Next Article in Journal / Special Issue
Cell Biological and Antibacterial Evaluation of a New Approach to Zirconia Implant Surfaces Modified with MTA
Previous Article in Journal
Recent Advances in Scaffolds for Guided Bone Regeneration
Previous Article in Special Issue
Electrospun Materials Based on Cellulose Acetate Loaded with Rosmarinic Acid with Antioxidant and Antifungal Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Use of Biomaterials in 3D Printing as a Solution to Microbial Infections in Arthroplasty and Osseous Reconstruction

by
Argyrios Periferakis
1,2,3,†,
Aristodemos-Theodoros Periferakis
1,3,†,
Lamprini Troumpata
1,
Serban Dragosloveanu
4,5,
Iosif-Aliodor Timofticiuc
1,
Spyrangelos Georgatos-Garcia
6,7,
Andreea-Elena Scheau
8,
Konstantinos Periferakis
2,9,
Ana Caruntu
10,11,
Ioana Anca Badarau
1,
Cristian Scheau
1,12,* and
Constantin Caruntu
1,13
1
Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
2
Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
3
Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
4
Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
5
Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
6
Tilburg Institute for Law, Technology, and Society (TILT), Tilburg University, 5037 DE Tilburg, The Netherlands
7
Corvers Greece IKE, 15124 Athens, Greece
8
Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
9
Pan-Hellenic Organization of Educational Programs (P.O.E.P.), 17236 Athens, Greece
10
Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
11
Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
12
Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
13
Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomimetics 2024, 9(3), 154; https://doi.org/10.3390/biomimetics9030154
Submission received: 26 January 2024 / Revised: 23 February 2024 / Accepted: 25 February 2024 / Published: 1 March 2024

Abstract

:
The incidence of microbial infections in orthopedic prosthetic surgeries is a perennial problem that increases morbidity and mortality, representing one of the major complications of such medical interventions. The emergence of novel technologies, especially 3D printing, represents a promising avenue of development for reducing the risk of such eventualities. There are already a host of biomaterials, suitable for 3D printing, that are being tested for antimicrobial properties when they are coated with bioactive compounds, such as antibiotics, or combined with hydrogels with antimicrobial and antioxidant properties, such as chitosan and metal nanoparticles, among others. The materials discussed in the context of this paper comprise beta-tricalcium phosphate (β-TCP), biphasic calcium phosphate (BCP), hydroxyapatite, lithium disilicate glass, polyetheretherketone (PEEK), poly(propylene fumarate) (PPF), poly(trimethylene carbonate) (PTMC), and zirconia. While the recent research results are promising, further development is required to address the increasing antibiotic resistance exhibited by several common pathogens, the potential for fungal infections, and the potential toxicity of some metal nanoparticles. Other solutions, like the incorporation of phytochemicals, should also be explored. Incorporating artificial intelligence (AI) in the development of certain orthopedic implants and the potential use of AI against bacterial infections might represent viable solutions to these problems. Finally, there are some legal considerations associated with the use of biomaterials and the widespread use of 3D printing, which must be taken into account.

1. Introduction

The emergence of additive manufacturing, most commonly known as 3D printing, has opened new possibilities in different scientific fields (e.g., [1,2,3,4,5,6]), including medicine (e.g., [7,8,9,10,11,12]). The 3D printing approach enables the building of a three-dimensional geometrical object layer-by-layer, guided by computer-aided design (CAD)/computer-aided manufacturing (CAM) software [13,14,15,16,17,18]. Improvements established in CAD/CAM software allow the producer to include the usual post-processing steps, such as milling, in the initial design, reducing manual post-processing stages that often increase errors in the final build [19]; the medical applications of 3D printing are ever-expanding, opening new frontiers in personalized medicine [20,21,22,23].
There are currently several 3D printing technologies available, classified under the ISO/ASTM52900-21 standard [24]. Amongst the many different applications of such techniques in medicine, a prominent one, perhaps the most prominent, is the development of prostheses for a variety of surgical procedures; such prostheses improve both the outcome and quality of life of the patients [25,26,27,28] by solving or at least mitigating some of the problems associated with surgical interventions.
Specifically in orthopedic surgeries, a notable problem involving the use of prostheses is the associated bacterial infections [29,30,31]. The use of artificial implants is already complex enough, given that they must have proper mechanical and structural properties, along with physicochemical compatibility with the natural bone tissue; different materials may be used for different applications, alone or combined [32,33,34,35]. However, regardless of all other considerations, the emergence of a microbial infection can lead to implant failure and even result in amputations and increases in mortality [36,37]. Such infections are most commonly the result of microbial biofilm formation on the surface of the implants [38,39,40]. Such bacterial biofilms oftentimes prove excessively resistant both to the host’s immune system and even to antibiotics [41,42]. Ideally, materials used in surgical prostheses must be both habitable by bone-forming cells and also have suitable anti-adhesive properties, so as to prevent biofilm formation [43]. Moreover, these materials must present an optimal rate of biodegradation, to create space for new bone formation and to exhibit osteogenic, osteoconductive, and osteoinductive properties for proper integration into the body, as a balance between the pore sizes of the build and the rate of biodegradation needs to be found [44,45,46,47,48,49].
In the context of this review, we will present the current consensus on the problem of orthopedic prostheses-associated bacterial infections and the current developments in the antimicrobial properties of biomaterials used in 3D printing to produce orthopedic materials.

2. Microbial Infections in Orthopedic Prostheses

The risk of microbial-related complications during orthopedic surgery is a major concern oftentimes necessitating pre-emptive systemic use of wide-spectrum antibiotics and proper debridement [50]. Despite this vast array of protective measures, the prevalence of surgical site infections remains significant, making up 12–16% of all nosocomial infections [51], and it has been suggested that absolute prevention might just not be feasible [52]. It was shown that the occurrence of surgical site infections, particularly for those patients who undergo multiple operations, and especially deep site infections, are more common in orthopedic surgery when compared with the traumatological department of the same clinic [53].
As such, surgeons go to great lengths to ensure the safety of established methods and to find new ones that pose less of a risk to patients. Indicatively, antibiotic bone cement is widely used to minimize the risk and recent findings indicate that antimicrobial tapes can be similarly effective [54]. Moreover, the implants used in oral and orthopedic surgery are made of alloys like stainless steel and titanium in order to prevent biofilm-associated infections [55,56]. This composition is essential in avoiding prosthetic infections which can necessitate long-term administration of antimicrobial regimens and even removal of the prosthetic, burdening both the patient and the healthcare system with additional hospitalizations [57]. Moreover, it was observed that different surfaces of titanium can induce anti-inflammatory responses mediated by the activation of M2-like macrophages that increase the level of interleukins 4 and 10 (different for smooth or rough titanium), creating a microenvironment with immunological properties, optimal for the healing response in patients with 3D-printed prostheses coated with titanium [58]. Although metal prostheses come with many advantages, from manufacturing to in-body responses, disadvantages can occur in the case of patients allergic to metals [59]. Research has likewise been conducted in regard to the efficacy of the incorporation of capsular traction sutures and it was concluded that they carry a low risk of colonization and thus can be used quite safely in hip arthroscopic surgery [60]. Other researchers have found out that the application of a cyanoacrylate-based skin sealant, called InteguSeal, seems to be beneficial during trauma surgery albeit without the results being conclusive [61]. At any rate, the standard operating room cleaning practices are most likely efficient in dealing with both infectious as well as non-infectious cases, as demonstrated by Balkissoon et al. [62] whose results suggest that conducting surgery on the former type of patient does not compromise a subsequent surgery on the latter that is conducted in the same room.
At the same time, other aspects of orthopedic surgery, such as the utilization of certain tourniquets [63] or sterile stockinettes [64], have been identified as possible sources of contamination. Similarly, a systematic review that examined implant contamination in spinal surgery by going over thirty-five studies deduced that even though intraoperative contamination can be reduced by taking certain safety measures, preoperative contamination through the utilization of single-use implants has not been shown to yield notable positive results [65]. Conversely, current intraoperative implant prophylaxis practices seem to not be as thorough as they could be, and thus new recommendations are being made [66].
The main risk stems from Staphylococcus epidermidis, Staphylococcus aureus, Staphylococcus pettenkoferi, and Micrococcus luteus bacteria [66] while Proteus mirabilis and Citrobacter koseri have also been implicated [60]. Staphylococcus aureus in particular may be responsible for septic arthritis and osteomyelitis, two severe conditions [29]. Corynebacterium spp. were also found to be present in a notable percentage of orthopedic patients belonging to a certain cohort, particularly C. striatum and C. tuberculostearicum [67]. The presence of Corynebacterium in an orthopedic setting has also been confirmed by the research of Walsh et al. [68] who traced it on tourniquets alongside coagulase-negative staphylococci, Aerococcus viridans, and even Bacillus spp. B. anthracis and B. cereus, which are pathogens that can cause lethal infections [69]. Their ability to form spores, thus protecting themselves from adverse environmental conditions and becoming impervious to the action of disinfectants, is a major factor contributing to the burden of disease [70,71]. In regard to the aforementioned M. luteus, infections caused by this germ are infrequent and occur mainly in immunocompromised patients, in the form of bloodstream infections [72]. Moreover, it has a notable presence on the mobile phones of medical personnel, ranking second after coagulase-negative staphylococci, with Bacillus spp. coming in at third place. This is an important finding as these devices can serve as a source of infection in orthopedic surgeries, potentially leading to surgical site infections [73].
Among the microorganisms mentioned, Staphylococcus aureus remains by far the most commonly encountered causative agent [74,75], accounting for two-thirds of all pathogens in orthopedic implant infections [29] and originating both from exogenous sources and due to the patient being a carrier of S. aureus when the surgery takes place, which actually constitutes a risk factor for infection [76]. It should be mentioned however that the diversity of microbes encountered is variable depending on the wound’s localization, with E. coli being responsible mainly for infections following visceral surgery [75]. Alarmingly, the research of Wolcott et al. [77] indicates that a plethora of other microorganisms can be involved as they identified anaerobic bacilli and most notably two previously uncharacterized Bacteroidales. A similar microorganism, B. fragilis, despite having a beneficial role for the host while in the gut [78,79], can cause infections when it finds its way out of the gastrointestinal tract [79], oftentimes resulting in notable bacteremia and abscess formation [78]; it is also commonly associated with polymicrobial infections [80]. Both its drug resistance [78,81] and its virulence, attributed in large part to its encapsulation, are notable; it therefore poses a significant threat [80].
The problem is exacerbated by the fact that several microbes, such as the extended-spectrum beta-lactamase Enterobacteriaceae, including the already mentioned E. coli and K. pneumoniae, account for many of these prosthetic infections and expose the patients to the risks of extensive antibiotic therapy and prosthesis removal, as discussed above, due to them being particularly resilient in the face of any attempts to eliminate them [82]. The latter is an opportunistic pathogen that is so widespread around the world that it makes up one-third of all Gram-negative bacterial infections [83]. Not only can it be the etiologic agent of severe nosocomial infections [84], but several strains have developed resistance to even last-line antibiotics [84,85].
However, there is great concern regarding S. aureus which is notorious for its MRSA strains that are characterized by significant morbidity and mortality and are very prevalent in the community as well as the nosocomial setting, wherein orthopedic patients find themselves; at the same time, these infections are very hard to treat [86]. Unfortunately, the same can be said about C. striatum which is becoming an important determinant of potentially lethal infections in the nosocomial setting, owing in large part to its biofilm formation capacity, with a number of MDR (multi-drug resistance) strains having been identified [87]. Other bacteria, like A. viridans, show a variable level of resistance, with several strains being impervious to the action of erythromycin, tetracycline, and minocycline, while resistance to other antibiotics like chloramphenicol and streptomycin was noted only in a single strain [88].

3. Biomaterials Compatible with Antibiotic Infusion

We can classify biomaterials as organic or inorganic, based on their nature; this is purely a classification scheme however, as it does not affect their suitability for 3D printing or their range of applications. While the chemical processing of organic materials is rather more complex, requiring polymerization of the organic compound to reach the final, 3D printable, synthetic form, the selection of the optimal biomaterial for 3D printing is performed in regard to its properties and the specific requirements of the application [24]. In this review, the inorganic materials discussed are β-tricalcium phosphate (β-TCP), biphasic calcium phosphate (BCP), hydroxyapatite, lithium disilicate, and zirconia; the organic materials are polyetheretherketone (PEEK), poly(propylenefumarate) (PPF), and poly(trimethylene carbonate) (PTMC). Their current uses as biomaterials are summarized in Table 1. The printing processes and the ways in which they are combined with antimicrobial substances are represented in the figure below (Figure 1).

3.1. Beta-Tricalcium Phosphate (β-TCP)

Of the four different forms of tricalcium phosphate, its beta form is of interest in 3D printing applications as it is both heat-stable and printable [125]. It is currently regarded as being of prime importance in bone graft construction [126]. Bone grafts made of β-TCP using 3D printing are suitably porous and strong [47]. Its potent bioactive properties comprise osteoconduction [47] and osteoinductivity [126], gradual biodegradation [48], and reasonably low cytotoxicity [127]. A few studies showed that compared with other biocompatible, 3D-printable materials, β-TCP presents rapid degradation in vivo, which may produce undesirable mechanical features, but these could be overcome with different combinations or metal loads [128].
This compound has already been successfully combined with antibiotics, namely gatifloxacin [129], ciprofloxacin [130], tetracycline [131], vancomycin [132,133,134], and gentamycin [135]. It also displays antimicrobial properties when combined with metals, namely zinc [136], boron nitrite nanotubes [137], iron [138], and silver alone [139,140] or as a hydrogel component [141]; notable antimicrobial properties were also observed when it was combined with chitosan [142,143]. Other combinations with glass [144,145,146] or other artificial compounds [147] have also been successfully tested for their antimicrobial capacity (Table 2).

3.2. Biphasic Calcium Phosphate (BCP)

This is a bioceramic, comprising hydroxyapatite and β-TCP; their ratio, which can vary depending on the needs, determines the properties of the final product [98]. The particulars of the bioactive properties of this material are a reflection of those of its constituents [44,148,149]. BCP exhibits good cytocompatibility and low cytotoxicity and is currently regarded as a prime choice for bone scaffold production [45,46,150]. The morphology of the builds and their influence on their bioactive properties is also a notable aspect, along with the dimension of the pores inside the construct [44,151].
An experimental BCP formulation has been proven capable of reliably eluting antibiotics [152]; combinations with silver ions [153,154] or chitosan [143,155] have also demonstrated antimicrobial properties. In the research of Chen et al. [143], even though no testing was performed on bacterial cells, their compound promoted osteoblast differentiation and activity; this can have important implications given the interplay between osteoblasts and bacterial infections [156,157,158,159] (Table 3).

3.3. Hydroxyapatite (HAP)

Hydroxyapatite, with the chemical formula C a 10 O H 2 P O 4 6 , is a basic component of the structure of human bones [160,161,162]. Apatite also occurs in nature [163,164,165], sometimes as inclusions in gems [166] or in association with other minerals [167,168]. As a biomaterial, it has very good properties [169], and it is hoped that in combination with metal implants, it will be able to increase the biointegration of the latter [170]. The potential of hydroxyapatite as a biomaterial is indeed immense [24] and is associated with its capacity to promote cellular integration and responsiveness [171].
When hydroxyapatite, as a biomaterial, was doped with nickel, tin, and molybdate ions [172], with zinc [173,174], cobalt [175], copper [176], titanium [177], tellurium [178], magnesium [179,180], silver nanoparticles [181,182], or a zinc and gallium combination [183], the results were promising, in that the addition of a small quantity of metals was enough to render the material active against several microorganisms. Another combination with a number of metals also proved effective [184] in this role. It has also been proven possible and successful to combine hydroxyapatite with ciprofloxacin [185] and with ciprofloxacin, dexamethasone, and metal ions [186] and chitosan [187,188]. Finally, some other combinations have been tested in this role, namely with baicalein [189], a plant flavonoid with noted antibacterial effects [190], a composite hydrogel–gelatin material with Ag nanoparticles [191], lactoferrin [192], a molecule with recently recognized promising properties [193], and alginic acid [194] (Table 4).

3.4. Polyetheretherketone, Poly(Propylene Fumarate), and Poly(Trimethylene Carbonate)

Polyetheretherketone (PEEK) has many favorable characteristics, which render it a suitable choice for use in orthopedic prostheses [195]. This material can be used for 3D printing [196,197,198] combined with computer-aided design (CAD) surgical planning, which has recently been gaining favor, especially in craniomaxillofacial reconstruction [199,200]. While it is stable from a chemical standpoint [201], its biological properties are associated with relatively poor osseointegration [202,203]. The addition of carbon fibers in PEEK can improve some of its properties [204,205]; nonetheless, it is still associated with some cytotoxicity [206,207].
There has been some research discussing PEEK implant infections [197,208] but strategies have already been tested on how to improve its antibacterial properties. It has been found that the surface modification of PEEK with sulfuric acid alone [209,210] or in combination with some metals [211,212] has a noted antibacterial effect in vitro; some such combinations even demonstrated this effect in vivo [209,212]. The sulfonation of composite materials containing PEEK exhibited promising antibacterial properties [213,214,215]; the coating of PEEK with antibiotic substances has also been applied successfully [216,217,218,219] (Table 5).
Poly(propylene fumarate) (PPF) has a fumaric acid base structure, opening up a number of potential medical applications [220]. Although it is neither osteoconductive nor osteoinductive, and therefore does not promote tissue regeneration, it has a number of other advantageous biological properties, such as great resorption [117,221]. When combined with various amounts of polyethylene glycol-functionalized graphene oxide (PEG-GO), it exhibits antibacterial action with no commensurate increase in cytotoxicity [222]. Nonetheless, there remain some considerations and challenges regarding its adaption as a biomaterial for 3D printers [117,223] (Table 5).
Poly(trimethylene carbonate) (PTMC), which is derived via ring-opening polymerization [120,224,225], exhibits increased compatibility with body fluids [225]; it has no intrinsic bioactivity but it can be suitably modified for medical engineering [122]. When combined with vinyl pyrrolidone (NVP), carboxymethylcellulose (CMC), and poly(lactic-co-glycolic acid) (PLGA), it also exhibits antimicrobial activity [226] (Table 5).
Table 5. Modifications of PEEK, PPF, and PTMC with antimicrobial properties.
Table 5. Modifications of PEEK, PPF, and PTMC with antimicrobial properties.
BiomaterialModificationDosage and
Compounds
SettingTested
Microorganism
YearReference
Polyetheretherketone (PEEK)Antibiotic coating and combinationsAg nanoparticles and gentamycin on PEEK surfaceIn vitroS. aureus, E. coli2018[216]
Dexamethasone and minocycline liposomes on PEEK surfaceIn vitro, in vivoS. mutans, P. gingivalis2019[217]
Gentamycin sulfate (5 mg/mL)In vitro, in vivoS. aureus, E. coli2020[218]
Dopamine hydrochloride (2 mg/mL) and gentamycin sulfate (3 mg/mL)In vitro, in vivoS. aureus, E. coli2021[219]
Composite material from sulfonation by concentrated sulfuric acidPEEK sulfonation by concentrated sulfuric acidIn vitroS. aureus, E. coli2020[213]
PEEK combination with nanoporous tantalum pentoxide and subsequent treatment by concentrated sulfuric acidIn vitro, in vivoS. aureus, E. coli2021[214]
PEEK combination with porous Ta nanoparticles and genisteinIn vitroS. aureus, E. coli2022[215]
Surface modificationPEEK sulfonation by concentrated sulfuric acidIn vitro, in vivoS. aureus, E. coli2016[209]
Creation of sulfonate PEEK biofilmsIn vitroS. mutans, E. faecalis2017[210]
Surface modification with concentrated sulfuric acid and ArIn vitroS. aureus, E. coli2018[211]
Surface modification with concentrated sulfuric acid and Cu nanoparticlesIn vitro, in vivoS. aureus (MRSA)2019[212]
Poly(propylene fumarate) (PPF)Combinations with other materials and compoundsPolyethylene glycol-functionalized graphene oxide (PEG-GO)In vitroS. aureus, S. epidermidis, P. aeruginosa, E. coli2016[222]
Poly(trimethylene
carbonate)
(PTMC)
N-vinyl pyrrolidone (NVP), carboxymethylcellulose (CMC) and poly(lactic-co-glycolic acid) (PLGA)In vitron/a–theorized antibacterial use2015[226]

3.5. Zirconia and Lithium Disilicate

Zircon dioxide, also known as zirconia Z r O 2 , occurs naturally as the mineral baddeleyite [227,228] and has excellent mechanical properties [229]; it is considered as both the most durable and aesthetically acceptable prosthesis [230,231,232]. Its biochemical and physicochemical properties justify its extensive use [231,233,234] considering its lack of bioactive properties [124]; nonetheless, there are some drawbacks associated with its 3D printing uses [231,235,236,237]. A few of the properties of zirconia, such as its low cytotoxicity and resistance to colonization of bacteria, and also good 3D printability, make this material relevant for review [234,238]. Zirconia has been tested for antibacterial action, when nanomodified [239], with a chitosan-containing surface modification [240], or when combined with Ag nanoparticles [241]; all such tests have proved successful.
Lithium disilicate is a glass-ceramic material with the chemical formula L i 2 S i 2 O 5 and has a biphasic crystalline structure [242]; it is currently mostly used in dental operations [106]. New 3D printing techniques have increased its usefulness and potential [243,244,245]. The combination of lithium with glass nanoparticles has exhibited some positive antibacterial results [105] (Table 6).

