Next Article in Journal
Population Pharmacokinetic and Pharmacodynamic Analysis of Dalbavancin for Long-Term Treatment of Subacute and/or Chronic Infectious Diseases: The Major Role of Therapeutic Drug Monitoring
Next Article in Special Issue
Multidrug-Resistant Bacterial Pathogens and Public Health: The Antimicrobial Effect of Cyanobacterial-Biosynthesized Silver Nanoparticles
Previous Article in Journal
Modulation of Multiple Gene Clusters’ Expression by the PAS-LuxR Transcriptional Regulator PteF
Previous Article in Special Issue
Antimicrobial Activity of Biogenic Silver Nanoparticles from Syzygium aromaticum against the Five Most Common Microorganisms in the Oral Cavity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Is Silver the New Gold? A Systematic Review of the Preclinical Evidence of Its Use in Bone Substitutes as Antiseptic

Orthopaedic and Traumatology Unit, IRRCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(8), 995; https://doi.org/10.3390/antibiotics11080995
Submission received: 3 July 2022 / Revised: 16 July 2022 / Accepted: 22 July 2022 / Published: 24 July 2022
(This article belongs to the Special Issue Silver and Gold Compounds as Antibiotics)

Abstract

:
Antibiotic-laden bone substitutes represent a viable option in the treatment of bone and joint infections with bone defects. In particular, the addition of silver ions or silver nanoparticles to bone substitutes to achieve local antiseptic activity could represent a further contribution, also helping to prevent bacterial resistance to antibiotics. An in-depth search of the main scientific databases was performed regarding the use of silver compounds for bone substitution. The available evidence is still limited to the preclinical level: 22 laboratory studies, 2 animal models, and 3 studies, with both in vitro and in vivo analysis, were found on the topic. Numerous biomaterials have been evaluated. In vitro studies confirmed that silver in bone substitutes retains the antibacterial activity already demonstrated in coatings materials. Cytotoxicity was generally found to be low and only related to silver concentrations higher than those sufficient to achieve antibacterial activity. Instead, there are only a few in vivo studies, which appear to confirm antibacterial efficacy, although there is insufficient evidence on the pharmacokinetics and safety profile of the compounds investigated. In conclusion, research on bone substitutes doped with silver is in its early stages, but the preliminary findings seem promising.

1. Introduction

Bone and joint infections (BJIs) represent an extremely heterogeneous group of diseases, including implant-associated infections (both in the fields of orthopaedics and odontology), septic arthritis, and osteomyelitis [1]. The increase in BJIs’ incidence shown in recent years, mainly due to the increase in joint replacements and the use of orthopaedic hardware, currently represents a growing social and economic issue for health systems [1,2]. Indeed, implant-related infection rate is reported to be 5% for primary cases, 6% for revision cases and increases to 43% for previously infected cases [3]. Osteomyelitis, defined as a bone inflammation caused by infection, may be the common endpoint of BJIs. Bacterial infections complicate the bone-healing process following fractures or surgical treatment, often resulting in significant bone loss [3]. Once occurred, bone infections are very challenging to treat, due to the difficulty of achieving a suitable antibiotic concentration in the affected area that may permit bacteria eradication [4]. Hence, the treatment of BJIs generally requires wide debridement with removal of all infected bone and soft tissues, irrigation, and, subsequently, dead-space filling [5,6]. Bone defects wider than 2 cm or circumferential losses involving more than 50% of bone are defined as critical-size bone defects (CSDs). CSDs usually progress to healing failure, even after optimal fixation [7]. In order to restore the continuity of the bone loss resulting from the surgical treatment, autologous, allogenic and artificial bone can be implanted [6,8]. Autograft substitutes are still considered the gold standard for bone repair and regeneration due to their osteogenic nature combined with no immunological side effects of the graft [7,9]; however, they are associated with donor site morbidity (hematomas, infection, and neurovascular injury) and longer operative time [10]. On the other hand, when using allografts, the principal concerns are related to mechanical resistance, limited osteoconduction and risk of infections [8]. Therefore, greater attention has increasingly been given to bone graft substitutes. They have been defined as “a synthetic or biologically organic combinations which can be inserted for the treatment of a bone defect instead of autogenous or allogenous bone” [10]. Theoretically, the bone substitutes mimic bone graft, combining advantages of natural and synthetic biomaterials [8,9] and supporting local bone healing [7]. The ideal bone substitute should be biocompatible, osteoconductive, osteoinductive, resorbable, thermally nonconductive, sterilizable, and available at a reasonable cost [10]. Bone substitutes can be broadly categorized into ceramics (nonresorbable and biodegradable), hydroxyapatite, β-tri-calcium phosphate, calcium sulfate, calcium carbonate, silicate (“Bioglass”), magnesium composites and calcium phosphate cements [7]. Current advances have been made with the development of tissue-engineered products, incorporating growth factors and stem cells [10].
Despite the development in biomaterials’ properties, risk of infection remains a major issue after implantation. Thus, antibacterial properties should be considered during the development and choice of a bone substitute, as well as biocompatibility and physic-chemical features [11,12]. This is in order to prevent infections of the bone substitutes, or to optimise their use in case of BJIs. In fact, the result of bacterial adhesion to implants or grafts usually progresses with complete removal [12]. Moreover, to prevent infection recurrence, systemic or local antibiotics should be administrated after surgery; nevertheless, inappropriate and excessive use of antibiotics, in addition to systemic side effects for the patient, increases the risk of the emergence of multidrug-resistant bacteria [6]. The addition of antimicrobial nanomaterials, such as silver, zinc, copper, carbon nanotube, graphene oxide, molybdenum disulfide and titanium oxide, into the biomaterials has significantly shown to inhibit microbial infection, determining also a lower tendency to develop bacterial resistance [9,13,14].
In particular, silver’s efficacy and safety has been reported in several in vitro and animal studies [15]. Silver ions (Ag) and silver nanoparticles (AgNPs) have garnered prominent consideration in recent years due to their broad spectrum of antibacterial properties, low bacterial resistance, and relatively low cytotoxicity [13]. For these reasons, the use of silver gained interest in the clinical practice with applications such as wound healing and cardiac and orthopaedic implant coating. Particularly in the orthopaedic field, the use of silver has proved to be effective in the treatment of megaprosthesis infections [15].
Although only preclinical studies about the use of silver combined with bone substitutes have been published, this issue may have important clinical implications for the prevention and treatment of BJIs. The aim of this review is to provide an overview of the evidence currently available in the literature.

