical state of Ag Release Tested bacteria Biocompatibility References

Ion implantation Ag ionic metallic/nanoparticles Not reported S. aureus Osteoblasts (if Ag b 1e17 ions cm−2, 20 keV) [51] [52]
(IBAD)

S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978


Plasma Immersion Ion Implantation Ag nanoparticles No (b10 ppb in 60 days) S. aureus, E. coli Osteoblasts [53] [54]
(PIII)
Ion beam assisted deposition (IBAD) Metallic/Ag nanoparticles No (b10 ppb cm2 in 70 days) for Ad doped S. aureus, E. coli Osteoblasts [55,56,57]
titanium.
Up to 15 days in water for Ag-np doped HAp
(max in the first hours 0.4–0.8 ppm in 24 h,
0.78–1.70 ppm in 172 h)
Plasma Electrolytic Oxidation (PEO) Ag-nanoparticles Not reported S. aureus Not reported [58,59,61]
Anodic spark deposition Ag nanoparticles In PBS max at 24 h (1.2 ng ∗ g−1 ∗ mm−2) and S. epidermidis Not reported [60]
constant up to 15 days (0.4 ng ∗ g−1 ∗ mm−2)
Electrophoresis TiO2–Ag nanoparticles Not reported Not reported Not reported [62]
Plasma Electrolytic Process (PEP) Ag-enriched HAp Not reported E. coli Not reported [63]
In situ synthesis of Ag-nps on TiO2 Ag nanoparticles In PBS maximum in the first days (0.15–0.45 ppm S. aureus, E. coli Moderate cytotoxicity for osteoblasts [64] [65,66,67]
by UV reduction of AgNO3 after 24 h) and slow down till 14 days
Ion exchange Ag ions/Ag nanoparticles 300–510 ppb in physiological saline solution, S. aureus, Meticillin Resistant Osteoblasts for Ag-doped bioactive glass [70,71,72,73]
320–440 pp. in PBS and 64,000–82,000 ppb in FBS, Staphylococcus aureus (MRSA) coatings
after 24 h for Ag-doped sodium titanate
Up to 4 μg/mm2 in SBF for Ag-doped bioactive glass
coatings
Sol–gel Ag nanoparticles Maximum after 1 h (0.4 ppm) in water for Ag E. coli, S. aureus Osteoblasts (0.8 wt.% Ag in HAp) [74,75] [76] [77]
nanoparticles containing TiO2 coatings [78] [79] [81]
Ag-doped HAp E. coli, S. albus [80]
Sputtering Ag-nanoparticles Artificial saliva, PBS max at 24 h S. aureus, Fibroblasts [82] [83] [84] [85]
A. actinomycetemcomitans [89]
Ag-nanorods HNO3 1 M, up to 20 days, maximum at 8 days E. coli Not reported [87]
(550 nM/ml) reduced for capped nanostructures
Plasma-spray Metallic/oxide in Ag–TiO2 coatings In water up to 28 days E. coli Osteoblasts [90]
(max at 24 h, 0.11 μg/(ml ∗ cm2)).
Ag-HAp on plasma spray Ti Max at 24 in SBF (up to 0.4 ppm) E. coli, P. aeruginosa, S. aureus Not reported [91]
AgO/SrO doped HAp In PBS up to 7 days (2 ppm) P. aeruginosa Osteoblasts (ok for Ag–Sr-HAp) [92]
Alloys Ti–Ag In PBS up to 7 days (4000 ppb) S. aureus Osteoblasts [93]
Controlled oxidation in Ag–H2O2 Ag nanoparticles In water 0.08–0.2 μg/ml in 24 h S. aureus Osteoblasts [95]
S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978 973

improved wear resistance was observed on the coated surfaces, com- favour rutile crystallization, and confirming other results already re-
pared to pure titanium. ported for silver containing titanium oxide films and previously
discussed [74,84,83]. A decrease in the film growth rate and in the
4.2.4. Sol–gel mean crystal size was observed, while a decrease in the grain growth
Copper doped TiO2 mono- and multi-layer coatings on Ti6Al4V sub- was also observed for silver containing titanium oxide [79]. Finally the
strates [46] and CuO/TiO2 composite nanofibres [102,34] have been ob- antibacterial behaviour of Cu-containing coatings was demonstrated
tained by means of the sol–gel technique. Titanium isopropoxide [102] against S. aureus (100% of bacterial reduction for a Cu content higher
and tetrabutoxytitanate [46] were used as TiO2 precursors, while copper than 1.5 mol%), as was the maintenance of coating properties for up to
acetate [46,102], copper nitrate [34] or copper nanoparticles [102] were 5 months of storage in ambient conditions with a 30% decrease for lon-
used as Cu ones. As far as coatings are concerned [46], the dip-coating ger times [104].
technique has been used to deposit the copper onto substrates and it The CVD technique has also been employed for the deposition of
was observed that the antibacterial action against S. aureus increased multi-layer titanium and copper oxide coatings [105]. A significant anti-
with the number of Cu-containing layers (up to 6log10 for 4 layers), bacterial activity against E. coli was noted under UV irradiation (N5log
but some signs of cellular suffering were observed, when the layer num- bacterial reduction after 20–40 min), and a significant effect of surface
ber was increased [46]. As far as fibres are concerned, electrospinning roughness on antibacterial behaviour was observed. In particular, the
was employed for [102,34] and a deleterious effect on the fibre structure effect of nanostructures on the improvement of the surface area and
was observed for copper acetate, while good results were obtained with Cu release was underlined. Cu release in distilled water was measured
Cu nanoparticles and copper nitrate. The TiO2 fibres were characterized in the range 3.3–5.5 ppm after 80 min immersion, depending on the
by anatase, while a rutile signal appeared for the Cu doped ones. A CuO coating type. Moreover, the ability of these coatings to self-regenerate
signal clearly appeared for fibres obtained using Cu nanoparticles [102], under UV irradiation was reported.
while it was not present for those obtained using copper nitrate [34]. In As far as copper enriched surfaces are concerned, the antibacterial
the latter formulation, Cu rich particles were observed by means of activity, analogously to silver enriched surfaces, is often attributed to
FESEM and microanalysis techniques. Antibacterial activity was tested the release of Cu2+ ions, except for Ti–Cu alloys [96,98] for which an ex-
against Klebsiella pneumonia for fibres obtained using Cu nanoparticles tremely low release was registered and antibacterial action attributed to
[102], and against S. aureus and E. coli for fibres obtained using Cu nitrate contact killing. Compared to silver, few data on copper release are
[34]. Increased antibacterial action was observed in the former case reported.
[102], when the copper content was increased, together with photocat- Moreover, no bioactivity data have been reported for Cu-enriched
alytic behaviour. Cell wall damage and DNA condensed deposits were surfaces.
visible for bacteria treated with Cu enriched fibres and with Cu nitrate A summary of the strategies utilized in the scientific literature for the
[34], which can be explained considering the mechanism of copper introduction of copper onto titanium surfaces is reported in Table 3. The
against micro-organisms. main features (Cu release, antibacterial activity and biocompatibility) of
Moreover TiO2–Cu composite nanoparticles (10 nm) have been ob- the modified surfaces are also reported.
tained by means of the UV photocatalytic reduction of copper chloride
containing TiO2 sol [103]. Anatase, ionic and metallic copper were iden- 4.3. Zinc introduction onto titanium surfaces
tified as the particle constituents. Significant antibacterial activity, in the
absence of light, against E. coli (complete inhibition with 21.4 μg/ml Only a few works can be found on the introduction of zinc to impart
nanoparticles) and S. aureus (complete inhibition with 14.3 μg/ml nano- antibacterial properties to titanium substrates. Similar techniques to the
particles) was observed for the composite particles. The indicated ones described for silver and copper doping, such as electrochemical
values are lower than the ones reported in the literature for pure titani- techniques, ion implantation and plasma spraying, have been used for
um and copper/copper oxide nanoparticles. It has been suggested that the introduction of zinc onto titanium substrates. Biocompatible and an-
negatively charged TiO2 particles could attract Cu ions and act as car- tibacterial surfaces have been obtained using these techniques.
riers, but no release tests are reported. Zn-doped titanium oxide coatings (5–10 μm thickness, well adhered
to the substrate) have been obtained on commercially pure titanium by
4.2.5. Chemical vapour deposition means of plasma electrolytic oxidation, introducing zinc acetate into the
Copper containing TiO2 coatings have been prepared on steel, silicon electrolyte solution [106]. Final concentration comprised between 4.6
and glass by means of pulsed Direct Liquid Injection Chemical Vapour and 9.3 at.% was obtained. XPS revealed that Zn entered the coating as
Deposition (DLI-CVD) using copper bis(2,2,6,6-tetramethyl-3,5- ZnO. Anatase and small amounts of rutile have been detected on Zn con-
heptadionate) and titanium tetra-iso-propoxide as Cu and TiO2 precur- taining coatings while only anatase was present onto Zn-free ones, thus
sors [104]. Anatase and metallic copper were individuated as the main suggesting a tendency to facilitate rutile crystallization for zinc, as al-
constituents of the coating; an early crystallization of rutile was also ready observed for silver and copper [74,82,83,104]. A significant in-
detected for the highest Cu content, suggesting the ability of copper to crease in surface wettability was observed upon oxidation, but no