4. Discussion

4.1. Critical Insight on Available Data Regarding Antimicrobial 3D-Printed Implants

From all the aforementioned studies, it is implied that the biomaterials utilized must have properties that can both mimic the characteristics of the replaced/reconstructed tissues and have antimicrobial properties so as to mitigate the risk of failure of the operation. The proper selection of materials and their most beneficial combination is paramount; such an endeavor can be undertaken by using a comprehensive approach to develop biomaterials for 3D printing [24]. Implant-associated infections are ever increasing as the sheer number of such surgeries [246], the relevant burden of disease [247,248], and the need for revision surgeries [249] also increases. Therefore, the need to develop new techniques, based on current technologies, is paramount [250].
Both biomaterials and techniques and methodologies associated with their production and application are increasing (e.g., [251,252,253,254,255,256,257,258,259]). As can be seen from the information heretofore presented, there exist numerous options, particularly for developing 3D printing-adapted biomaterials with antimicrobial properties. Many experiments are focused on the combination of existing biomaterials with metal nanoparticles. Indeed, the antimicrobial potential of metal nanoparticles has been studied in detail by numerous researchers (e.g., [260,261,262,263,264,265,266,267,268,269]). Based on recent evidence, metal nanoparticles may also have an important role to play in the diagnosis and even treatment of cancer [270,271]; this can be important in cases of bone degeneration and even fracturing due to cancer [272,273]. A typical example is the case of osteosarcoma, a primary bone malignancy [274,275] where sometimes the only therapeutical avenues include allografting and autografting along with metallic prostheses [276]. Despite all these research efforts, it must be noted that there is still a lack of a complete understanding of the potentially toxic effects of some metal nanoparticles [277,278].
In contrast to inorganic materials that are more commonly used in 3D printing, recent studies have focused on organic materials, resulting from polymerization, which have notable properties, such as poly(methyl methacrylate) (PMMA), PTMC, PEEK, and PPF [24]. PPF, unlike the other materials presented in this review, does not exhibit bioactive properties, but it compensates through its superior mechanical properties and the possibility of creating a structure with unique geometries and the optimal porosity that can later be coated or loaded with antibiotics [279].
On the other hand, PMMA and PTMC (methacrylate-based polymers) present better utility in building a 3D model that mimics soft tissues [280,281]. The use of antibiotics together with these materials has not yet been researched, but adding antimicrobial substances in the final structures, for example, as a 3D-printed meniscus, could be a possibility in the future [120].
PEEK scaffolds are known to imitate the mechanical aspects of cancellous bone and also exhibit angiogenic properties, which can be enhanced with different metal-coatings such as magnesium [282]. Compared with other materials, there are no significant differences regarding the mechanical or bioactive properties but depending on the purposes of the research or the compatibility with 3D printers, a larger array of materials offers more flexibility for projects.
Quite a number of studies have focused on combinations of biomaterials with chitosan, an abundant biopolymer derived from a number of organisms [283], which has many positive properties [284,285,286,287] in addition to its more important, in the context of this paper, antimicrobial ones [288,289]. Given that the most recent research regarding the antimicrobial potential of chitosan-containing combinations and nanoparticles has demonstrated encouraging results [290,291,292,293], one can only imagine the potential of its incorporation in the prosthetics and implants field.
Another avenue, which of course has been extensively studied, is the combination of biomaterials with antibiotics. This is only natural, given that antibiotics still represent the most potent medical intervention against bacterial infections [294,295]. While the combination of biomaterials with antibiotics has been steadily gaining traction [296,297,298,299], there are still some problems with such applications. An anticipated problem is the resistance to antibiotics which is characterized by the ineffectiveness against an infection by resistant bacteria, or the creation of resistance due to the pre-emptive use of antibiotics.
We have mentioned that the most common pathogens in orthopedic implant infections are Staphylococcus aureus, Escherichia coli, and Klebsiella pneumoniae. For S. aureus, over 30% of strains are reportedly resistant to some common antibiotics [300]; indeed, there are numerous mechanisms reported to be associated with such antibiotic resistance in this bacterial species [301]. For E. coli, there is likewise a trend of emerging resistance based on recent studies [302,303] and the same can be said for K. pneumoniae [304,305]. So, a problem arises regarding the selection of antibiotics to be incorporated into the prostheses. What if there are resistant bacterial strains? Perhaps a solution would be the pre-emptive use of very powerful antibiotics such as vancomycin [306]; however, there are already bacterial strains resistant even to this drug [307,308,309], and the injudicious use of vancomycin may be by itself a cause of resistance emergence [310].
Regarding the enterococci, E. faecium and E. faecalis are the most relevant species from a clinical point of view [311] since they account for a notable part of the infections encountered in the nosocomial setting [312]. Not only can such cases be potentially life-threatening [313,314], but our means of curing them are being limited as vancomycin-resistant enterococci (VRE) strains are emerging [315]. Similarly, bacteria of the Pseudomonas aeruginosa species are responsible for a considerable number of nosocomial infections [316], both localized and systemic, which not only can be life-threatening [317], but may also be difficult to handle as resistant P. aeruginosa strains are becoming more prevalent [318,319]. Streptococcus anginosus, the official name of a group of bacteria commonly referred to as S. milleri, shows notable variety regarding its hemolytic, physiological, and serological characteristics, making its identification challenging in the laboratory setting [320]. It is clinically relevant as it can cause severe infections, particularly purulent ones [320]. Porphyromonas is most commonly associated with periodontitis, but it can also cause severe systemic infections [321,322] and has even been implicated in cognitive impairment [323] and carcinogenesis [321,324,325].
Streptococcus mutans is mostly known as an important cause of dental plaque formation, with its ability to form biofilms playing a critical role in its pathogenicity [326,327]. However, it can cause other serious conditions, such as life-threatening endocarditis [326] and carcinogenesis [327]. Salmonella is a major etiological agent of foodborne pathologies that is a cause for concern for global public health [328,329]. It has a characteristic diversity when it comes to serovariability, having over 2600 serotypes [330], as well as antigenic variability [331]. Its virulence and mortality rates are not to be underestimated [332], as many strains exhibit antibiotic resistance [333,334]. S. enteritidis is among the most frequently encountered species, and is mainly found in chicken eggs [335]. Shigella dysenteriae is a common causative agent of diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome [336], with a wide arsenal of virulence factors at its disposal [336,337]. Antibiotic resistance is a concern in this case as well [338]. Finally, the rarely mentioned Actinobacillus actinomycetemcomitans is normally a part of the physiological flora of the oral cavity; it is, however, capable of causing periodontitis as well as systemic pathologies [339,340,341], such as coronary artery disease in the case of serotypes b and c [340]. It is also notable for its ability to evade the immune system [342] and its very potent leukotoxin [339].
Still on the subject of infections, we must note that while bacteria account for the majority of orthopedic infections, there are other pathogens of concern. For example, a case report by Soukup et al. [343] mentions the appearance of toxocariasis as a post-surgical complication after transthoracic spine surgery. But, apart from such rare incidents, other applications may prove useful; a prominent case might be the surgical removal of cysts of Echinococcus granulosus from the spine [344]. However, the surgical removal of the cysts may sometimes present complications [345]; the removal might only be partial [346] or the resultant spillage may lead to secondary echinococcosis [347,348]. In such cases, prostheses associated with proper drugs, namely albendazole, mebendazole, and perhaps praziquantel [349], may be useful. Another incidence of parasitic infection may occur in patients who are immunosuppressed in the course of rheumatoid arthritis treatment—such a case has been reported by Trigkidis et al. [350]. Perhaps, in such particular cases, and given that rheumatoid arthritis frequently necessitates the use of orthopedic prostheses [351], the incorporation of antimonial drugs, which have a proven anti-leishmanial effect [352], may prove useful.
A minor consideration, compared to bacterial infections, is fungal infections in orthopedic implants. Still, research has been conducted regarding the role that microorganisms like Candida spp. and Aspergillus spp. play in hip prosthetic joint infections, a condition most commonly associated with Staphylococcus spp. [353,354]. Fungi can indeed be the causative agent of such an infection, mostly owing to Candida spp., and, occasionally, they even coinfect the patient alongside bacterial pathogens [353,354]. Although this complication arises infrequently, it causes a severe condition that requires multidisciplinary action to be properly dealt with [353,354]. The combination of biomaterials with common antifungal agents, such as triazoles and amphotericin formulations [355,356], should be studied in the future.
In general, the development of biomaterials suitable for bioprinting can reduce, or even hopefully eliminate, the need for bone allografts and the management of the associated immune response [357,358]. Perhaps the most potent material for this purpose is hydroxyapatite [359], with BCP and β-TCP being less resilient to mechanical stress, despite having good properties overall [360]. As there are still other biomaterials better suitable for soft tissue replacement, and still other biomaterials with untapped potential [361,362], it is very possible that, in the near future, novel approaches for producing biomaterials with a potent antimicrobial action will arise.

4.2. Current Challenges in 3D Printing with Mixtures Containing Antimicrobial Substances

Regarding 3D printing models with antimicrobial activity, we can divide the final construct into two main groups: models with incorporated antimicrobial particles, and models with antimicrobial substance coatings or loading. In the second group, the first step is printing the 3D model separately, and in the post-processing steps, the active substances are added as a coating or loaded into the micro-pores of the construct. So, the only challenges that can occur in these cases are strictly structural aspects and later biocompatibility and bioactivity within the body, as the functionality of the build or the activity of the antimicrobial substances should not affect each other.
Many of the 3D printing technologies require high-temperature treatment of the material at the time of the printing [363]; in that case, the initial mix that contains the antimicrobial particles has to withstand these temperatures, and to still be able to exhibit antimicrobial activity after printing. Most antibiotics are thermolabile, and after thermic treatment of the mix, the bioactivity of the substances can decrease dramatically [364]. In that case, we can conclude that fused deposition modeling (FDM), a popular 3D printing technique, that uses temperatures over 80 °C is not the most efficient method in building parts with antimicrobial activity. Even if the thermic problem is managed, it is already known that the mechanical properties of the construct with antibiotics are significantly decreased compared with the constructs with no antimicrobial substances added [365].
However, metals and nanoparticles with antimicrobial activity, such as zinc, iron, copper, magnesium, and their oxides are known to maintain their antimicrobial activity after printing with the standard temperatures of different 3D printing technologies such as FDM [366]. Even though temperature is not a problem when printing with incorporated metals or metal oxides, dispersing them homogenously can still be a challenge, which can alter the final product, inducing filling defects or agglomeration of particles and causing structural instability [366].
Another method, called inkjet printing, requires high-temperature treatment only in the post-processing steps, which can be skipped if needed; in this way, the functionality of the antibiotics can be maintained [367]. With inkjet printing, it was already demonstrated that adding antibiotics does not alter the mechanical properties of the final build, but there are not many biocompatible materials that can be used with this technology; shortly, new materials may be available [368]. Antibiotics or nanoparticles with antimicrobial activity that are UV sensible cannot be printed by vat photopolymerization; however, positively charged quaternary ammonium compounds and silver–halloysite in combination with methacrylate-based polymers such as PTMC and PMMA reported good results in dentistry applications [367].
From our team’s experience with 3D printing, especially with stereolithography (SLA) technology, finding the optimal mixture of the biocompatible material and the antibiotic or antimicrobial particles is the most important goal. Different technologies require different solubilities, viscosities, and temperatures, and also, different post-processing steps. Finding ways to solubilize the antimicrobial substance in a way that the optimal viscosity is maintained and the mechanical properties of the final constructs (that usually contain pores of different sizes) are not altered are the main challenges that should be further studied.

4.3. Legal Considerations for 3D-Printed Antibiotic-Integrated Medical Implants

Even the most promising applications inevitably bring associated challenges. In this particular situation, aside from the previously discussed considerations, there is a medico-legal dimension to take into account. In today’s medical practice, legal proceedings regarding medical responsibility and liability have become an integral aspect of the profession. This intricacy is exacerbated by factors that go beyond the conventional patient care environment, notably the incorporation of advanced technologies [369], like 3D printing. Therefore, present-day healthcare professionals need to familiarize themselves with the applicable regulatory principles and guidelines, especially when engaging with such technologies in pursuit of innovation. In the European Union (EU), medical liability is predominantly regulated by national laws; however, specific EU directives and regulations outline the overarching principles.
The EU Clinical Trials Regulation [370] delineates the regulations for clinical studies and trials, defined in Article 2(2) as inclusive of “therapeutic strategies that deviate from the normal clinical practice of the Member State concerned”. Ensuring transparency through precise and comprehensive reporting, proportional balancing of risks and benefits, obtaining informed consent, and adhering to safety standards are pivotal for safeguarding research participants. The Medical Devices Regulation [371] focuses on the concept of medical devices, defined in Article 2(1), and relevant terms such as an “accessory of medical device” [Art. 2(2)], “implantable device” [Art. 2(5)], etc. It establishes safety and performance requirements through mandatory measures, including risk classification, conformity assessments by manufacturers, clinical evaluations, and heightened scrutiny. Furthermore, the EU Patient Rights Directive [372] and the European Charter of Patients’ Rights [373] establish a framework to protect patients’ rights in the European Union, encompassing the right to information and informed consent, access to medical records, privacy, and the right to redress in case of harm. This latter right is particularly significant, empowering patients to voice complaints and seek redress in instances of medical malpractice or dissatisfaction with healthcare services.
As for the applicability of the legal or quasi-legal documents above and their provisions in the context of the present article and to provide an extra layer of information on the connection between what is written in this article and the legal status quo, the following apply:
(a) Under no circumstance, based on the current data, can the use of antimicrobial material in 3D printing in the field of Arthroplasty and Osseous Reconstruction be considered as “normal clinical practice”, meaning the day-to-day typical medical approach. Therefore, the definition of the clinical trial as mentioned in the EU Clinical Trials Regulation seems to encompass the aforementioned notion, providing the necessary framework for the implementation of the Regulation.
(b) Additionally, the Medical Devices Regulation focusing on medical devices includes in its provisions the concept of “implanted devices”, a classification which largely, if not exactly, reflects the essence of this article, meaning both the materials used and the 3D-printed orthopedic implants under discussion.
(c) As for the EU Patient Rights Directive and the European Charter of Patients’ Rights, once patients are involved in the whole procedure, they are applicable by default, and no further clarifications are required since the patients’ rights and their protection hold great significance in the EU legal framework.

4.4. Future Directions and Emerging Trends

The approach of using antibiotic-laden biomaterials in a protective manner is a trending idea among research groups worldwide. However, such use has been known to be either ineffective at times, or even to promote biofilm formation and resistant infection occurrence [374]. Based on the increased capabilities and potential of medical prosthetics [375], prostheses could be outfitted with methods of releasing suitable antibiotics after the source of infection has been precisely identified and the relevant resistance profile has been determined. In addition, there exists nowadays the possibility of incorporating artificial intelligence (AI) in the development of orthopedic implants [375] and given that AI is also under research for use in combatting antibiotic resistance [376,377,378], the integration of relevant AI schemes into orthopedic implants in the future might be a viable solution.
Another avenue that can be explored to enhance the antimicrobial properties of the discussed biomaterials is phytochemicals [379,380]. Recent research has highlighted the antimicrobial potential of numerous phytochemicals (e.g., [381,382,383,384,385,386]); many purified plant compounds have been found to have antimicrobial properties, such as capsaicin [387] and other capsaicinoids [388,389], curcumin [390,391,392], kaempferol and its derivatives [393,394], catechins [395,396], turmeric [397], fucoidan [398], and other plant compounds (e.g., [399,400]). Phytochemicals have already been used as coatings for a variety of materials and for a variety of purposes. Importantly, phytochemicals can be incorporated into artificial materials to lower their potential toxicity [401]. Furthermore, the research on combinations of phytochemicals and nanoparticles has yielded promising results (e.g., [402,403,404]); nanoparticles are already being applied as drug delivery systems [405], and they have also been already combined successfully with prostheses as outlined in this paper.
Finally, a further potential avenue that warrants further exploration is the combination of the materials discussed herein with antibiotic pearls for antibiotic applications. This has been shown to ameliorate the prognosis in prosthetic surgeries by eliminating biofilms and enabling extended antibiotic action both qualitatively and temporally through the use of calcium sulphate antibiotic-added beads [406,407]. The usefulness of these beads as adjuvants had previously been mentioned and can also be corroborated by the findings of Agarwal and Healy [408]. Joint infections after arthroplasty of the knee are of particular interest as it is in this context that the debridement, antibiotic bead, and retention of the implant (DABRI) method was compared to the debridement, antibiotics, and implant retention (DAIR) method and was found to be similarly effective [409]. We would also like to note, that, in the context of antibiotic pearls in particular, and of the potential combinations mentioned in this paper in general, the potential of adverse effects, especially associated with drug pairing, is a noteworthy constraint; given that such interactions could affect absorption or toxicity [410,411,412,413], caution toward administration is deemed necessary [414].

5. Conclusions

The antimicrobial properties of materials adapted for 3D printing are a promising research field, and there are still many compounds and combinations that can be tested. The current tests mostly revolve around combinations of existing biomaterials with antibiotics, metal nanoparticles, and chitosan. Future research must be centered around addressing the relevant problem of antibiotic resistance and the possibility, however small, of fungal or parasitic infection.
The combination of biomaterials with phytochemicals of known antibacterial potential also represents a promising avenue of research. Last, but not least, there is a need for accurate and in-depth information on medical liability frameworks in conjunction with all the relevant EU legal documents; it is also vital to refer to the individual national rules of the EU member states.