2. Results

A total of 271 studies were found through the electronic search and 8 studies were added after cross-referenced research on the bibliography of the examined full-text articles. After a preliminary analysis, a total of 27 studies were included in this systematic review [3,4,6,8,9,11,12,13,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34]. To date, there are no clinical studies on the use of bone substitutes containing adjuvant silver. Twenty-two laboratory studies [4,6,8,9,11,12,13,16,17,18,19,20,22,23,24,25,26,27,28,29,30,31,32,34], two animal models [21,33] and three studies in which both in vitro and in vivo analysis were performed [3,6,31] were found on the topic.
A wide range of biomaterials were evaluated as possible carriers of silver in bone. The tests used to appraise the in vitro or in vivo antibacterial activity of the compounds in the various studies included: cultures from bone samples, agar diffusion, halo test, agar dilution, broth microdilution, spread plate method, bacterial count through scanning electron microscope, confocal laser scanning microscopy, epi-fluorescence microscopy, histopathological examinations, RT-PCR bacterial DNA measurement, and radiological examination. All in vitro studies reported a partial or total inhibition of the bacterial growth. All in vivo studies that directly investigated the antibacterial effect of silver compounds confirmed their efficacy in the treatment of osteomyelitis. The data on cell and tissue toxicity are inconsistent; however, in all the studies, the antibacterial activity of the compounds tested was reported at nontoxic concentrations. Extended data from the included studies are reported in Table 1.

3. Discussion

Silver antimicrobial activity relies on several mechanisms. Principally, it stops cells’ respiratory chain, affecting the cells’ energy generation, due to its affinity to the sulfhydryl and thiol groups [35]. Additionally, silver leads to a release of potassium [36], binds DNA and RNA, disrupting the cells’ translation and transcription processes [37], and produces intracellular reactive oxygen species [38]. Consequently, silver has the ability to eliminate a broad spectrum of pathogens that can be found at implant sites [39]. Surfaces or materials containing silver particles act by directly releasing ions into the water solution in which they lie. More recently, silver technology has focused on the use of nanoparticles. AgNPs seems to be more reactive, with a stronger antibiofilm potential than their bulk metal counterparts, partially due to the increased active surface area [40,41]. AgNPs are usually 1 nm to 20 nm in size. Because of their small dimensions, the surface area is taken advantage of, passing more into cell membranes, thereby contributing to augmented antimicrobial action [42]. Furthermore, antibacterial mechanisms of AgNPs have been hypothesized that do not depend on the release of ions but are related to the interaction between silver and other substrates. For example, interaction with some titanium alloys can lead to the production of an electron cloud around the surface of the compounds [35]. This cathodic reaction, which produces a proton depletion region, would appear to reduce the transmembrane proton electrochemical gradient and lead to bacterial death by interfering with ATP synthesis [35]. In addition, to date, there is limited evidence that AgNPs possess osteoconductive capabilities, promoting the proliferation of mesenchymal stem cells and their osteogenic differentiation in vitro, as well as enhancing bone fracture healing in animal models [43].
The clinical use of silver for antibacterial purposes in implantable devices has been mainly investigated as a coating material [15]. Therefore, there is already evidence regarding the efficacy and safety profile of this application. Studies largely agree on time and concentration influencing the bactericidal effect of silver in bone substitutes, with higher Ag concentration and longer exposure time associated with better antibacterial responses [3,22]. Accordingly, a cytotoxic effect of silver at excessively high concentrations is to be expected, by means of the same antibacterial mechanisms, to which, however, eukaryotic cells are less sensitive at low concentrations because of their extremely better antioxidant and DNA repair activity [44]. It has been reported that silver has toxic effects for humans at a high blood concentration of 300–1200 ppb [45]. Instead, silver concentrations below 200 ppb can be considered as normal [37]. Instead, regarding AgNPs’ safety profile, various studies suggested that there is a large gap, at least by an order of magnitude, between the toxic and antibacterial doses of AgNPs [46,47]. All available clinical data on the pharmacokinetics of silver are related to its use in coating materials, especially regarding serum levels, which have never been found to exceed the threshold of toxicity. Nevertheless, several side effects have been related to silver in clinical studies, including diffuse argyria, kidney and liver damage, leukopenia, and peripheral neuropathies [36,48,49]. No embryotoxic side effects in humans are described [50].
In contrast to silver used in coatings, this review highlighted that there are currently no clinical data on the use of silver as an adjuvant in biomaterials for bone substitution. However, there are some in vivo studies using animal models and a substantial number of laboratory studies investigating the antibacterial efficacy of silver on a plethora of different biomaterials.
All in vitro studies included in this review supported that even when used as a biomaterial constituent, silver appears to preserve its antibacterial activity [3,4,6,8,9,11,12,13,16,17,18,19,20,22,23,24,25,26,27,28,29,30,31,32,34]. This evidence emerged regardless of the type of analysis used by individual studies to highlight the effect of silver. In particular, the impact of silver was found to be cross-cutting for both Gram+ and Gram- bacteria. Some studies hypothesised that due to the different composition of the membranes, silver-doped materials could have more antibacterial effect against Gram+ [14,25,26]. However, other studies did not report differential antibacterial effect, showing a complete response in both cases [3,27,29]. Unfortunately, no study included in this review conducted quantitative evaluations specifically aimed at investigating this aspect using different bacterial species clustered according to membrane characteristics. Three out of five studies with animal models directly investigated the in vivo antibacterial effect of silver compounds. All confirmed the efficacy of using biomaterials containing adjuvant silver in the treatment of S. aureus osteomyelitis [21,31,33]. In detail: (1) Zhang et al. investigated silver application in two hydroxyapatite compounds with different silver concentrations [33]; (2) calcium phosphate beads doped with silver ions were used in the study by Kose et al. [21]; (3) Weng et al. instead considered the use of silver nanoparticle (AgNP)-loaded nano-hydroxyapatite-reduced graphene oxide scaffolds [31]. In all in vivo studies, it was also confirmed that the presence of silver did not affect the osteoconductive and osteoinductive activity of the bone substitutes employed [3,6,21,31,33].
The ever-increasing resistance of human pathogens to antibiotics makes silver a convenient alternative to be harnessed as an antibacterial weapon. Indeed, bacterial resistance hardly arises in the presence of silver ions in the environment [51,52], while no relevant data describing bacterial resistance to AgNPs have been reported yet [53]. Therefore, it would be interesting to examine the antibacterial effect of silver compared with antibiotics in bone substitutes. However, only few conflicting data are available: Kose et al. showed a better result for silver compared to vancomycin in murine models [21]; on the other side, two studies showed a better antibacterial effect of bone substitutes doped with doxycycline or gentamicin alone [22,27].
Little can be stated about the safety profile of silver use in biomaterials. Indeed, it might be expected that the pharmacokinetics of silver used as a constituent material in bone substitutes would differ radically from its use in coatings. This could lead to the ineffective release of silver in vivo, exceeding toxicity thresholds, or even irregular or excessively phasic kinetics that alternate between these two possibilities. Several in vitro studies have evaluated the kinetics of silver release from biomaterials in water or in different solutions of simulated body fluid. However, on the one hand, the extreme heterogeneity of the materials tested, and on the other hand, the radical differences in environment compared to a real setting, fully restrict the deducible conclusions. In fact, only in vivo assessments may allow approximating the pharmacokinetics. Similar remarks can be made about cytotoxicity from in vitro studies, with results varying widely due to the broad spectrum of materials analysed. However, in all in vitro studies, the antibacterial activity of the compounds analysed was achieved at noncytotoxic concentrations. From this point of view, with regard to the in vivo studies, Zhang et al. found that the silver concentration of the bone tissue was found to be over 2 ppm in the n-HA/PU10 group with local toxic risk increasing when the silver concentration exceeds 1 ppm in tissue, as recommended by the safety guidelines [33]. Moreover, the same compound has shown the possibility of inducing liver toxicity [33]. In contrast, the studies by Kose et al. and Yuan et al. found no local and systemic toxicity in the animal models used, both investigating calcium phosphate derivates doped with silver ions or nanoparticles, respectively [3,21]. Shimabukuro et al. reported that the inflammatory effect of silver phosphate (Ag3PO4) in a bone substitute composed of carbonate apatite (CO3Ap) was concentration-dependent [6]. Similarly, the study by Schneider et al., not included in this review as it did not directly investigate the antibacterial effect, showed no inflammatory reactions to cotton-wool-like silver-doped calcium phosphate nanocomposites in sheep [54]. Another study, not included in this review for the same reason, performed by Wnukiewicz et al., examined the soft-tissue reaction to corundum ceramic with colloidal silver in rabbits, finding no difference with the control group [55].