Table 3
Techniques for the preparation of copper enriched titanium substrates and the related properties.

Technique Chemical state of Cu Release Tested bacteria Biocompatibility References

Ion implantation Ionic Not reported S. aureus Not reported [51] [97]
Alloys Ti–Cu Up to 72 h in physiological S. aureus, E. coli Fibroblasts (ok 1% Cu) [96] [98]
solution (0.05 mg/l)
Magnetron sputtering Metallic/ionic Up to 10 days in DMEM S. epidermidis, S. aureus, E. coli Some cytotoxic effect on osteoblasts [99] [100]
(up to 6 mmol/l in 24 h)
Plasma surface alloying Cu-Ni alloy Not reported E. coli, S. aureus Not reported [101]
Sol–gel Oxide Not reported S. aureus, K. pneumonia, E. coli Fibroblasts, signs of cytotoxicity for [46,102] [32]
high Cu content
UV reduction CuCl2 TiO2–Cu nanoparticles Not reported E. coli, S. aureus Not reported [103]
DLI-CVD Metallic Not reported S. aureus Not reported [104]
CVD Oxide Not reported E. coli Not reported [105]
974 S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978

alteration was introduced upon Zn addition. Zinc containing coatings dissolution in a Tris–HCl buffer, but the coating ability to induce hy-
are completely biocompatible for rat bone marrow stromal cells, droxyapatite precipitation in a simulated body fluid was maintained
which also present an increased proliferation and ALP production after 28 days of soaking. Zn release in Tris–HCl buffer was observed up
[106]. A significant reduction in the viability of E. coli (40–100% reduc- to 14 days (at about 1.5 ppm after 1 day and 4 ppm after 14 days). Zn
tion) and S. aureus (96–100% reduction) for Zn containing coatings, showed significant antibacterial behaviour against S. aureus (93% reduc-
which increased with the Zn content, was reported. A Zn release in a tion) [110] and E. coli (93% reduction) [35]. A detailed study on the zinc
Tris–HCl solution, proportional to the Zn content in the coating, was mechanism of action against E. coli has been performed, which revealed
observed for up to 14 days (up to 3.6 ppm in 14 days); the values that Zn ions induced significant damage of the cell wall and membrane,
were all below the cytotoxicity limits for Zn ions (at about 10 ppm). as well as DNA condensation and a loss of the internal cell organization
Moreover, using a similar electrochemical technique (anodic spark de- [35]. As widely discussed for Ag+ and Cu2+ ions, Zn2+ ions can easily at-
position), Huo et al. [107] obtained titanium oxide nanotubes (with differ- tach to bacterial cells, thanks to their positive charge (the opposite of
ent diameters of 30 and 80 nm) on commercially pure titanium, and they the bacterial one) and to their ability to tightly bond S-containing mol-
then enriched them with different amounts of Zn (up to 60.2 μg/cm2), ecules [35].
through a hydrothermal process. ZnTiO3 was observed on the Zn- According to the data reported for silver and copper, the antibacteri-
enriched surfaces, and a small increase in the nanotube wall thickness al action of zinc enriched surfaces is attributed to the release of Zn2+
was observed after the introduction of Zn. The Zn release in PBS increased ions, while no other mechanism has been suggested for these surfaces.
with the Zn content; it reached a maximum after 1 day (up to 0.06 ppm Bioactivity has been described only for Zn-enriched calcium silicate
after 24 h immersion), then it decreased with time, and was almost ab- coatings [110,35].
sent after 1 month. The nanotubes increased protein absorption on the A summary of the strategies utilized in the scientific literature for the
surface; this phenomenon depended to a great extent on the surface introduction of zinc onto titanium surfaces is reported in Table 4. The
nano-roughness, and the highest Zn containing surfaces (which are main features (Zn release, antibacterial activity and biocompatibility)
slightly smoother) therefore induced less protein absorption. The modi- of the modified surfaces are also reported.
fied surfaces were antibacterial against S. aureus, both adhered bacteria
and planktonic ones were significantly reduced (57–97% reduction of
planktonic bacteria and 77–100% reduction of adhered ones at day 1). 4.4. Other strategies adopted to impart antibacterial properties to titanium
The antibacterial behaviour increased with the Zn content and decreased surfaces
with time (45–78% reduction of planktonic bacteria and 43–87% reduc-
tion of adhered ones after 7 days, according to the release tests). A more The addition of fluorine [111,112], iodine [113,114], iron [115],
rapid decrease in antibacterial activity was observed in planktonic bacte- nitrogen [116] and cobalt [117] has also been considered to impart
ria, compared to the surface adhered ones, suggesting a double bactericid- antibacterial properties to titanium surfaces.
al mechanism of doped surfaces: Zn release and ROS production induced The ion implantation of F induces antibacterial activity against
by surface bonded zinc. Differently from the observation on Ag-enriched P. gingivalis and A. actinomycetemicomitans, even though no fluorine re-
nanotubes [65], no cytotoxic effects were observed for Zn doped surfaces, lease has been observed in a physiological saline solution after 7 days
while an increase in cellular spreading and mineral nodule formation was [111]. The activity was attributed to the formation of a metal-fluoride
observed for the Zn containing materials. complex on the surface. The F-modified samples resulted biocompatible
Plasma Immersion Ion Implantation deposition (PIIID) has been also for fibroblast cells. Unlike almost all of the studies in this field, the Ag
applied in order to enrich a c. p. Ti gr4 surface with zinc [108]. Varying and Zn enriched surfaces with Ion Beam Mixing and N-ones doped
the deposition time, different Zn concentrations were obtained (1.1– with TiN coating or N implantation did not show the ability to kill the
2.1 at.%). Increasing antibacterial activity against Steptococcus mutans, tested bacteria. Anodic oxidation in a fluorine-containing electrolyte
one of the main constituents of dental plaque, (69–96% reduction of ad- has also been considered in order to introduce F onto titanium surfaces
hered bacteria) was observed, as the Zn content was increased. [112]. The oxidation conditions were optimized to obtain a compact
Furthermore TiO2, Ti(Zn)O2 and ZnO have been deposited on com- layer and to separate the chemical effect from the topographical one.
mercially pure titanium (c. p. Ti) by cathodic arc deposition [109]. A po- The ability of F-enriched surfaces to inhibit 6 clinical strains of
rous layer was obtained for Ti(Zn)O2 (with a surface zinc content of S. aureus and S. epidermidis was demonstrated. This property was attrib-
7.6 at.%), and a fibrous one for ZnO (with a surface zinc content of uted to enzymatic activity inhibition by TiF4 complexes.
45.7 at.%). An increase in the water contact angle was observed for the The introduction of iodine onto titanium surfaces by means of anodic
coated samples (at about 84°, 85° and 89° for TiO2, Ti(Zn)O2 and ZnO, oxidation in an I-containing electrolyte has also been performed [113,
respectively) compared to c. p. Ti (at about 78°). A reduction in 114]. This process resulted in effective antibacterial activity against
S. aureus adhesion and viability was observed for the Zn coating (78% S. aureus and E. coli, without in vitro (fibroblast) or in vivo (rabbit) cyto-
for Ti(Zn)O2 and 87% for ZnO). Completely biocompatible behaviour toxicity [113]. Moreover, these I-enriched implants reduced inflamma-
was instead verified for the Ti(Zn)O2 coatings, while a certain cytotoxic tion and infection and improved bone integration. Clinical trials on
effect on osteoblasts was observed on the ZnO ones. humans [114] have demonstrated good antibacterial activity, without
Plasma spray coatings of Zn-enriched calcium silicate (Ca2ZnSi2O7), cytotoxic effects or thyroid abnormalities. After 1 year of implantation,
obtained by means of the sol–gel method, have also been considered 20–30% of the introduced iodine was still present on the material sur-
on Ti6Al4V substrates [110,35]. Zn addition reduced calcium silicate face [114]. A broad spectrum of iodine activity was reported, and the