Author Contributions

Conceptualization, A.P., C.S. and C.C.; investigation, A.P., A.-T.P., L.T., K.P. and I.A.B.; resources, S.D., I.-A.T., S.G.-G., A.-E.S. and A.C.; writing—original draft preparation, A.P., A.-T.P., L.T., S.D., I.-A.T., S.G.-G., A.-E.S., K.P., A.C., I.A.B., C.S. and C.C.; writing—review and editing, A.P., A.-T.P., A.-E.S., A.C., C.S. and C.C.; supervision, A.C., I.A.B., C.S. and C.C. 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.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mohammed, J.S. Applications of 3D printing technologies in oceanography. Methods Oceanogr. 2016, 17, 97–117. [Google Scholar] [CrossRef]
  2. Sun, Y.; Li, Q. The application of 3D printing in mathematics education. In Proceedings of the 2017 12th International Conference on Computer Science and Education (ICCSE), Houston, TX, USA, 22–25 August 2017; pp. 47–50. [Google Scholar]
  3. Lee, J.-Y.; An, J.; Chua, C.K. Fundamentals and applications of 3D printing for novel materials. Appl. Mater. Today 2017, 7, 120–133. [Google Scholar] [CrossRef]
  4. Thakar, C.M.; Parkhe, S.S.; Jain, A.; Phasinam, K.; Murugesan, G.; Ventayen, R.J.M. 3D Printing: Basic principles and applications. Mater. Today Proc. 2022, 51, 842–849. [Google Scholar] [CrossRef]
  5. Rezaei, H.; Matin, A.A. 3D-printed solid phase microextraction fiber based on Co-Al layered double hydroxide nanosheets; application in determination of phenolic acids in fruit juice samples. Food Chem. 2024, 437, 137894. [Google Scholar] [CrossRef] [PubMed]
  6. Gao, X.; Liu, K.; Su, C.; Zhang, W.; Dai, Y.; Parkin, I.P.; Carmalt, C.J.; He, G. From bibliometric analysis: 3D printing design strategies and battery applications with a focus on zinc-ion batteries. SmartMat 2024, 5, e1197. [Google Scholar] [CrossRef]
  7. Ventola, C.L. Medical Applications for 3D Printing: Current and Projected Uses. Pharm. Ther. 2014, 39, 704–711. [Google Scholar]
  8. Chimene, D.; Lennox, K.K.; Kaunas, R.R.; Gaharwar, A.K. Advanced bioinks for 3D printing: A materials science perspective. Ann. Biomed. Eng. 2016, 44, 2090–2102. [Google Scholar] [CrossRef] [PubMed]
  9. Bakhtiar, S.M.; Butt, H.A.; Zeb, S.; Quddusi, D.M.; Gul, S.; Dilshad, E. Chapter 10—3D Printing Technologies and Their Applications in Biomedical Science. In Omics Technologies and Bio-Engineering; Barh, D., Azevedo, V., Eds.; Academic Press: Cambridge, MA, USA, 2018; pp. 167–189. [Google Scholar] [CrossRef]
  10. Jakus, A.E. Chapter 1—An Introduction to 3D Printing—Past, Present, and Future Promise. In 3D Printing in Orthopaedic Surgery; Dipaola, M., Wodajo, F.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 1–15. [Google Scholar]
  11. Tian, Y.; Chen, C.; Xu, X.; Wang, J.; Hou, X.; Li, K.; Lu, X.; Shi, H.; Lee, E.-S.; Jiang, H.B. A review of 3D printing in dentistry: Technologies, affecting factors, and applications. Scanning 2021, 2021, 9950131. [Google Scholar] [CrossRef] [PubMed]
  12. Li, J.; Liang, D.; Chen, X.; Sun, W.; Shen, X. Applications of 3D printing in tumor treatment. Biomed. Technol. 2024, 5, 1–13. [Google Scholar] [CrossRef]
  13. Anadioti, E.; Musharbash, L.; Blatz, M.B.; Papavasiliou, G.; Kamposiora, P. 3D printed complete removable dental prostheses: A narrative review. BMC Oral. Health 2020, 20, 343. [Google Scholar] [CrossRef] [PubMed]
  14. Salmi, M. Additive Manufacturing Processes in Medical Applications. Materials 2021, 14, 191. [Google Scholar] [CrossRef]
  15. Ahmed, K.S.; Ibad, H.; Suchal, Z.A.; Gosain, A.K. Implementation of 3D Printing and Computer-Aided Design and Manufacturing (CAD/CAM) in Craniofacial Reconstruction. J. Craniofac. Surg. 2022, 33, 1714–1719. [Google Scholar] [CrossRef]
  16. Christakopoulos, F.; van Heugten, P.M.H.; Tervoort, T.A. Additive Manufacturing of Polyolefins. Polymers 2022, 14, 5147. [Google Scholar] [CrossRef]
  17. Dzogbewu, T.C.; Fianko, S.K.; Amoah, N.; Afrifa Jnr, S.; de Beer, D. Additive manufacturing in South Africa: Critical success factors. Heliyon 2022, 8, e11852. [Google Scholar] [CrossRef]
  18. Alghazzawi, T.F. Advancements in CAD/CAM technology: Options for practical implementation. J. Prosthodont. Res. 2016, 60, 72–84. [Google Scholar] [CrossRef]
  19. Ottoni, R.; Marocho, S.M.S.; Griggs, J.A.; Borba, M. CAD/CAM versus 3D-printing/pressed lithium disilicate monolithic crowns: Adaptation and fatigue behavior. J. Dent. 2022, 123, 104181. [Google Scholar] [CrossRef]
  20. Vaz, V.M.; Kumar, L. 3D printing as a promising tool in personalized medicine. Aaps. Pharmscitech. 2021, 22, 1–20. [Google Scholar] [CrossRef] [PubMed]
  21. Amekyeh, H.; Tarlochan, F.; Billa, N. Practicality of 3D printed personalized medicines in therapeutics. Front. Pharmacol. 2021, 12, 646836. [Google Scholar] [CrossRef] [PubMed]
  22. Tan, G.; Ioannou, N.; Mathew, E.; Tagalakis, A.D.; Lamprou, D.A.; Yu-Wai-Man, C. 3D printing in Ophthalmology: From medical implants to personalised medicine. Int. J. Pharm. 2022, 625, 122094. [Google Scholar] [CrossRef] [PubMed]
  23. Serrano, D.R.; Kara, A.; Yuste, I.; Luciano, F.C.; Ongoren, B.; Anaya, B.J.; Molina, G.; Diez, L.; Ramirez, B.I.; Ramirez, I.O. 3D printing technologies in personalized medicine, nanomedicines, and biopharmaceuticals. Pharmaceutics 2023, 15, 313. [Google Scholar] [CrossRef] [PubMed]
  24. Timofticiuc, I.-A.; Călinescu, O.; Iftime, A.; Dragosloveanu, S.; Caruntu, A.; Scheau, A.-E.; Badarau, I.A.; Didilescu, A.C.; Caruntu, C.; Scheau, C. Biomaterials Adapted to Vat Photopolymerization in 3D Printing: Characteristics and Medical Applications. J. Funct. Biomater. 2024, 15, 7. [Google Scholar] [CrossRef] [PubMed]
  25. Zoabi, A.; Redenski, I.; Oren, D.; Kasem, A.; Zigron, A.; Daoud, S.; Moskovich, L.; Kablan, F.; Srouji, S. 3D Printing and Virtual Surgical Planning in Oral and Maxillofacial Surgery. J. Clin. Med. 2022, 11, 2385. [Google Scholar] [CrossRef] [PubMed]
  26. Vasiliadis, A.V.; Koukoulias, N.; Katakalos, K. From Three-Dimensional (3D)- to 6D-Printing Technology in Orthopedics: Science Fiction or Scientific Reality? J. Funct. Biomater. 2022, 13, 101. [Google Scholar] [CrossRef] [PubMed]
  27. Cristea, S.; Predescu, V.; Dragosloveanu, S.; Cuculici, S.; Marandici, N. Surgical Approaches for Total Knee Arthroplasty. In Arthroplasty—A Comprehensive Review; Bagaria, V., Ed.; IntechOpen: London, UK, 2016; pp. 25–47. [Google Scholar]
  28. Alshamrani, A.; Alhotan, A.; Owais, A.; Ellakwa, A. The Clinical Potential of 3D-Printed Crowns Reinforced with Zirconia and Glass Silica Microfillers. J. Funct. Biomater. 2023, 14, 267. [Google Scholar] [CrossRef] [PubMed]
  29. Ribeiro, M.; Monteiro, F.J.; Ferraz, M.P. Infection of orthopedic implants with emphasis on bacterial adhesion process and techniques used in studying bacterial-material interactions. Biomatter 2012, 2, 176–194. [Google Scholar] [CrossRef] [PubMed]
  30. Song, Z.; Borgwardt, L.; Høiby, N.; Wu, H.; Sørensen, T.S.; Borgwardt, A. Prosthesis infections after orthopedic joint replacement: The possible role of bacterial biofilms. Orthop. Rev. 2013, 5, 65–71. [Google Scholar] [CrossRef]
  31. Kaufman, M.G.; Meaike, J.D.; Izaddoost, S.A. Orthopedic prosthetic infections: Diagnosis and orthopedic salvage. Semin. Plast Surg. 2016, 30, 66–72. [Google Scholar] [CrossRef]
  32. Duan, K.; Wang, R. Surface modifications of bone implants through wet chemistry. J. Mater. Chem. 2006, 16, 2309–2321. [Google Scholar] [CrossRef]
  33. Paital, S.R.; Dahotre, N.B. Calcium phosphate coatings for bio-implant applications: Materials, performance factors, and methodologies. Mater. Sci. Eng. R Rep. 2009, 66, 1–70. [Google Scholar] [CrossRef]
  34. Chevalier, J.; Gremillard, L. Ceramics for medical applications: A picture for the next 20 years. J. Eur. Ceram. Soc. 2009, 29, 1245–1255. [Google Scholar] [CrossRef]
  35. Vallet-Regí, M. Evolution of bioceramics within the field of biomaterials. Comptes Rendus Chim. 2010, 13, 174–185. [Google Scholar] [CrossRef]
  36. Anderson, J.M.; Marchant, R.E. Biomaterials: Factors Favoring Colonization and Infection. In Infections Associated with Indwelling Medical Devices; Waldvogel, F.A., Bisno, A.L., Eds.; ASM Press: Washington, DC, USA, 2000; pp. 89–109. [Google Scholar] [CrossRef]
  37. Ercan, B.; Kummer, K.M.; Tarquinio, K.M.; Webster, T.J. Decreased Staphylococcus aureus biofilm growth on anodized nanotubular titanium and the effect of electrical stimulation. Acta Biomater. 2011, 7, 3003–3012. [Google Scholar] [CrossRef]
  38. Trampuz, A.; Osmon, D.R.; Hanssen, A.D.; Steckelberg, J.M.; Patel, R. Molecular and antibiofilm approaches to prosthetic joint infection. Clin. Orthop. Relat. Res. 2003, 414, 69–88. [Google Scholar] [CrossRef] [PubMed]
  39. Zilberman, M.; Elsner, J.J. Antibiotic-eluting medical devices for various applications. J. Control. Release 2008, 130, 202–215. [Google Scholar] [CrossRef]
  40. Gbejuade, H.O.; Lovering, A.M.; Webb, J.C. The role of microbial biofilms in prosthetic joint infections. Acta Orthop. 2015, 86, 147–158. [Google Scholar] [CrossRef] [PubMed]
  41. Davies, D. Understanding biofilm resistance to antibacterial agents. Nat. Rev. Drug Discov. 2003, 2, 114–122. [Google Scholar] [CrossRef]
  42. Gristina, A.; Naylor, P.; Myrvik, Q. Infections from biomaterials and implants: A race for the surface. Med. Prog. Through Technol. 1988, 14, 205–224. [Google Scholar]
  43. Montanaro, L.; Campoccia, D.; Arciola, C. Nanostructured materials for inhibition of bacterial adhesion in orthopedic implants: A minireview. Int. J. Artif. Organs 2008, 31, 771–776. [Google Scholar] [CrossRef] [PubMed]
  44. Seo, Y.-W.; Park, J.-Y.; Lee, D.-N.; Jin, X.; Cha, J.-K.; Paik, J.-W.; Choi, S.-H. Three-dimensionally printed biphasic calcium phosphate blocks with different pore diameters for regeneration in rabbit calvarial defects. Biomater. Res. 2022, 26, 25. [Google Scholar] [CrossRef]
  45. Jeon, S.-H.; Song, Y.W.; Cha, J.-K.; Paik, J.-W.; Han, S.-S.; Choi, S.-H. Scanning electron microscopic evaluation of the internal fit accuracy of 3D-printed biphasic calcium phosphate block: An ex vivo pilot study. Materials 2021, 14, 1557. [Google Scholar] [CrossRef]
  46. Yang, Z.; Xie, L.; Zhang, B.; Zhang, G.; Huo, F.; Zhou, C.; Liang, X.; Fan, Y.; Tian, W.; Tan, Y. Preparation of BMP-2/PDA-BCP bioceramic scaffold by DLP 3D printing and its ability for inducing continuous bone formation. Front. Bioeng. Biotechnol. 2022, 10, 854693. [Google Scholar] [CrossRef]
  47. Schmidleithner, C.; Malferrari, S.; Palgrave, R.; Bomze, D.; Schwentenwein, M.; Kalaskar, D.M. Application of high resolution DLP stereolithography for fabrication of tricalcium phosphate scaffolds for bone regeneration. Biomed. Mater. 2019, 14, 045018. [Google Scholar] [CrossRef]
  48. Wang, D.; Hou, J.; Xia, C.; Wei, C.; Zhu, Y.; Qian, W.; Qi, S.; Wu, Y.; Shi, Y.; Qin, K. Multi-element processed pyritum mixed to β-tricalcium phosphate to obtain a 3D-printed porous scaffold: An option for treatment of bone defects. Mater. Sci. Eng. C 2021, 128, 112326. [Google Scholar] [CrossRef]
  49. Barbur, I.; Opris, H.; Crisan, B.; Cuc, S.; Colosi, H.A.; Baciut, M.; Opris, D.; Prodan, D.; Moldovan, M.; Crisan, L. Statistical comparison of the mechanical properties of 3D-printed resin through triple-jetting technology and conventional PMMA in orthodontic occlusal splint manufacturing. Biomedicines 2023, 11, 2155. [Google Scholar] [CrossRef] [PubMed]
  50. Zalavras, C.G.; Marcus, R.E.; Levin, L.S.; Patzakis, M.J. Management of open fractures and subsequent complications. Instr. Course Lect. 2008, 57, 51–63. [Google Scholar] [CrossRef] [PubMed]
  51. Emori, T.G.; Gaynes, R.P. An overview of nosocomial infections, including the role of the microbiology laboratory. Clin. Microbiol. Rev. 1993, 6, 428–442. [Google Scholar] [CrossRef] [PubMed]
  52. Cooper, R.A. Surgical site infections: Epidemiology and microbiological aspects in trauma and orthopaedic surgery. Int. Wound. J. 2013, 10 (Suppl. S1), 3–8. [Google Scholar] [CrossRef]
  53. Grujović, Z.; Ilić, M.; Milicić, B. The level of microbial contamination and frequency of surgical site infections at the department of orthopedic and traumatologic surgery of the clinical hospital center in Kragujevac. Med. Pregl. 2005, 58, 287–291. [Google Scholar] [CrossRef] [PubMed]
  54. Hesselvig, A.B.; Arpi, M.; Madsen, F.; Bjarnsholt, T.; Odgaard, A. Does an Antimicrobial Incision Drape Prevent Intraoperative Contamination? A Randomized Controlled Trial of 1187 Patients. Clin. Orthop Relat. Res. 2020, 478, 1007–1015. [Google Scholar] [CrossRef] [PubMed]
  55. Veerachamy, S.; Yarlagadda, T.; Manivasagam, G.; Yarlagadda, P.K. Bacterial adherence and biofilm formation on medical implants: A review. Proc. Inst. Mech. Eng. H 2014, 228, 1083–1099. [Google Scholar] [CrossRef]
  56. Tapscott, D.C.; Wottowa, C. Orthopedic Implant Materials. In StatPearls; StatPearls Publishing: St. Petersburg, FL, USA, 2023. [Google Scholar]
  57. Ferraris, S.; Spriano, S. Antibacterial titanium surfaces for medical implants. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 61, 965–978. [Google Scholar] [CrossRef]
  58. Hotchkiss, K.M.; Reddy, G.B.; Hyzy, S.L.; Schwartz, Z.; Boyan, B.D.; Olivares-Navarrete, R. Titanium surface characteristics, including topography and wettability, alter macrophage activation. Acta Biomater. 2016, 31, 425–434. [Google Scholar] [CrossRef]
  59. Takeuchi, Y.; Tanaka, M.; Tanaka, J.; Kamimoto, A.; Furuchi, M.; Imai, H. Fabrication systems for restorations and fixed dental prostheses made of titanium and titanium alloys. J. Prosthodont. Res. 2019, 64, 1–5. [Google Scholar] [CrossRef]
  60. Patten, I.S.; Sun, Y.; Maldonado, D.R.; Lee, M.S.; Banffy, M.B. Microbial Colonization of Capsular Traction Sutures in Hip Arthroscopic Surgery. Orthop. J. Sports Med. 2023, 11, 23259671231166705. [Google Scholar] [CrossRef]
  61. Daeschlein, G.; Napp, M.; Assadian, O.; Bluhm, J.; Krueger, C.; von Podewils, S.; Gümbel, D.; Hinz, P.; Haase, H.; Dohmen, P.M.; et al. Influence of preoperative skin sealing with cyanoacrylate on microbial contamination of surgical wounds following trauma surgery: A prospective, blinded, controlled observational study. Int. J. Infect. Dis. 2014, 29, 274–278. [Google Scholar] [CrossRef] [PubMed]
  62. Balkissoon, R.; Nayfeh, T.; Adams, K.L.; Belkoff, S.M.; Riedel, S.; Mears, S.C. Microbial surface contamination after standard operating room cleaning practices following surgical treatment of infection. Orthopedics 2014, 37, e339–e344. [Google Scholar] [CrossRef] [PubMed]
  63. Mufarrih, S.H.; Qureshi, N.Q.; Rashid, R.H.; Ahmed, B.; Irfan, S.; Zubairi, A.J.; Noordin, S. Microbial Colonization of Pneumatic Tourniquets in the Orthopedic Operating Room. Cureus 2019, 11, e5308. [Google Scholar] [CrossRef] [PubMed]
  64. Boekel, P.; Blackshaw, R.; Van Bavel, D.; Riazi, A.; Hau, R. Sterile stockinette in orthopaedic surgery: A possible pathway for infection. ANZ J. Surg. 2012, 82, 838–843. [Google Scholar] [CrossRef] [PubMed]
  65. Schömig, F.; Perka, C.; Pumberger, M.; Ascherl, R. Implant contamination as a cause of surgical site infection in spinal surgery: Are single-use implants a reasonable solution?—A systematic review. BMC Musculoskelet. Disord. 2020, 21, 634. [Google Scholar] [CrossRef] [PubMed]
  66. Agarwal, A.; Lin, B.; Wang, J.C.; Schultz, C.; Garfin, S.R.; Goel, V.K.; Anand, N.; Agarwal, A.K. Efficacy of Intraoperative Implant Prophylaxis in Reducing Intraoperative Microbial Contamination. Glob. Spine J. 2019, 9, 62–66. [Google Scholar] [CrossRef] [PubMed]
  67. Kalt, F.; Schulthess, B.; Sidler, F.; Herren, S.; Fucentese, S.F.; Zingg, P.O.; Berli, M.; Zinkernagel, A.S.; Zbinden, R.; Achermann, Y. Corynebacterium Species Rarely Cause Orthopedic Infections. J. Clin. Microbiol. 2018, 56, 18. [Google Scholar] [CrossRef]
  68. Walsh, E.F.; Ben-David, D.; Ritter, M.; Mechrefe, A.; Mermel, L.A.; DiGiovanni, C. Microbial colonization of tourniquets used in orthopedic surgery. Orthopedics 2006, 29, 709–713. [Google Scholar] [CrossRef]
  69. Spencer, R.C. Bacillus anthracis. J. Clin. Pathol. 2003, 56, 182–187. [Google Scholar] [CrossRef]
  70. Brown, K.L. Control of bacterial spores. Br. Med. Bull. 2000, 56, 158–171. [Google Scholar] [CrossRef]
  71. Cote, C.K.; Heffron, J.D.; Bozue, J.A.; Welkos, S.L. Chapter 102—Bacillus anthracis and Other Bacillus Species. In Molecular Medical Microbiology, 2nd ed.; Tang, Y.-W., Sussman, M., Liu, D., Poxton, I., Schwartzman, J., Eds.; Academic Press: Boston, MA, USA, 2015; pp. 1789–1844. [Google Scholar] [CrossRef]
  72. Zhu, M.; Zhu, Q.; Yang, Z.; Liang, Z. Clinical Characteristics of Patients with Micrococcus luteus Bloodstream Infection in a Chinese Tertiary-Care Hospital. Pol. J. Microbiol. 2021, 70, 321–326. [Google Scholar] [CrossRef]
  73. Qureshi, N.Q.; Mufarrih, S.H.; Irfan, S.; Rashid, R.H.; Zubairi, A.J.; Sadruddin, A.; Ahmed, I.; Noordin, S. Mobile phones in the orthopedic operating room: Microbial colonization and antimicrobial resistance. World J. Orthop. 2020, 11, 252–264. [Google Scholar] [CrossRef] [PubMed]
  74. National Nosocomial Infections Surveillance (NNIS). National Nosocomial Infections Surveillance (NNIS) System Report, data summary from January 1992 through June 2004, issued October 2004. Am. J. Infect. Control 2004, 32, 470–485. [Google Scholar] [CrossRef]
  75. Misteli, H.; Widmer, A.F.; Rosenthal, R.; Oertli, D.; Marti, W.R.; Weber, W.P. Spectrum of pathogens in surgical site infections at a Swiss university hospital. Swiss Med. Wkly 2011, 140, w13146. [Google Scholar] [CrossRef] [PubMed]
  76. Berthelot, P.; Grattard, F.; Cazorla, C.; Passot, J.P.; Fayard, J.P.; Meley, R.; Bejuy, J.; Farizon, F.; Pozzetto, B.; Lucht, F. Is nasal carriage of Staphylococcus aureus the main acquisition pathway for surgical-site infection in orthopaedic surgery? Eur. J. Clin. Microbiol. Infect. Dis. 2010, 29, 373–382. [Google Scholar] [CrossRef] [PubMed]
  77. Wolcott, R.D.; Gontcharova, V.; Sun, Y.; Zischakau, A.; Dowd, S.E. Bacterial diversity in surgical site infections: Not just aerobic cocci any more. J. Wound. Care 2009, 18, 317–323. [Google Scholar] [CrossRef] [PubMed]
  78. Wexler, H.M. Bacteroides: The good, the bad, and the nitty-gritty. Clin. Microbiol. Rev. 2007, 20, 593–621. [Google Scholar] [CrossRef]
  79. Patrick, S. A tale of two habitats: Bacteroides fragilis, a lethal pathogen and resident in the human gastrointestinal microbiome. Microbiology 2022, 168, 1156. [Google Scholar] [CrossRef]