4. Materials and Methods

An in-depth search of the scientific research was performed according to 2020 PRISMA (preferred reporting items for systematic reviews and meta-analyses) guidelines [56]. The search algorithm according to these guidelines is shown in Figure 1.
A search regarding the existing evidence on the use of silver compounds and silver nanoparticles combined with biomaterials for bone substitution with no restriction on date of publication, up to the end of June 2022, was performed on the PubMed (https://pubmed.ncbi.nlm.nih.gov/ (accessed on 30 June 2022)), Scopus (https://www.scopus.com (accessed on 30 June 2022)), and Web of Science (www.webofscience.com (accessed on 30 June 2022)) databases. Various combinations of the following keywords were used: “silver compound”, “silver nanoparticles”, “bone substitutes”, “bone biomaterials”. The inclusion criteria were as follows: original research reporting preclinical results on in vitro testing and in vivo animal models of the antibacterial activity, pharmacokinetics and pharmacodynamics of silver combined with biomaterials used for bone substitution. Only studies in English were retained. Articles that were considered relevant during the electronic search were retrieved in full-text, and a cross-referencing hand-search of their bibliography was performed, in order to find further related articles. Reviews and meta-analysis were also analysed, in order to broaden the search for studies that might have been missed through the electronic search.
A formal assessment of the quality of the articles was not conducted, as there is no clear evidence of validated tools for the evaluation of preclinical laboratory studies. Only descriptive statistics were used for this study as the type of data provided.
The following data were independently extracted by all the investigators and summarized in Table 1: study type, material tested, type of antimicrobial activity evaluations, microorganisms tested, and findings about toxicity.

5. Conclusions

The introduction of bone substitutes doped with ionic silver or silver nanoparticles into clinical practice would provide a valuable further contribution to the management of challenging diseases such as osteomyelitis and peri-prosthetic or implant-related infections, as well as to the prevention of bacterial resistance to antibiotics. Numerous materials have already been evaluated for this purpose, but the available evidence is still limited to the preclinical level. In vitro studies have confirmed that when silver is added to bone substitutes, it retains the antibacterial activity already demonstrated in coatings materials. The antibacterial effect against Gram+ might be higher than against Gram-. However, conclusive data are lacking as well as it is unclear whether silver could provide greater efficacy than antibiotic loading. The cytotoxicity of silver compounds has generally been shown to be low and only related to concentrations of silver significantly higher than those sufficient to achieve antibacterial activity. On the other hand, there are only a few in vivo studies which appear to confirm antibacterial efficacy, although there is insufficient evidence on the pharmacokinetics and safety profile of the biomaterials investigated. In conclusion, research on bone substitutes doped with silver is in its early stages but the preliminary findings seem promising.