Table 4
Techniques for the preparation of zinc enriched titanium substrates and the related properties.

Technique Chemical State of Zn Release Tested bacteria Biocompatibility References

Plasma electrolytic oxidation Oxide Up to 14 days in TRIS–HCl (max 3.6 ppm) E. coli, S. aureus Rat bone marrow stromal cells [106]
Anodic spark deposition ZnTiO3 In PBS max at 24 h, up to 0.06 ppm, complete S. aureus Mesenchymal stem cells [107]
after 1 month
PIIID Ionic Not reported S. mutans Not reported [108]
Cathodic arc deposition Ti(Zn)O2, ZnO Not reported S. aureus Osteoblasts (ok Ti(Zn)O2, some [109]
cytotoxicity for ZnO
Plasma spray Ca2ZnSi2O7 Tris–HCl up to 14 days (4 ppm) S. aureus, E. coli Osteoblasts [110] [35]
S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978 975

mechanism was attributed to an oxidizing action, related to protein de- functionalization with VEGF [127] or RGD [128] has been performed
naturation, and to consequent micro-organism destruction [114]. to improve the material bone integration. Carboxymethylchitosan
Moreover Fe containing titanium oxide films have been produced on (CMCS) has been bonded to titanium via several strategies (e.g. silane,
a glass substrate, by means of the sol–gel dip coating technique [115]. dopamine, polydopamine) [129]. Its resistance to several sterilization
Fe3+ ions entered the TiO2 crystal lattice, where they substituted the and ageing conditions was investigated, and it was demonstrated that
Ti4 + ions, thanks to the similarity between the ionic radii (Fe3 + = covalent bonding improved molecule resistance.
0.063 nm, Ti4+ = 0.068 nm). The Fe-doped TiO2 showed antibacterial Chitosan coating on TiO2 nanotubes loaded with selenium has been
activity against E. coli under fluorescent irradiation. considered in order to couple the anticancer activity of Se to the antibac-
N-doped TiO2 films have also been obtained on stainless steel by terial activities of chitosan [130].
means of oxidation of a TiN layer [116]. These films demonstrated anti- Antimicrobial peptides (AMPs) have also been considered, and re-
bacterial activity against S. aureus, under a fluorescent lamp. Increased cently reviewed [131], thanks to their wide spectrum of activity, high ef-
corrosion resistance was observed for the modified samples. ficacy at low concentrations, specificity, synergistic action with
Finally Co-enriched titanium oxide nanofibres (350–450 nm) have conventional antibiotics and low resistance development. AMPs are
been obtained by means of the electrospinning technique [117]. Co small (15–20 amino acids) and positively charged and can therefore
was observed as cobalt oxide, and it induced antibacterial activity bind to a bacterial surface, which is generally negatively charged. The
against S. aureus and Salmonella typhimurium. The proposed Co antibac- antibacterial activity of AMPs, after grafting onto solid supports, is cur-
terial action mechanism is analogous to the Ag, Cu and Zn one: the pos- rently under investigation; the necessity of a proper spacer has been
itive ion binds to the negatively charged bacterium, causing bacterial suggested [131].
cell walls to rupture and enzyme inactivation. On the other hand no Cecropin B peptide has been grafted onto a polished titanium surface
cellular tests were performed on these materials to verify their via an intermediate polydopamine layer [132] in order to induce anti-
biocompatibility. bacterial activity against S. aureus, B. subtilis, P. aeruginosa and E. coli.
Surface functionalization with organic molecules has also been con- Biocompatibility was verified for osteoblast cells, together with an
sidered to obtain antibacterial titanium surfaces. anti-inflammatory effect on macrophages.
In addition to the systemic administration of antibiotics, their
coupling with biomaterials has been considered in order to obtain 5. Conclusions and future outlooks
local delivery. The most common example of biomaterial-antibiotic
coupling is that of antibiotic loaded bone cements [118]. The surface modification of titanium substrates seems a challenging
As far as titanium implants are concerned, the possibility of intro- strategy to prevent the development of prosthetic infections. The
ducing the drug directly onto a metal surface has also been considered. grafting of an antimicrobial agent directly onto an implant surface pre-
Antoci et al. [119,120] covalently grafted vancomycin onto silanized vents bacterial colonization and allows the localized delivery of a thera-
titanium surfaces. The specificity of the antibiotic for gram positive bac- peutic agent to help prevent or treat an infection. The presence of the
teria was demonstrated after the grafting. The covalently bounded mol- therapeutic principle directly at the implant site can improve the effica-
ecule was not released in solution, but remained stable for up to 3 weeks cy of the treatment in case of biofilm development on implant surface.
in serum. Moreover this way of application can reduce the side effects of systemic
Vancomycin has also been incorporated in a sol–gel silica coating on application by reducing the amount of the antibacterial agent and by its
a Ti6Al4V alloy [121]. The antibiotic maintained its structure upon load- local delivery (only were needed).
ing and did not alter the silica structure. However, the coating degrada- Numerous solutions have been proposed in the scientific literature,
tion rate increased with the vancomycin content. An antibiotic release and have been reviewed in the present paper. The considered solutions
of up to 7 days was obtained with this technique. are mainly based on inorganic antibacterial agents (such as silver, cop-
Moreover vancomycin or tobramycin loading in hyaluronic acid/ per or zinc), which have been introduced in order to overcome the
polylactic acid hydrogel has been performed for the coating of titanium growing problem of bacterial resistance to antibiotics.
[122]. The coated samples were resistant to autoclave sterilization and The main open problems related to this strategy are the time limited
biocompatible for human dermal fibroblasts. release of the antibacterial agent (generally reaching a maximum in
Furthermore polymeric nanoparticles have been functionalized with 1 day and then rapidly decreasing over the next 10–30 days), the anti-
gentamicin sulphate through pH-sensitive bonding [123]. This strategy bacterial effect that increases with the concentration of the antibacterial
allows an antibiotic to only be released in the typical acidic pH of infec- agent, coupled to cytotoxic behaviour at the highest concentration and
tions. Functionalized nanoparticles were coupled with silanized titani- the limited resistance (e.g. chemical, mechanical) of the modified layer.
um by means of covalent bonding. Actually no titanium based implant with antibacterial properties is
Gentamicin has also been introduced onto titanium surfaces by available on the medical devices market, probably due to the aforemen-
means of gentamicin loaded bioresorbable PDLLA coatings [124]. The tioned open problems. Moreover the presence of an active principle that
coatings were antibacterial against Bacillus subtilis and biocompatible can be released from the material surface can complicate the classifica-
for osteoblasts. Antibiotic release was very quick (60% in the first min- tion procedure of the related medical device increasing time and costs of
ute), both in vitro and in vivo. commercialization. However, the problem of prosthetic infection is a
Finally a patented spray technique has been employed for the depo- critical issue for dental and orthopaedic implantology. In fact, prolonged
sition of gentamicin on Ti6Al4V blasted substrates [125]. A PLGA coating antibiotic therapies, the consequent development of antibiotic resistant
was also deposited to protect the antibiotic during implantation proce- bacterial strains, the sometimes consequent need to remove and to sub-
dures. The polymeric coating degraded, thus allowing gentamicin re- stitute the implant and increasing costs for the healthcare system are
lease. An effective antibacterial action was demonstrated against 30 the main consequences of prosthetic infections.
clinically isolated bacterial strains. In this context the development of titanium based surfaces with
Chitosan [126] and chitosan, coupled with hyaluronic acid, [127,128] effective and prolonged antibacterial action, a good bone integration
have also been explored as antibacterial molecules. Their effective ability and stability in the physiological environment can represent a
action against S. aureus has been demonstrated [127,128], and it has in promising solution to the problem.
particular been observed that chitosan presents an antibacterial action, The future research in the biomaterial field should be directed to the
while hyaluronic acid can only reduce bacterial adhesion [127]. Since development of innovative metallic surfaces able to improve and fasten
a certain reduction in osteoblast adhesion has been recorded for chito- tissue integration and offer a prolonged antibacterial action. The devel-
san and hyaluronic acid modified titanium [127,128], a further opment of surface modifications able to guarantee a prolonged release
976 S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978