  80. Brook, I. Pathogenicity of the Bacteroides fragilis group. Ann. Clin. Lab. Sci. 1989, 19, 360–376. [Google Scholar] [PubMed]
  81. Ghotaslou, R.; Yekani, M.; Memar, M.Y. The role of efflux pumps in Bacteroides fragilis resistance to antibiotics. Microbiol. Res. 2018, 210, 1–5. [Google Scholar] [CrossRef] [PubMed]
  82. Martínez-Pastor, J.C.; Vilchez, F.; Pitart, C.; Sierra, J.M.; Soriano, A. Antibiotic resistance in orthopaedic surgery: Acute knee prosthetic joint infections due to extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae. Eur. J. Clin. Microbiol. Infect. Dis. 2010, 29, 1039–1041. [Google Scholar] [CrossRef] [PubMed]
  83. Navon-Venezia, S.; Kondratyeva, K.; Carattoli, A. Klebsiella pneumoniae: A major worldwide source and shuttle for antibiotic resistance. FEMS Microbiol. Rev. 2017, 41, 252–275. [Google Scholar] [CrossRef]
  84. Effah, C.Y.; Sun, T.; Liu, S.; Wu, Y. Klebsiella pneumoniae: An increasing threat to public health. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 1. [Google Scholar] [CrossRef]
  85. Surleac, M.; Czobor Barbu, I.; Paraschiv, S.; Popa, L.I.; Gheorghe, I.; Marutescu, L.; Popa, M.; Sarbu, I.; Talapan, D.; Nita, M.; et al. Whole genome sequencing snapshot of multi-drug resistant Klebsiella pneumoniae strains from hospitals and receiving wastewater treatment plants in Southern Romania. PLoS ONE 2020, 15, e0228079. [Google Scholar] [CrossRef]
  86. Turner, N.A.; Sharma-Kuinkel, B.K.; Maskarinec, S.A.; Eichenberger, E.M.; Shah, P.P.; Carugati, M.; Holland, T.L.; Fowler, V.G., Jr. Methicillin-resistant Staphylococcus aureus: An overview of basic and clinical research. Nat. Rev. Microbiol. 2019, 17, 203–218. [Google Scholar] [CrossRef]
  87. Silva-Santana, G.; Silva, C.M.F.; Olivella, J.G.B.; Silva, I.F.; Fernandes, L.M.O.; Sued-Karam, B.R.; Santos, C.S.; Souza, C.; Mattos-Guaraldi, A.L. Worldwide survey of Corynebacterium striatum increasingly associated with human invasive infections, nosocomial outbreak, and antimicrobial multidrug-resistance, 1976–2020. Arch. Microbiol. 2021, 203, 1863–1880. [Google Scholar] [CrossRef]
  88. Buu-Hoï, A.; Le Bouguénec, C.; Horaud, T. Genetic basis of antibiotic resistance in Aerococcus viridans. Antimicrob. Agents Chemother. 1989, 33, 529–534. [Google Scholar] [CrossRef] [PubMed]
  89. Knop, C.; Sitte, I.; Canto, F.; Reinhold, M.; Blauth, M. Successful posterior interlaminar fusion at the thoracic spine by sole use of beta-tricalcium phosphate. Arch. Orthop. Trauma Surg. 2006, 126, 204–210. [Google Scholar] [CrossRef] [PubMed]
  90. Damron, T.A. Use of 3D beta-tricalcium phosphate (Vitoss) scaffolds in repairing bone defects. Nanomedicene 2007, 2, 763–775. [Google Scholar] [CrossRef] [PubMed]
  91. Liu, B.; Lun, D.X. Current application of β-tricalcium phosphate composites in orthopaedics. Orthop. Surg. 2012, 4, 139–144. [Google Scholar] [CrossRef]
  92. Buser, Z.; Brodke, D.S.; Youssef, J.A.; Meisel, H.J.; Myhre, S.L.; Hashimoto, R.; Park, J.B.; Tim Yoon, S.; Wang, J.C. Synthetic bone graft versus autograft or allograft for spinal fusion: A systematic review. J. Neurosurg. Spine 2016, 25, 509–516. [Google Scholar] [CrossRef]
  93. Dragosloveanu, Ş.; Dragosloveanu, C.D.M.; Stanca, H.T.; Cotor, D.C.; Andrei, A.C.; Dragosloveanu, C.I.; Stoica, C.I. Tricalcium phosphate and hydroxyapatite treatment for benign cavitary bone lesions: A prospective clinical trial. Exp. Ther. Med. 2020, 20, 215. [Google Scholar] [CrossRef]
  94. Arbez, B.; Libouban, H. Behavior of macrophage and osteoblast cell lines in contact with the β-TCP biomaterial (beta-tricalcium phosphate). Morphologie 2017, 101, 154–163. [Google Scholar] [CrossRef]
  95. Rh Owen, G.; Dard, M.; Larjava, H. Hydoxyapatite/beta-tricalcium phosphate biphasic ceramics as regenerative material for the repair of complex bone defects. J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 2493–2512. [Google Scholar] [CrossRef]
  96. Lowe, B.; Ottensmeyer, M.P.; Xu, C.; He, Y.; Ye, Q.; Troulis, M.J. The Regenerative Applicability of Bioactive Glass and Beta-Tricalcium Phosphate in Bone Tissue Engineering: A Transformation Perspective. J. Funct. Biomater. 2019, 10, 10016. [Google Scholar] [CrossRef]
  97. Ebrahimi, M.; Botelho, M.G.; Dorozhkin, S.V. Biphasic calcium phosphates bioceramics (HA/TCP): Concept, physicochemical properties and the impact of standardization of study protocols in biomaterials research. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 71, 1293–1312. [Google Scholar] [CrossRef]
  98. Kim, S.E.; Park, K. Recent Advances of Biphasic Calcium Phosphate Bioceramics for Bone Tissue Regeneration. Adv. Exp. Med. Biol. 2020, 1250, 177–188. [Google Scholar] [CrossRef] [PubMed]
  99. Re, F.; Borsani, E.; Rezzani, R.; Sartore, L.; Russo, D. Bone Regeneration Using Mesenchymal Stromal Cells and Biocompatible Scaffolds: A Concise Review of the Current Clinical Trials. Gels 2023, 9, 389. [Google Scholar] [CrossRef]
  100. Zhou, H.; Lee, J. Nanoscale hydroxyapatite particles for bone tissue engineering. Acta Biomater. 2011, 7, 2769–2781. [Google Scholar] [CrossRef]
  101. Siddiqui, H.A.; Pickering, K.L.; Mucalo, M.R. A Review on the Use of Hydroxyapatite-Carbonaceous Structure Composites in Bone Replacement Materials for Strengthening Purposes. Materials 2018, 11, 1813. [Google Scholar] [CrossRef]
  102. Mosaad, K.E.; Shoueir, K.R.; Saied, A.H.; Dewidar, M.M. New Prospects in Nano Phased Co-substituted Hydroxyapatite Enrolled in Polymeric Nanofiber Mats for Bone Tissue Engineering Applications. Ann. Biomed. Eng. 2021, 49, 2006–2029. [Google Scholar] [CrossRef] [PubMed]
  103. Ishikawa, K.; Hayashi, K. Carbonate apatite artificial bone. Sci. Technol. Adv. Mater. 2021, 22, 683–694. [Google Scholar] [CrossRef]
  104. Costa-Pinto, A.R.; Lemos, A.L.; Tavaria, F.K.; Pintado, M. Chitosan and Hydroxyapatite Based Biomaterials to Circumvent Periprosthetic Joint Infections. Materials 2021, 14, 804. [Google Scholar] [CrossRef]
  105. El-Kady, A.M.; Farag, M.M.; El-Rashedi, A.M. Bioactive glass nanoparticles designed for multiple deliveries of lithium ions and drugs: Curative and restorative bone treatment. Eur. J. Pharm. Sci. 2016, 91, 243–250. [Google Scholar] [CrossRef] [PubMed]
  106. Kraipok, A.; Mamanee, T.; Ruangsuriya, J.; Nawarat, P.; Leenakul, W. Phase Formation, Mechanical Strength, and Bioactive Properties of Lithium Disilicate Glass-Ceramics with Different Al2O3 Contents. Materials 2022, 15, 8283. [Google Scholar] [CrossRef]
  107. Vadapalli, S.; Sairyo, K.; Goel, V.K.; Robon, M.; Biyani, A.; Khandha, A.; Ebraheim, N.A. Biomechanical rationale for using polyetheretherketone (PEEK) spacers for lumbar interbody fusion-A finite element study. Spine 2006, 31, E992–E998. [Google Scholar] [CrossRef]
  108. Mastronardi, L.; Ducati, A.; Ferrante, L. Anterior cervical fusion with polyetheretherketone (PEEK) cages in the treatment of degenerative disc disease. Preliminary observations in 36 consecutive cases with a minimum 12-month follow-up. Acta Neurochir. 2006, 148, 307–312; discussion 312. [Google Scholar] [CrossRef] [PubMed]
  109. Topuz, K.; Colak, A.; Kaya, S.; Simşek, H.; Kutlay, M.; Demircan, M.N.; Velioğlu, M. Two-level contiguous cervical disc disease treated with peek cages packed with demineralized bone matrix: Results of 3-year follow-up. Eur. Spine J. 2009, 18, 238–243. [Google Scholar] [CrossRef] [PubMed]
  110. Najeeb, S.; Zafar, M.S.; Khurshid, Z.; Siddiqui, F. Applications of polyetheretherketone (PEEK) in oral implantology and prosthodontics. J. Prosthodont. Res. 2016, 60, 12–19. [Google Scholar] [CrossRef]
  111. Stratton-Powell, A.A.; Pasko, K.M.; Brockett, C.L.; Tipper, J.L. The Biologic Response to Polyetheretherketone (PEEK) Wear Particles in Total Joint Replacement: A Systematic Review. Clin. Orthop. Relat. Res. 2016, 474, 2394–2404. [Google Scholar] [CrossRef] [PubMed]
  112. Deng, Y.; Tang, W.; Li, Z. Repairing a Facial Cleft by Polyether-Ether-Ketone Implant Combined with Titanium Mesh. J. Craniofac. Surg. 2018, 29, e582–e585. [Google Scholar] [CrossRef] [PubMed]
  113. Papathanasiou, I.; Kamposiora, P.; Papavasiliou, G.; Ferrari, M. The use of PEEK in digital prosthodontics: A narrative review. BMC Oral. Health 2020, 20, 217. [Google Scholar] [CrossRef] [PubMed]
  114. Saracco, M.; Fulchignoni, C.; Velluto, C.; Rocchi, L. Safety and reliability of carbon-peek plate for the treatment of distal radius fractures: A review of the literature. Orthop. Rev. 2021, 13, 28362. [Google Scholar] [CrossRef]
  115. Mozaffari, K.; Rana, S.; Chow, A.; Mahgerefteh, N.; Duong, C.; Sheppard, J.P.; Phillips, H.W.; Jarrahy, R.; Yang, I. Customized polyetheretherketone (PEEK) implants are associated with similar hospital length of stay compared to autologous bone used in cranioplasty procedures. J. Neurol. Sci. 2022, 434, 120169. [Google Scholar] [CrossRef]
  116. Luo, Y.; Dolder, C.K.; Walker, J.M.; Mishra, R.; Dean, D.; Becker, M.L. Synthesis and Biological Evaluation of Well-Defined Poly(propylene fumarate) Oligomers and Their Use in 3D Printed Scaffolds. Biomacromolecules 2016, 17, 690–697. [Google Scholar] [CrossRef]
  117. Cai, Z.; Wan, Y.; Becker, M.L.; Long, Y.Z.; Dean, D. Poly(propylene fumarate)-based materials: Synthesis, functionalization, properties, device fabrication and biomedical applications. Biomaterials 2019, 208, 45–71. [Google Scholar] [CrossRef]
  118. Kleinfehn, A.P.; Lammel Lindemann, J.A.; Razvi, A.; Philip, P.; Richardson, K.; Nettleton, K.; Becker, M.L.; Dean, D. Modulating Bioglass Concentration in 3D Printed Poly(propylene fumarate) Scaffolds for Post-Printing Functionalization with Bioactive Functional Groups. Biomacromolecules 2019, 20, 4345–4352. [Google Scholar] [CrossRef]
  119. Liu, X.; George, M.N.; Park, S.; Miller Ii, A.L.; Gaihre, B.; Li, L.; Waletzki, B.E.; Terzic, A.; Yaszemski, M.J.; Lu, L. 3D-printed scaffolds with carbon nanotubes for bone tissue engineering: Fast and homogeneous one-step functionalization. Acta Biomater. 2020, 111, 129–140. [Google Scholar] [CrossRef]
  120. van Bochove, B.; Hannink, G.; Buma, P.; Grijpma, D.W. Preparation of Designed Poly(trimethylene carbonate) Meniscus Implants by Stereolithography: Challenges in Stereolithography. Macromol. Biosci. 2016, 16, 1853–1863. [Google Scholar] [CrossRef] [PubMed]
  121. Guerra, A.J.; Lara-Padilla, H.; Becker, M.L.; Rodriguez, C.A.; Dean, D. Photopolymerizable Resins for 3D-Printing Solid-Cured Tissue Engineered Implants. Curr. Drug. Targets 2019, 20, 823–838. [Google Scholar] [CrossRef] [PubMed]
  122. Brossier, T.; Volpi, G.; Vasquez-Villegas, J.; Petitjean, N.; Guillaume, O.; Lapinte, V.; Blanquer, S. Photoprintable Gelatin-graft-Poly(trimethylene carbonate) by Stereolithography for Tissue Engineering Applications. Biomacromolecules 2021, 22, 3873–3883. [Google Scholar] [CrossRef] [PubMed]
  123. Manicone, P.F.; Rossi Iommetti, P.; Raffaelli, L. An overview of zirconia ceramics: Basic properties and clinical applications. J. Dent. 2007, 35, 819–826. [Google Scholar] [CrossRef] [PubMed]
  124. Piconi, C.; Sprio, S. Oxide Bioceramic Composites in Orthopedics and Dentistry. J. Compos. Sci. 2021, 5, 206. [Google Scholar] [CrossRef]
  125. Chaair, H.; Labjar, H.; Britel, O. Synthesis of β-tricalcium phosphate. Morphologie 2017, 101, 120–124. [Google Scholar] [CrossRef] [PubMed]
  126. Bohner, M.; Santoni, B.L.G.; Döbelin, N. β-tricalcium phosphate for bone substitution: Synthesis and properties. Acta Biomater. 2020, 113, 23–41. [Google Scholar] [CrossRef]
  127. Zhang, H.; Zhang, H.; Xiong, Y.; Dong, L.; Li, X. Development of hierarchical porous bioceramic scaffolds with controlled micro/nano surface topography for accelerating bone regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 130, 112437. [Google Scholar] [CrossRef]
  128. Qi, D.; Su, J.; Li, S.; Zhu, H.; Cheng, L.; Hua, S.; Yuan, X.; Jiang, J.; Shu, Z.; Shi, Y. 3D printed magnesium-doped β-TCP gyroid scaffold with osteogenesis, angiogenesis, immunomodulation properties and bone regeneration capability in vivo. Biomater. Adv. 2022, 136, 212759. [Google Scholar] [CrossRef]
  129. Miyai, T.; Ito, A.; Tamazawa, G.; Matsuno, T.; Sogo, Y.; Nakamura, C.; Yamazaki, A.; Satoh, T. Antibiotic-loaded poly-epsilon-caprolactone and porous beta-tricalcium phosphate composite for treating osteomyelitis. Biomaterials 2008, 29, 350–358. [Google Scholar] [CrossRef]
  130. Nakhaee, F.M.; Rajabi, M.; Bakhsheshi-Rad, H.R. In-vitro assessment of β-tricalcium phosphate/bredigite-ciprofloxacin (CPFX) scaffolds for bone treatment applications. Biomed. Mater. 2021, 16, 590. [Google Scholar] [CrossRef]
  131. Li, Q.L.; Wu, Y.X.; Zhang, Y.X.; Mao, J.; Zhang, Z.X. Enhancing osteogenic differentiation of MC3T3-E1 cells during inflammation using UPPE/β-TCP/TTC composites via the Wnt/β-catenin pathway. RSC Adv. 2024, 14, 1527–1537. [Google Scholar] [CrossRef]
  132. Makarov, C.; Berdicevsky, I.; Raz-Pasteur, A.; Gotman, I. In vitro antimicrobial activity of vancomycin-eluting bioresorbable β-TCP-polylactic acid nanocomposite material for load-bearing bone repair. J. Mater. Sci. Mater. Med. 2013, 24, 679–687. [Google Scholar] [CrossRef]
  133. Qiu, X.; Li, S.; Li, X.; Xiao, Y.; Li, S.; Fen, Q.; Kang, X.; Zhen, P. Experimental study of β-TCP scaffold loaded with VAN/PLGA microspheres in the treatment of infectious bone defects. Colloids Surf. B Biointerfaces 2022, 213, 112424. [Google Scholar] [CrossRef] [PubMed]
  134. Manchón, A.; Alkhraisat, M.H.; Rueda-Rodriguez, C.; Pintado, C.; Prados-Frutos, J.C.; Torres, J.; Lopez Cabarcos, E. Silicon bioceramic loaded with vancomycin stimulates bone tissue regeneration. J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 2307–2315. [Google Scholar] [CrossRef] [PubMed]
  135. Prat-Poiret, N.; Langlais, F.; Bonnaure, M.; Cormier, M.; Lancien, G. Tricalcium phosphate and gentamycin. In vitro and in vivo antibiotic diffusion, rehabilitation in bone site in sheep. Chirurgie 1996, 121, 298–308. [Google Scholar] [PubMed]
  136. Fadeeva, I.V.; Goldberg, M.A.; Preobrazhensky, I.I.; Mamin, G.V.; Davidova, G.A.; Agafonova, N.V.; Fosca, M.; Russo, F.; Barinov, S.M.; Cavalu, S.; et al. Improved cytocompatibility and antibacterial properties of zinc-substituted brushite bone cement based on β-tricalcium phosphate. J. Mater. Sci. Mater. Med. 2021, 32, 99. [Google Scholar] [CrossRef] [PubMed]
  137. Rau, J.V.; Fosca, M.; Fadeeva, I.V.; Kalay, S.; Culha, M.; Raucci, M.G.; Fasolino, I.; Ambrosio, L.; Antoniac, I.V.; Uskoković, V. Tricalcium phosphate cement supplemented with boron nitride nanotubes with enhanced biological properties. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 114, 111044. [Google Scholar] [CrossRef] [PubMed]
  138. Uskoković, V.; Graziani, V.; Wu, V.M.; Fadeeva, I.V.; Fomin, A.S.; Presniakov, I.A.; Fosca, M.; Ortenzi, M.; Caminiti, R.; Rau, J.V. Gold is for the mistress, silver for the maid: Enhanced mechanical properties, osteoinduction and antibacterial activity due to iron doping of tricalcium phosphate bone cements. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 94, 798–810. [Google Scholar] [CrossRef]
  139. Yuan, J.; Wang, B.; Han, C.; Huang, X.; Xiao, H.; Lu, X.; Lu, J.; Zhang, D.; Xue, F.; Xie, Y. Nanosized-Ag-doped porous β-tricalcium phosphate for biological applications. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 114, 111037. [Google Scholar] [CrossRef]
  140. Nayak, V.V.; Tovar, N.; Hacquebord, J.H.; Duarte, S.; Panariello, B.H.D.; Tonon, C.; Atria, P.J.; Coelho, P.G.; Witek, L. Physiochemical and bactericidal activity evaluation: Silver-augmented 3D-printed scaffolds-An in vitro study. J. Biomed. Mater. Res. B Appl. Biomater. 2022, 110, 195–209. [Google Scholar] [CrossRef]
  141. Makvandi, P.; Ali, G.W.; Della Sala, F.; Abdel-Fattah, W.I.; Borzacchiello, A. Hyaluronic acid/corn silk extract based injectable nanocomposite: A biomimetic antibacterial scaffold for bone tissue regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 107, 110195. [Google Scholar] [CrossRef]
  142. Tai, H.Y.; Chou, S.H.; Cheng, L.P.; Yu, H.T.; Don, T.M. Asymmetric composite membranes from chitosan and tricalcium phosphate useful for guided bone regeneration. J. Biomater. Sci. Polym. Ed. 2012, 23, 1153–1170. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, Y.H.; Tai, H.Y.; Fu, E.; Don, T.M. Guided bone regeneration activity of different calcium phosphate/chitosan hybrid membranes. Int. J. Biol. Macromol. 2019, 126, 159–169. [Google Scholar] [CrossRef] [PubMed]
  144. Spirandeli, B.R.; Ribas, R.G.; Amaral, S.S.; Martins, E.F.; Esposito, E.; Vasconcellos, L.M.R.; Campos, T.M.B.; Thim, G.P.; Trichês, E.S. Incorporation of 45S5 bioglass via sol-gel in β-TCP scaffolds: Bioactivity and antimicrobial activity evaluation. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 131, 112453. [Google Scholar] [CrossRef] [PubMed]
  145. Amaral, S.S.; Lima, B.S.d.S.; Avelino, S.O.M.; Spirandeli, B.R.; Campos, T.M.B.; Thim, G.P.; Trichês, E.d.S.; Prado, R.F.d.; Vasconcellos, L.M.R.d. β-TCP/S53P4 Scaffolds Obtained by Gel Casting: Synthesis, Properties, and Biomedical Applications. Bioengineering 2023, 10, 597. [Google Scholar] [CrossRef] [PubMed]
  146. Alves, A.P.N.; Arango-Ospina, M.; Oliveira, R.; Ferreira, I.M.; de Moraes, E.G.; Hartmann, M.; de Oliveira, A.P.N.; Boccaccini, A.R.; de Sousa Trichês, E. 3D-printed β-TCP/S53P4 bioactive glass scaffolds coated with tea tree oil: Coating optimization, in vitro bioactivity and antibacterial properties. J. Biomed. Mater. Res. B Appl. Biomater. 2023, 111, 881–894. [Google Scholar] [CrossRef]
  147. Qian, Y.; Zhou, X.; Sun, H.; Yang, J.; Chen, Y.; Li, C.; Wang, H.; Xing, T.; Zhang, F.; Gu, N. Biomimetic Domain-Active Electrospun Scaffolds Facilitating Bone Regeneration Synergistically with Antibacterial Efficacy for Bone Defects. ACS Appl. Mater. Interfaces 2018, 10, 3248–3259. [Google Scholar] [CrossRef] [PubMed]
  148. De Oliveira, J.F.; De Aguiar, P.F.; Rossi, A.M.; Soares, G.A. Effect of process parameters on the characteristics of porous calcium phosphate ceramics for bone tissue scaffolds. Artif. Organs 2003, 27, 406–411. [Google Scholar] [CrossRef]
  149. Kato, E.; Lemler, J.; Sakurai, K.; Yamada, M. Biodegradation property of beta-tricalcium phosphate-collagen composite in accordance with bone formation: A comparative study with Bio-Oss Collagen® in a rat critical-size defect model. Clin. Implant. Dent. Relat. Res. 2014, 16, 202–211. [Google Scholar] [CrossRef]