Author Contributions

Conceptualization, M.F. (Michele Fiore), A.B. and M.D.P.; methodology, M.F. (Matteo Filippini); software, A.S. and L.M.; investigation, A.B. and C.G.; data curation, M.F. (Matteo Filippini) and C.R.; writing—original draft preparation, A.B., C.G. and C.R.; writing—review and editing, M.F. (Michele Fiore) and A.S.; supervision, M.D.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data reported in this study are available in the literature.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sambri, A.; Spinnato, P.; Tedeschi, S.; Zamparini, E.; Fiore, M.; Zucchini, R.; Giannini, C.; Caldari, E.; Crombe, A.; Viale, P.; et al. Bone and Joint Infections: The Role of Imaging in Tailoring Diagnosis to Improve Patients’ Care. J. Pers. Med. 2021, 11, 1317. [Google Scholar] [CrossRef] [PubMed]
  2. Mandracchia, V.J.; Sanders, S.M.; Jaeger, A.J.; Nickles, W.A. Management of osteomyelitis. Clin. Podiatr. Med. Surg. 2004, 21, 335–351. [Google Scholar] [CrossRef]
  3. Yuan, J.; Wang, B.; Han, C.; Huang, X.; Xiao, H.; Lu, X.; Lu, J.; Zhang, D.; Xue, F.; Xie, Y. Nanosized-Ag-doped porous beta-tricalcium phosphate for biological applications. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 114, 111037. [Google Scholar] [CrossRef] [PubMed]
  4. Paterson, T.E.; Shi, R.; Tian, J.; Harrison, C.J.; De Sousa Mendes, M.; Hatton, P.V.; Li, Z.; Ortega, I. Electrospun Scaffolds Containing Silver-Doped Hydroxyapatite with Antimicrobial Properties for Applications in Orthopedic and Dental Bone Surgery. J. Funct. Biomater. 2020, 11, 58. [Google Scholar] [CrossRef] [PubMed]
  5. Sambri, A.; Fiore, M.; Tedeschi, S.; De Paolis, M. The Need for Multidisciplinarity in Modern Medicine: An Insight into Orthopaedic Infections. Microorganisms 2022, 10, 756. [Google Scholar] [CrossRef]
  6. Shimabukuro, M.; Hayashi, K.; Kishida, R.; Tsuchiya, A.; Ishikawa, K. No-Observed-Effect Level of Silver Phosphate in Carbonate Apatite Artificial Bone on Initial Bone Regeneration. ACS Infect. Dis. 2022, 8, 159–169. [Google Scholar] [CrossRef]
  7. Busch, A.; Wegner, A.; Haversath, M.; Jager, M. Bone Substitutes in Orthopaedic Surgery: Current Status and Future Perspectives. Z. Orthop. Unfallchir. 2021, 159, 304–313. [Google Scholar] [CrossRef]
  8. Correia, T.R.; Figueira, D.R.; de Sá, K.D.; Miguel, S.P.; Fradique, R.G.; Mendonca, A.G.; Correia, I.J. 3D Printed scaffolds with bactericidal activity aimed for bone tissue regeneration. Int. J. Biol. Macromol. 2016, 93, 1432–1445. [Google Scholar] [CrossRef]
  9. Dalavi, P.A.; Prabhu, A.; Shastry, R.P.; Venkatesan, J. Microspheres containing biosynthesized silver nanoparticles with alginate-nano hydroxyapatite for biomedical applications. J. Biomater. Sci. Polym. Ed. 2020, 31, 2025–2043. [Google Scholar] [CrossRef]
  10. Campana, V.; Milano, G.; Pagano, E.; Barba, M.; Cicione, C.; Salonna, G.; Lattanzi, W.; Logroscino, G. Bone substitutes in orthopaedic surgery: From basic science to clinical practice. J. Mater. Sci. Mater. Med. 2014, 25, 2445–2461. [Google Scholar] [CrossRef]
  11. Bostancioglu, R.B.; Peksen, C.; Genc, H.; Gurbuz, M.; Karel, F.B.; Koparal, A.S.; Dogan, A.; Kose, N.; Koparal, A.T. Analyses of the modulatory effects of antibacterial silver doped calcium phosphate-based ceramic nano-powder on proliferation, survival, and angiogenic capacity of different mammalian cells in vitro. Biomed. Mater. 2015, 10, 045024. [Google Scholar] [CrossRef]
  12. Afzal, M.A.; Kalmodia, S.; Kesarwani, P.; Basu, B.; Balani, K. Bactericidal effect of silver-reinforced carbon nanotube and hydroxyapatite composites. J. Biomater. Appl. 2013, 27, 967–978. [Google Scholar] [CrossRef] [PubMed]
  13. Bee, S.L.; Bustami, Y.; Ul-Hamid, A.; Lim, K.; Abdul Hamid, Z.A. Synthesis of silver nanoparticle-decorated hydroxyapatite nanocomposite with combined bioactivity and antibacterial properties. J. Mater. Sci. Mater. Med. 2021, 32, 106. [Google Scholar] [CrossRef] [PubMed]
  14. Kolmas, J.; Groszyk, E.; Kwiatkowska-Rozycka, D. Substituted hydroxyapatites with antibacterial properties. BioMed Res. Int. 2014, 2014, 178123. [Google Scholar] [CrossRef] [PubMed]
  15. Fiore, M.; Sambri, A.; Zucchini, R.; Giannini, C.; Donati, D.M.; De Paolis, M. Silver-coated megaprosthesis in prevention and treatment of peri-prosthetic infections: A systematic review and meta-analysis about efficacy and toxicity in primary and revision surgery. Eur. J. Orthop. Surg. Traumatol. 2021, 31, 201–220. [Google Scholar] [CrossRef]
  16. Deng, L.; Deng, Y.; Xie, K. AgNPs-decorated 3D printed PEEK implant for infection control and bone repair. Colloids Surf. B Biointerfaces 2017, 160, 483–492. [Google Scholar] [CrossRef]
  17. Gong, J.; Yang, L.; He, Q.; Jiao, T. In vitro evaluation of the biological compatibility and antibacterial activity of a bone substitute material consisting of silver-doped hydroxyapatite and Bio-Oss((R)). J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 410–420. [Google Scholar] [CrossRef]