of the active principle in sufficient amount for bacterial killing but in the Grade 2 and Ti6Al4V alloy for dental and orthopedic devices, Colloids Surf. B 88
(2011) 648–655.
biocompatible range represent and extremely challenging topic. On the [30] P. Stephens Antibiotic resistance now ‘global threat’, WHO warns, http://www.bbc.
other hand the opportunity to develop surfaces able to reduce bacterial com/news/health-27204988, (accessed 7th May 2014)
adhesion and exert a local contact killing action for bacteria, without the [31] M.L.W. Knetsch, L.H. Koole, New strategies in the development of antimicrobial
coatings: the example of increasing usage of silver and silver nanoparticles, Poly-
release of any substance, represent a promising strategy in order to mer 3 (2011) 340–366.
overcome classification and toxicity concerns. [32] J.A. Lemire, J.J. Harrison, R.J. Turner, Antimicrobial activity of metals: mechanisms,
molecular targets and applications, Nat. Rev. 11 (2013) 371.
[33] L.G. Ovington, The truth about silver, Ostomy Wound Manage. 50 (2004) 1S–10S.
References [34] M.S. Hassan, T. Amna, H.Y. Kim, M.S. Khil, Enhanced bactericidal effect of novel
CuO/TiO2 composite nanorods and a mechanism thereof, Compos. Part B 45
[1] M.F. Davis, S.A. Iverson, P. Baron, A. Vasse, E.K. Silbergeld, E. Lautenbach, D.O. (2013) 904–910.
Morris, Household transmission of methicillin-resistant Staphylococcus aureus [35] K. Li, Y. Xie, L. Huang, H. Ji, X. Zheng, Antibacterial mechanism of plasma sprayed
and other staphylococci, Lancet Infect. Dis. 12 (2012) 703–716. Ca2ZnSi2O7 coating against Escherichia coli, J. Mater. Sci. Mater. Med. 24 (2013)
[2] R.R.W. Brady, J. Verran, N.N. Damani, A.P. Gibb, Review of mobile communication de- 171–178.
vices as potential reservoirs of nosocomial pathogens, J. Hosp. Infect. 71 (2009) [36] M. Rai, A. Yadav, A. Gade, Silver nanoparticles as a new generation of antimicro-
295–300. bials, Biotechnol. Adv. 27 (2009) 76–83.
[3] M. Kousta, M. Mataragas, P. Skandamis, E.H. Drosinos, Prevalence and sources of [37] X. Chen, H.J. Schluesener, Nanosilver: a nanoproduct in medical application,
cheese contamination with pathogens at farm and processing levels, Food Control Toxicol. Lett. 176 (2008) 1–12.
21 (2010) 805–815. [38] S.W.P. Wijnhoven, W.J.G.M. Peijnenburg, C.A. Herberts, W.I. Hagens, A.G. Oomen,
[4] A.F. Engelsman, H.C. van der Mei, R.J. Ploeg, H.J. Busscher, The phenomenon of in- E.H.W. Heugens, B. Roszek, J. Bisschops, I. Gosens, D. Van de Meent, S. Dekkers,
fection with abdominal wall reconstruction, Biomaterials 28 (2007) 2314–2327. W.H. De Jong, M. Van Zijverden, A.J.A.M. Sips, R.E. Geertsma, Nano-silver — a re-
[5] P. Eggimann, H. Sax, D. Pittet, Catheter-related infections, Microbes Infect. 6 (2004) view of available data and knowledge gaps in human and environmental risk as-
1033–1042. sessment, Nanotoxicology 3 (2009) 109–138.
[6] L.G. Harris, R.G. Richards, Staphylococci and implant surfaces: a review, Injury 37 [39] Q.L. Feng, J. Wu, G.Q. Chen, F.Z. Cui, T.N. Kim, J.O. Kim, A mechanicistic study of the
(2006) S3–S14. antibacterial effect of silver ions on Escherichia coli and Staphylococcus aureus, J.
[7] R. Van Houdt, K. Mijnendonckx, N. Leys, Microbial contamination monitoring and Biomed. Mater. Res. 52 (2000) 662–668.
control during human space missions, Planet. Space Sci. 60 (2012) 115–120. [40] F. Mirzajani, A. Ghassempour, A. Aliahmadi, M.A. Esmaeili, Antibacterial effect
[8] C. Balagna, S. Ferraris, S. Perero, M. Miola, F. Baino, A. Coggiola, D. Dolcino, A. of silver nanoparticles on Staphylococcus aureus, Res. Microbiol. 162 (2011)
Battiato, M. Ch, E. Vittone, E. Vernè, M. Ferraris, Silver nanocluster/silica composite 542–549.
coatings obtained by sputtering for antibacterial applications, in: J. Njuguna (Ed.), [41] I. Sondi, B. Salopek-Sondi, Silver nanoparticles as antimicrobial agent: a case study
Structural Nanocomposites, Engineering Materials, Springer-Verlag Berlin Heidel- on E. coli as model for Gram-negative bacteria, J. Colloid Interface Sci. 275 (2004)
berg 2013, pp. 225–247. 177–182.
[9] E. Moran, I. Byren, B.L. Atkins, The diagnosis and management of prosthetic joint [42] T.J. Berger, J.A. Spadaro, S.E. Chapin, R.O. Becker, Electrically generated silver ions:
infections, J. Antimicrob. Chemother. 65 (Suppl. 3) (2010) iii45–iii54. quantitative effects on bacterial and mammalian cells, Antimicrob. Agents
[10] P.J. Papagelopoulos, A.A. Partsinevelos, G.S. Themistocleous, A.F. Mavrogenis, D.S. Chemother. 9 (1976) 357–358.
Korres, P.N. Soucacos, Complications after tibia plateau fracture surgery, injury, [43] L. Kvitek, A. Panacek, R. Prucek, J. Soukupova, M. Vanickova, M. Kolar, R. Zboril, An-
Int. J. Care Inj. 37 (2006) 475–484. tibacterial activity and toxicity of silver — nanosilver versus ionic silver, J. Phys.
[11] D. Campoccia, L. Montanaro, C.R. Arciola, The significance of infection related to ortho- Conf. Ser. 304 (2011) 012029.
pedic devices and issues of antibiotic resistance, Biomaterials 27 (2006) 2331–2339. [44] W.L. Du, Y.L. Xu, Z.R. Xu, C.L. Fan, Preparation, characterization and antibacterial
[12] P.A. Norowski, J.D. Bumgardner, Biomaterial and antibiotic strategies for peri- properties against E coli K88 of chitosan nanoparticles loaded copper ions, Nano-
implantitis, J. Biomed. Mater. Res. B Appl. Biomater. 88B (2009) 530–543. technology 19 (2008) 085707.
[13] L. Zhao, P.K. Chu, Y. Zhang, Z. Wu, Antibacterial coatings on titanium implants, J. [45] W.L. Du, S.S. Niu, Y.L. Xu, Z.R. Xu, C.L. Fan, Antibacterial activity of chitosan
Biomed. Mater. Res. B Appl. Biomater. 91B (2009) 470–480. tripolyphosphate nanoparticles loaded with various metal ions, Cabohydr. Polym.
[14] A. Simchi, E. Tamjid, F. Pishbin, A.R. Boccaccini, Recent progress in inorganic and 75 (2009) 385–389.