  150. Wang, Y.; Wang, K.; Li, X.; Wei, Q.; Chai, W.; Wang, S.; Che, Y.; Lu, T.; Zhang, B. 3D fabrication and characterization of phosphoric acid scaffold with a HA/β-TCP weight ratio of 60:40 for bone tissue engineering applications. PLoS ONE 2017, 12, e0174870. [Google Scholar] [CrossRef]
  151. Su, J.; Hua, S.; Chen, A.; Chen, P.; Yang, L.; Yuan, X.; Qi, D.; Zhu, H.; Yan, C.; Xiao, J. Three-dimensional printing of gyroid-structured composite bioceramic scaffolds with tuneable degradability. Biomater. Adv. 2022, 133, 112595. [Google Scholar] [CrossRef] [PubMed]
  152. Iooss, P.; Le Ray, A.M.; Grimandi, G.; Daculsi, G.; Merle, C. A new injectable bone substitute combining poly(epsilon-caprolactone) microparticles with biphasic calcium phosphate granules. Biomaterials 2001, 22, 2785–2794. [Google Scholar] [CrossRef] [PubMed]
  153. Menotti, F.; Scutera, S.; Coppola, B.; Longo, F.; Mandras, N.; Cavallo, L.; Comini, S.; Sparti, R.; Fiume, E.; Cuffini, A.M.; et al. Tuning of Silver Content on the Antibacterial and Biological Properties of Poly(ɛ-caprolactone)/Biphasic Calcium Phosphate 3D-Scaffolds for Bone Tissue Engineering. Polymers 2023, 15, 3618. [Google Scholar] [CrossRef] [PubMed]
  154. Comini, S.; Sparti, R.; Coppola, B.; Mohammadi, M.; Scutera, S.; Menotti, F.; Banche, G.; Cuffini, A.M.; Palmero, P.; Allizond, V. Novel Silver-Functionalized Poly(ε-Caprolactone)/Biphasic Calcium Phosphate Scaffolds Designed to Counteract Post-Surgical Infections in Orthopedic Applications. Int. J. Mol. Sci. 2021, 22, 10176. [Google Scholar] [CrossRef]
  155. Sukpaita, T.; Chirachanchai, S.; Chanamuangkon, T.; Nampuksa, K.; Monmaturapoj, N.; Sumrejkanchanakij, P.; Pimkhaokham, A.; Ampornaramveth, R.S. Novel Epigenetic Modulation Chitosan-Based Scaffold as a Promising Bone Regenerative Material. Cells 2022, 11, 3217. [Google Scholar] [CrossRef]
  156. Marriott, I. Apoptosis-associated uncoupling of bone formation and resorption in osteomyelitis. Front. Cell Infect. Microbiol. 2013, 3, 101. [Google Scholar] [CrossRef]
  157. Josse, J.; Velard, F.; Gangloff, S.C. Staphylococcus aureus vs. Osteoblast: Relationship and Consequences in Osteomyelitis. Front. Cell Infect. Microbiol. 2015, 5, 85. [Google Scholar] [CrossRef]
  158. Mouton, W.; Josse, J.; Jacqueline, C.; Abad, L.; Trouillet-Assant, S.; Caillon, J.; Bouvard, D.; Bouchet, M.; Laurent, F.; Diot, A. Staphylococcus aureus internalization impairs osteoblastic activity and early differentiation process. Sci. Rep. 2021, 11, 17685. [Google Scholar] [CrossRef] [PubMed]
  159. Zhang, X.; Man, K.-W.; Li, G.H.-Y.; Tan, K.C.; Kung, A.W.-C.; Cheung, C.-L. Osteoporosis is a novel risk factor of infections and sepsis: A cohort study. eClinicalMedicine 2022, 49, 101488. [Google Scholar] [CrossRef] [PubMed]
  160. Boskey, A.L.; Posner, A.S. Bone structure, composition, and mineralization. Orthop. Clin. N. Am. 1984, 15, 597–612. [Google Scholar] [CrossRef]
  161. Mondal, S.; Park, S.; Choi, J.; Vu, T.T.H.; Doan, V.H.M.; Vo, T.T.; Lee, B.; Oh, J. Hydroxyapatite: A journey from biomaterials to advanced functional materials. Adv. Colloid. Interface Sci. 2023, 321, 103013. [Google Scholar] [CrossRef]
  162. Radulescu, D.E.; Vasile, O.R.; Andronescu, E.; Ficai, A. Latest Research of Doped Hydroxyapatite for Bone Tissue Engineering. Int. J. Mol. Sci. 2023, 24, 13157. [Google Scholar] [CrossRef]
  163. Hughes, J.M.; Cameron, M.; Crowley, K.D. Structural variations in natural F, OH, and Cl apatites. Am. Mineral. 1989, 74, 870–876. [Google Scholar]
  164. Liu, J.-W.; Li, L.; Li, S.-R.; Santosh, M.; Yuan, M.-W. Apatite as a fingerprint of granite fertility and gold mineralization: Evidence from the Xiaoqinling Goldfield, North China Craton. Ore Geol. Rev. 2022, 142, 104720. [Google Scholar] [CrossRef]
  165. Liu, J.-W.; Li, L.; Li, S.-R.; Santosh, M.; Yuan, M.-W.; Alam, M.; Yan, S.-F. Sulphur in apatite: A potential monitor of the magmatic redox state in the world-class gold fields of the North China Craton. Int. Geol. Rev. 2024, 66, 1–19. [Google Scholar] [CrossRef]
  166. Voudouris, P.; Mavrogonatos, C.; Graham, I.; Giuliani, G.; Melfos, V.; Karampelas, S.; Karantoni, V.; Wang, K.; Tarantola, A.; Zaw, K.; et al. Gem Corundum Deposits of Greece: Geology, Mineralogy and Genesis. Minerals 2019, 9, 49. [Google Scholar] [CrossRef]
  167. Melfos, V.; Voudouris, P.; Melfou, M.; Sánchez, M.G.; Papadopoulou, L.; Filippidis, A.; Spry, P.G.; Schaarschmidt, A.; Klemd, R.; Haase, K.M.; et al. Mineralogical Constraints on the Potassic and Sodic-Calcic Hydrothermal Alteration and Vein-Type Mineralization of the Maronia Porphyry Cu-Mo ± Re ± Au Deposit in NE Greece. Minerals 2020, 10, 182. [Google Scholar] [CrossRef]
  168. Baziotis, I.; Xydous, S.; Asimow, P.D.; Mavrogonatos, C.; Flemetakis, S.; Klemme, S.; Berndt, J. The potential of phosphorus in clinopyroxene as a geospeedometer: Examples from mantle xenoliths. Geochim. Cosmochim. Acta 2019, 266, 307–331. [Google Scholar] [CrossRef]
  169. Hubbe, U.; Beiser, S.; Kuhn, S.; Stark, T.; Hoess, A.; Cristina-Schmitz, H.; Vasilikos, I.; Metzger, M.C.; Rothweiler, R. A fully ingrowing implant for cranial reconstruction: Results in critical size defects in sheep using 3D-printed titanium scaffold. Biomater. Adv. 2022, 136, 212754. [Google Scholar] [CrossRef] [PubMed]
  170. Morimoto, T.; Hirata, H.; Eto, S.; Hashimoto, A.; Kii, S.; Kobayashi, T.; Tsukamoto, M.; Yoshihara, T.; Toda, Y.; Mawatari, M. Development of Silver-Containing Hydroxyapatite-Coated Antimicrobial Implants for Orthopaedic and Spinal Surgery. Medicina 2022, 58, 519. [Google Scholar] [CrossRef] [PubMed]
  171. Martinez, J.S.; Peterson, S.; Hoel, C.A.; Erno, D.J.; Murray, T.; Boyd, L.; Her, J.H.; McLean, N.; Davis, R.; Ginty, F.; et al. High resolution DLP stereolithography to fabricate biocompatible hydroxyapatite structures that support osteogenesis. PLoS ONE 2022, 17, e0272283. [Google Scholar] [CrossRef] [PubMed]
  172. Mollaei, M.; Varshosaz, J. Preparation and characterization of hydroxyapatite nanoparticles doped with nickel, tin, and molybdate ions for their antimicrobial effects. Drug Dev. Ind. Pharm. 2023, 49, 168–178. [Google Scholar] [CrossRef]
  173. de Lima, C.O.; de Oliveira, A.L.M.; Chantelle, L.; Silva Filho, E.C.; Jaber, M.; Fonseca, M.G. Zn-doped mesoporous hydroxyapatites and their antimicrobial properties. Colloids Surf. B Biointerfaces 2021, 198, 111471. [Google Scholar] [CrossRef]
  174. Padilla-Gainza, V.; Rodríguez-Tobías, H.; Morales, G.; Ledezma-Pérez, A.; Alvarado-Canché, C.; Loera-Valencia, R.; Rodríguez, C.; Gilkerson, R.; De Leo, C.T.; Lozano, K. Development of zinc oxide/hydroxyapatite/poly(D,L-lactic acid) fibrous scaffold for tissue engineering applications. Biomater. Adv. 2022, 133, 112594. [Google Scholar] [CrossRef]
  175. Ajduković, Z.R.; Mihajilov-Krstev, T.M.; Ignjatović, N.L.; Stojanović, Z.; Mladenović-Antić, S.B.; Kocić, B.D.; Najman, S.; Petrović, N.D.; Uskoković, D.P. In Vitro Evaluation of Nanoscale Hydroxyapatite-Based Bone Reconstructive Materials with Antimicrobial Properties. J. Nanosci. Nanotechnol. 2016, 16, 1420–1428. [Google Scholar] [CrossRef]
  176. Shi, F.; Liu, Y.; Zhi, W.; Xiao, D.; Li, H.; Duan, K.; Qu, S.; Weng, J. The synergistic effect of micro/nano-structured and Cu(2+)-doped hydroxyapatite particles to promote osteoblast viability and antibacterial activity. Biomed. Mater. 2017, 12, 035006. [Google Scholar] [CrossRef]
  177. Acharjee, D.; Mandal, S.; Samanta, S.K.; Roy, M.; Kundu, B.; Roy, S.; Basak, P.; Nandi, S.K. In Vitro and In Vivo Bone Regeneration Assessment of Titanium-Doped Waste Eggshell-Derived Hydroxyapatite in the Animal Model. ACS Biomater. Sci. Eng. 2023, 9, 4673–4685. [Google Scholar] [CrossRef]
  178. Yahia, I.S.; Shkir, M.; AlFaify, S.; Ganesh, V.; Zahran, H.Y.; Kilany, M. Facile microwave-assisted synthesis of Te-doped hydroxyapatite nanorods and nanosheets and their characterizations for bone cement applications. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 72, 472–480. [Google Scholar] [CrossRef] [PubMed]
  179. Predoi, D.; Iconaru, S.L.; Predoi, M.V.; Stan, G.E.; Buton, N. Synthesis, Characterization, and Antimicrobial Activity of Magnesium-Doped Hydroxyapatite Suspensions. Nanomaterial 2019, 9, 1295. [Google Scholar] [CrossRef] [PubMed]
  180. Dragosloveanu, Ş.; Cotor, D.C.; Dragosloveanu, C.D.M.; Stoian, C.; Stoica, C.I. Preclinical study analysis of massive magnesium alloy graft for calcaneal fractures. Exp. Ther. Med. 2021, 22, 731. [Google Scholar] [CrossRef] [PubMed]
  181. 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]
  182. Sofi, H.S.; Akram, T.; Shabir, N.; Vasita, R.; Jadhav, A.H.; Sheikh, F.A. Regenerated cellulose nanofibers from cellulose acetate: Incorporating hydroxyapatite (HAp) and silver (Ag) nanoparticles (NPs), as a scaffold for tissue engineering applications. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 118, 111547. [Google Scholar] [CrossRef] [PubMed]
  183. Shokri, M.; Kharaziha, M.; Tafti, H.A.; Eslaminejad, M.B.; Aghdam, R.M. Synergic role of zinc and gallium doping in hydroxyapatite nanoparticles to improve osteogenesis and antibacterial activity. Biomater. Adv. 2022, 134, 112684. [Google Scholar] [CrossRef] [PubMed]
  184. George, S.M.; Nayak, C.; Singh, I.; Balani, K. Multifunctional Hydroxyapatite Composites for Orthopedic Applications: A Review. ACS Biomater. Sci. Eng. 2022, 8, 3162–3186. [Google Scholar] [CrossRef] [PubMed]
  185. Padmanabhan, V.P.; Kulandaivelu, R.; Panneer, D.S.; Vivekananthan, S.; Sagadevan, S.; Lett, J.A. Surfactant Assisted Hydroxyapatite Nanoparticles: Drug Loading and In Vitro Leaching Kinetics and Antimicrobial Properties. J. Nanosci. Nanotechnol. 2019, 19, 7198–7204. [Google Scholar] [CrossRef]
  186. Ullah, I.; Hussain, Z.; Ullah, S.; Zahra, Q.U.A.; Zhang, Y.; Mehmood, S.; Liu, X.; Kamya, E.; Waseem Ghani, M.; Mansoorianfar, M.; et al. An osteogenic, antibacterial, and anti-inflammatory nanocomposite hydrogel platform to accelerate bone reconstruction. J. Mater. Chem. B 2023, 11, 5830–5845. [Google Scholar] [CrossRef]
  187. Mututuvari, T.M.; Harkins, A.L.; Tran, C.D. Facile synthesis, characterization, and antimicrobial activity of cellulose-chitosan-hydroxyapatite composite material: A potential material for bone tissue engineering. J. Biomed. Mater. Res. A 2013, 101, 3266–3277. [Google Scholar] [CrossRef]
  188. Ignjatović, N.; Wu, V.; Ajduković, Z.; Mihajilov-Krstev, T.; Uskoković, V.; Uskoković, D. Chitosan-PLGA polymer blends as coatings for hydroxyapatite nanoparticles and their effect on antimicrobial properties, osteoconductivity and regeneration of osseous tissues. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 60, 357–364. [Google Scholar] [CrossRef]
  189. Palierse, E.; Hélary, C.; Krafft, J.M.; Génois, I.; Masse, S.; Laurent, G.; Alvarez Echazu, M.I.; Selmane, M.; Casale, S.; Valentin, L.; et al. Baicalein-modified hydroxyapatite nanoparticles and coatings with antibacterial and antioxidant properties. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 118, 111537. [Google Scholar] [CrossRef]
  190. Hu, Z.; Guan, Y.; Hu, W.; Xu, Z.; Ishfaq, M. An overview of pharmacological activities of baicalin and its aglycone baicalein: New insights into molecular mechanisms and signaling pathways. Iran. J. Basic Med. Sci. 2022, 25, 14–26. [Google Scholar] [CrossRef]
  191. Sobczak-Kupiec, A.; Malina, D.; Piatkowski, M.; Krupa-Zuczek, K.; Wzorek, Z.; Tyliszczak, B. Physicochemical and biological properties of hydrogel/gelatin/hydroxyapatite PAA/G/HAp/AgNPs composites modified with silver nanoparticles. J. Nanosci. Nanotechnol. 2012, 12, 9302–9311. [Google Scholar] [CrossRef]
  192. Shi, P.; Wang, Q.; Yu, C.; Fan, F.; Liu, M.; Tu, M.; Lu, W.; Du, M. Hydroxyapatite nanorod and microsphere functionalized with bioactive lactoferrin as a new biomaterial for enhancement bone regeneration. Colloids Surf. B Biointerfaces 2017, 155, 477–486. [Google Scholar] [CrossRef]
  193. Kowalczyk, P.; Kaczyńska, K.; Kleczkowska, P.; Bukowska-Ośko, I.; Kramkowski, K.; Sulejczak, D. The Lactoferrin Phenomenon-A Miracle Molecule. Molecules 2022, 27, 2941. [Google Scholar] [CrossRef]
  194. Sikkema, R.; Keohan, B.; Zhitomirsky, I. Alginic Acid Polymer-Hydroxyapatite Composites for Bone Tissue Engineering. Polymers 2021, 13, 3070. [Google Scholar] [CrossRef] [PubMed]
  195. Zheng, Z.; Liu, P.; Zhang, X.; Jingguo, X.; Yongjie, W.; Zou, X.; Mei, X.; Zhang, S.; Zhang, S. Strategies to improve bioactive and antibacterial properties of polyetheretherketone (PEEK) for use as orthopedic implants. Mater. Today Bio 2022, 16, 100402. [Google Scholar] [CrossRef] [PubMed]
  196. Parthasarathy, J. 3D modeling, custom implants and its future perspectives in craniofacial surgery. Ann. Maxillofac. Surg. 2014, 4, 9–18. [Google Scholar] [CrossRef] [PubMed]
  197. Punchak, M.; Chung, L.K.; Lagman, C.; Bui, T.T.; Lazareff, J.; Rezzadeh, K.; Jarrahy, R.; Yang, I. Outcomes following polyetheretherketone (PEEK) cranioplasty: Systematic review and meta-analysis. J. Clin. Neurosci. 2017, 41, 30–35. [Google Scholar] [CrossRef] [PubMed]
  198. Sharma, N.; Aghlmandi, S.; Cao, S.; Kunz, C.; Honigmann, P.; Thieringer, F.M. Quality Characteristics and Clinical Relevance of In-House 3D-Printed Customized Polyetheretherketone (PEEK) Implants for Craniofacial Reconstruction. J. Clin. Med. 2020, 9, 2818. [Google Scholar] [CrossRef]
  199. Zhao, L.; Patel, P.K.; Cohen, M. Application of virtual surgical planning with computer assisted design and manufacturing technology to cranio-maxillofacial surgery. Arch. Plast. Surg. 2012, 39, 309–316. [Google Scholar] [CrossRef]
  200. Dragosloveanu, S.; Petre, M.-A.; Gherghe, M.E.; Nedelea, D.-G.; Scheau, C.; Cergan, R. Overall Accuracy of Radiological Digital Planning for Total Hip Arthroplasty in a Specialized Orthopaedics Hospital. J. Clin. Med. 2023, 12, 4503. [Google Scholar] [CrossRef]
  201. Kurtz, S.M.; Devine, J.N. PEEK biomaterials in trauma, orthopedic, and spinal implants. Biomaterials 2007, 28, 4845–4869. [Google Scholar] [CrossRef]
  202. Noiset, O.; Schneider, Y.J.; Marchand-Brynaert, J. Fibronectin adsorption or/and covalent grafting on chemically modified PEEK film surfaces. J. Biomater. Sci. Polym. Ed. 1999, 10, 657–677. [Google Scholar] [CrossRef] [PubMed]
  203. Panayotov, I.V.; Orti, V.; Cuisinier, F.; Yachouh, J. Polyetheretherketone (PEEK) for medical applications. J. Mater. Sci. Mater. Med. 2016, 27, 118. [Google Scholar] [CrossRef]
  204. Sandler, J.; Windle, A.H.; Werner, P.; Altstädt, V.; Es, M.; Shaffer, M.S. Carbon-nanofibre-reinforced poly (ether ether ketone) fibres. J. Mater. Sci. 2003, 38, 2135–2141. [Google Scholar] [CrossRef]
  205. Bembey, A.K.; Oyen, M.L.; Ko, C.-C.; Bushby, A.J.; Boyde, A. Elastic modulus and mineral density of dentine and enamel in natural caries lesions. MRS Online Proc. Libr. (OPL) 2005, 874, L5–L15. [Google Scholar] [CrossRef]
  206. Li, Y.; Wang, D.; Qin, W.; Jia, H.; Wu, Y.; Ma, J.; Tang, B. Mechanical properties, hemocompatibility, cytotoxicity and systemic toxicity of carbon fibers/poly(ether-ether-ketone) composites with different fiber lengths as orthopedic implants. J. Biomater. Sci. Polym. Ed. 2019, 30, 1709–1724. [Google Scholar] [CrossRef]
  207. Qin, W.; Li, Y.; Ma, J.; Liang, Q.; Tang, B. Mechanical properties and cytotoxicity of hierarchical carbon fiber-reinforced poly (ether-ether-ketone) composites used as implant materials. J. Mech. Behav. Biomed. Mater. 2019, 89, 227–233. [Google Scholar] [CrossRef]
  208. Chen, Y.; Zhang, L.; Qin, T.; Wang, Z.; Li, Y.; Gu, B. Evaluation of neurosurgical implant infection rates and associated pathogens: Evidence from 1118 postoperative infections. Neurosurg. Focus 2019, 47, E6. [Google Scholar] [CrossRef] [PubMed]
  209. Ouyang, L.; Zhao, Y.; Jin, G.; Lu, T.; Li, J.; Qiao, Y.; Ning, C.; Zhang, X.; Chu, P.K.; Liu, X. Influence of sulfur content on bone formation and antibacterial ability of sulfonated PEEK. Biomaterials 2016, 83, 115–126. [Google Scholar] [CrossRef] [PubMed]
  210. Montero, J.F.; Tajiri, H.A.; Barra, G.M.; Fredel, M.C.; Benfatti, C.A.; Magini, R.S.; Pimenta, A.L.; Souza, J.C. Biofilm behavior on sulfonated poly(ether-ether-ketone) (sPEEK). MMater. Sci. Eng. C Mater. Biol. Appl. 2017, 70, 456–460. [Google Scholar] [CrossRef] [PubMed]
  211. Wang, S.; Deng, Y.; Yang, L.; Shi, X.; Yang, W.; Chen, Z.G. Enhanced antibacterial property and osteo-differentiation activity on plasma treated porous polyetheretherketone with hierarchical micro/nano-topography. J. Biomater. Sci. Polym. Ed. 2018, 29, 520–542. [Google Scholar] [CrossRef] [PubMed]
  212. Liu, W.; Li, J.; Cheng, M.; Wang, Q.; Qian, Y.; Yeung, K.W.K.; Chu, P.K.; Zhang, X. A surface-engineered polyetheretherketone biomaterial implant with direct and immunoregulatory antibacterial activity against methicillin-resistant Staphylococcus aureus. Biomaterials 2019, 208, 8–20. [Google Scholar] [CrossRef]
  213. Niu, Y.; Guo, L.; Hu, F.; Ren, L.; Zhou, Q.; Ru, J.; Wei, J. Macro-Microporous Surface with Sulfonic Acid Groups and Micro-Nano Structures of PEEK/Nano Magnesium Silicate Composite Exhibiting Antibacterial Activity and Inducing Cell Responses. Int. J. Nanomed. 2020, 15, 2403–2417. [Google Scholar] [CrossRef]
  214. Mei, S.; Wang, F.; Hu, X.; Yang, K.; Xie, D.; Yang, L.; Wu, Z.; Wei, J. Construction of a hierarchical micro & nanoporous surface for loading genistein on the composite of polyetheretherketone/tantalum pentoxide possessing antibacterial activity and accelerated osteointegration. Biomater. Sci. 2021, 9, 167–185. [Google Scholar] [CrossRef]
  215. Luo, S.; Wang, P.; Ma, M.; Pan, Z.; Lu, L.; Yin, F.; Cai, J. Genistein loaded into microporous surface of nano tantalum/PEEK composite with antibacterial effect regulating cellular response in vitro, and promoting osseointegration in vivo. J. Mech. Behav. Biomed. Mater. 2022, 125, 104972. [Google Scholar] [CrossRef] [PubMed]
  216. Yan, J.; Zhou, W.; Jia, Z.; Xiong, P.; Li, Y.; Wang, P.; Li, Q.; Cheng, Y.; Zheng, Y. Endowing polyetheretherketone with synergistic bactericidal effects and improved osteogenic ability. Acta Biomater. 2018, 79, 216–229. [Google Scholar] [CrossRef]
  217. Xu, X.; Li, Y.; Wang, L.; Li, Y.; Pan, J.; Fu, X.; Luo, Z.; Sui, Y.; Zhang, S.; Wang, L.; et al. Triple-functional polyetheretherketone surface with enhanced bacteriostasis and anti-inflammatory and osseointegrative properties for implant application. Biomaterials 2019, 212, 98–114. [Google Scholar] [CrossRef]
  218. Xue, Z.; Wang, Z.; Sun, A.; Huang, J.; Wu, W.; Chen, M.; Hao, X.; Huang, Z.; Lin, X.; Weng, S. Rapid construction of polyetheretherketone (PEEK) biological implants incorporated with brushite (CaHPO4 2H2O) and antibiotics for anti-infection and enhanced osseointegration. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 111, 110782. [Google Scholar] [CrossRef] [PubMed]
  219. Sun, A.; Lin, X.; Xue, Z.; Huang, J.; Bai, X.; Huang, L.; Lin, X.; Weng, S.; Chen, M. Facile surface functional polyetheretherketone with antibacterial and immunoregulatory activities for enhanced regeneration toward bacterium-infected bone destruction. Drug. Deliv. 2021, 28, 1649–1663. [Google Scholar] [CrossRef] [PubMed]
  220. Diez-Pascual, A.M. Tissue Engineering Bionanocomposites Based on Poly(propylene fumarate). Polymers 2017, 9, 260. [Google Scholar] [CrossRef] [PubMed]
  221. Wang, M.O.; Piard, C.M.; Melchiorri, A.; Dreher, M.L.; Fisher, J.P. Evaluating changes in structure and cytotoxicity during in vitro degradation of three-dimensional printed scaffolds. Tissue Eng. Part A 2015, 21, 1642–1653. [Google Scholar] [CrossRef] [PubMed]
  222. Díez-Pascual, A.M.; Díez-Vicente, A.L. Poly(propylene fumarate)/Polyethylene Glycol-Modified Graphene Oxide Nanocomposites for Tissue Engineering. ACS Appl. Mater. Interfaces 2016, 8, 17902–17914. [Google Scholar] [CrossRef]
  223. Le Fer, G.; Becker, M.L. 4D Printing of Resorbable Complex Shape-Memory Poly(propylene fumarate) Star Scaffolds. ACS Appl. Mater. Interfaces 2020, 12, 22444–22452. [Google Scholar] [CrossRef]
  224. Fukushima, K. Biodegradable functional biomaterials exploiting substituted trimethylene carbonates and organocatalytic transesterification. Polym. J. 2016, 48, 1103–1114. [Google Scholar] [CrossRef]
  225. Zhu, J.; Luo, X.; Li, X. Ring-Opening Polymerization of Trimethylene Carbonate with Phosphazene Organocatalyst. Polymers 2023, 15, 720. [Google Scholar] [CrossRef]
  226. Henslee, A.M.; Shah, S.R.; Wong, M.E.; Mikos, A.G.; Kasper, F.K. Degradable, antibiotic releasing poly(propylene fumarate)-based constructs for craniofacial space maintenance applications. J. Biomed. Mater. Res. A 2015, 103, 1485–1497. [Google Scholar] [CrossRef]
  227. Smith, D.K.; Newkirk, W. The crystal structure of baddeleyite (monoclinic ZrO2) and its relation to the polymorphism of ZrO2. Acta Crystallogr. 1965, 18, 983–991. [Google Scholar] [CrossRef]
  228. Kisi, E.H.; Howard, C.J.; Hill, R.J. Crystal Structure of Orthorhombic Zirconia in Partially Stabilized Zirconia. J. Am. Ceram. Soc. 1989, 72, 1757–1760. [Google Scholar] [CrossRef]
  229. Kalavathi, V.; Kumar Bhuyan, R. A detailed study on zirconium and its applications in manufacturing process with combinations of other metals, oxides and alloys—A review. Mater. Today Proc. 2019, 19, 781–786. [Google Scholar] [CrossRef]
  230. Zhang, Y.; Lawn, B.R. Novel Zirconia Materials in Dentistry. J. Dent. Res. 2018, 97, 140–147. [Google Scholar] [CrossRef] [PubMed]
  231. Branco, A.C.; Colaço, R.; Figueiredo-Pina, C.G.; Serro, A.P. Recent Advances on 3D-Printed Zirconia-Based Dental Materials: A Review. Materials 2023, 16, 1860. [Google Scholar] [CrossRef] [PubMed]
  232. Kongkiatkamon, S.; Rokaya, D.; Kengtanyakich, S.; Peampring, C. Current classification of zirconia in dentistry: An updated review. PeerJ 2023, 11, e15669. [Google Scholar] [CrossRef]
  233. Komissarenko, D.A.; Sokolov, P.S.; Evstigneeva, A.D.; Shmeleva, I.A.; Dosovitsky, A.E. Rheological and Curing Behavior of Acrylate-Based Suspensions for the DLP 3D Printing of Complex Zirconia Parts. Materials 2018, 11, 2350. [Google Scholar] [CrossRef]
  234. Son, M.; Raju, K.; Lee, J.; Jung, J.; Jeong, S.; Kim, J.I.; Cho, J. 3D Printing of CNT- and YSZ-Added Dental Resin-Based Composites by Digital Light Processing and Their Mechanical Properties. Materials 2023, 16, 1873. [Google Scholar] [CrossRef] [PubMed]
  235. Marsico, C.; Øilo, M.; Kutsch, J.; Kauf, M.; Arola, D. Vat Polymerization-Printed Partially Stabilized Zirconia: Mechanical Properties, Reliability and Structural defects. Addit. Manuf. 2020, 36, 101450. [Google Scholar] [CrossRef]
  236. Kang, J.H.; Sakthiabirami, K.; Kim, H.A.; Hosseini Toopghara, S.A.; Jun, M.J.; Lim, H.P.; Park, C.; Yun, K.D.; Park, S.W. Effects of UV Absorber on Zirconia Fabricated with Digital Light Processing Additive Manufacturing. Materials 2022, 15, 8726. [Google Scholar] [CrossRef]
  237. Jang, J.G.; Kang, J.H.; Joe, K.B.; Sakthiabirami, K.; Jang, K.J.; Jun, M.J.; Oh, G.J.; Park, C.; Park, S.W. Evaluation of Physical Properties of Zirconia Suspension with Added Silane Coupling Agent for Additive Manufacturing Processes. Materials 2022, 15, 1337. [Google Scholar] [CrossRef]
  238. Coppola, B.; Montanaro, L.; Palmero, P. DLP Fabrication of Zirconia Scaffolds Coated with HA/β-TCP Layer: Role of Scaffold Architecture on Mechanical and Biological Properties. J. Funct. Biomater. 2022, 13, 148. [Google Scholar] [CrossRef]
  239. Zhu, Y.; Liu, K.; Deng, J.; Ye, J.; Ai, F.; Ouyang, H.; Wu, T.; Jia, J.; Cheng, X.; Wang, X. 3D printed zirconia ceramic hip joint with precise structure and broad-spectrum antibacterial properties. Int. J. Nanomed. 2019, 14, 5977–5987. [Google Scholar] [CrossRef]
  240. Wu, T.; Zhou, Q.; Hong, G.; Bai, Z.; Bian, J.; Xie, H.; Chen, C. A chlorogenic acid-chitosan complex bifunctional coating for improving osteogenesis differentiation and bactericidal properties of zirconia implants. Colloids Surf. B Biointerfaces 2023, 230, 113484. [Google Scholar] [CrossRef]
  241. Goldschmidt, G.M.; Krok-Borkowicz, M.; Zybała, R.; Pamuła, E.; Telle, R.; Conrads, G.; Schickle, K. Biomimetic in situ precipitation of calcium phosphate containing silver nanoparticles on zirconia ceramic materials for surface functionalization in terms of antimicrobial and osteoconductive properties. Dent. Mater. 2021, 37, 10–18. [Google Scholar] [CrossRef]
  242. Biskri, Z.E.; Rached, H.; Bouchear, M.; Rached, D. Computational study of structural, elastic and electronic properties of lithium disilicate (Li2Si2O5) glass-ceramic. J. Mech. Behav. Biomed. Mater. 2014, 32, 345–350. [Google Scholar] [CrossRef]
  243. Pieger, S.; Salman, A.; Bidra, A.S. Clinical outcomes of lithium disilicate single crowns and partial fixed dental prostheses: A systematic review. J. Prosthet. Dent. 2014, 112, 22–30. [Google Scholar] [CrossRef] [PubMed]
  244. Chen, Z.; Sun, X.; Shang, Y.; Xiong, K.; Xu, Z.; Guo, R.; Cai, S.; Zheng, C. Dense ceramics with complex shape fabricated by 3D printing: A review. J. Adv. Ceram. 2021, 10, 195–218. [Google Scholar] [CrossRef]
  245. Unkovskiy, A.; Beuer, F.; Metin, D.S.; Bomze, D.; Hey, J.; Schmidt, F. Additive Manufacturing of Lithium Disilicate with the LCM Process for Classic and Non-Prep Veneers: Preliminary Technical and Clinical Case Experience. Materials 2022, 15, 6034. [Google Scholar] [CrossRef] [PubMed]
  246. Shirai, T.; Tsuchiya, H.; Terauchi, R.; Tsuchida, S.; Shimomura, S.; Kajino, Y.; Takahashi, K. Iodine-supported implants in prevention and treatment of surgical site infections for compromised hosts: A prospective study. J. Orthop. Surg. Res. 2023, 18, 388. [Google Scholar] [CrossRef]
  247. Bhandari, M.; Smith, J.; Miller, L.E.; Block, J.E. Clinical and Economic Burden of Revision Knee Arthroplasty. Clin. Med. Insights Arthritis Musculoskelet. Disord. 2012, 5, S10859. [Google Scholar] [CrossRef]
  248. Pirisi, L.; Pennestrì, F.; Viganò, M.; Banfi, G. Prevalence and burden of orthopaedic implantable-device infections in Italy: A hospital-based national study. BMC Infect. Dis. 2020, 20, 337. [Google Scholar] [CrossRef]
  249. Khan, M.; Osman, K.; Green, G.; Haddad, F.S. The epidemiology of failure in total knee arthroplasty. Bone Jt. J. 2016, 98-B, 105–112. [Google Scholar] [CrossRef]
  250. Rahyussalim, A.J.; Marsetio, A.F.; Saleh, I.; Kurniawati, T.; Whulanza, Y. The Needs of Current Implant Technology in Orthopaedic Prosthesis Biomaterials Application to Reduce Prosthesis Failure Rate. J. Nanomater. 2016, 2016, 5386924. [Google Scholar] [CrossRef]
  251. Chanachai, S.; Chaichana, W.; Insee, K.; Benjakul, S.; Aupaphong, V.; Panpisut, P. Physical/Mechanical and Antibacterial Properties of Orthodontic Adhesives Containing Calcium Phosphate and Nisin. J. Funct. Biomater. 2021, 12, 73. [Google Scholar] [CrossRef] [PubMed]
  252. Topolnitskiy, E.; Chekalkin, T.; Marchenko, E.; Yasenchuk, Y.; Kang, S.-B.; Kang, J.-H.; Obrosov, A. Evaluation of Clinical Performance of TiNi-Based Implants Used in Chest Wall Repair after Resection for Malignant Tumors. J. Funct. Biomater. 2021, 12, 60. [Google Scholar] [CrossRef] [PubMed]
  253. Scheau, C.; Didilescu, A.C.; Caruntu, C. Medical Application of Functional Biomaterials—The Future Is Now. J. Funct. Biomater. 2022, 13, 244. [Google Scholar] [CrossRef] [PubMed]
  254. Miranda, I.; Souza, A.; Sousa, P.; Ribeiro, J.; Castanheira, E.M.S.; Lima, R.; Minas, G. Properties and Applications of PDMS for Biomedical Engineering: A Review. J. Funct. Biomater. 2022, 13, 2. [Google Scholar] [CrossRef] [PubMed]
  255. Streza, A.; Antoniac, A.; Manescu, V.; Ciocoiu, R.; Cotrut, C.-M.; Miculescu, M.; Miculescu, F.; Antoniac, I.; Fosca, M.; Rau, J.V.; et al. In Vitro Studies Regarding the Effect of Cellulose Acetate-Based Composite Coatings on the Functional Properties of the Biodegradable Mg3Nd Alloys. Biomimetics 2023, 8, 526. [Google Scholar] [CrossRef] [PubMed]
  256. Jeyachandran, S.; Chellapandian, H.; Ali, N. Advancements in Composite Materials and Their Expanding Role in Biomedical Applications. Biomimetics 2023, 8, 518. [Google Scholar] [CrossRef]
  257. Haque, F.; Luscher, A.F.; Mitchell, K.-A.S.; Sutradhar, A. Optimization of Fixations for Additively Manufactured Cranial Implants: Insights from Finite Element Analysis. Biomimetics 2023, 8, 498. [Google Scholar] [CrossRef]
  258. Bakina, O.; Svarovskaya, N.; Ivanova, L.; Glazkova, E.; Rodkevich, N.; Evstigneev, V.; Evstigneev, M.; Mosunov, A.; Lerner, M. New PMMA-Based Hydroxyapatite/ZnFe2O4/ZnO Composite with Antibacterial Performance and Low Toxicity. Biomimetics 2023, 8, 488. [Google Scholar] [CrossRef]
  259. Kozelskaya, A.I.; Verzunova, K.N.; Akimchenko, I.O.; Frueh, J.; Petrov, V.I.; Slepchenko, G.B.; Bakina, O.V.; Lerner, M.I.; Brizhan, L.K.; Davydov, D.V.; et al. Antibacterial Calcium Phosphate Coatings for Biomedical Applications Fabricated via Micro-Arc Oxidation. Biomimetics 2023, 8, 444. [Google Scholar] [CrossRef]
  260. Mikhailova, E.O. Selenium Nanoparticles: Green Synthesis and Biomedical Application. Molecules 2023, 28, 8125. [Google Scholar] [CrossRef]
  261. Mohanta, Y.K.; Mishra, A.K.; Panda, J.; Chakrabartty, I.; Sarma, B.; Panda, S.K.; Chopra, H.; Zengin, G.; Moloney, M.G.; Sharifi-Rad, M. Promising applications of phyto-fabricated silver nanoparticles: Recent trends in biomedicine. Biochem. Biophys. Res. Commun. 2023, 688, 149126. [Google Scholar] [CrossRef]
  262. Burlec, A.F.; Corciova, A.; Boev, M.; Batir-Marin, D.; Mircea, C.; Cioanca, O.; Danila, G.; Danila, M.; Bucur, A.F.; Hancianu, M. Current Overview of Metal Nanoparticles’ Synthesis, Characterization, and Biomedical Applications, with a Focus on Silver and Gold Nanoparticles. Pharmaceuticals 2023, 16, 1410. [Google Scholar] [CrossRef]
  263. Wahab, S.; Salman, A.; Khan, Z.; Khan, S.; Krishnaraj, C.; Yun, S.I. Metallic Nanoparticles: A Promising Arsenal against Antimicrobial Resistance-Unraveling Mechanisms and Enhancing Medication Efficacy. Int. J. Mol. Sci. 2023, 24, 14897. [Google Scholar] [CrossRef]
  264. Silva, J.M.; Teixeira, A.B.; Reis, A.C. Silver-based gels for oral and skin infections: Antimicrobial effect and physicochemical stability. Future Microbiol. 2023, 18, 985–996. [Google Scholar] [CrossRef]
  265. Berhe, M.G.; Gebreslassie, Y.T. Biomedical Applications of Biosynthesized Nickel Oxide Nanoparticles. Int. J. Nanomed. 2023, 18, 4229–4251. [Google Scholar] [CrossRef]
  266. Ramos-Zúñiga, J.; Bruna, N.; Pérez-Donoso, J.M. Toxicity Mechanisms of Copper Nanoparticles and Copper Surfaces on Bacterial Cells and Viruses. Int. J. Mol. Sci. 2023, 24, 10503. [Google Scholar] [CrossRef]
  267. Kamyab, H.; Chelliapan, S.; Hayder, G.; Yusuf, M.; Taheri, M.M.; Rezania, S.; Hasan, M.; Yadav, K.K.; Khorami, M.; Farajnezhad, M.; et al. Exploring the potential of metal and metal oxide nanomaterials for sustainable water and wastewater treatment: A review of their antimicrobial properties. Chemosphere 2023, 335, 139103. [Google Scholar] [CrossRef]
  268. Shandhiya, M.; Janarthanan, B.; Sharmila, S. A comprehensive review on antibacterial analysis of natural extract-based metal and metal oxide nanoparticles. Arch. Microbiol. 2024, 206, 52. [Google Scholar] [CrossRef]
  269. Meng, Y.Q.; Shi, Y.N.; Zhu, Y.P.; Liu, Y.Q.; Gu, L.W.; Liu, D.D.; Ma, A.; Xia, F.; Guo, Q.Y.; Xu, C.C.; et al. Recent trends in preparation and biomedical applications of iron oxide nanoparticles. J. Nanobiotechnol. 2024, 22, 24. [Google Scholar] [CrossRef]
  270. Lan, H.; Jamil, M.; Ke, G.; Dong, N. The role of nanoparticles and nanomaterials in cancer diagnosis and treatment: A comprehensive review. Am. J. Cancer Res. 2023, 13, 5751–5784. [Google Scholar]
  271. Malik, M.A.; Hashmi, A.A.; Al-Bogami, A.S.; Wani, M.Y. Harnessing the power of gold: Advancements in anticancer gold complexes and their functionalized nanoparticles. J. Mater. Chem. B 2024, 12, 552–576. [Google Scholar] [CrossRef]
  272. Scully, S.P.; Ghert, M.A.; Zurakowski, D.; Thompson, R.C.; Gebhardt, M.C. Pathologic fracture in osteosarcoma: Prognostic importance and treatment implications. J. Bone Jt. Surg. Am. 2002, 84, 49–57. [Google Scholar] [CrossRef]
  273. Dragosloveanu, Ș.; Dragosloveanu, C.D.M.; Cotor, D.C.; Stoica, C.I. Short vs. long intramedullary nail systems in trochanteric fractures: A randomized prospective single center study. Exp. Ther. Med. 2022, 23, 106. [Google Scholar] [CrossRef]
  274. Lindsey, B.A.; Markel, J.E.; Kleinerman, E.S. Osteosarcoma Overview. Rheumatol. Ther. 2017, 4, 25–43. [Google Scholar] [CrossRef]
  275. Misaghi, A.; Goldin, A.; Awad, M.; Kulidjian, A.A. Osteosarcoma: A comprehensive review. Sicot. J. 2018, 4, 12. [Google Scholar] [CrossRef]
  276. Tiwari, A. Current concepts in surgical treatment of osteosarcoma. J. Clin. Orthop. Trauma 2012, 3, 4–9. [Google Scholar] [CrossRef]
  277. Grande, F.; Tucci, P. Titanium Dioxide Nanoparticles: A Risk for Human Health? Mini Rev. Med. Chem. 2016, 16, 762–769. [Google Scholar] [CrossRef]
  278. Zhang, X.; Song, Y.; Gong, H.; Wu, C.; Wang, B.; Chen, W.; Hu, J.; Xiang, H.; Zhang, K.; Sun, M. Neurotoxicity of Titanium Dioxide Nanoparticles: A Comprehensive Review. Int. J. Nanomed. 2023, 18, 7183–7204. [Google Scholar] [CrossRef]
  279. Dilla, R.A.; Motta, C.M.; Snyder, S.R.; Wilson, J.A.; Wesdemiotis, C.; Becker, M.L. Synthesis and 3D printing of PEG–poly (propylene fumarate) diblock and triblock copolymer hydrogels. ACS Macro Lett. 2018, 7, 1254–1260. [Google Scholar] [CrossRef]
  280. Fukushima, K. Poly (trimethylene carbonate)-based polymers engineered for biodegradable functional biomaterials. Biomater. Sci. 2016, 4, 9–24. [Google Scholar] [CrossRef]
  281. Frazer, R.Q.; Byron, R.T.; Osborne, P.B.; West, K.P. PMMA: An essential material in medicine and dentistry. J. Long-Term Eff. Med. Implant. 2005, 15, 629–639. [Google Scholar] [CrossRef]
  282. Wei, X.; Zhou, W.; Tang, Z.; Wu, H.; Liu, Y.; Dong, H.; Wang, N.; Huang, H.; Bao, S.; Shi, L. Magnesium surface-activated 3D printed porous PEEK scaffolds for in vivo osseointegration by promoting angiogenesis and osteogenesis. Bioact. Mater. 2023, 20, 16–28. [Google Scholar] [CrossRef]
  283. Aranaz, I.; Alcántara, A.R.; Civera, M.C.; Arias, C.; Elorza, B.; Heras Caballero, A.; Acosta, N. Chitosan: An Overview of Its Properties and Applications. Polymers 2021, 13, 3256. [Google Scholar] [CrossRef]
  284. Ueno, H.; Mori, T.; Fujinaga, T. Topical formulations and wound healing applications of chitosan. Adv. Drug Deliv. Rev. 2001, 52, 105–115. [Google Scholar] [CrossRef]
  285. Azuma, K.; Osaki, T.; Minami, S.; Okamoto, Y. Anticancer and anti-inflammatory properties of chitin and chitosan oligosaccharides. J. Funct. Biomater. 2015, 6, 33–49. [Google Scholar] [CrossRef]
  286. Avelelas, F.; Horta, A.; Pinto, L.F.V.; Cotrim Marques, S.; Marques Nunes, P.; Pedrosa, R.; Leandro, S.M. Antifungal and Antioxidant Properties of Chitosan Polymers Obtained from Nontraditional Polybius henslowii Sources. Mar. Drugs 2019, 17, 239. [Google Scholar] [CrossRef]
  287. Sarkar, S.; Das, D.; Dutta, P.; Kalita, J.; Wann, S.B.; Manna, P. Chitosan: A promising therapeutic agent and effective drug delivery system in managing diabetes mellitus. Carbohydr. Polym. 2020, 247, 116594. [Google Scholar] [CrossRef]
  288. Shih, P.Y.; Liao, Y.T.; Tseng, Y.K.; Deng, F.S.; Lin, C.H. A Potential Antifungal Effect of Chitosan Against Candida albicans Is Mediated via the Inhibition of SAGA Complex Component Expression and the Subsequent Alteration of Cell Surface Integrity. Front. Microbiol. 2019, 10, 602. [Google Scholar] [CrossRef] [PubMed]
  289. Ke, C.L.; Deng, F.S.; Chuang, C.Y.; Lin, C.H. Antimicrobial Actions and Applications of Chitosan. Polymers 2021, 13, 904. [Google Scholar] [CrossRef]
  290. Alqarni, L.S.; Alghamdi, A.M.; Elamin, N.Y.; Rajeh, A. Enhancing the optical, electrical, dielectric properties and antimicrobial activity of chitosan/gelatin incorporated with Co-doped ZnO nanoparticles: Nanocomposites for use in energy storage and food packaging. J. Mol. Struct. 2024, 1297, 137011. [Google Scholar] [CrossRef]
  291. Jeon, S.; Kim, I.; Jeong, Y.J.; Kim, Y.; Chung, J.J.; Kim, S.W. Comparative Analysis of Antibacterial and Wound Healing Activities of Chitosan and Povidone-Iodine–Based Hydrogels. Ann. Plast. Surg. 2024, 92, 240–244. [Google Scholar] [CrossRef]
  292. Li, C.; Wang, K.; Li, F.; Xie, D. Green fabrication, characterization and antimicrobial activities of AgO/Ag/carboxymethyl chitosan-graphene oxide films. Arab. J. Chem. 2024, 17, 105380. [Google Scholar] [CrossRef]
  293. Zhou, Q.; Lan, W.; Xie, J. Phenolic acid-chitosan derivatives: An effective strategy to cope with food preservation problems. Int. J. Biol. Macromol. 2024, 254, 127917. [Google Scholar] [CrossRef]
  294. Kotsiftopoulos, C. The rational use of antibiotics medicine. Arch. Med. 2017, 2, 36. [Google Scholar]
  295. Kourkouta, L.; Koukourikos, K.; Iliadis, C.; Plati, P.; Dimitriadou, A. History of antibiotics. Sumer. J. Med. Healthc. 2018, 1, 51–55. [Google Scholar]
  296. Norowski, P.A., Jr.; Bumgardner, J.D. Biomaterial and antibiotic strategies for peri-implantitis: A review. J. Biomed. Mater. Res. B Appl. Biomater. 2009, 88, 530–543. [Google Scholar] [CrossRef]
  297. Pritchard, E.M.; Valentin, T.; Panilaitis, B.; Omenetto, F.; Kaplan, D.L. Antibiotic-releasing silk biomaterials for infection prevention and treatment. Adv. Funct. Mater. 2013, 23, 854–861. [Google Scholar] [CrossRef]
  298. Inzana, J.A.; Schwarz, E.M.; Kates, S.L.; Awad, H.A. Biomaterials approaches to treating implant-associated osteomyelitis. Biomaterials 2016, 81, 58–71. [Google Scholar] [CrossRef]
  299. Sadowska, J.M.; Genoud, K.J.; Kelly, D.J.; O’Brien, F.J. Bone biomaterials for overcoming antimicrobial resistance: Advances in non-antibiotic antimicrobial approaches for regeneration of infected osseous tissue. Mater. Today 2021, 46, 136–154. [Google Scholar] [CrossRef]
  300. Tălăpan, D.; Sandu, A.-M.; Rafila, A. Antimicrobial Resistance of Staphylococcus aureus Isolated between 2017 and 2022 from Infections at a Tertiary Care Hospital in Romania. Antibiotics 2023, 12, 974. [Google Scholar] [CrossRef]
  301. Foster, T.J. Antibiotic resistance in Staphylococcus aureus. Current status and future prospects. FEMS Microbiol. Rev. 2017, 41, 430–449. [Google Scholar] [CrossRef]
  302. Olorunmola, F.O.; Kolawole, D.O.; Lamikanra, A. Antibiotic resistance and virulence properties in Escherichia coli strains from cases of urinary tract infections. Afr. J. Infect. Dis. 2013, 7, 1–7. [Google Scholar] [CrossRef]
  303. Wang, S.; Zhao, S.; Zhou, Y.; Jin, S.; Ye, T.; Pan, X. Antibiotic resistance spectrum of E. coli strains from different samples and age-grouped patients: A 10-year retrospective study. BMJ Open 2023, 13, e067490. [Google Scholar] [CrossRef]
  304. Ghanavati, R.; Kazemian, H.; Asadollahi, P.; Heidari, H.; Irajian, G.; Navab-Moghadam, F.; Razavi, S. Characterization of Antimicrobial Resistance Patterns of Klebsiella pneumoniae Isolates Obtained from Wound Infections. Infect. Disord. Drug. Targets 2021, 21, 119–124. [Google Scholar] [CrossRef]
  305. Ameshe, A.; Engda, T.; Gizachew, M. Antimicrobial Resistance Patterns, Extended-Spectrum Beta-Lactamase Production, and Associated Risk Factors of Klebsiella Species among UTI-Suspected Patients at Bahir Dar City, Northwest Ethiopia. Int. J. Microbiol. 2022, 2022, 8216545. [Google Scholar] [CrossRef]
  306. Moellering, R.C., Jr. Vancomycin: A 50-Year Reassessment. Clin. Infect. Dis. 2006, 42, S3–S4. [Google Scholar] [CrossRef]
  307. Johnson, A.P.; Uttley, A.; Woodford, N.; George, R. Resistance to vancomycin and teicoplanin: An emerging clinical problem. Clin. Microbiol. Rev. 1990, 3, 280–291. [Google Scholar] [CrossRef]
  308. Tenover, F.C.; Biddle, J.W.; Lancaster, M.V. Increasing resistance to vancomycin and other glycopeptides in Staphylococcus aureus. Emerg. Infect. Dis. 2001, 7, 327. [Google Scholar] [CrossRef]
  309. Miller, W.R.; Murray, B.E.; Rice, L.B.; Arias, C.A. Resistance in vancomycin-resistant enterococci. Infect. Dis. Clin. 2020, 34, 751–771. [Google Scholar] [CrossRef]
  310. Álvarez, R.; López Cortés, L.E.; Molina, J.; Cisneros, J.M.; Pachón, J. Optimizing the clinical use of vancomycin. Antimicrob. Agents Chemother. 2016, 60, 2601–2609. [Google Scholar] [CrossRef]
  311. Noskin, G.A.; Peterson, L.R.; Warren, J.R. Enterococcus faecium and Enterococcus faecalis bacteremia: Acquisition and outcome. Clin. Infect. Dis. 1995, 20, 296–301. [Google Scholar] [CrossRef]
  312. Schaberg, D.R.; Culver, D.H.; Gaynes, R.P. Major trends in the microbial etiology of nosocomial infection. Am. J. Med. 1991, 91, S72–S75. [Google Scholar] [CrossRef]
  313. Kaye, D. Enterococci: Biologic and epidemiologic characteristics and in vitro susceptibility. Arch. Intern. Med. 1982, 142, 2006–2009. [Google Scholar] [CrossRef]
  314. Maki, D.G.; Agger, W.A. Enterococcal Bacteremia: Clinical Features, the Risk of Endocarditis, and Management. Medicine 1988, 67, 248. [Google Scholar] [CrossRef]
  315. Cetinkaya, Y.; Falk, P.; Mayhall, C.G. Vancomycin-resistant enterococci. Clin. Microbiol. Rev. 2000, 13, 686–707. [Google Scholar] [CrossRef]
  316. Buhl, M.; Peter, S.; Willmann, M. Prevalence and risk factors associated with colonization and infection of extensively drug-resistant Pseudomonas aeruginosa: A systematic review. Expert. Rev. Anti. Infect. Ther. 2015, 13, 1159–1170. [Google Scholar] [CrossRef]
  317. Klockgether, J.; Tümmler, B. Recent advances in understanding Pseudomonas aeruginosa as a pathogen. F1000Res 2017, 6, 1261. [Google Scholar] [CrossRef]
  318. Hancock, R.E. Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria. Clin. Infect. Dis. 1998, 27 (Suppl. S1), S93–S99. [Google Scholar] [CrossRef]
  319. Pang, Z.; Raudonis, R.; Glick, B.R.; Lin, T.J.; Cheng, Z. Antibiotic resistance in Pseudomonas aeruginosa: Mechanisms and alternative therapeutic strategies. Biotechnol. Adv. 2019, 37, 177–192. [Google Scholar] [CrossRef]
  320. Ruoff, K.L. Streptococcus anginosus (“Streptococcus milleri”): The unrecognized pathogen. Clin. Microbiol. Rev. 1988, 1, 102–108. [Google Scholar] [CrossRef]
  321. Villalobos, V.; Garrido, M.; Reyes, A.; Fernández, C.; Diaz, C.; Torres, V.A.; González, P.A.; Cáceres, M. Aging envisage imbalance of the periodontium: A keystone in oral disease and systemic health. Front. Immunol. 2022, 13, 1044334. [Google Scholar] [CrossRef]
  322. Bregaint, S.; Boyer, E.; Fong, S.B.; Meuric, V.; Bonnaure-Mallet, M.; Jolivet-Gougeon, A. Porphyromonas gingivalis outside the oral cavity. Odontology 2022, 110, 1–19. [Google Scholar] [CrossRef]
  323. Zhang, J.; Yu, C.; Zhang, X.; Chen, H.; Dong, J.; Lu, W.; Song, Z.; Zhou, W. Porphyromonas gingivalis lipopolysaccharide induces cognitive dysfunction, mediated by neuronal inflammation via activation of the TLR4 signaling pathway in C57BL/6 mice. J. Neuroinflamm. 2018, 15, 37. [Google Scholar] [CrossRef]
  324. Wang, X.; Jia, Y.; Wen, L.; Mu, W.; Wu, X.; Liu, T.; Liu, X.; Fang, J.; Luan, Y.; Chen, P.; et al. Porphyromonas gingivalis Promotes Colorectal Carcinoma by Activating the Hematopoietic NLRP3 Inflammasome. Cancer Res. 2021, 81, 2745–2759. [Google Scholar] [CrossRef]
  325. Lamont, R.J.; Fitzsimonds, Z.R.; Wang, H.; Gao, S. Role of Porphyromonas gingivalis in oral and orodigestive squamous cell carcinoma. Periodontol 2000 2022, 89, 154–165. [Google Scholar] [CrossRef]
  326. Lemos, J.A.; Palmer, S.R.; Zeng, L.; Wen, Z.T.; Kajfasz, J.K.; Freires, I.A.; Abranches, J.; Brady, L.J. The Biology of Streptococcus mutans. Microbiol. Spectr. 2019, 7, 1128. [Google Scholar] [CrossRef]
  327. Cui, T.; Luo, W.; Xu, L.; Yang, B.; Zhao, W.; Cang, H. Progress of Antimicrobial Discovery against the Major Cariogenic Pathogen Streptococcus mutans. Curr. Issues Mol. Biol. 2019, 32, 601–644. [Google Scholar] [CrossRef]
  328. Crump, J.A.; Luby, S.P.; Mintz, E.D. The global burden of typhoid fever. Bull. World Health Organ. 2004, 82, 346–353. [Google Scholar]
  329. Popa, G.L.; Papa, M.I. Salmonella spp. infection—A continuous threat worldwide. Germs 2021, 11, 88–96. [Google Scholar] [CrossRef]
  330. Allerberger, F.; Liesegang, A.; Grif, K.; Khaschabi, D.; Prager, R.; Danzl, J.; Höck, F.; Ottl, J.; Dierich, M.P.; Berghold, C.; et al. Occurrence of Salmonella enterica serovar Dublin in Austria. Wien. Med. Wochenschr. 2003, 153, 148–152. [Google Scholar] [CrossRef]
  331. Elias, A.; Viana, J.X.; Rangel, H.; Osles, A.G. Antigenic variation in Salmonella typhimurium. Proc. Soc. Exp. Biol. Med. 1974, 145, 392–396. [Google Scholar] [CrossRef]
  332. Chiu, C.H.; Wu, T.L.; Su, L.H.; Chu, C.; Chia, J.H.; Kuo, A.J.; Chien, M.S.; Lin, T.Y. The emergence in Taiwan of fluoroquinolone resistance in Salmonella enterica serotype choleraesuis. N. Engl. J. Med. 2002, 346, 413–419. [Google Scholar] [CrossRef]
  333. Martin, L.J.; Fyfe, M.; Doré, K.; Buxton, J.A.; Pollari, F.; Henry, B.; Middleton, D.; Ahmed, R.; Jamieson, F.; Ciebin, B.; et al. Increased Burden of Illness Associated with Antimicrobial-Resistant Salmonella enterica Serotype Typhimurium Infections. J. Infect. Dis. 2004, 189, 377–384. [Google Scholar] [CrossRef]
  334. Hussain, A.; Satti, L.; Hanif, F.; Zehra, N.M.; Nadeem, S.; Bangash, T.M.; Peter, A. Typhoidal Salmonella strains in Pakistan: An impending threat of extensively drug-resistant Salmonella Typhi. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 2145–2149. [Google Scholar] [CrossRef]
  335. Gantois, I.; Ducatelle, R.; Pasmans, F.; Haesebrouck, F.; Gast, R.; Humphrey, T.J.; Van Immerseel, F. Mechanisms of egg contamination by Salmonella Enteritidis. FEMS Microbiol. Rev. 2009, 33, 718–738. [Google Scholar] [CrossRef]
  336. Pakbin, B.; Brück, W.M.; Brück, T.B. Molecular Mechanisms of Shigella Pathogenesis; Recent Advances. Int. J. Mol Sci. 2023, 24, 2448. [Google Scholar] [CrossRef]
  337. Mattock, E.; Blocker, A.J. How Do the Virulence Factors of Shigella Work Together to Cause Disease? Front. Cell. Infect. Microbiol. 2017, 7, 64. [Google Scholar] [CrossRef]
  338. Panhotra, B.R.; Desai, B. Resistant Shigella dysenteriae. Lancet 1983, 2, 1420. [Google Scholar] [CrossRef] [PubMed]
  339. Johansson, A. Aggregatibacter actinomycetemcomitans leukotoxin: A powerful tool with capacity to cause imbalance in the host inflammatory response. Toxins 2011, 3, 242–259. [Google Scholar] [CrossRef] [PubMed]
  340. Pietiäinen, M.; Kopra, K.A.E.; Vuorenkoski, J.; Salminen, A.; Paju, S.; Mäntylä, P.; Buhlin, K.; Liljestrand, J.M.; Nieminen, M.S.; Sinisalo, J.; et al. Aggregatibacter actinomycetemcomitans serotypes associate with periodontal and coronary artery disease status. J. Clin. Periodontol. 2018, 45, 413–421. [Google Scholar] [CrossRef] [PubMed]
  341. Han, E.C.; Choi, S.Y.; Lee, Y.; Park, J.W.; Hong, S.H.; Lee, H.J. Extracellular RNAs in periodontopathogenic outer membrane vesicles promote TNF-α production in human macrophages and cross the blood-brain barrier in mice. FASEB J. 2019, 33, 13412–13422. [Google Scholar] [CrossRef] [PubMed]
  342. Oscarsson, J.; Claesson, R.; Lindholm, M.; Höglund Åberg, C.; Johansson, A. Tools of Aggregatibacter actinomycetemcomitans to Evade the Host Response. J. Clin. Med. 2019, 8, 1079. [Google Scholar] [CrossRef] [PubMed]
  343. Soukup, J.; Cerny, J.; Cegan, M.; Kelbich, P.; Novotny, T. Toxocariasis as a Rare Parasitic Complication of a Transthoracic Spine Surgery Procedure. Medicina 2021, 57, 1328. [Google Scholar] [CrossRef] [PubMed]
  344. Sioutis, S.; Reppas, L.; Bekos, A.; Soulioti, E.; Saranteas, T.; Koulalis, D.; Sapkas, G.; Mavrogenis, A.F. Echinococcosis of the spine. EFORT Open Rev. 2021, 6, 288–296. [Google Scholar] [CrossRef]
  345. Buttenschoen, K.; Carli Buttenschoen, D. Echinococcus granulosus infection: The challenge of surgical treatment. Langenbeck’s Arch. Surg. 2003, 388, 218–230. [Google Scholar] [CrossRef]
  346. Junghanss, T.; da Silva, A.M.; Horton, J.; Chiodini, P.L.; Brunetti, E. Clinical management of cystic echinococcosis: State of the art, problems, and perspectives. Am. J. Trop. Med. Hyg. 2008, 79, 301–311. [Google Scholar] [CrossRef]
  347. Moro, P.; Schantz, P.M. Echinococcosis: A review. Int. J. Infect. Dis. 2009, 13, 125–133. [Google Scholar] [CrossRef]
  348. Brunetti, E.; Praticò, L.; Neumayr, A.; Maestri, M.; Tamarozzi, F. Update on treatment for cystic echinococcosis of the liver. Curr. Treat. Options Infect. Dis. 2016, 8, 153–164. [Google Scholar] [CrossRef]
  349. McManus, D.P.; Gray, D.J.; Zhang, W.; Yang, Y. Diagnosis, treatment, and management of echinococcosis. BMJ Br. Med. J. 2012, 344, e3866. [Google Scholar] [CrossRef]
  350. Trigkidis, K.; Geladari, E.; Kokkinakis, E.; Vallianou, N. Visceral Leishmaniasis in a patient with rheumatoid arthritis undergoing treatment with methotrexate: Case report and review of the literature. Eur. J. Rheumatol. 2017, 4, 139–141. [Google Scholar] [CrossRef]
  351. Hsieh, P.H.; Huang, K.C.; Shih, H.N. Prosthetic joint infection in patients with rheumatoid arthritis: An outcome analysis compared with controls. PLoS ONE 2013, 8, e71666. [Google Scholar] [CrossRef]
  352. Periferakis, A.; Caruntu, A.; Periferakis, A.-T.; Scheau, A.-E.; Badarau, I.A.; Caruntu, C.; Scheau, C. Availability, Toxicology and Medical Significance of Antimony. Int. J. Environ. Res. Public Health 2022, 19, 4669. [Google Scholar] [CrossRef] [PubMed]
  353. Gross, C.E.; Della Valle, C.J.; Rex, J.C.; Traven, S.A.; Durante, E.C. Fungal Periprosthetic Joint Infection: A Review of Demographics and Management. J. Arthroplast. 2021, 36, 1758–1764. [Google Scholar] [CrossRef]
  354. Koutserimpas, C.; Naoum, S.; Giovanoulis, V.; Raptis, K.; Alpantaki, K.; Dretakis, K.; Vrioni, G.; Samonis, G. Fungal Periprosthetic Hip Joint Infections. Diagnostics 2022, 12, 2341. [Google Scholar] [CrossRef]
  355. Chen, S.C.; Sorrell, T.C. Antifungal agents. Med. J. Aust. 2007, 187, 404. [Google Scholar] [CrossRef] [PubMed]
  356. Gupta, A.K.; Tomas, E. New antifungal agents. Dermatol. Clin. 2003, 21, 565–576. [Google Scholar] [CrossRef]
  357. Graham, S.M.; Leonidou, A.; Aslam-Pervez, N.; Hamza, A.; Panteliadis, P.; Heliotis, M.; Mantalaris, A.; Tsiridis, E. Biological therapy of bone defects: The immunology of bone allo-transplantation. Expert. Opin. Biol. Ther. 2010, 10, 885–901. [Google Scholar] [CrossRef] [PubMed]
  358. Ebrahimi, A.; Hosseini, S.A.; Rahim, F. Immunosuppressive therapy in allograft transplantation: From novel insights and strategies to tolerance and challenges. Cent. Eur. J. Immunol. 2014, 39, 400–409. [Google Scholar] [CrossRef]
  359. Baumer, V.; Gunn, E.; Riegle, V.; Bailey, C.; Shonkwiler, C.; Prawel, D. Robocasting of Ceramic Fischer-Koch S Scaffolds for Bone Tissue Engineering. J. Funct. Biomater. 2023, 14, 251. [Google Scholar] [CrossRef] [PubMed]
  360. Su, X.; Wang, T.; Guo, S. Applications of 3D printed bone tissue engineering scaffolds in the stem cell field. Regen. Ther. 2021, 16, 63–72. [Google Scholar] [CrossRef] [PubMed]
  361. Hwang, K.S.; Choi, J.W.; Kim, J.H.; Chung, H.Y.; Jin, S.; Shim, J.H.; Yun, W.S.; Jeong, C.M.; Huh, J.B. Comparative Efficacies of Collagen-Based 3D Printed PCL/PLGA/β-TCP Composite Block Bone Grafts and Biphasic Calcium Phosphate Bone Substitute for Bone Regeneration. Materials 2017, 10, 421. [Google Scholar] [CrossRef] [PubMed]
  362. Khan, M.U.A.; Aslam, M.A.; Bin Abdullah, M.F.; Hasan, A.; Shah, S.A.; Stojanović, G.M. Recent perspective of polymeric biomaterial in tissue engineering—A review. Mater. Today Chem. 2023, 34, 101818. [Google Scholar] [CrossRef]
  363. Tack, P.; Victor, J.; Gemmel, P.; Annemans, L. 3D-printing techniques in a medical setting: A systematic literature review. Biomed. Eng. Online 2016, 15, 115. [Google Scholar] [CrossRef] [PubMed]
  364. Roca, M.; Villegas, L.; Kortabitarte, M.; Althaus, R.; Molina, M. Effect of heat treatments on stability of β-lactams in milk. J. Dairy Sci. 2011, 94, 1155–1164. [Google Scholar] [CrossRef]
  365. Tappa, K.; Jammalamadaka, U.; Weisman, J.A.; Ballard, D.H.; Wolford, D.D.; Pascual-Garrido, C.; Wolford, L.M.; Woodard, P.K.; Mills, D.K. 3D printing custom bioactive and absorbable surgical screws, pins, and bone plates for localized drug delivery. J. Funct. Biomater. 2019, 10, 17. [Google Scholar] [CrossRef]
  366. Abdullah, T.; Qurban, R.O.; Bolarinwa, S.O.; Mirza, A.A.; Pasovic, M.; Memic, A. 3D printing of metal/metal oxide incorporated thermoplastic nanocomposites with antimicrobial properties. Front. Bioeng. Biotechnol. 2020, 8, 568186. [Google Scholar] [CrossRef]
  367. Ballard, D.H.; Tappa, K.; Boyer, C.J.; Jammalamadaka, U.; Hemmanur, K.; Weisman, J.A.; Alexander, J.S.; Mills, D.K.; Woodard, P.K. Antibiotics in 3D-printed implants, instruments and materials: Benefits, challenges and future directions. J. 3D Print. Med. 2019, 3, 83–93. [Google Scholar] [CrossRef]
  368. Huang, W.; Zheng, Q.; Sun, W.; Xu, H.; Yang, X. Levofloxacin implants with predefined microstructure fabricated by three-dimensional printing technique. Int. J. Pharm. 2007, 339, 33–38. [Google Scholar] [CrossRef]
  369. Ferrara, S.D.; Baccino, E.; Bajanowski, T.; Boscolo-Berto, R.; Castellano, M.; De Angel, R.; Pauliukevičius, A.; Ricci, P.; Vanezis, P.; Vieira, D.N.; et al. Malpractice and medical liability. European Guidelines on Methods of Ascertainment and Criteria of Evaluation. Int. J. Leg. Med. 2013, 127, 545–557. [Google Scholar] [CrossRef]
  370. The European Parliament and the Council of the European Union. Regulation (EU) No 536/2014 of the European Parliament and of the Council of 16 April 2014 on Clinical Trials on Medicinal Products for Human Use, and Repealing Directive 2001/20/EC Text with EEA relevance. In OJL158; The European Parliament and the Council of the European Union: Brussels, Belgium, 2014; pp. 1–76. [Google Scholar]
  371. The European Parliament and the Council of the European Union. Regulation (EU) 2017/745 of the European Parliament and of the Council of 5 April 2017 on medical devices, amending Directive 2001/83/EC, Regulation (EC) No 178/2002 and Regulation (EC) No 1223/2009 and repealing Council Directives 90/385/EEC and 93/42/EEC (Text with EEA relevance). In OJL117; The European Parliament and the Council of the European Union: Brussels, Belgium, 2017; pp. 1–175. [Google Scholar]
  372. The European Parliament and the Council of the European Union. Directive 2011/24/EU of the European Parliament and of the Council of 9 March 2011 on the application of patients’ rights in cross-border healthcare. In OJL88; The European Parliament and the Council of the European Union: Brussels, Belgium, 2011; pp. 45–65. [Google Scholar]
  373. Active Citizenship Network. European Charter of Patients’ Rights; Active Citizenship Network: Rome, Italy, 2002. [Google Scholar]
  374. Campoccia, D.; Montanaro, L.; Speziale, P.; Arciola, C.R. Antibiotic-loaded biomaterials and the risks for the spread of antibiotic resistance following their prophylactic and therapeutic clinical use. Biomaterials 2010, 31, 6363–6377. [Google Scholar] [CrossRef]
  375. Kulkarni, P.G.; Paudel, N.; Magar, S.; Santilli, M.F.; Kashyap, S.; Baranwal, A.K.; Zamboni, P.; Vasavada, P.; Katiyar, A.; Singh, A.V. Overcoming Challenges and Innovations in Orthopedic Prosthesis Design: An Interdisciplinary Perspective. Biomed. Mater. Devices 2024, 2, 58–69. [Google Scholar] [CrossRef]
  376. Fanelli, U.; Pappalardo, M.; Chinè, V.; Gismondi, P.; Neglia, C.; Argentiero, A.; Calderaro, A.; Prati, A.; Esposito, S. Role of Artificial Intelligence in Fighting Antimicrobial Resistance in Pediatrics. Antibiotics 2020, 9, 767. [Google Scholar] [CrossRef]
  377. Lv, J.; Deng, S.; Zhang, L. A review of artificial intelligence applications for antimicrobial resistance. Biosaf. Health 2021, 3, 22–31. [Google Scholar] [CrossRef]
  378. Rabaan, A.A.; Alhumaid, S.; Mutair, A.A.; Garout, M.; Abulhamayel, Y.; Halwani, M.A.; Alestad, J.H.; Bshabshe, A.A.; Sulaiman, T.; AlFonaisan, M.K.; et al. Application of Artificial Intelligence in Combating High Antimicrobial Resistance Rates. Antibiotics 2022, 11, 784. [Google Scholar] [CrossRef]
  379. Yanez, M.; Blanchette, J.; Jabbarzadeh, E. Modulation of Inflammatory Response to Implanted Biomaterials Using Natural Compounds. Curr. Pharm. Des. 2017, 23, 6347–6357. [Google Scholar] [CrossRef]
  380. Marrazzo, P.; O’Leary, C. Repositioning Natural Antioxidants for Therapeutic Applications in Tissue Engineering. Bioengineering 2020, 7, 104. [Google Scholar] [CrossRef]
  381. Košćak, L.; Lamovšek, J.; Đermić, E.; Godena, S. The Antibacterial Effect of Selected Essential Oils and Their Bioactive Constituents on Pseudomonas savastanoi pv. savastanoi: Phytotoxic Properties and Potential for Future Olive Disease Control. Microorganisms 2023, 1, 2735. [Google Scholar] [CrossRef]
  382. Molokoane, T.L.; Kemboi, D.; Siwe-Noundou, X.; Famuyide, I.M.; McGaw, L.J.; Tembu, V.J. Extractives from Artemisia afra with Anti-Bacterial and Anti-Fungal Properties. Plants 2023, 12, 3369. [Google Scholar] [CrossRef]
  383. Nissanka, M.C.; Weerasekera, M.M.; Dilhari, A.; Dissanayaka, R.; Rathnayake, S.; Wijesinghe, G.K. Phytomedicinal properties of Hygrophila schulli (Neeramulliya). Iran. J. Basic Med. Sci. 2023, 26, 979–986. [Google Scholar] [CrossRef]
  384. Gulati, R.K.; Jain, N.; Singh, A.; Jain, A.; Jindal, A.; Kumawat, M.K.; Paiwal, K. Comparative Evaluation of the Antimicrobial Efficacy of Chemical and Phytomedicinal Agents When Used As Intracanal Irrigants: An In Vitro Study. Cureus 2023, 15, e48754. [Google Scholar] [CrossRef]
  385. Ismail, J.; Shebaby, W.N.; Daher, J.; Boulos, J.C.; Taleb, R.; Daher, C.F.; Mroueh, M. The Wild Carrot (Daucus carota): A Phytochemical and Pharmacological Review. Plants 2023, 13, 10093. [Google Scholar] [CrossRef]
  386. Salaria, D.; Rolta, R.; Lal, U.R.; Dev, K.; Kumar, V. A comprehensive review on traditional applications, phytochemistry, pharmacology, and toxicology of Thymus serpyllum. Indian J. Pharmacol. 2023, 55, 385–394. [Google Scholar] [CrossRef] [PubMed]
  387. Periferakis, A.T.; Periferakis, A.; Periferakis, K.; Caruntu, A.; Badarau, I.A.; Savulescu-Fiedler, I.; Scheau, C.; Caruntu, C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients 2023, 15, 4097. [Google Scholar] [CrossRef] [PubMed]
  388. Alonso-Villegas, R.; González-Amaro, R.M.; Figueroa-Hernández, C.Y.; Rodríguez-Buenfil, I.M. The Genus Capsicum: A Review of Bioactive Properties of Its Polyphenolic and Capsaicinoid Composition. Molecules 2023, 28, 4239. [Google Scholar] [CrossRef] [PubMed]
  389. Yasin, M.; Li, L.; Donovan-Mak, M.; Chen, Z.H.; Panchal, S.K. Capsicum Waste as a Sustainable Source of Capsaicinoids for Metabolic Diseases. Foods 2023, 12, 907. [Google Scholar] [CrossRef] [PubMed]
  390. Bayram Sarıipek, F. Biopolymeric nanofibrous scaffolds of poly(3-hydroxybuthyrate)/chitosan loaded with biogenic silver nanoparticle synthesized using curcumin and their antibacterial activities. Int. J. Biol. Macromol. 2023, 256, 128330. [Google Scholar] [CrossRef]
  391. Luo, L.; Wang, M.; Su, W.; Zhuo, J.; Zhang, L.; Zhu, W.; Zhang, W.; Wang, R.; Wang, J. Thermal-Driven Curcumin Release Film with Dual-Mode Synergistic Antibacterial Behavior for Efficient Tangerine Preservation. J. Agric. Food. Chem. 2024. [Google Scholar] [CrossRef]
  392. Ding, P.; Ding, X.; Li, J.; Guo, W.; Okoro, O.V.; Mirzaei, M.; Sun, Y.; Jiang, G.; Shavandi, A.; Nie, L. Facile preparation of self-healing hydrogels based on chitosan and PVA with the incorporation of curcumin-loaded micelles for wound dressings. Biomed. Mater. 2024, 19, 025021. [Google Scholar] [CrossRef] [PubMed]
  393. Periferakis, A.; Periferakis, K.; Badarau, I.A.; Petran, E.M.; Popa, D.C.; Caruntu, A.; Costache, R.S.; Scheau, C.; Caruntu, C.; Costache, D.O. Kaempferol: Antimicrobial Properties, Sources, Clinical, and Traditional Applications. Int. J. Mol Sci. 2022, 23, 15054. [Google Scholar] [CrossRef] [PubMed]
  394. Periferakis, A.; Periferakis, A.T.; Troumpata, L.; Periferakis, K.; Scheau, A.E.; Savulescu-Fiedler, I.; Caruntu, A.; Badarau, I.A.; Caruntu, C.; Scheau, C. Kaempferol: A Review of Current Evidence of Its Antiviral Potential. Int. J. Mol Sci. 2023, 24, 16299. [Google Scholar] [CrossRef]
  395. Song, J.M.; Lee, K.H.; Seong, B.L. Antiviral effect of catechins in green tea on influenza virus. Antivir. Res. 2005, 68, 66–74. [Google Scholar] [CrossRef] [PubMed]
  396. Isaacs, C.E.; Wen, G.Y.; Xu, W.; Jia, J.H.; Rohan, L.; Corbo, C.; Di Maggio, V.; Jenkins, E.C., Jr.; Hillier, S. Epigallocatechin gallate inactivates clinical isolates of herpes simplex virus. Antimicrob. Agents Chemother. 2008, 52, 962–970. [Google Scholar] [CrossRef] [PubMed]
  397. Golestannezhad, N.; Divsalar, A.; Badalkhani-Khamseh, F.; Rasouli, M.; Seyedarabi, A.; Ghalandari, B.; Ding, X.; Goli, F.; Bekeschus, S.; Movahedi, A.A.M.; et al. Oxali-palladium nanoparticle synthesis, characterization, protein binding, and apoptosis induction in colorectal cancer cells. J. Mater. Sci. Mater. Med. 2024, 35, 4. [Google Scholar] [CrossRef]
  398. Saravanakumar, K.; Li, Z.; Kim, Y.; Park, S.; Keon, K.; Lee, C.M.; Ahn, G.; Cho, N. Fucoidan-coated cotton dressing functionalized with biomolecules capped silver nanoparticles (LB-Ag NPs-FN-OCG) for rapid healing therapy of infected wounds. Environ. Res. 2023, 246, 118004. [Google Scholar] [CrossRef]
  399. Ieven, M.; Vlietinck, A.J.; Vanden Berghe, D.A.; Totte, J.; Dommisse, R.; Esmans, E.; Alderweireldt, F. Plant antiviral agents. III. Isolation of alkaloids from Clivia miniata Regel (Amaryllidaceae). J. Nat. Prod. 1982, 45, 564–573. [Google Scholar] [CrossRef]
  400. Ren, G.; Ding, G.; Zhang, H.; Wang, H.; Jin, Z.; Yang, G.; Han, Y.; Zhang, X.; Li, G.; Li, W. Antiviral activity of sophoridine against enterovirus 71 in vitro. J. Ethnopharmacol. 2019, 236, 124–128. [Google Scholar] [CrossRef]
  401. Lee, J.; Lee, J.H.; Lee, S.Y.; Park, S.A.; Kim, J.H.; Hwang, D.; Kim, K.A.; Kim, H.S. Antioxidant Iron Oxide Nanoparticles: Their Biocompatibility and Bioactive Properties. Int. J. Mol. Sci. 2023, 24, 15901. [Google Scholar] [CrossRef] [PubMed]
  402. Ghosh, S.; Patil, S.; Ahire, M.; Kitture, R.; Kale, S.; Pardesi, K.; Cameotra, S.S.; Bellare, J.; Dhavale, D.D.; Jabgunde, A. Synthesis of silver nanoparticles using Dioscorea bulbifera tuber extract and evaluation of its synergistic potential in combination with antimicrobial agents. Int. J. Nanomed. 2012, 7, 483–496. [Google Scholar]
  403. Chuan, L.; Zhang, J.; Yu-Jiao, Z.; Shu-Fang, N.; Jun, C.; Qian, W.; Shao-Ping, N.; Ze-Yuan, D.; Ming-Yong, X.; Shu, W. Biocompatible and biodegradable nanoparticles for enhancement of anti-cancer activities of phytochemicals. Chin. J. Nat. Med. 2015, 13, 641–652. [Google Scholar]
  404. Nath, R.; Roy, R.; Barai, G.; Bairagi, S.; Manna, S.; Chakraborty, R. Modern Developments of Nano Based Drug Delivery System by Combined with Phytochemicals-Presenting New Aspects. Int. J. Sci. Res. Sci. Technol. 2021, 8, 107–129. [Google Scholar] [CrossRef]
  405. Matei, A.-M.; Caruntu, C.; Tampa, M.; Georgescu, S.R.; Matei, C.; Constantin, M.M.; Constantin, T.V.; Calina, D.; Ciubotaru, D.A.; Badarau, I.A. Applications of nanosized-lipid-based drug delivery systems in wound care. Appl. Sci. 2021, 11, 4915. [Google Scholar] [CrossRef]
  406. Calanna, F.; Chen, F.; Risitano, S.; Vorhies, J.S.; Franceschini, M.; Giori, N.J.; Indelli, P.F. Debridement, antibiotic pearls, and retention of the implant (DAPRI): A modified technique for implant retention in total knee arthroplasty PJI treatment. J. Orthop. Surg. 2019, 27, 2309499019874413. [Google Scholar] [CrossRef]
  407. Indelli, P.F.; Ghirardelli, S.; Valpiana, P.; Bini, L.; Festini, M.; Iannotti, F. Debridement, Antibiotic Pearls, and Retention of the Implant (DAPRI) in the Treatment of Early Periprosthetic Joint Infections: A Consecutive Series. Pathogens 2023, 12, 605. [Google Scholar] [CrossRef]
  408. Agarwal, S.; Healey, B. The use of antibiotic impregnated absorbable calcium sulphate beads in management of infected joint replacement prostheses. J. Arthrosc. Jt. Surg. 2014, 1, 72–75. [Google Scholar] [CrossRef]
  409. Piovan, G.; Farinelli, L.; Screpis, D.; Marocco, S.; Motta, L.; Palazzolo, G.; Natali, S.; Zorzi, C. The role of antibiotic calcium sulfate beads in acute periprosthetic knee infection: A retrospective cohort study. Arthroplasty 2022, 4, 42. [Google Scholar] [CrossRef]
  410. Bint, A.J.; Burtt, I. Adverse antibiotic drug interactions. Drugs 1980, 20, 57–68. [Google Scholar] [CrossRef]
  411. Aronson, J. Serious drug interactions. Practitioner 1993, 237, 789–791. [Google Scholar]
  412. Zebrowska-Łupina, I.; Szymczyk, G.; Wróbel, A. Adverse effects of interactions of antibiotics with other drugs. Pol. Merkur. Lek. 2000, 9, 623–626. [Google Scholar]
  413. Ament, P.W.; Bertolino, J.G.; Liszewski, J.L. Clinically significant drug interactions. Am. Fam. Physician 2000, 61, 1745–1754. [Google Scholar] [PubMed]
  414. Kuscu, F.; Ulu, A.; Inal, A.S.; Suntur, B.M.; Aydemir, H.; Gul, S.; Ecemis, K.; Komur, S.; Kurtaran, B.; Ozkan Kuscu, O.; et al. Potential Drug-Drug Interactions with Antimicrobials in Hospitalized Patients: A Multicenter Point-Prevalence Study. Med. Sci. Monit. 2018, 24, 4240–4247. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Antimicrobial substances can be either applied as a coating after printing the final construct (1) or inoculated directly into the initial mix (2). The figure also presents a summary of the properties of the models resulting from this method. The 3D printing technique given as an example in this figure is based on the principles of vat-photopolymerization (DLP, SLA).
Figure 1. Antimicrobial substances can be either applied as a coating after printing the final construct (1) or inoculated directly into the initial mix (2). The figure also presents a summary of the properties of the models resulting from this method. The 3D printing technique given as an example in this figure is based on the principles of vat-photopolymerization (DLP, SLA).
Biomimetics 09 00154 g001
Table 1. Current principal functions of biomaterials for 3D printing discussed in this paper.
Table 1. Current principal functions of biomaterials for 3D printing discussed in this paper.
BiomaterialCurrent UsesReferences
Beta-tricalcium phosphate (β-TCP)Bone defect filling and repairing, bone tissue engineering and bone scaffold manufacturing, bone grafts[89,90,91,92,93,94,95,96]
Biphasic calcium phosphate (BCP)Bone scaffold manufacturing, bone grafts manufacturing, tissue engineering[47,48,97,98,99]
HydroxyapatiteBone tissue engineering and bone scaffold manufacturing, joint replacement surgeries[100,101,102,103,104]
Lithium disilicate glassBone scaffold manufacturing, dental applications[105,106]
Polyetheretherketone (PEEK)Spinal cages, skull/maxillofacial defect and dental implants, joint replacements, fracture healing support plates, spinal fusions[107,108,109,110,111,112,113,114,115]
Poly(propylene fumarate) (PPF)Bone tissue engineering, biocompatible scaffolds[116,117,118,119]
Poly(trimethylene carbonate) (PTMC)Bone tissue engineering, bone tissue implants[120,121,122]
ZirconiaHip head prostheses, orthopedic implants, dental implants[123,124]
Table 2. Modifications of β-TCP with antimicrobial properties.
Table 2. Modifications of β-TCP with antimicrobial properties.
ModificationDosage and CompoundsSettingTested
Microorganism
YearReference
Antibiotic coating and combinationsGentamycinIn vitro, in vivon/a1996[135]
260 ± 48 μg of gatifloxacine hydrate per ceramic diskIn vitro, in vivoS. milleri, B. fragilis2008[129]
1 wt.% vancomycin hydrochlorideIn vitroS. aureus (MRSA)2013[132]
5 mg/mL concentration of vancomycin solutionIn vitro, in vivoS. aureus2018[134]
1–5 wt.% ciprofloxacinIn vitroS. aureus2021[130]
300 mg vancomycin hydrochloride per 1 mL waterIn vitro, in vivoS. aureus2022[133]
1 wt.% tetracyclineIn vitroP. gingivalis2024[131]
Metal coatings and combinations0.49 and 1.09 wt.% FeIn vitroE. coli, S. enteritidis, P. aeruginosa, S. aureus2019[138]
1 wt.% B nitrate microtubulesIn vitroS. aureus2020[137]
Ag nanoparticles as part of β-TCP hydrogelIn vitroS. aureus, B. subtilis, P. aeruginosa, E. coli2020[141]
5 and 10 wt.% nanosized AgIn vitro, in vivoS. aureus, E. coli2020[139]
1.4 wt.% ZnIn vitroE. faecium, E. coli, P. aeruginosa2021[136]
0.1, 1, 10 wt.% AgIn vitroS. aureus (MRSA)2022[140]
Combination with chitosan2 wt.% chitosan solution
(3.0 g TCP based on
10.0 g chitosan)
In vitron/a–theorized antibacterial use2012[142]
3 g of chitosan per membraneIn vitron/a–theorized antibacterial use2019[143]
Combinations with glass or other materials2.5 wt.% β-TCP added into a PP (core layer) solutionIn vitroS. aureus, S. mutans2018[147]
Ceramic suspensions with solids content of 30% wt.% In vitroS. aureus, E. coli, C. albicans2021[144]
Transparent bioglass sol used to impregnate the β-TCP scaffoldsIn vitro, in vivoC. albicans, P. aeruginosa, S. aureus2023[145]
Bioactive glass S53P4In vitroS. aureus2023[146]
n/a—not available.
Table 3. Modifications of BCP with antimicrobial properties.
Table 3. Modifications of BCP with antimicrobial properties.
ModificationDosage and CompoundsSettingTested
Microorganism
YearReference
Antibiotic coating and combinationsVancomycin in 90 mg loaded microparticlesIn vitron/a2001[152]
Metal coatings and combinations1.06 wt.% AgIn vitroS. aureus2021[154]
Variable concentration of Ag ionsIn vitroS. aureus, S. epidemidis, E. coli2023[153]
Combination with chitosan3 g of chitosan in each membraneIn vitro, in vivon/a2019[143]
4 w/v% chitosanIn vitron/a2022[155]
n/a—not available.
Table 4. Modifications of hydroxyapatite with antimicrobial properties.
Table 4. Modifications of hydroxyapatite with antimicrobial properties.
ModificationDosage and CompoundsSettingTested
Microorganism
YearReferences
Antibiotic coating and combinationsCiprofloxacin 30 wt.%In vitroS. aureus, E. coli2019[185]
CiprofloxacinIn vivo, in vitroGram negative and Gram-negative bacteria2023[186]
Metal coatings and combinationsCo replacement at 5% and 12%In vitroS. aureus, E. coli2016[175]
0.04, 0.08, 0.16, 0.24 wt.% Te contentIn vitroB. subtilis, S. aureus, Micrococcus sp., P. aeruginosa, Klebsiella sp., S. dysenteriae, Candida albicans2017[178]
Cu addition to specific molar ratioIn vitroS. aureus, E. coli2017[176]
Mg addition to specific molar ratioIn vitroS. aureus, E. faecalis, E. coli, P. aeruginosa, Candida albicans2019[179]
Ag nanoparticles in different concentrationsIn vitroS. aureus2021[181]
Zn doping at 0.25, 0.5 and 1.0 mmol/LIn vitroS. aureus, E. coli2021[173]
Ag ions in various concentrationsIn vitroS. aureus, E. coli2021[182]
Doping with Ga and ZnIn vitroS. aureus, E. coli2022[183]
Various metalsIn vitro, in vivoVarious microbes2022[184]
ZnO 5 wt.%In vitroS. aureus, E. coli2022[174]
Ni, Sn, and Mo ions in 500, 1000 and 2000 ppmIn vitroS. aureus, P. aeruginosa2023[172]
Ti dopingIn vitro, in vivoVarious microbes2023[177]
Combination with chitosanCellulose–chitosan–hydroxyapatite composite materialIn vitroS. aureus (MRSA), VRE, E. coli, P. aeruginosa2013[187]
Chitosan and HAP gel at 4:6 mass ratio In vitro, in vivoS. aureus, S. epidermidis, P. aeruginosa, C. albicans2016[188]
Combinations with other materials and compoundsAg nanoparticles at 5%In vitron/a–theorized antibacterial use2012[191]
10 mL lactoferrin per 50 mg of hydroxyapatiteIn vitron/a–theorized antibacterial use2017[192]
63 mg/g of baicaleinIn vitroS. epidermidis2021[189]
Different combinations of HAP and algaeIn vitroGram-negative, gram-positive bacteria2021[194]
Table 6. Modifications of zirconia and lithium disilicate with antimicrobial properties.
Table 6. Modifications of zirconia and lithium disilicate with antimicrobial properties.
ModificationDosage and CompoundsSettingTested
Microorganism
YearReference
Lithium combination with glass nanoparticlesDifferent concentrations of Li2O were used to replace Na2O in the glass structureIn vitron/a–theorized antibacterial use2016[105]
Zirconia with antibacterial nanomodificationAn aqueous solution of a mixture of 3Y-ZrO2 nanopowder and ammonium citrate (dispersant)In vitro, in vivoE. coli, S. aureus2019[239]
Zirconia with Ag nanoparticles3 g/L silver nanoparticlesIn vitroE. coli, S. aureus2021[241]
Zirconia surface modification with a chitosan-containing compound5 distinct groups, each with a different mixtureIn vitroP. gingivalis, A. actinomycetemcomitans2023[240]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Periferakis, A.; Periferakis, A.-T.; Troumpata, L.; Dragosloveanu, S.; Timofticiuc, I.-A.; Georgatos-Garcia, S.; Scheau, A.-E.; Periferakis, K.; Caruntu, A.; Badarau, I.A.; et al. Use of Biomaterials in 3D Printing as a Solution to Microbial Infections in Arthroplasty and Osseous Reconstruction. Biomimetics 2024, 9, 154. https://doi.org/10.3390/biomimetics9030154

AMA Style

Periferakis A, Periferakis A-T, Troumpata L, Dragosloveanu S, Timofticiuc I-A, Georgatos-Garcia S, Scheau A-E, Periferakis K, Caruntu A, Badarau IA, et al. Use of Biomaterials in 3D Printing as a Solution to Microbial Infections in Arthroplasty and Osseous Reconstruction. Biomimetics. 2024; 9(3):154. https://doi.org/10.3390/biomimetics9030154

Chicago/Turabian Style

Periferakis, Argyrios, Aristodemos-Theodoros Periferakis, Lamprini Troumpata, Serban Dragosloveanu, Iosif-Aliodor Timofticiuc, Spyrangelos Georgatos-Garcia, Andreea-Elena Scheau, Konstantinos Periferakis, Ana Caruntu, Ioana Anca Badarau, and et al. 2024. "Use of Biomaterials in 3D Printing as a Solution to Microbial Infections in Arthroplasty and Osseous Reconstruction" Biomimetics 9, no. 3: 154. https://doi.org/10.3390/biomimetics9030154

Article Metrics

Back to TopTop