  18. Jacquart, S.; Siadous, R.; Henocq-Pigasse, C.; Bareille, R.; Roques, C.; Rey, C.; Combes, C. Composition and properties of silver-containing calcium carbonate-calcium phosphate bone cement. J. Mater. Sci. Mater. Med. 2013, 24, 2665–2675. [Google Scholar] [CrossRef] [Green Version]
  19. Jegatheeswaran, S.; Sundrarajan, M. PEGylation of novel hydroxyapatite/PEG/Ag nanocomposite particles to improve its antibacterial efficacy. Mater. Sci. Eng. C Mater. Biol. Appl. 2015, 51, 174–181. [Google Scholar] [CrossRef]
  20. Jiang, J.; Li, L.; Li, K.; Li, G.; You, F.; Zuo, Y.; Li, Y.; Li, J. Antibacterial nanohydroxyapatite/polyurethane composite scaffolds with silver phosphate particles for bone regeneration. J. Biomater. Sci. Polym. Ed. 2016, 27, 1584–1598. [Google Scholar] [CrossRef]
  21. Kose, N.; Asfuroglu, Z.M.; Kose, A.; Sahinturk, V.; Gurbuz, M.; Dogan, A. Silver ion-doped calcium phosphate-based bone-graft substitute eliminates chronic osteomyelitis: An experimental study in animals. J. Orthop. Res. 2021, 39, 1390–1401. [Google Scholar] [CrossRef] [PubMed]
  22. Sampath Kumar, T.S.; Madhumathi, K.; Rubaiya, Y.; Doble, M. Dual mode antibacterial activity of ion substituted calcium phosphate nanocarriers for bone infections. Front. Bioeng. Biotechnol. 2015, 3, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lim, P.N.; Shi, Z.; Neoh, K.G.; Ho, B.; Tay, B.Y.; Thian, E.S. The effects of silver, silicon-containing apatite towards bacteria and cell responses. Biomed. Mater. 2014, 9, 015010. [Google Scholar] [CrossRef]
  24. Nam, K.Y. Characterization and antimicrobial efficacy of Portland cement impregnated with silver nanoparticles. J. Adv. Prosthodont. 2017, 9, 217–223. [Google Scholar] [CrossRef] [Green Version]
  25. Sethmann, I.; Volkel, S.; Pfeifer, F.; Kleebe, H.J. Development of Phosphatized Calcium Carbonate Biominerals as Bioactive Bone Graft Substitute Materials, Part II: Functionalization with Antibacterial Silver Ions. J. Funct. Biomater. 2018, 9, 69. [Google Scholar] [CrossRef] [PubMed]
  26. Sonamuthu, J.; Samayanan, S.; Jeyaraman, A.R.; Murugesan, B.; Krishnan, B.; Mahalingam, S. Influences of ionic liquid and temperature on the tailorable surface morphology of F-apatite nanocomposites for enhancing biological abilities for orthopedic implantation. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 84, 99–107. [Google Scholar] [CrossRef] [PubMed]
  27. Srinivasan, S.; Kumar, P.T.; Nair, S.V.; Nair, S.V.; Chennazhi, K.P.; Jayakumar, R. Antibacterial and bioactive alpha- and beta-chitin hydrogel/nanobioactive glass ceramic/nano silver composite scaffolds for periodontal regeneration. J. Biomed. Nanotechnol. 2013, 9, 1803–1816. [Google Scholar] [CrossRef] [PubMed]
  28. Miola, M.; Ferraris, S.; Di Nunzio, S.; Robotti, P.F.; Bianchi, G.; Fucale, G.; Maina, G.; Cannas, M.; Gatti, S.; Masse, A.; et al. Surface silver-doping of biocompatible glasses to induce antibacterial properties. Part II: Plasma sprayed glass-coatings. J. Mater. Sci. Mater. Med. 2009, 20, 741–749. [Google Scholar] [CrossRef] [Green Version]
  29. Verne, E.; Miola, M.; Vitale Brovarone, C.; Cannas, M.; Gatti, S.; Fucale, G.; Maina, G.; Masse, A.; Di Nunzio, S. Surface silver-doping of biocompatible glass to induce antibacterial properties. Part I: Massive glass. J. Mater. Sci. Mater. Med. 2009, 20, 733–740. [Google Scholar] [CrossRef]
  30. Vollmer, N.L.; Spear, J.R.; Ayers, R.A. Antimicrobial activity and biologic potential of silver-substituted calcium phosphate constructs produced with self-propagating high-temperature synthesis. J. Mater. Sci. Mater. Med. 2016, 27, 104. [Google Scholar] [CrossRef]
  31. Weng, W.; Li, X.; Nie, W.; Liu, H.; Liu, S.; Huang, J.; Zhou, Q.; He, J.; Su, J.; Dong, Z.; et al. One-Step Preparation of an AgNP-nHA@RGO Three-Dimensional Porous Scaffold and Its Application in Infected Bone Defect Treatment. Int. J. Nanomed. 2020, 15, 5027–5042. [Google Scholar] [CrossRef] [PubMed]
  32. Wilcock, C.J.; Stafford, G.P.; Miller, C.A.; Ryabenkova, Y.; Fatima, M.; Gentile, P.; Mobus, G.; Hatton, P.V. Preparation and Antibacterial Properties of Silver-Doped Nanoscale Hydroxyapatite Pastes for Bone Repair and Augmentation. J. Biomed. Nanotechnol. 2017, 13, 1168–1176. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, D.; Liu, W.; Wu, X.D.; He, X.; Lin, X.; Wang, H.; Li, J.; Jiang, J.; Huang, W. Efficacy of novel nano-hydroxyapatite/polyurethane composite scaffolds with silver phosphate particles in chronic osteomyelitis. J. Mater. Sci. Mater. Med. 2019, 30, 59. [Google Scholar] [CrossRef]
  34. Zhang, L.; Jia, G.; Tang, M.; Chen, C.; Niu, J.; Huang, H.; Kang, B.; Pei, J.; Zeng, H.; Yuan, G. Simultaneous enhancement of anti-corrosion, biocompatibility, and antimicrobial activities by hierarchically-structured brushite/Ag3PO4-coated Mg-based scaffolds. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 111, 110779. [Google Scholar] [CrossRef]
  35. Cao, H.; Liu, X.; Meng, F.; Chu, P.K. Biological actions of silver nanoparticles embedded in titanium controlled by micro-galvanic effects. Biomaterials 2011, 32, 693–705. [Google Scholar] [CrossRef] [PubMed]