composite coatings with bactericidal capability for orthopaedic applications, [46] F. Heidenau, W. Mittelmeir, R. Detsch, M. Haenle, F. Stenzel, G. Ziegler, H.
Nanomed.-Nanotechnol. 7 (2011) 22–39. Gollwitzer, A novel antibacterial titania coating: metal ion toxicity and in vitro sur-
[15] P. Cheang, K.A. Khor, Addressing processing problems associated with plasma face colonization, J. Mater. Sci. Mater. Med. 16 (2005) 883–888.
spraying of hydroxyapatite coatings, Biomaterials 17 (1996) 537–544. [47] M.J. Hajipour, K.M. Fromm, A.A. Ashkarran, D.J. de Aberasturi, I.R. de Larramendi, T.
[16] K. Hori, S. Matsumoto, Bacterial adhesion: from mechanism to control, Biochem. Rojo, V. Serpooshan, W.J. Parak, M. Mahmoudi, Antibacterial properties of nano-
Eng. J. 48 (2010) 424–434. particles, Trends Biotechnol. 30 (2012) 499–511.
[17] D. Lindsay, A. von Holy, Bacterial biofilms within the clinical setting: what [48] J. Chapman, E. Weir, F. Regan, Period four metal nanoparticles on the inhibition of
healthcare professionals should know, J. Hosp. Infect. 64 (2006) 313–325. biofouling, Colloids Surf. B 78 (2010) 208–216.
[18] C.R. Arciola, D. Campoccia, P. Speziale, L. Montanaro, J.W. Costerton, Biofilm forma- [49] J.P. Ruparelia, A.K. Chatterjee, S.P. Duttagupta, S. Mukherji, Strain specificity in antimi-
tion in Staphylococcus implant infections. A review of molecular machanisms and crobial activity of silver and copper nanoparticles, Acta Biomater. 4 (2008) 707–716.
implications for biofilm-resistant materials, Biomaterials 33 (2012) 5967–5982. [50] G. Ren, D. Hu, E.W.C. Cheng, M.A. Vargas-Reus, P. Reip, R.P. Allaker, Characteriza-
[19] P. Watnick, R. Kolter, Biofilm city of microbes, J. Bacteriol. 182 (2000) 2675–2679. tion of copper oxide nanoparticles for antimicrobial applications, Int. J. Antimicrob.
[20] H.Y. An, R.J. Friedman, Concise review of mechanisms of bacterial adhesion to bio- Agents 33 (2009) 587–590.
material surfaces, J. Biomed. Mater. Res. B 43 (1998) 338–348. [51] Y.Z. Wan, S. Raman, F. He, Y. Huang, Surface modification of medical metals by ion
[21] K. Subramani, R.E. Jung, A. Molenberg, C.H.F. Hammerle, Biofilm on dental im- implantation of silver and copper, Vacuum 81 (2007) 1114–1118.
plants: a review of the literature, Int. J. Oral Maxillofac. Implants 24 (2009) [52] J. Fiedler, A. Kolitsch, B. Kleffner, D. Henke, S. Stenger, R.E. Brenner, Copper and sil-
616–626. ver ion implantation of aluminium oxide-blasted titanium surfaces: proliferative
[22] S. Hou, H. Gu, C. Smith, D. Ren, Microtopographic patterns affect Escherichia coli response of osteoblasts and antibacterial effects, Int. J. Artif. Organs 34 (2011)
biofilm formation on poly(dimethylsiloxane) surfaces, Langmuir 27 (2011) 882–888.
2686–2691. [53] H. Cao, X. Liu, F. Meng, P.K. Chu, Biological action of silver nanoparticles embedded
[23] B. Chehroudi, D.M. Brunette, Subcutaneous microfabricated surfaces inhibit epithe- in titanium controlled by micro-galvanic effects, Biomaterials 32 (2011) 693–705.
lial recession and promote long term survival of percutaneous implants, Biomate- [54] J. Li, Y. Qiao, Z. Ding, X. Liu, Microstructure and properties of Ag/N dual ions im-
rials 23 (2002) 229–237. planted titanium, Surf. Coat. Technol. 205 (2011) 5430–5436.
[24] V. Frojd, P. Linderback, A. Wennerberg, L.C. de Paz, G. Svensater, J.R. Davies, Effect [55] F. Zhang, G.K. Wolf, X. Wang, X. Liu, Surface properties of silver doped titanium
of nanoporous TiO2 coating and anodized Ca2+ modification of titanium surfaces oxide films, Surf. Coat. Technol. 148 (2001) 65–70.
on early microbial biofilm formation, Oral Health 11 (2011) 8. [56] H. Cao, Y. Qiao, X. Liu, T. Lu, T. Cui, F. Meng, P.K. Chu, Electron storage mediated
[25] M. Quirynen, C.M.L. Bollen, W. Papaioannou, J. Van Eldere, D. van Steenberghe, The dark antibacterial action of bound silver nanoparticles: smaller is not always bet-
influence of titanium abutment surface roughness on plaque accumulation and ter, Acta Biomater. 9 (2013) 5100–5110.
gingivitis: short term observations, Int. J. Oral Maxillofac. Implants 11 (1996) [57] X. Bai, S. Sandukas, M. Appleford, J.L. Ong, A. Rabiei, Antibacterial effect and cyto-
169–178. toxicity of Ag-doped functionally graded hydroxyapatite coatings, J. Biomed.
[26] C. Wagner, S. Aytac, G.M. Hansch, Biofilm growth on implants: bacteria prefer plas- Mater. Res. B Appl. Biomater. 100B (2012) 553–561.
ma coats, Int. J. Artif. Organs 34 (2011) 811–817. [58] B.S. Necula, I. Apachitei, F.D. Tichelaar, L.E. Fratina-Apachitei, J. Duszczyk, An elec-
[27] A. Gristina, Biomaterial-Centered Infection: Microbial Adhesion Versus Tissue Inte- tron microscopical study on the growth of TiO2–Ag antibacterial coatings on
gration Science, 2371987 1588–1595. Ti6Al7Nb biomedical alloy, Acta Biomater. 7 (2011) 2751–2757.
[28] M. Walkowiak-Przybyto, L. Klimek, W. Okroj, W. Jakubowski, M. Chwitka, A. [59] B.S. Necula, L.E. Fratila-Apachitei, S.A.J. Zaat, I. Apachitei, J. Duszczyk, In vitro anti-
Czajka, B. Walkowiak, Adhesion activation and aggregation of blood platelets bacterial activity of porous TiO2–Ag composite layers against methicillin-resistant
and biofilm formation on the surfaces of titanium alloys Ti6Al4V and Ti6Al7Nb, J. Staphylococcus aureus, Acta Biomater. 5 (2009) 3573–3580.
Biomed. Mater. Res. A 100A (2012) 768–775. [60] C. Gasqueres, G. Schneider, R. Nusko, G. Maier, E. Dingeldein, A. Eliezer, Innovative
[29] C. Giordano, E. Saino, L. Rimondini, M.P. Piedeferri, L. Visai, A. Cigada, R. Chiesa, antibacterial coating by anodic spark deposition, Surf. Coat. Technol. 206 (2012)
Electrochemically induced anatase inhibits bacterial colonization on Titanium 3410–3414.
S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978 977