  36. Tweden, K.S.; Cameron, J.D.; Razzouk, A.J.; Holmberg, W.R.; Kelly, S.J. Biocompatibility of silver-modified polyester for antimicrobial protection of prosthetic valves. J. Heart Valve Dis. 1997, 6, 553–561. [Google Scholar] [PubMed]
  37. Wan, A.T.; Conyers, R.A.; Coombs, C.J.; Masterton, J.P. Determination of silver in blood, urine, and tissues of volunteers and burn patients. Clin. Chem. 1991, 37, 1683–1687. [Google Scholar] [CrossRef]
  38. Russell, A.D.; Hugo, W.B. Antimicrobial activity and action of silver. Prog. Med. Chem. 1994, 31, 351–370. [Google Scholar] [CrossRef]
  39. Hetrick, E.M.; Schoenfisch, M.H. Reducing implant-related infections: Active release strategies. Chem. Soc. Rev. 2006, 35, 780–789. [Google Scholar] [CrossRef]
  40. Palanisamy, N.K.; Ferina, N.; Amirulhusni, A.N.; Mohd-Zain, Z.; Hussaini, J.; Ping, L.J.; Durairaj, R. Antibiofilm properties of chemically synthesized silver nanoparticles found against Pseudomonas aeruginosa. J. Nanobiotechnol. 2014, 12, 2. [Google Scholar] [CrossRef] [Green Version]
  41. Afkhami, F.; Ahmadi, P.; Chiniforush, N.; Sooratgar, A. Correction to: Effect of different activations of silver nanoparticle irrigants on the elimination of Enterococcus faecalis. Clin. Oral Investig. 2022, 26, 1103. [Google Scholar] [CrossRef] [PubMed]
  42. Yin, I.X.; Zhang, J.; Zhao, I.S.; Mei, M.L.; Li, Q.; Chu, C.H. The Antibacterial Mechanism of Silver Nanoparticles and Its Application in Dentistry. Int. J. Nanomed. 2020, 15, 2555–2562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zhang, R.; Lee, P.; Lui, V.C.; Chen, Y.; Liu, X.; Lok, C.N.; To, M.; Yeung, K.W.; Wong, K.K. Silver nanoparticles promote osteogenesis of mesenchymal stem cells and improve bone fracture healing in osteogenesis mechanism mouse model. Nanomedicine 2015, 11, 1949–1959. [Google Scholar] [CrossRef] [PubMed]
  44. AshaRani, P.V.; Low Kah Mun, G.; Hande, M.P.; Valiyaveettil, S. Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano 2009, 3, 279–290. [Google Scholar] [CrossRef]
  45. Clement, J.L.; Jarrett, P.S. Antibacterial silver. Met.-Based Drugs 1994, 1, 467–482. [Google Scholar] [CrossRef]
  46. Pauksch, L.; Hartmann, S.; Rohnke, M.; Szalay, G.; Alt, V.; Schnettler, R.; Lips, K.S. Biocompatibility of silver nanoparticles and silver ions in primary human mesenchymal stem cells and osteoblasts. Acta Biomater. 2014, 10, 439–449. [Google Scholar] [CrossRef]
  47. Necula, B.S.; van Leeuwen, J.P.; Fratila-Apachitei, L.E.; Zaat, S.A.; Apachitei, I.; Duszczyk, J. In vitro cytotoxicity evaluation of porous TiO(2)-Ag antibacterial coatings for human fetal osteoblasts. Acta Biomater. 2012, 8, 4191–4197. [Google Scholar] [CrossRef]
  48. Brutel de la Riviere, A.; Dossche, K.M.; Birnbaum, D.E.; Hacker, R. First clinical experience with a mechanical valve with silver coating. J. Heart Valve Dis. 2000, 9, 123–129. [Google Scholar]
  49. Lansdown, A.B. Critical observations on the neurotoxicity of silver. Crit. Rev. Toxicol. 2007, 37, 237–250. [Google Scholar] [CrossRef]
  50. Shavlovski, M.M.; Chebotar, N.A.; Konopistseva, L.A.; Zakharova, E.T.; Kachourin, A.M.; Vassiliev, V.B.; Gaitskhoki, V.S. Embryotoxicity of silver ions is diminished by ceruloplasmin—Further evidence for its role in the transport of copper. Biometals 1995, 8, 122–128. [Google Scholar] [CrossRef]
  51. Percival, S.L.; Bowler, P.G.; Russell, D. Bacterial resistance to silver in wound care. J. Hosp. Infect. 2005, 60, 1–7. [Google Scholar] [CrossRef] [PubMed]
  52. Landsdown, A.B.; Williams, A. Bacterial resistance to silver in wound care and medical devices. J. Wound Care 2007, 16, 15–19. [Google Scholar] [CrossRef] [PubMed]
  53. Gallo, J.; Panacek, A.; Prucek, R.; Kriegova, E.; Hradilova, S.; Hobza, M.; Holinka, M. Silver Nanocoating Technology in the Prevention of Prosthetic Joint Infection. Materials 2016, 9, 337. [Google Scholar] [CrossRef] [PubMed]
  54. Schneider, O.D.; Mohn, D.; Fuhrer, R.; Klein, K.; Kampf, K.; Nuss, K.M.; Sidler, M.; Zlinszky, K.; von Rechenberg, B.; Stark, W.J. Biocompatibility and Bone Formation of Flexible, Cotton Wool-like PLGA/Calcium Phosphate Nanocomposites in Sheep. Open Orthop. J. 2011, 5, 63–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wnukiewicz, W.; Rutowski, R.; Zboromirska-Wnukiewicz, B.; Reichert, P.; Gosk, J. Evaluation of Soft Tissue Reaction to Corundum Ceramic Implants Infiltrated with Colloidal Silver. Adv. Clin. Exp. Med. 2016, 25, 129–133. [Google Scholar] [CrossRef] [Green Version]
  56. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
Figure 1. PRISMA 2020 flow diagram and the selection of studies.
Figure 1. PRISMA 2020 flow diagram and the selection of studies.
Antibiotics 11 00995 g001
Table 1. Data from included studies.
Table 1. Data from included studies.
StudyType of StudyMaterial TestedAntimicrobial Activity EvaluationsBacteria TestedReported ResultsToxicity
Afzal, 2012 [12]In vitroHydroxyapatite–silver (Ag-HA) and carbon nanotube–silver (CNT-Ag) compositesBacterial count through SEMEscherichia coli
Staphylococcus epidermidis
Partial response.N/A
Bee, 2020 [13]In vitroAntibacterial silver-nanoparticle-decorated hydroxyapatite (HAp/AgNP)Agar diffusionStaphylococcus aureusZone of inhibition of bacterial growth.N/A
Bostancıoğlu, 2015 [11]In vitroSilver-doped calcium-phosphate-based inorganic powder (ABT)Agar diffusion
Agar dilution
Escherichia coli
Pseudomonas aeruginosa
Staphylococcus aureus
Partial response or total response depending on dilution and concentration.Concentration-dependent cytotoxicity on V79 379A and HUVEC lines. ABT is noncytotoxic and bears good biocompatibility even at 1000 μg mL−1 of ABT with the highest content of silver.
Correia, 2016 [8]In vitroTricalcium phosphate (TCP)/sodium alginate scaffold doped with AgNPAgar diffusionStaphylococcus aureusHalo of 0.820 cm with 1 cm scaffold.No cytotoxicity on osteoblast cells.
Dalavi, 2020 [9]In vitroAlginate-nanohydroxyapatite doped with chitooligosaccharide-coated silver nanoparticles (COS-Ag-Alg-HA)Broth microdilutionStaphylococcus aureusTotal response at higher concentration than 77.2% using 3 mg/mL of microsphere.No cytotoxicity on human osteosarcoma osteoblast-like MG-63 cells.
Deng, 2017 [16]In vitroPEEK doped with Ag + nanoparticlesAgar diffusionStaphylococcus aureus
Escherichia coli
Halo of 14 mm of inhibition for both the bacteria with 0.9 mm scaffold.Initial low proliferation rate of human osteosarcoma osteoblast-like MG-63 cells.
Gong, 2017 [17]In vitroSilver-doped hydroxyapatite (Ag-HA) + Bio-OssRT-PCR bacterial DNA measurementPorphyromonas gingivalis
Fusobacterium nucleatum
Partial response, with decreasing of bacterial DNA at 2 h, 4 h, and 24 h compared to control group in which no inhibition was seen.AgHA showed obvious cytotoxicity against periodontal fibroblasts and rat bone-marrow stromal cells, with relative survival rates of <80%. Bio-Oss only showed survival rates exceeding 95% of periodontal.
Jacquart, 2013 [18]In vitroCalcium carbonate–calcium phosphate bone cement doped with silver (Ag-CaCO3-CaP)Broth microdilutionStaphylococcus aureus
Escherichia coli
Complete response.No cytotoxicity on human bone marrow stroma cells.
Jegatheeswaran, 2015 [19]In vitroPolyethylene-glycol/hydroxyapatite doped with silver (Ag-HAp-PEG)Epi-fluorescence microscopyEscherichia coliPartial response with increasing bacteria death in analyses at 6 and 12 h.N/A
Jiang, 2016 [20]In vitroHydroxyapatite/polyurethane composite scaffolds doped with silver phosphate particles (Ag3PO4-n-HA/PU)Agar diffusionStaphylococcus aureus
Escherichia coli
The bacteriostatic rate resulted time and weight percentage of Ag incorporated depending.Scaffolds with no more than 5 wt% appear to have no cytotoxicity on human osteosarcoma osteoblast-like MG-63 cells. Higher concentration (>5%) would weaken cytocompatibility.
Kose, 2020 [21]In vivo (rabbit)Calcium phosphate (CP) with silver ionsRadiological examination
Bacterial cultures from bone samples
Histopathological examinations
Staphylococcus aureusNo MRSA was found at cultures, no X-ray signs of osteomyelitis and no sign of chronic inflammation in histological analysis, compared to the control groups.No inflammatory reactions.
Sampath Kumar, 2015 [22]In vitroCalcium-deficient hydroxyapatite (CDHA) carrier of doxycycline and Ag+ ionsMIC/MBC studies and time-kill assayStaphylococcus aureus
Escherichia coli
When compared with doxycycline, the antibiotic release provided the initial high antibacterial activity, while the sustained ion release provided a long-term antibacterial activity. No cytotoxicity on L6 myoblast cells.
Lim, 2014 [23] In vitroSilver and silicon-containing apatite (Ag,Si-HA)Bacterial count through SEMStaphylococcus aureus
Escherichia coli
No bacteria growth compared to negative control: complete response.MSCs treated with Ag,Si-HA showed an initial low proliferation rate compared to controls, and faster proliferation after day 3.
Nam, 2017 [24]In vitroPortland cement doped with silver nanoparticles (SNPC)Agar diffusionStreptococcus mutans
Streptococcus sobrinus
1.0% wt of SNPC has no antibacterial effect; 3.0 wt% SNPC inhibited S. sorbinus by 1.9 ± 0.5 mm, while no inhibition halos were shown for S. mutans at the same dose. SNPC of 5.0 wt% significantly inhibited S. sorbinus (halo diameter 4.2 ± 0.3 mm) and S. mutans (halo diameter 2.2 ± 0.4 mm).N/A
Paterson, 2020 [4]In vitroPolycaprolactone scaffolds with silver-doped hydroxyapatite (Ag-nHA)Agar diffusionStaphylococcus aureus
Escherichia coli
The scaffold reduced the viable bacteria count to undetectable levels by 48 h for E. coli and 96 h for S. aureus: complete response.Silver-doped nHA to enhance MSC differentiation down an osteogenic path. Scaffolds containing 10 mol.% silver may be toxic for MSCs.
Sethmann, 2018 [25]In vitroPhosphatized Calcium Carbonate biomineral (PCCB) doped with Ag + silver ionsAgar diffusionPseudomonas aeruginosa
Staphylococcus aureus
Samples treated with an AgNO3 solution with 10 mmol/L showed nearly the same antibacterial performance as samples treated with 100 mmol/L. Halo of 1.1–1.2 mm for Gram- and 3 mm for Gram+.N/A