[61] B.S. Necula, I. Apachitei, L.E. Fratila-Apachitei, E.J. van Langelaan, J. Duszczyk, Tita- [89] H.L. Huang, Y.Y. Chang, J.C. Weng, Y.C. Chen, C.H. Lai, T.M. Shieh, Anti-bacterial per-
nium bone implants with superimposed micro/nano-scale porosity and antibacte- formance of zirconia coatings on titanium implants, Thin Solid Films 528 (2013)
rial capability, Appl. Surf. Sci. 273 (2013) 310–314. 151–156.
[62] M.J. Santillan, N.E. Quaranta, A.R. Boccaccini, Titania and titania-silver nano com- [90] B. Li, X. Liu, F. Meng, J. Chang, C. Ding, Preparation and antibacterial properties of
posite coatings grown by electrophoretic deposition from aqueosu suspension, plasma sprayed nano-titania/silver coatings, Mater. Chem. Phys. 118 (2009)
Surf. Coat. Technol. 205 (2010) 2562–2571. 99–104.
[63] V. Kotharu, R. Nagumothu, C.B. Arumugam, M. Veerappan, S. Sankaran, M.A. [91] Y. Chen, X. Zheng, Y. Xie, H. Ji, C. Ding, Antibacterial properties of vacuum plasma
Davoodbasha, T. Nooruddin, Fabrication of corrosion resistant, bioactive and anti- sprayed titanium coatings after chemical treatment, Surf. Coat. Technol. 204
bacterial silver substituted hydroxyapatite/titania composite coating on Cp Ti, (2009) 685–690.
Ceram. Int. 38 (2012) 731–740. [92] G.A. Fielding, M. Roy, A. Bandyopaldhyay, S. Bose, Antibacterial and biological char-
[64] R. van Grieken, J. Marugan, C. Sordo, P. Martinez, C. Pablos, Photocatalytic inactiva- acteristics of silver containing and strontium doped plasma spray hydroxyapatite
tion of bacteria in water using suspended and immobilized silver–TiO2, Appl. Catal. coating, Acta Biomater. 8 (2012) 3144–3152.
B Environ. 93 (2009) 112–118. [93] M.K. Kang, S.K. Moon, J.S. Kwon, K.M. Kim, K.N. Kim, Antibacterial effect of sand
[65] L. Zhao, H. Wang, K. Huo, L. Cui, W. Zhang, H. Ni, Y. Zhang, Z. Wu, PK Chu antibac- blasted, large-grit, acid etiched treated Ti–Ag alloys, Mater. Res. Bull. 47 (2012)
terial nano-structured titania coating incorporated with silver nanoparticles, Bio- 2952–2955.
materials 32 (2011) 5706–5716. [94] Y.F. Zheng, B.B. Zhang, B.L. Wang, Y.B. Wang, L. Li, Q.B. Yang, L.S. Cui, Introduction of
[66] I. Piwonski, K. Kadziola, A. Kisielewska, K. Soliwoda, M. Wolszczak, K. Lisowska, N. antibacterial function into biomedical TiNi shape memory alloy by the addition of
Wronska, A. Felczak, The effect of the deposition parameters on size, distribution element Ag, Acta Biomater. 7 (2011) 2758–2767.
and antimicrobial properties of photoinduced silver nanoparticles on titania coat- [95] S. Ferraris, A. Venturello, M. Miola, A. Cochis, L. Rimondini, S. Spriano, Antibacterial
ings, Appl. Surf. Sci. 257 (2011) 7076–7082. and bioactive nanostructured titanium surfaces for bone integration, Appl. Surf. Sci.
[67] C. Zhao, B. Feng, Y. Li, J. Tan, X. Lu, J. Weng, Preparation and antibacterial activity of 311 (2014) 279–291.
titanium nanotubes loaded with Ag nanoparticles in the dark and under UV light, [96] T. Shirai, H. Tsuchiya, T. Shimizu, K. Ohtani, Y. Zen, K. Tomita, Prevention of pin
Appl. Surf. Sci. 280 (2013) 8–14. tract infection with titanium-copper alloys, J. Biomed. Mater. Res. B Appl. Biomater.
[68] C.Y. Flores, C. Diaz, A. Rubert, G.A. Benitez, M.S. Moreno, M.A. Fernandez lorenzo de 91B (2009) 373–380.
Mele, R.C. Salvarezza, P.L. Schilardi, C. Vericat, Spontaneous adsorption of silver [97] Y.Z. Wan, G.Y. Xiong, H. Liang, S. Raman, F. He, Y. Huang, Modification of medical
nanoparticles on Ti/TiO2 surfaces. Antibacterial effect on Pseudomonas aeruginosa, metals by ion implantation of copper, Appl. Surf. Sci. 253 (2007) 9426–9429.
J. Colloid Interface Sci. 350 (2010) 402–408. [98] E. Zhang, F. Li, H. Wang, J. Liu, C. Wang, M. Li, K. Yang, A new antibacterial titani-
[69] X. Zhang, Z. Li, X. Yuan, Z. Cui, H. Bao, X. Li, Y. Liu, X. Yang, Cytotoxicity and antibac- um–copper sintered alloy: preparation and antibacterial property, Mater. Sci.
terial property of titanium alloy coated with silver nanoparticle-containing Eng. C 33 (2013) 4280–4287.
polyelectrlyte multilayer, Mater. Sci. Eng. C 33 (2013) 2816–2820. [99] V. Stranak, H. Wulff, H. Rebl, C. Zietz, K. Arndt, R. Bogdanowicz, B. Nebe, R. Bader, A.
[70] Y. Inoue, M. Uota, T. Torikai, T. Watari, I. Noda, T. Hotokebuchi, M. Yada, Antibacte- Podbielski, Z. Hubicka, R. Hippler, Deposition of thin titanium–copper films with
rial properties of nanostructured silver titanate thin films formed on a titanium antimicrobial effect by advanced magnetron sputtering methods, Mater. Sci. Eng.
plate, J. Biomed. Mater. Res. 92A (2010) 1171–1180. C 31 (2011) 1512–1519.
[71] S.-B. Lee, U. Otgonbayar, Y.H. Lee, K.M. Kim, K.N. Kim, Silver ion exchanged sodium [100] X.B. Tian, Z.M. Wang, S.Q. Yang, Z.J. Luo, R.K.Y. Fu, P.K. Chu, Antibacterial copper-
titanate and resulting effect on antibacterial efficacy, Surf. Coat. Technol. 205 containing titanium nitride films produced by dual magnetron sputtering, Surf.
(2010) S172–S176. Coat. Technol. 201 (2007) 8606–8609.
[72] M. Miola, S. Ferraris, S. Di Nunzio, P. Robotti, G. Bianchi, G. Fucale, G. Maina, M. [101] X. Zhang, Y. Ma, N. Lin, X. Huang, R. Hang, A. Fan, B. Tang, Microstructure, antibac-
Cannas, S. Gatti, A. Masse’, C. Vitale-Brovarone, E. Verne’, Surface silver-doping of terial properties and wear resistance of plasma Cu–Ni surface modified titanium,
biocompatible glasses to induce antibacterial properties. Part II: plasma sprayed Surf. Coat. Technol. 232 (2013) 515–520.
glass-coatings, J. Mater. Sci. Mater. Med. 20 (2009) 741–749. [102] A. Yousef, N.A.M. Barakat, T. Amma, S.S. Al-Deyab, M.S. Hassan, A. Abdel-hay, H.Y.
[73] E. Verné, S. Ferraris, M. Miola, G. Fucale, G. Maina, P. Robotti, G. Bianchi, G. Kim, Inactivation of klebisiella peumoniae by CuO/TiO2 nanofibers: a multifunc-
Martinasso, R.A. Canuto, C. Vitale-Brovarone, Synthesis and characterization of a tional nano material via one-step electrospinning, Ceram. Int. 38 (2012)
bioactive and antibacterial glass-ceramic (II): plasma spray coatings on metallic 4525–4532.
substrates, Adv. Appl. Ceram. 107 (2008) 245–253. [103] S. Chen, Y. Guo, S. Chen, Z. Ge, H. Yang, J. Tang, Fabrication of Cu/TiO2 nanocompos-
[74] J. Garcia-Serrano, E. Gomez-Hernandez, M. Ocampo-Fernandez, U. Pal, Effect of Ag ite: toward an enhanced antibacterial performance in the absence of light, Mater.
doping on the crystallization and phase transition of TiO2 nanoparticles, Curr. Appl. Lett. 83 (2012) 154–157.
Phys. 9 (2009) 1097–1105. [104] J. Mungkalasiri, L. Bedel, F. Emieux, J. Dorè, F.N.R. Renaud, F. Maury, DLI-CVD of
[75] B. Braconnier, C.A. Paez, S. Lambert, C. Aliè, C. Henrist, D. Poelman, J.P. Pirard, R. TiO2–Cu antibacterial thin films: growth and characterization, Surf. Coat. Technol.
Cloots, B. Heinrichs, Ag- and SiO2-doped TiO2 with enhanced thermal stability, Mi- 204 (2009) 887–892.
croporous Mesoporous Mater. 122 (2009) 247–254. [105] H.M. Yates, L.A. Brook, I.B. Ditta, P. Evans, H.A. Foster, D.W. Sheel, A. Steele, Photo-
[76] L. Chai, S. Wei, B. Peng, Z. Li, Effect of thermal treating temperature on characteris- induced self-cleaning and biocidal behaviour of titania and copper oxide multi-
tics of silver-doped titania, Trans. Nonferrous Metals Soc. China 18 (2008) layers, J. Photochem. Photobiol. A 197 (2008) 197–205.
980–985. [106] H. Hu, W. Wang, Y. Qiao, X. Jiang, X. Liu, C. Ding, Antibacterial activity and increases
[77] B. Yu, K.M. Leung, Q. Guo, W.M. Lau, J. Yang, Synthesis of Ag–TiO2 composite nano bone marrow stem cell functions of Zn-incorporated TiO2 coatings on titanium,
thin film for antimicrobial application, Nanotechnology 22 (2011) 115603 (9pp). Acta Biomater. 8 (2012) 904–915.
[78] L. Mai, D. Wang, S. Zhang, Y. Xie, C. Huang, Z. Zhang, Synthesis and bactericidal abil- [107] K. Huo, X. Zhang, H. Wang, L. Zhao, X. Liu, P.K. Chu, Osteogenic activity and antibac-
ity of Ag/TiO2 composite films deposited on titanium plate, Appl. Surf. Sci. 257 terial effects on titanium surfaces modified with Zn-incorporated nanotube arrays,
(2010) 974–978. Biomaterials 34 (2013) 3467–3478.
[79] S.A. Tomas, A. Luna-Resendis, L.C. Cortes-Cuautli, D. Jacinto, Optical and morpho- [108] J. Xu, G. Ding, J. Li, S. Yang, B. Fang, H. Sun, Y. Zhou, Zinc-ion implanted and depos-
logical characterization of photocatalytic TiO2 thin films doped with silver, Thin ited titanium surfaces reduces adhesion of Streptococcus mutans, Appl. Surf. Sci.
Solid Films 518 (2009) 1337–1340. 256 (2010) 7540–7544.
[80] J. Qu, X. Lu, D. Li, Y. Ding, Y. Leng, J. Weng, S. Qu, B. Feng, F. Watari, Silver/hydroxy- [109] M.T. Tsai, Y.Y. Chang, H.L. Huang, J.T. Hsu, Y.C. Chen, A.Y.J. Wu, Characterization and
apatite composite coatings on porous titanium surfaces by sol–gel method, J. antibacterial performance of bioactive Ti–Zn–O coatings deposited on titanium im-
Biomed. Mater. Res. B Appl. Biomater. 97B (2011) 40–48. plants, Thin Solid Films 528 (2013) 143–150.
[81] K.D. Secinti, H. Ozlap, A. Attar, M.F. Sargon, Nanoparticle silver ion coatings inhibit [110] K. Li, J. Yu, Y. Xie, L. Huang, X. Ye, X. Zheng, Chemical stability and antimicrobial ac-
biofilm formation on titanium implants, J. Clin. Neurosci. 18 (2011) 391–395. tivity of plasma sprayed bioactive Ca2ZnSi2O7 coating, J. Mater. Sci. Mater. Med. 22
[82] F. Meng, Z. Sun, A mechanism for enhanced hydrophilicity of silver nanoparticles (2011) 2781–2789.
modified TiO2 thin films deposited by RF magnetron sputtering, Appl. Surf. Sci. [111] M. Yoshinari, Y. Oda, T. Kato, K. Okuda, Influence of surface modification to titani-
255 (2009) 6715–6720. um on antibacterial activity on vitro, Biomaterials 22 (2001) 2043–2048.
[83] F. Meng, Z. Sun, Enhanced photocatalytic activity of silver nanoparticles modified [112] M.A. Arenas, C. Perez-Jorge, A. Conde, E. Matykina, J.M. Hernandez-Lopez, R. Perez-
TiO2 thin films prepared by RF magnetro sputtering, Mater. Chem. Phys. 118 Tanoira, J.J. de Damborenea, E. Gomez-Barrena, J. Esteba, Doped TiO2 anodic layers
(2009) 349–353. of enhanced antibacterial properties, Colloids Surf. B 105 (2013) 106–112.
[84] D.H. Song, S.H. Uhm, S.B. Lee, J.G. Han, K.N. Kim, Antimicrobial silver-containing ti- [113] T. Shirai, T. Shimizu, K. Ohtani, Y. Zen, M. Takaya, H. Tsuchiya, Antibacterial iodine-
tanium oxide nano composite coatings by reactive magnetron sputtering, Thin supported titanium implants, Acta Biomater. 7 (2011) 1928–1933.
Solid Films 519 (2011) 7079–7085. [114] H. Tsuchiya, T. Shiarai, H. Nishida, H. Murakami, T. Kabata, N. Yamamoto, K.
[85] D.H. Song, S.H. Uhm, S.E. Kim, J.S. Kwon, J.G. Han, K.N. Kim, Synthesis of titanium Watanabe, J. Nakase, Innovative antimicrobial coating of titanium implants with
oxide thin films containing antibacterial silver nanoparticles by a reactive magne- iodine, J. Orthop. Sci. 17 (2012) 595–604.
tron co-sputtering system for application in biomedical implants, Mater. Res. Bull. [115] W. Zhang, Y. Chen, S. Yu, S. Chen, Y. Yin, Preparation and antibacterial behavior of
47 (2012) 2994–2998. Fe3+-doped nanostructured TiO2 thin films, Thin Solid Films 516 (2008)
[86] H.L. Huang, Y.Y. Chang, M.C. Lai, C.R. Lin, C.H. Lai, T.M. Shieh, Antibacterial TaN–Ag 4690–4694.
coatings on titanium dental implants, Surf. Coat. Technol. 205 (2010) 1636–1641. [116] H. Wang, B. Tang, X. Li, Y. Ma, Antibacterial properties and corrosion resistance of
[87] O. Akhavan, E. Ghaderi, Capping antibacterial Ag nanorods aligned on Ti interlayer nitrogen-doped TiO2 coatings on stainless steel, J. Mater. Sci. Technol. 27 (2011)
by mesoporous TiO2 layer, Surf. Coat. Technol. 203 (2009) 3123–3128. 309–316.
[88] N.A. Trujillo, R.A. Oldinski, H. Ma, J.D. Bryers, J.D. Williams, K.C. Popat, Antibacterial [117] T. Amna, M.S. Hassan, M. Pandurangan, M.S. Khil, H.K. Lee, I.H. Hwang, Character-
effects of silver-doped hydroxyapatite thin films sputter deposited on titanium, ization and potent bactericidal effect of cobalt doped titanium dioxide nanofibers,
Mater. Sci. Eng. C 32 (2012) 2135–2144. Ceram. Int. 39 (2013) 3189–3193.
978 S. Ferraris, S. Spriano / Materials Science and Engineering C 61 (2016) 965–978

[118] A. Bistolfi, G. Massazza, E. Vernè, A. Massè, D. Deledda, S. Ferraris, M. Miola, F. [126] T.Y. Lim, W. Wang, Z. Shi, C.K. Pho, K.G. Neoh, Human bone marrow-derived mes-
Galetto, M. Crova, Antibiotic loaded cement in orthopedic surgery, A Review, enchymal stem cells and osteoblast differentiation on titanium with surface-
ISRN ORTHOPEDICS, 2011 (8 pages). grafted chitosan and immobilized bone morphogenetic protein-2, J. Mater. Sci.
[119] V. Antoci, C.S. Adams, J. Parvizi, H.M. Davidson, R.J. Composto, T.A. Freeman, E. Mater. Med. 20 (2009) 1–10.
Wickstrom, P. Ducheyne, D. Jungkind, I.M. Shapiro, N.J. Hickok, The inhibition of [127] X. Hu, K.G. Neoh, Z. Shi, E.T. Kang, C. Poh, W. Wang, An in vitro assessment of tita-
Staphylococcus epidermidis biofilm formation by vancomycin-modified titanium nium functionalized with polysaccharides conjugated with vascular endothelial
alloy and implications for the treatment of periprosthetic infection, Biomaterials growth factor for enhanced osseointegration and inhibition of bacterial adhesion,
29 (2008) 4684–4690. Biomaterials 31 (2010) 8854–8863.
[120] B. Jose, V. Antoci, A.R. Zeiger, E. Wickstrom, N.J. Hickok, Vancomycin covalently [128] P.H. Chua, K.G. Neoh, E.T. Kang, W. Wang, Surface functionalization of titanium
bonded to titanium beads kills Staphylococcus aureus, Chem. Biol. 12 (2005) with hyaluronic acid/chitosan polyelectrolyte multilayers and RGD for promoting
1041–1048. osteoblast functions and inhibiting bacterial adhesion, Biomaterials 29 (2008)
[121] S. Radin, P. Ducheyne, Controlled release of vancomycin from thin sol–gel films on 1412–1421.
titanium alloy fracture plate material, Biomaterials 28 (2007) 1721–1729. [129] D. Zheng, K.G. Neoh, Z. Shi, E.T. Kang, Assessment of stability of surface anchors for
[122] G. Pitarresi, F.S. Palumbo, F. Calascibetta, C. Fiorica, M. Di Stefano, G. Giammona, antibacterial coatings and immobilized growth factors on titanium, J. Colloid Inter-
Medicated hydrogels of hyaluronic acid derivatives for use in orthopedic field, face Sci. 406 (2013) 238–246.
Int. J. Pharm. 449 (2013) 84–94. [130] X. Chen, K. Cai, J. Fang, Y. Hou, J. Li, Z. Luo, Y. Hu, L. Tang, Fabrication of selenium-
[123] L. Pichavant, G. Amador, C. Jacqueline, B. Brouillaud, V. Heroguez, M.C. Durrieu, pH- deposited and chitosan coated titania nanotubes with anticancer and antibacterial
controlled delivery of gentamicine sulphate from orthopaedic devices preventing properties, Colloids Surf. B 103 (2013) 149–157.
nosocomial infections, J. Control. Release 162 (2012) 373–381. [131] F. Costa, I.F. Carvalho, R.C. Montelaro, P. Gomes, M.C.L. Martins, Covalent immobi-
[124] H. Vester, B. Wildemann, G. Schmidmaier, U. Stockle, M. Lucke, Gentamycin deliv- lization of antimicrobial peptides (AMPs) onto biomaterial surfaces, Acta Biomater.
ered from a PDLLA coating of metallic implants. In vivo and in vitro characteriza- 7 (2011) 1431–1440.
tion for local prophylaxis of implant-related osteomyelitis, Inj. Int. J. Care Inj. 41 [132] D. Xu, W. Yang, Y. Hu, Z. Luo, J. Li, Y. Hou, Y. Liu, K. Cai, Surface functionalization of
(2010) 1053–1059. titanium substrates with cecropin B to improve their cytocompatibility and reduce
[125] D. Neut, R.J.B. Dijkstra, J.I. Thompson, H.C. van der Mei, H.J. Busscher, A gentamicin- inflammation responses, Colloids Surf. B 110 (2013) 225–235.
releasing coating for cementless hip prostheses — longitudinal evaluation of effica-
cy using in vitro bio-optical imaging and its wide-spectrum antibacterial activity, J.
Biomed. Mater. Res. A 100 (2012) 3220–3226.

You might also like