Shimabukuro, 2021 [6]In vitro + in vivo (rabbit)Silver phosphate in carbonate apatite (Ag3PO4-CO3Ap)Agar diffusion immunofluorescenceStaphylococcus epidermidisAntibacterial effect if concentration of Ag3Po4 is more than 0.1 wt %. Complete response.Ag3PO4 content of 0.1–0.95 wt % may show antibacterial properties without cytotoxicity. Higher concentrations showed increasing toxicity for MC3T3-E1 cells. Ag3PO4 content of 0.1–0.3 wt % in the samples did not affect bone formation in vivo.
Sonamuthu, 2018 [26]In vitroFluorinate-hydroxyapatite/polyvinyl alcohol doped with silver nanoparticles (AgNp-fHA)Agar diffusion
CLSM
Broth microdilution
Staphylococcus aureus
Escherichia coli
Antibacterial activity is time- and concentration-dependent. More effect on Gram + due to the different composition of membrane; complete response G+ and G- partial response in CLSM.No cytotoxicity on human osteosarcoma osteoblast-like MG-63 cells.
Sowmya-Srinavasan, 2013 [27]In vitroBioactive alpha- and beta-chitin hydrogel/nanobioactive glass ceramic doped with silverAgar diffusionStaphylococcus aureus
Escherichia coli
Antibacterial activity of Ag dose dependent, similar effect between G+ and G-, but less effective than gentamicin alone.No cytotoxicity on human primary osteoblasts and
human periodontal ligament cells.
Verné, 2009 [29] + Miola, 2009 [28]In vitroSiO-CaO-NaO-AlO doped with silver (Ag-SCNA)Agar diffusion
Broth microdilution
Staphylococcus aureus
Escherichia coli
Same antimicrobial activity against G+ and G-, halo of 2 mm.No cytotoxicity on fibroblasts. Slightly lower proliferation rate compared to control cells.
Vollmer, 2016 [30]In vitroCalcium phosphate (CaP) doped with silverAgar diffusion
Bacterial count through SEM
Escherichia coliAntimicrobial activity with halo in agar diffusion (no dimensions reported) and characteristics of poor health of bacteria at SEM compared to control.No cytotoxicity on human osteoblasts.
Weng, 2020 [31]In vitro + in vivo (rabbit)Loaded nano-hydroxyapatite-reduced graphene oxide doped with Ag nanoparticles (AgNp-AHRG)Agar diffusion
Kirby–Bauer diffusion
WBC count
CRP
Radiological examination
Staphylococcus aureusAntibacterial activity in vitro and the halo zone is dependent on the concentration of Ag. In vivo, it significantly reduced the levels of inflammatory markers, such as leukocytes and CRP, after implantation in the infected site. In subsequent observations, the healing of the bone in the implanted group was significantly improved compared to the untreated group.Concentration-dependent cytotoxicity on bone marrow stromal cells. No cytotoxicity for 1% and 2% silver AgNp-AHRG scaffolds.
Wilcock, 2017 [32]In vitroHydroxyapatite paste silver doped (Ag-nHA)Agar diffusionPseudomonas aeruginosa
Staphylococcus aureus
Antibacterial activity dependent on Ag concentration.N/A
Yuan, 2016 [3]In vitro + in vivo (rabbit)Porous β-tricalcium phosphate with Ag nanoparticles (AgNp- βTCP)Agar diffusion
Bacterial count through SEM
Staphylococcus aureus
Escherichia coli
Antibacterial activity dependent on concentration. Difference in activity between G+ and G- was not reported. At SEM, there is some bacteria visible, but no biofilm was seen.No local and systemic toxicity.
Zhang, 2019 [33]In vivo (rabbit)Nano-hydroxyapatite/polyurethane composite scaffolds doped with silver phosphate particles (Ag/n-HA/PU)WBC count
Radiological examination
Histopathological examinations
Staphylococcus aureusRadiological healing of infection with no difference between 3% wt and 10% wt concentration as well as no difference in histological analysis for trabeculae formation.Local toxicity for highest concentration of silver (Ag/n-HA/10PU).
Zhang, 2020 [34]In vitroBrushite/Ag3PO4-coated Mg-based scaffolds (Mg-DCPD-Ag)Spread plate method
Bacterial count through SEM
Staphylococcus aureus
Escherichia coli
Staphylococcus epidermidis
Antibacterial activity with complete response depending on concentration of Ag.Cytotoxicity for highest concentration of silver (Mg-DCPD-0.46 Ag)
Abbreviations: SEM, scanning electron microscope; MRSA, methicillin-resistant Staphylococcus aureus; WBC, white blood cells; CLSM, confocal laser scanning microscopy; MSC, mesenchymal Stem Cell.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fiore, M.; Bruschi, A.; Giannini, C.; Morante, L.; Rondinella, C.; Filippini, M.; Sambri, A.; De Paolis, M. Is Silver the New Gold? A Systematic Review of the Preclinical Evidence of Its Use in Bone Substitutes as Antiseptic. Antibiotics 2022, 11, 995. https://doi.org/10.3390/antibiotics11080995

AMA Style

Fiore M, Bruschi A, Giannini C, Morante L, Rondinella C, Filippini M, Sambri A, De Paolis M. Is Silver the New Gold? A Systematic Review of the Preclinical Evidence of Its Use in Bone Substitutes as Antiseptic. Antibiotics. 2022; 11(8):995. https://doi.org/10.3390/antibiotics11080995

Chicago/Turabian Style

Fiore, Michele, Alessandro Bruschi, Claudio Giannini, Lorenzo Morante, Claudia Rondinella, Matteo Filippini, Andrea Sambri, and Massimiliano De Paolis. 2022. "Is Silver the New Gold? A Systematic Review of the Preclinical Evidence of Its Use in Bone Substitutes as Antiseptic" Antibiotics 11, no. 8: 995. https://doi.org/10.3390/antibiotics11080995

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop