Next Article in Journal
Preparation and Performance of a Cr/CrN/TiAlCN Composite Coating on a GCr15 Bearing Steel Surface
Previous Article in Journal
Tribological Properties and Surface Wettability of Coatings Produced on the Mg-AZ31B Alloy by Plasma Electrolytic Oxidation
Previous Article in Special Issue
Characterisation of TiCN Coatings for Biomedical Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress on the Preparation Process and Material Structure of 3D-Printed Dental Implants and Their Clinical Applications

School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Coatings 2024, 14(7), 781; https://doi.org/10.3390/coatings14070781
Submission received: 13 May 2024 / Revised: 19 June 2024 / Accepted: 20 June 2024 / Published: 21 June 2024
(This article belongs to the Special Issue Bioactive Coatings on Elements Used in the Oral Cavity Environment)

Abstract

:
Additive manufacturing, commonly known as 3D printing technology, has become a prominent topic of research globally in recent years and is playing an increasingly important role in various industries. Particularly within the healthcare sector, the use of 3D printing technology is gaining prominence, with a special focus on the manufacturing and application of dental implants. As research in this field progresses, the preparation methods, material selection, and technological innovations for dental implants are evolving, promising a future where the manufacturing process of dental implants becomes even more refined and efficient. Through thorough research in materials science, it is possible to develop dental implant materials that have better biocompatibility with the human body and improved mechanical properties. Additionally, advancements in surface modification technology can further enhance the strength and stability of the bond between dental implants and bone tissue. These advancements not only expand treatment options for patients but also greatly improve the long-term success rate of dental implants. In the field of dental implants, the success of the implant depends on the interactions between the materials used, the cells involved, and the bone tissue. Therefore, there is an urgent need to explore the molecular mechanisms of such interactions in depth. In this study, we provide a comprehensive review of the application of 3D printing technology in the fabrication of dental implants. This includes an examination of the process methods, surface coating technology, and a comparison of the shapes and structures of different dental implants, along with their advantages and disadvantages. Furthermore, this paper analyzes the intrinsic mechanisms of successful dental implant placement in clinical practice, and it highlights the latest progress in the clinical application of 3D-printed dental implants. Undeniably, the use of 3D-printed dental implants not only offers patients more precise and personalized treatment plans but also brings revolutionary changes to the development of the medical industry.

1. Introduction

Oral problems are a growing concern in today’s society. A healthy mouth not only promotes good eating habits but also affects a person’s appearance [1]. Prosthetic restoration of missing teeth can be achieved by means of cemented dentures, removable dentures, and dental implants, with dental implants being the most desirable restorative method. In 1969, Brånemark et al. [2] first proposed the concept of osseointegration by summarizing the good results obtained from implant trials in male canines, and since the concept of osseointegration was proposed, the development of dental implants has had a relatively clear direction.
Currently, dental implants continue to gain attention in dentistry [3]. Simply, dental implants consist of the implant body and its supporting superstructure (crown and abutment), also known as an artificial tooth root, made of artificial material and surgically implanted into the jawbone in the area of the missing tooth. Dental implants provide an effective solution for restoring chewing function, offering superior strength and stability compared to other traditional restoration materials. In addition, implants can greatly beautify the patient’s appearance, are more convenient in daily life than traditional dentures, protect the remaining natural teeth, prevent bone loss, and restore facial bone structure. It is important to understand that dental implants provide a restoration that is very similar in function, structure, and aesthetics to a natural tooth, and the structural comparison between a healthy tooth and a dental implant is shown in Figure 1.
In recent years, attention has been drawn to the rise of 3D printing technology, which is capable of creating complex 3D structures directly from computational models, a feature that has led to the search for alternatives to the traditional mechanical fabrication of dental implants [4]. The processes used for 3D printing of implants are numerous [5]. The most used 3D printing technology processes are stereolithography (SLA), selective laser sintering (SLS), fused deposition molding (FDM), etc., and the principles and methods of the various processes are not the same. Relying on 3D printing technology, various types of implants already exist, with different implant structures, combination methods, and materials used. For example, many investigations and clinical studies have identified titanium as a biomaterial for oral rehabilitation, along with other materials such as zirconia, polyether ether ketone, and carbon nanotubes [6].
Successful implant placement is the result of a combination of factors, and although implantology is rapidly evolving and many surgical protocols are being improved, there are several potential complications that can compromise the stability of dental implants. Studies have proposed that a large part of early implant failure is due to bacterial infection, but of course, this undesirable outcome is the result of a number of cofactors [7,8].
Osseointegration, which pertains to oral implants, is a fundamental component of modern reconstructive dentistry [9]. Osseointegration was originally defined as direct contact between vital bone and weight-bearing implants observed at the histological level [10]. Insua et al. [11] presented an update on the molecular mechanisms occurring during osseointegration, focusing on the relevance of osteoblasts and immune cells in the process of bone maintenance. However, the biology and metabolism of peri-implant bone healing and its impact on bone loss at the peri-implant margin has been less well studied.
In this review, we examine the latest developments in clinically 3D-printed dental implants and whether they can be more effectively personalized to treat complex clinical problems. We highlight process methods, surface modifications, new types of dental implant, and advances in the clinical treatment and prognosis of dental implants with 3D printing technology to provide ideas for further research.

2. Three-Dimensionally (3D)-Printed Dental Implant Technology and Surface Modification

2.1. The Process Methods of 3D-Printed Dental Implants

2.1.1. Selective Laser Melting

Selective laser melting (SLM) is an advanced 3D printing technique that uses a high-power laser to fuse metallic powders layer by layer. This process allows for the production of precise dental implant structures, overcoming the challenges associated with manufacturing metal parts [12]. The processing flow of SLM technology is illustrated in Figure 2 [13]. Commonly, materials like titanium and its alloys are employed in SLM-based dental implants due to their biocompatibility and resistance to corrosion. For instance, Qin et al. [14] conducted a study demonstrating the successful fabrication of titanium dental implants using SLM technology, resulting in a precise fit and osseointegration. The study emphasized the significance of optimizing laser parameters and powder characteristics to achieve the desired implant properties. One major advantage of SLM is its ability to create personalized implant designs based on individual patient anatomy. By utilizing patient-specific 3D imaging data, SLM can manufacture implants with customized geometries, improving the overall success rate and patient satisfaction [15]. SLM has attracted significant attention in dental implantology due to its capability to produce complex geometries with excellent biocompatibility and mechanical properties [16]. However, SLM also faces challenges. For instance, the high cost of SLM machines and materials may hinder its widespread adoption in some dental practices. Additionally, post-processing steps may be necessary to enhance the surface finish and eliminate residual stresses, thereby adding complexity to the manufacturing process [17].

2.1.2. Electron Beam Melting

Electron beam melting (EBM) is an advanced 3D printing technique that shows great promise in the field of dental implant fabrication. This innovative technology uses a high-power electron beam to selectively melt and fuse metallic powder particles layer by layer, resulting in the precise manufacturing of complex dental implant structures [18]. Figure 3 illustrates the EBM process [18]. EBM is commonly employed to produce dental implants using biocompatible materials like titanium and its alloys. One of EBM’s significant advantages is its ability to create implants with excellent mechanical properties and biocompatibility. The implants manufactured through EBM exhibit high strength, corrosion resistance, and osseointegration potential, making them ideal for long-term implantation [19]. The customization potential of EBM is also noteworthy. By utilizing patient-specific 3D imaging data, dental professionals can tailor implant designs to match individual anatomical requirements. This personalized approach enhances the fit, stability, and overall success rate of dental implants [20]. Moreover, EBM allows for the fabrication of lattice structures within the implant. These lattice structures not only reduce the implant’s weight but also promote bone ingrowth, facilitating faster healing and osseointegration [21]. Several studies have demonstrated the successful application of EBM in dental implantology. For instance, Li et al. [22] conducted a study utilizing EBM-fabricated titanium dental implants. The results showed the EBM implants not only diminished stress shielding but also demonstrated suitable osteoconductive properties. While EBM offers significant advantages, it is crucial to consider certain challenges. The equipment and materials required for EBM can be relatively expensive, which may limit its widespread adoption in smaller dental practices. Additionally, post-processing steps may be necessary to remove residual stresses and achieve the desired surface finish [23].

2.1.3. Selective Laser Sintering

Selective laser sintering (SLS) is a highly anticipated 3D printing technology that uses a high-power laser to selectively sinter metal powder particles layer by layer. This achieves the bonding of high-melting-point metals and ceramics [24]. The process is illustrated in Figure 4 [24]. Compared to other technologies, parts produced through laser sintering have several advantages, including good performance, fast production speed, a wide range of materials, and low cost. In the field of oral implants, SLS commonly employs biocompatible materials such as titanium and its alloys. Implants made using this method have shown excellent stability and osteogenic activity [25]. Furthermore, SLS technology’s high precision and chromatographic resolution enable the fabrication of microstructures. This is crucial in simulating the shape and function of natural teeth, which helps to enhance patients’ oral comfort and chewing effectiveness. Patients not only receive natural-looking implants but also benefit from an improved oral experience. We have summarized the current research on these three process methods in Table 1.

2.2. Implant Surface Modification Methods

2.2.1. Physical Modification

Sandblasting, plasma treatment, and laser modification are commonly used physical techniques for modifying implant surfaces [37].
Sandblasting involves using compressed air to spray particles of blast material, such as steel grit or emery, onto the implant surface to alter its roughness [38]. Gil et al. demonstrated that sandblasting increases the contact area between the implant and surrounding bone tissue, promoting the adhesion and proliferation of osteoblasts and improving the implant’s ability to integrate with the bone [39]. Additionally, sandblasting is often combined with acid etching to remove any residual blast material [40,41,42,43]. In a retrospective clinical study, Buser et al. [44] evaluated the outcomes of 511 SLA (sandblasted, large grit, acid-etched) implants in 303 partially edentulous patients over a 10-year period. The authors reported a success rate of 97.0% and a 10-year implant survival rate of 98.8%. The incidence of peri-implantitis was as low as 1.8%.
Plasma treatment can involve both spraying and implantation [45]. Plasma spraying uses a plasma arc generated by ionized inert gases as a heat source to melt or semi-melt materials such as ceramics, alloys, or metals, which are then sprayed onto the pre-treated implant surface to form a firmly adhered coating [46]. Hydroxyapatite (HA) coatings, commonly used in clinical practice, are created by spraying HA particles onto the implant surface at high temperatures and then rapidly cooling them [47,48,49,50]. Lu et al. [51] demonstrated that a tantalum-doped HA-coated implant, created through plasma spraying, not only had improved roughness and wettability but also promoted osteogenesis and differentiation of bone marrow mesenchymal stem cells. Plasma implantation involves injecting ion beams directly into the implant, causing changes in its surface composition, structure, and properties [52,53]. Jin et al. [54] used ion implantation to inject Zn and Ag ions into titanium, forming a Zn/Ag film that greatly enhanced the osteogenic activity and antibacterial ability of the material. Wan et al. [55] injected Cu and Ag ions into titanium alloy separately, and the modified samples exhibited good antibacterial properties against Escherichia coli and Staphylococcus aureus.
Laser modification is a technique that has gained notable attention in recent years. This method employs laser micromachining to create precise nano- and micro-scale features on implant surfaces. Implants with collar surfaces treated through laser micromachining to produce nano-channels have demonstrated improved integration and stability [56,57]. The advantages of laser modification of implants are extensively documented, including enhanced osseointegration, improved mechanical interlocking between the implant and bone, and decreased bacterial adhesion [58,59]. A study by Koodaryan R et al. [60] showed that laser microtexturing of the dental implant collar significantly improved crestal bone levels and peri-implant health. Botos et al. [61] found that the application of laser-microtextured grooves to the implant collar resulted in shallower probing depths and less peri-implant crestal bone loss than that seen around implants with machined collars. These findings indicate that implants with laser-modified surfaces exhibit a superior peri-implant tissue response, thereby contributing to the long-term success and stability of the implants [62].

2.2.2. Chemical Modification

Chemical modification techniques for implants can be classified into two main categories: electrochemical modification, and acid–base solution treatment [63].
In terms of electrochemical modification techniques, commonly used methods include anodic oxidation, micro-arc oxidation, and electrophoretic deposition [64,65,66,67].
Anodic oxidation is a method that involves the formation of an oxide film on the implant surface through discharge oxidation [68]. This oxide film can alter the implant’s surface color, corrosion resistance, hardness, and other properties [69,70,71,72]. It is worth noting that the anodic oxidation method is often used to create titanium dioxide nanotube arrays. These nanotubes have a hollow tubular structure, which provides a large specific surface area and high adsorption capacity, greatly improving the implant’s bioactivity [73,74,75,76,77]. In a retrospective study conducted by Wagenberg et al. [78], 312 anodic oxidized surface implants were analyzed among 1187 implants that were immediately placed after tooth extraction. Radiographic comparisons over 2–12 years (with an average of 7.4 years) revealed that these implants experienced significantly less mesial–distal bone loss (0.4 mm) compared to machined implants (0.6 mm) following the same immediate placement protocol.
Micro-arc oxidation (MAO) is a modification method that utilizes the transient high-temperature effect generated by arc light discharge to form an oxide film with a dense inner layer and porous outer layer on the implant surface. This technique is considered to be an upgrade to anodic oxidation technology [79]. The micro/nano bioactive titanium coating formed through MAO modification promotes the adhesion of osteoblasts on the implant surface and enhances the osteogenic activity of the implant [80,81,82]. Furthermore, MAO can be combined with other coating methods. Huang et al. [83] conducted a study investigating the surface morphology, chemical properties, and cellular interactions of coatings prepared through MAO and hydrothermal treatment. The in vitro and in vivo results indicated that the encapsulated implant exhibited enhanced protein adsorption, osteoblast activity, adhesion, and differentiation. Moreover, it facilitated early osseointegration, improved bioactivity, and enhanced osseointegration compared to implants with MAO alone. Hu et al. [84] combined ultrasound with MAO to create coatings on the surface of Ti-Cu alloys, which demonstrated strong long-term antimicrobial properties and were non-toxic. Electrophoretic deposition (EPD) is a modification method that deposits charged particles onto the surface of an implant using an electric field [85]. EPD not only ensures a uniform coating but also controls the thickness of the coating [86]. Common coatings applied using EPD include HA, graphene oxide (GO), and Ag [87,88,89,90]. Juliadmi et al. [91] used EPD to deposit HA coatings of natural origin onto implant surfaces, resulting in good surface coverage and improved coating performance. EPD is also capable of coating more complex shapes. Nicoli et al. [92] utilized EPD technology to apply a novel semi-transparent coating on a 3D-printed Ti alloy mesh for guided alveolar bone regeneration.
In terms of acid and alkaline solution treatments, commonly used modifications are acid etching and alkaline heat treatment [93,94].
Acid etching is a common method that enhances the biocompatibility of implants [95]. By immersing the metal implant in hydrofluoric, sulfuric, nitric, or mixed acid solutions, a chemical reaction occurs, resulting in increased surface roughness of the implant [96,97]. Yan et al. [98] discovered that acid-etched Ti alloy implants did not significantly affect hydrophilicity, but they did promote macrophage adherence and polarization while reducing reactive oxygen species (ROS) levels. However, it is important to control the reaction time and conditions during acid etching treatment to prevent overreaction and damage to the implant surface [99].
Alkaline heat treatment is a modification method that involves immersing the metal implant in a strong alkaline solution for a certain period, followed by heat treatment at 300–800 °C to obtain a porous oxide layer [100]. Nishio et al. [101] observed that alkaline heat treatment greatly increased the surface roughness of titanium, and that the resulting micrometer-sized porous structure provided nucleation sites for apatite deposition when immersed in a simulated body fluid. Zhang et al. [102] conducted continuous alkaline heat treatment, pre-calcification, and simulated body fluid immersion to obtain HA-coated porous titanium. Implanting porous titanium and HA-coated porous titanium into a tibial defect model in New Zealand white rabbits, it was found that the latter effectively reduced the fracture risk and enhanced bone healing.

3. The Structure, Material, and Shape of the Implant

3.1. Classification of the Implant Structure

First, according to the structure of the implant, there are three types of implant: one-stage implants, two-stage non-embedded implants, and two-stage embedded implants. Two-stage implants do not support the attachment of fixed screws, so the single-stage small-diameter implants are generally considered to be more robust than the two-stage implants [103]. For the three types of implant, see Figure 5 below, and the advantages and disadvantages of the three implant types are compared in Table 2.

3.1.1. One-Stage Implants

The base and threaded parts of an implant are manufactured as one piece, which allows for simultaneous processing and eliminates the need for later assembly. The placement of the implant in the clinic only requires a single surgical procedure, which is relatively minor and minimizes the patient’s response to the implant. After surgery, the abutment is exposed to the oral cavity through the gums. The abutment may experience external forces that cause micromotion, which can affect the integration of the implant with the bone [104]. By performing only one operation for a short duration, the pain can be reduced. There is no need for additional surgery, resulting in less pain and shorter healing time for the patient. Overall, this treatment reduces the cost of treatment. Although this approach involves a staged procedure, there is still a microgap between the implant and the prosthetic abutment at the level of the bone crest [105].
However, because the abutment is directly exposed to the oral cavity, it is susceptible to external fluctuations that can influence the ability of the implant to fuse with the surrounding bone tissue within a specific time period. Ensuring successful implantation in a single operation can be challenging [106]. Kawakita et al. used CT scans to examine the bone morphology around the first mandibular molars and maxillary incisors. They found that selecting an appropriate implant diameter and length based on the bone morphology helped improve the success rate of the implants. Based on 10 years of clinical experience with the prototype implant system developed in the mid-1980s by the ITI® Dental Implant System (Straumann, Switzerland), three basic implant shapes are available: solid screw, cannulated screw, and hollow cylinder implants [107]. These implants were evaluated not only for survival but also for their prospective success rates. Detailed analysis showed that solid screw implants had the best results compared to cannulated screw and hollow cylinder implants. When comparing implants of different lengths, the 12 mm long implants had the highest 8-year success rate [108].

3.1.2. Two-Stage Non-Embedded Implants

A two-stage non-embedded implant denture can complete the implantation in one operation, eliminating the need for a second operation to repair the upper part of the structure at the oral junction. This significantly reduces the entire treatment cycle [109]. The denture is connected to healthy teeth through a dental bridge, which holds it to the alveolar bone. The stability of the denture depends on the condition of the surrounding teeth or other forms of support. Soft tissue healing occurs simultaneously with bone tissue healing, promoting the closure of the soft tissue. Compared to underwater implants, non-underwater implants have a higher abutment connection plane and only require one connection suture. This creates a more favorable biological width and height of the gingival margin, eliminating the need for two surgeries and reducing the treatment time. However, this implant approach requires high levels of oral hygiene to prevent plaque formation around the healing abutment during implant healing, which can cause inflammation in the peri-implant soft tissue [110]. Additionally, this type of implant requires good dental conditions and may not be suitable for everyone.
Zhang et al. [111] summarized the success rate of non-embedded implant dentures. In addition to correct treatment design, good perioperative care measures are also crucial to ensure the success of the implant surgery and form a strong implant–bone interface. This provides a solid bone support system for future implant denture repairs and enables good chewing function.

3.1.3. Two-Stage Embedded Implants

A two-stage embedded implant denture involves two separate surgeries to complete the implant placement [112]. In the first stage, an artificial implant is fully embedded in the alveolar bone, and a healing period follows. In the second stage, an incision is made to expose the implant in the gums, and the final crown connector is repaired. This type of implant, with a strong structural implant integrated into the bone, provides stability similar to that of a natural root. The embedded implant is thus more stable and capable of supporting normal chewing and bite force. Additionally, this two-stage process separates the healing environment of the implant from the external environment, preventing bacterial attacks and reducing the risk of infection. It also allows for independent healing of the implant, without being affected by bite force. This promotes initial stability and minimizes the chances of osseointegration failure or retention of fibrous bone. However, it is important to note that the two-stage implant denture requires a second operation, resulting in a longer treatment time. Moreover, the second operation involves another incision of the soft tissue, which may potentially cause secondary damage to the soft tissue.

3.2. Classification of Implant Length

According to the classification of implant length, there are four categories: ultra-short, short, standard, and long (Table 3). Implants measuring 6 mm or less are classified as ultra-short, those measuring greater than 6 mm and less than 10 mm are classified as short, those measuring 10 mm or more but less than 13 mm are classified as standard, and those measuring 13 mm or more are classified as long [113].

3.3. Implant Diameter Classification

Implants are classified based on their diameter and can be categorized into four groups: ultra-narrow, narrow, standard, and wide (Table 4). Implants with a diameter of less than 3.0 mm are considered ultra-narrow, while those measuring 3.0 mm to 3.75 mm are classified as narrow. Implants less than 5 mm in diameter are considered standard, and those equal to or greater than 5 mm are classified as wide [113].

3.4. New Dental Implant Materials

3.4.1. PMMA-ZrO Biomimetic Nanocomposites

PMMA (polymethylmethacrylate) is a commonly used polymer material in the medical field [114]. It has good biocompatibility and stability. ZrO (zirconia), on the other hand, is a ceramic material with excellent mechanical properties and chemical stability, often used for high-end dental and orthopedic implants. By combining PMMA and ZrO, a biomimetic nanocomposite material can be created, which fully exploits the advantages of both materials [115]. This material inherits the biocompatibility and stability of PMMA while also possessing the high mechanical strength and chemical stability of ZrO.

3.4.2. Zirconia–Glass Ceramic Composite Material

The zirconia–glass ceramic composite is a combination of ZrO (Li2Si2O5) and a glass ceramic material (lithium silicate or lithium disilicate). This combination results in a material that combines the high strength, high hardness, and good biocompatibility of zirconia with the light transmittance and natural appearance of the glass phase [116]. Compared to traditional total zirconia ceramic materials, this composite material provides a more natural appearance and better light transmission, making restored teeth more realistic. Additionally, this material is easier to process and repair, offering more flexibility and convenience. It has good biocompatibility and is widely used in dental restoration, such as creating tooth crowns and implant bridges. Its excellent performance ensures that restored teeth not only look realistic but also function effectively, meeting the daily needs of patients.
With the continuous development of materials science and oral clinical medicine, the application prospects for new dental implant materials will continue to expand. In the future, this material is expected to play an even greater role in improving the aesthetic effect of restorations, enhancing biocompatibility, and extending the service life.

3.4.3. Titanium Alloy Dental Implant Material

The use of titanium (Ti) alloy as a dental implant material offers numerous advantages due to its excellent performance. It not only prevents damage to healthy teeth but also protects the integrity of the surrounding teeth [117]. Additionally, it restores the ability to chew in the area where a tooth is missing, allowing patients to eat and digest normally. Titanium implants can also serve as brackets, providing firmness to the teeth and simulating the pressure stimulation that real teeth provide. Moreover, they help maintain the stability and shape of the jawbone. Titanium has been the preferred material for dental implants for many years due to its favorable mechanical and biological properties, resulting in high clinical success rates. As surface development and temperature enhancement continue to progress, modifications are being made to improve the speed and degree of osseointegration, therefore enhancing the choices and outcomes of clinical treatment. Li et al. found that the tested titanium material exhibited reduced bacterial counts and reduced biofilm development compared to the control materials [118], Ti and PEEK. Furthermore, the experiment confirmed that the survival and success of a Ti implant in the implant area corresponded to those of an implant in natural bone.

3.4.4. Glass and Ceramic Materials

Glass and ceramic implant materials have good biocompatibility, bioreactivity, aesthetic effects, and bonding performance [119], and they are not easy to denature after sterilization, while also having strong compression strength, hardness, wear resistance, and stable chemical properties, but the materials are brittle and easy to fold. They can meet the patient’s aesthetic and functional needs and are a worthwhile choice for restorative dentistry. In addition, they have rich sources, simple production processes, low cost, and their mechanical properties and process integrity also meet the clinical requirements. The temperature system and master phase of glass and ceramic dental materials were characterized by Wang et al. [120], who discussed how to determine the main phase of the glass and ceramic materials according to the performance requirements of the dental materials and use it to select the glass composition and the corresponding crystallization temperature system. Compared with metal and other alloy materials, their color is closer to that of natural teeth, and most of them have a guiding osteogenesis effect, which has been a hot spot in the development of biomaterials in recent years.

3.5. Different Structures of the Tooth Root

The design of the root profile of the implant directly affects the initial stability of the implant. This is determined by the shape of the portion of the implant placed approximately 3–5 mm above the alveolar socket. The design of the root profile, thread design, and self-tapping characteristics all contribute to this stability [121].
In a study by Zhong et al. [122], double-root 3D tomography, micro-computed tomography (micro-CT), and histological analysis of a 3D printed implant were conducted. It was found that the stability and bone remodeling around the fixation of the 3D-printed double-root implant were comparable.
Implants are made from a wide variety of materials. In Table 5, we have summarized the advantages and disadvantages of different types of implants.

4. Progress in the Clinical and Molecular Mechanisms of Dental Implants

4.1. Development of Dental Implant Surgery

Generally, prior to dental implant surgery, oral CT is required along with a routine oral examination to ensure the success of the procedure [123]. In practice, the dental implant procedure is not complicated, but it does take some time. First, the dental implant can only be placed three months after the tooth extraction [1,124]; the procedure begins with anesthesia, and then the mucosa is incised on the gingiva of the missing tooth area, the alveolar bone is exposed, the implant socket is prepared, and the implant is placed. After the completion of the implant surgery, approximately three months of waiting is necessary to allow for osseointegration between the alveolar bone and the implant. Subsequently, a 3D model is created, and the most suitable simulated crown is calculated, which is then fitted onto the implant.
New 3D printing techniques have been rapidly advancing, and current surgical protocols are evolving quickly. Recent studies have demonstrated that piezoelectric surgery can minimize trauma and enhance postoperative healing more effectively than traditional drilling techniques. For instance, Maglione et al. [125] found that the ultrasonic piezoelectric approach yielded positive results in implant surgery when compared to the conventional technique. By comparing operative time and postoperative pain between the two methods (conventional drilling and ultrasonic implantation), they observed significant differences. In another study, Fujiwara et al. [126] experimentally compared the osseointegration level and marginal bone in implants using either conventional drilling or piezoelectric devices. They concluded that utilizing piezoelectric devices for implant placement is a safe procedure.
As digital technology continues to develop, personalized treatment is receiving more attention in dental implant surgery, and robot-assisted surgery may emerge as a new trend. Robotic surgery offers higher precision and stability, reducing the influence of human factors and enhancing the success rate of surgeries.

4.2. Postoperative Evaluation and Patient Expectations of Implants

Implant failures can be classified into early and late failures. Many risk factors for early implant failure listed in clinical studies are not systemic diseases, such as diabetes and hypertension, or bone-related factors, such as placement site, implant diameter, and length. Instead, they are related to bone augmentation, such as the technique and graft material used. Clauser et al. [127] conducted a systematic evaluation and meta-analysis, which found a significant association between bone enlargement procedures and early implant failure. Munakata et al. [128] summarized the factors related to early implant failure and concluded that various surgical choices of graft materials and appropriate surgical techniques can help prevent it. Late failure is mainly caused by bacterial infection. Other causes include excessive occlusal forces, poor plaque control, and overloading due to an insufficient number of implants or the effects of implant diameter and length [129,130,131].
Furthermore, implant dentistry is a relatively new field in oral healthcare, and patients often have limited experience or understanding before undergoing implant treatment. Yao et al. [132] conducted a study and found that many patients highlighted the high cost of implants in their postoperative evaluations. Many also expressed a desire for enhanced therapeutic function and improved aesthetics. McCrea et al. [133] designed a post-treatment questionnaire and conducted a statistical analysis, concluding that there was a direct relationship between patients’ expectations of treatment outcomes and their satisfaction upon the completion of treatment. Significant relationships were found between appearance, comfort, and overall satisfaction, with p = 0.001. Moreover, a highly significant relationship was found between comfort level and the patient’s overall experience, with p = 0.001. These studies suggest that the quality of communication between the patient and the healthcare provider plays a crucial role in determining patient satisfaction. Kashbour et al. [134] conducted face-to-face and telephone interviews with over 40 patients to explore their feelings and thoughts about dental implant surgery. They found that many patients are increasingly interested in implants as a solution for missing teeth; therefore, it is essential to establish a complete and accurate understanding of implants with patients.
The postoperative evaluation of dental implants has a significant impact on the further development of the field. Therefore, it is important to assess patient satisfaction with the treatment and identify areas for improvement in both the implant itself and the procedure.

4.3. Mechanisms of Implant Osseointegration

The maxillofacial skeleton is a highly dynamic system that requires a delicate balance between bone resorption and bone formation. It is important to understand the complex interactions among dental implants, bone, and the immune system. Osseointegration, for example, is considered to be an immune process that can result in the formation of new bone on the surface of the implant. It is not simply a single bone response. The success of an osseointegrated implant largely depends on the process of bone formation and remodeling [135].
Biologically, the fate of the implant is determined by the interaction between bone cells, immune cells, and the implant surface. Luis et al. [136] proposed that dendritic cells and macrophages, which are immune sentinels in the peri-implant environment, play a crucial role in determining the longevity of the implant. Wang et al. [137] conducted a study on dental implant osseointegration and found that macrophages play a dual role in regulating the bone healing process and the immune response to implant placement. On the other hand, research has shown that an ion-treated implant can modulate pro-regenerative immune responses and optimize osseointegration. For example, high concentrations of magnesium (Mg) on the implant surface have been shown to reduce the secretion of pro-inflammatory cytokines such as TNF-α, IL-1b, IL-6, and PEG2 [138].
In recent years, several studies have suggested a connection between hypertension and periodontal tissue metabolism and disease. Saravi et al. [139] conducted a retrospective analysis comparing a group of patients taking β-blockers and renin–angiotensin system (RAS) inhibitors with a control group. The results showed that bone remodeling and osseointegration were enhanced in the group taking RAS inhibitors (Figure 6). Therefore, the use of anti-hypertensive medication may reduce the risk of dental implant failure due to enhanced bone remodeling.
The mechanisms of oral osseointegration are complex and involve various cellular and molecular processes specific to the oral and maxillofacial structures [140]. A study has also shown that porous titanium surfaces can recruit bone progenitor cells, which, when differentiated into osteoblasts, produce woven bone with the influence of bone morphogenetic proteins, vascular endothelial growth factor, and other specific osteoprotegerins [141]. Recognizing this complexity is important for designing specialized implant structures and achieving successful therapeutic outcomes in implant placement and prevention of oral diseases.

5. Conclusions

This review offers a unique perspective on the use of 3D printing technology in designing innovative dental implants. It examines the potential future advancements in the clinical application of dental implants. It begins by providing an overview of the 3D printing workflow, with a particular focus on implant coating methods and the preparation of implants using various materials, such as metals and bioceramics. It also highlights the different implant configurations available. The subsequent section summarizes recent research on the clinical application of dental implants, covering surgical approaches, patient expectations, and osseointegration studies that push the boundaries of knowledge and technology. These findings can serve as a foundation for future studies, which can incorporate emerging technologies and employ the best techniques to create superior implants. This will undoubtedly drive rapid progress in dental implant research, leading to more precise clinical treatments in the near future.

Funding

This research was funded by the Natural Science Foundation of Sichuan Province, China (2022NSFSC1510); the Medical Scientific Research Project of Chengdu City, China (2021043); and the Innovation Team Project of the Clinical Medical College and Affiliated Hospital of Chengdu University (CDFYCX202208).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhao, X.; Qiao, S.C.; Shi, J.Y. Evaluation of the clinical and aesthetic outcomes of Straumann Standard Plus implant supported single crowns placed in nonaugmented healed sites in the anterior maxilla: A 5–8-year retrospective study. Clin. Oral Implant. Res. 2016, 27, 106–112. [Google Scholar] [CrossRef]
  2. Brånemark, P.I.; Adell, R.; Breine, U.; Hansson, B.O.; Lindström, J.; Ohlsson, A. Intra-osseous anchorage of dental prostheses: I. Experimental studies. Scand. J. Plast. Reconstr. Surg. 1969, 3, 81–100. [Google Scholar] [CrossRef]
  3. Peng, T.Y.; Lin, D.J.; Mine, Y.; Tasi, C.Y.; Li, P.J.; Shih, Y.H.; Chiu, K.C.; Wang, T.H.; Hsia, S.M.; Shieh, T.M. Biofilm Formation on the Surface of (Poly)Ether-Ether-Ketone and In Vitro Antimicrobial Efficacy of Photodynamic Therapy on Peri-Implant Mucositis. Polymers 2021, 13, 940. [Google Scholar] [CrossRef]
  4. Yang, F.; Chen, C.; Zhou, Q.; Gong, Y.; Li, R.; Li, C.; Klämpfl, F.; Freund, S.; Wu, X.; Sun, Y.; et al. Laser beam melting 3D printing of Ti6Al4V based porous structured dental implant: Fabrication, biocompatibility analysis and photoelastic study. Sci. Rep. 2017, 7, 45360. [Google Scholar] [CrossRef]
  5. Wang, Z.; Yang, Y. Application of 3D Printing in Implantable Medical Devices. BioMed Res. Int. 2021, 2021, 6653967. [Google Scholar] [CrossRef]
  6. Cao, J.; Lu, Y.; Chen, H.; Zhang, L.; Xiong, C. Preparation, properties and in vitro cellular response of multi-walled carbon nanotubes/bioactive glass/poly(etheretherketone) biocomposite for bone tissue engineering. Int. J. Polym. Mater. Polym. Biomater. 2018, 68, 433–441. [Google Scholar] [CrossRef]
  7. Baqain, Z.H.; Moqbel, W.Y.; Sawair, F.A. Early dental implant failure: Risk factors. Br. J. Oral Maxillofac. Surg. 2012, 50, 239–243. [Google Scholar] [CrossRef]
  8. Nchingolo, F.; Tatullo, M.; Abenavoli, F.M.; Marrelli, M.; Inchingolo, A.D.; Palladino, A.; Inchingolo, A.M.; Dipalma, G. Oral piercing and oral diseases: A short time retrospective study. Int. J. Med. Sci. 2011, 8, 649–652. [Google Scholar]
  9. Setzer, F.C.; Kim, S. Comparison of long-term survival of implant and endodontically treated teeth. Dent. Res. 2014, 93, 19–26. [Google Scholar] [CrossRef]
  10. Albrektsson, T.; Brånemark, P.I.; Hansson, H.A.; Lindström, J. Osseointegrated titanium implant. requirements for ensuring a long-lasting, direct bone-to-implant anchorage in man. Acta Orthop. Scand. 1981, 52, 155–170. [Google Scholar] [CrossRef]
  11. Insua, A.; Monje, A.; Wang, H.L.; Miron, R.J. Basis of bone metabolism around dental implant during osseointegration and peri-implant bone loss. J. Biomed. Mater. Res. Part A 2017, 105, 2075–2089. [Google Scholar] [CrossRef]
  12. Gao, B.; Zhao, H.; Peng, L.; Sun, Z. A Review of Research Progress in Selective Laser Melting (SLM). Micromachines 2023, 14, 57. [Google Scholar] [CrossRef]
  13. Bremen, S.; Meiners, W.; Diatlov, A. Selective laser melting: A manufacturing technology for the future. Laser Tech. J. 2012, 9, 33–38. [Google Scholar] [CrossRef]
  14. Qin, Z.G.; He, Y.; Gao, J.J.; Dong, Z.H.; Long, S.; Cheng, L.J.; Shi, Z. Surface modification improving the biological activity and osteogenic ability of 3D printing porous dental implants. Front. Mater. 2023, 10, 2296–8016. [Google Scholar] [CrossRef]
  15. Zhang, J.; Song, B.; Wei, Q.; Bourell, D.L.; Shi, Y. A review of selective laser melting of aluminum alloys: Processing, microstructure, property and developing trends. J. Mater. Sci. Technol. 2019, 35, 270–284. [Google Scholar] [CrossRef]
  16. Vandenbroucke, B.; Kruth, J.P. Selective laser melting of biocompatible metals for rapid manufacturing of medical parts. Rapid Prototyp. J. 2007, 13, 196–203. [Google Scholar] [CrossRef]
  17. Zadpoor, A.A.; Malda, J. Additive manufacturing of biomaterials, tissues, and organs. Ann. Biomed. Eng. 2017, 45, 1–11. [Google Scholar] [CrossRef]
  18. Tamayo, J.A.; Riascos, M.; Vargas, C.A.; Baena, L.M. Additive manufacturing of Ti6Al4V alloy via electron beam melting for the development of implant for the biomedical industry. Heliyon 2021, 7, e06892. [Google Scholar] [CrossRef]
  19. Galati, M.; Luca, I. A literature review of powder-based electron beam melting focusing on numerical simulations. Addit. Manuf. 2018, 19, 1–20. [Google Scholar] [CrossRef]
  20. Suska, F.; Kjeller, G.; Tarnow, P.; Hryha, E.; Nyborg, L.; Snis, A.; Palmquist, A. Electron Beam Melting Manufacturing Technology for Individually Manufactured Jaw Prosthesis: A Case Report. J. Oral Maxillofac. Surg. 2016, 74, 1706.e1–1706.e15. [Google Scholar] [CrossRef]
  21. Cheong, V.S.; Fromme, P.; Mumith, A.; Coathup, M.J.; Blunn, G.W. Novel adaptive finite element algorithms to predict bone ingrowth in additive manufactured porous implants. J. Mech. Behav. Biomed. Mater. 2018, 87, 230–239. [Google Scholar] [CrossRef]
  22. Li, X.; Feng, Y.F.; Wang, C.T.; Li, G.C.; Lei, W.; Zhang, Z.Y.; Wang, L. Evaluation of biological properties of electron beam melted Ti6Al4V implant with biomimetic coating in vitro and in vivo. PLoS ONE 2012, 7, e52049. [Google Scholar] [CrossRef]
  23. Lan, L.; Jin, X.; Gao, S.; He, B.; Rong, Y. Microstructural evolution and stress state related to mechanical properties of electron beam melted Ti-6Al-4V alloy modified by laser shock peening. J. Mater. Sci. Technol. 2020, 50, 153–161. [Google Scholar] [CrossRef]
  24. Fina, F.; Madla, C.M.; Goyanes, A.; Zhang, J.; Gaisford, S.; Basit, A.W. Fabricating 3D printed orally disintegrating printlets using selective laser sintering. Int. J. Pharm. 2018, 541, 101–107. [Google Scholar] [CrossRef]
  25. Ou, P.; Liu, J.; Hao, C.; He, R.; Chang, L.; Ruan, J. Cytocompatibility, stability and osteogenic activity of powder metallurgy Ta-xZr alloys as dental implant materials. J. Biomater. Appl. 2021, 35, 790–798. [Google Scholar] [CrossRef]
  26. Ran, Q.C.; Yang, W.H.; Hu, Y.; Shen, X.K.; Yu, Y.L.; Xiang, Y.; Cai, K.Y. Osteogenesis of 3D printed porous Ti6Al4V implant with different pore sizes. J. Mech. Behav. Biomed. Mater. 2018, 84, 1–11. [Google Scholar] [CrossRef]
  27. Zhou, Y.; Tang, C.Z.; Deng, J.L.; Xu, R.G.; Yang, Y.; Deng, F.L. Micro/Nano Topography of Selective Laser Melting Titanium Inhibits Osteoclastogenesis via Mediation of Macrophage Polarization. Biochem. Biophys. Res. Commun. 2021, 581, 53–59. [Google Scholar] [CrossRef]
  28. Xu, R.; Hu, X.; Yu, X.; Wan, S.; Wu, F.; Ouyang, J.; Deng, F. Micro-/nano-topography of selective laser melting titanium enhances adhesion and proliferation and regulates adhesion-related gene expressions of human gingival fibroblasts and human gingival epithelial cells. Int. J. Nanomed. 2018, 13, 5045–5057. [Google Scholar] [CrossRef]
  29. Fukuda, A.; Takemoto, M.; Saito, T.; Fujibayashi, S.; Neo, M.; Pattanayak, D.K.; Matsushita, T.; Sasaki, K.; Nishida, N.; Kokubo, T.; et al. Osteoinduction of porous Ti implant with a channel structure fabricated by selective laser melting. Acta Biomater. 2011, 7, 2327–2336. [Google Scholar] [CrossRef]
  30. Sun, X.T.; Lin, H.S.; Zhang, C.Y.; Liu, Y.; Jin, J.; Di, S. A biomimetic hierarchical structure on selective laser melting titanium with enhanced hydrophilic/hydrophobic surface. J. Alloys Compd. 2022, 895, 162585. [Google Scholar] [CrossRef]
  31. Ren, B.; Wan, Y.; Liu, C.; Wang, H.; Yu, M.; Zhang, X.; Huang, Y. Improved osseointegration of 3D printed Ti-6Al-4V implant with a hierarchical micro/nano surface topography: An in vitro and in vivo study. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 118, 111505. [Google Scholar] [CrossRef] [PubMed]
  32. Goto, M.; Matsumine, A.; Yamaguchi, S.; Takahashi, H.; Akeda, K.; Nakamura, T.; Asanuma, K.; Matsushita, T.; Kokubo, T.; Sudo, A. Osteoconductivity of bioactive Ti-6Al-4V implant with lattice-shaped interconnected large pores fabricated by electron beam melting. J. Biomater. Appl. 2021, 35, 1153–1167. [Google Scholar] [CrossRef] [PubMed]
  33. Thomsen, P.; Malmström, J.; Emanuelsson, L.; René, M.; Snis, A. Electron beam-melted, free-form-fabricated titanium alloy implant: Material surface characterization and early bone response in rabbits. J. Biomed. Mater. Res. B Appl. Biomater. 2009, 90, 35–44. [Google Scholar] [CrossRef] [PubMed]
  34. Harada, Y.; Ishida, Y.; Miura, D.; Watanabe, S.; Aoki, H.; Miyasaka, T.; Shinya, A. Mechanical Properties of Selective Laser Sintering Pure Titanium and Ti-6Al-4V, and Its Anisotropy. Materials 2020, 13, 5081. [Google Scholar] [CrossRef] [PubMed]
  35. Li, J.; Li, Z.; Shi, Y.; Wang, H.; Li, R.; Tu, J.; Jin, G. In vitro and in vivo comparisons of the porous Ti6Al4V alloys fabricated by the selective laser melting technique and a new sintering technique. J. Mech. Behav. Biomed. Mater. 2019, 91, 149–158. [Google Scholar] [CrossRef] [PubMed]
  36. Chang Tu, C.; Tsai, P.I.; Chen, S.Y.; Kuo, M.Y.; Sun, J.S.; Chang, J.Z. 3D laser-printed porous Ti6Al4V dental implants for compromised bone support. J. Formos. Med. Assoc. 2020, 119, 420–429. [Google Scholar] [CrossRef]
  37. Wang, Q.; Zhou, P.; Liu, S.; Attarilar, S.; Ma, R.L.; Zhong, Y.; Wang, L. Multi-Scale Surface Treatments of Titanium Implants for Rapid Osseointegration: A Review. Nanomaterials 2020, 10, 1244. [Google Scholar] [CrossRef] [PubMed]
  38. Smeets, R.; Stadlinger, B.; Schwarz, F.; Beck-Broichsitter, B.; Jung, O.; Precht, C.; Kloss, F.; Gröbe, A.; Heiland, M.; Ebker, T. Impact of Dental Implant Surface Modifications on Osseointegration. Biomed. Res. Int. 2016, 2016, 6285620. [Google Scholar] [CrossRef] [PubMed]
  39. Gil, F.J.; Pérez, R.A.; Olmos, J.; Herraez-Galindo, C.; Gutierrez-Pérez, J.L.; Torres-Lagares, D. The effect of using Al2O3 and TiO2 in sandblasting of titanium dental implant. J. Mater. Res. 2022, 37, 2604–2613. [Google Scholar] [CrossRef]
  40. Robles-Ruíz, J.J.; Ciamponi, A.L.; Medeiros, I.S.; Kanashiro, L.K. Effect of lingual enamel sandblasting with aluminum oxide of different particle sizes in combination with phosphoric acid etching on indirect bonding of lingual brackets. Angle Orthod. 2014, 84, 1068–1073. [Google Scholar] [CrossRef] [PubMed]
  41. Wennerberg, A.; Galli, S.; Albrektsson, T. Current knowledge about the hydrophilic and nanostructured SLActive surface. Clin. Cosmet. Investig. Dent. 2011, 3, 59–67. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, E.W.; Wang, Y.B.; Shuai, K.G.; Gao, F.; Bai, Y.J.; Cheng, Y.; Xiong, X.L.; Zheng, F.; Wei, S.C. In vitro and in vivo evaluation of SLA titanium surfaces with further alkali or hydrogen peroxide and heat treatment. Biomed. Mater. 2011, 6, 025001. [Google Scholar] [CrossRef] [PubMed]
  43. Chang, Y.R.; Xu, F.F.; Li, J.; You, Y.H.; Liu, C.; Yin, L.H. Surface Morphology and Surface Properties of Ti and TiZr Implant Materials. Zhonghua Kou Qiang Yi Xue Za Zhi 2019, 54, 118–123. [Google Scholar] [PubMed]
  44. Buser, D.; Janner, S.F.M.; Wittneben, J.G.; Brägger, U.; Ramseier, C.A.; Salvi, G.E. 10-Year survival and success rates of 511 titanium implant with a sandblasted and acid-etched surface: A retrospective study in 303 partially edentulous patients. Clin. Implant. Dent. Relat. Res. 2012, 14, 839–851. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, H.; Jeon, H.J.; Jung, A.; Kim, J.; Kim, J.Y.; Lee, S.H.; Kim, H.; Yeom, M.S.; Choe, W.; Gweon, B.; et al. Improvement of osseointegration efficacy of titanium implant through plasma surface treatment. Biomed. Eng. Lett. 2022, 12, 421–432. [Google Scholar] [CrossRef] [PubMed]
  46. Schafer, S.; Swain, T.; Parra, M.; Slavin, B.V.; Mirsky, N.A.; Nayak, V.V.; Witek, L.; Coelho, P.G. Nonthermal Atmospheric Pressure Plasma Treatment of Endosteal Implant for Osseointegration and Antimicrobial Efficacy: A Comprehensive Review. Bioengineering 2024, 11, 320. [Google Scholar] [CrossRef] [PubMed]
  47. Hameed, P.; Gopal, V.; Bjorklund, S.; Ganvir, A.; Sen, D.; Markocsan, N.; Manivasagam, G. Axial Suspension Plasma Spraying: An ultimate technique to tailor Ti6Al4V surface with HAp for orthopaedic applications. Colloids Surf. B Biointerfaces 2019, 173, 806–815. [Google Scholar] [CrossRef] [PubMed]
  48. Groot, K.; Geesink, R.; Klein, C.P.; Serekian, P. Plasma sprayed coatings of hydroxylapatite. J. Biomed. Mater. Res. 1987, 21, 1375–1381. [Google Scholar] [CrossRef] [PubMed]
  49. Filiaggi, M.J.; Pilliar, R.M.; Coombs, N.A. Post-plasma-spraying heat treatment of the HA coating/Ti-6A1-4V implant system. J. Biomed. Mater. Res. 1993, 27, 191–198. [Google Scholar] [CrossRef] [PubMed]
  50. Unabia, R.B.; Candidato, R.T., Jr.; Pawłowski, L.; Salvatori, R.; Bellucci, D.; Cannillo, V. In vitro studies of solution precursor plasma-sprayed copper-doped hydroxyapatite coatings with increasing copper content. J. Biomed. Mater. Res. B Appl. Biomater. 2020, 108, 2579–2589. [Google Scholar] [CrossRef] [PubMed]
  51. Lu, R.J.; Wang, X.; He, H.X.; E, L.L.; Li, Y.; Zhang, G.L.; Li, C.J.; Ning, C.Y.; Liu, H.C. Tantalum-incorporated hydroxyapatite coating on titanium implant: Its mechanical and in vitro osteogenic properties. J. Mater. Sci. Mater. Med. 2019, 30, 111. [Google Scholar] [CrossRef] [PubMed]
  52. Han, W.; Shuobo, F.; Zhong, Q.; Qi, S. Influence of Dental Implant Surface Modifications on Osseointegration and Biofilm Attachment. Coatings 2022, 12, 1654. [Google Scholar] [CrossRef]
  53. Sotova, C.; Yanushevich, O.; Kriheli, N.; Grigoriev, S.; Evdokimov, V.; Kramar, O.; Nozdrina, M.; Peretyagin, N.; Undritsova, N.; Popelyshkin, E. Dental Implant: Modern Materials and Methods of Their Surface Modification. Materials 2023, 16, 7383. [Google Scholar] [CrossRef] [PubMed]
  54. Jin, G.; Qin, H.; Cao, H.; Qian, S.; Zhao, Y.; Peng, X.; Zhang, X.; Liu, X.; Chu, P.K. Synergistic effects of dual Zn/Ag ion implantation in osteogenic activity and antibacterial ability of titanium. Biomaterials 2014, 35, 7699–7713. [Google Scholar] [CrossRef] [PubMed]
  55. Wan, Y.Z.; Raman, S.; He, F.; Huang, Y. Surface modification of medical metals by ion implantation of silver and copper. Vacuum 2007, 81, 1114–1118. [Google Scholar] [CrossRef]
  56. Wang, C.; Hu, H.; Li, Z.; Shen, Y.; Xu, Y.; Zhang, G.; Zeng, X.; Deng, J.; Zhao, S.; Ren, T.; et al. Enhanced Osseointegration of Titanium Alloy Implants with Laser Microgrooved Surfaces and Graphene Oxide Coating. ACS Appl. Mater. Interfaces 2019, 11, 39470–39483. [Google Scholar] [CrossRef] [PubMed]
  57. Tzanakakis, E.-G.C.; Skoulas, E.; Pepelassi, E.; Koidis, P.; Tzoutzas, I.G. The Use of Lasers in Dental Materials: A Review. Materials 2021, 14, 3370. [Google Scholar] [CrossRef] [PubMed]
  58. Doll, K.; Fadeeva, E.; Stumpp, N.S.; Grade, S.; Chichkov, B.N.; Stiesch, M. Reduced bacterial adhesion on titanium surfaces micro-structured by ultra-short pulsed laser ablation. BioNanoMaterials 2016, 17, 53–57. [Google Scholar] [CrossRef]
  59. Kligman, S.; Ren, Z.; Chung, C.H.; Perillo, M.A.; Chang, Y.C.; Koo, H.; Zheng, Z.; Li, C. The Impact of Dental Implant Surface Modifications on Osseointegration and Biofilm Formation. J. Clin. Med. 2021, 10, 1641. [Google Scholar] [CrossRef]
  60. Koodaryan, R.; Hafezeqoran, A. Effect of laser-microtexturing on bone and soft tissue attachments to dental implants: A systematic review and meta-analysis. J. Dent. Res. Dent. Clin. Dent. Prospect. 2021, 15, 290–296. [Google Scholar] [CrossRef] [PubMed]
  61. Botos, S.; Yousef, H.; Zweig, B.; Flinton, R.; Weiner, S. The effects of laser microtexturing of the dental implant collar on crestal bone levels and peri-implant health. Int. J. Oral Maxillofac. Implant. 2011, 26, 492–498. [Google Scholar]
  62. Nammour, S.; El Mobadder, M.; Namour, M.; Namour, A.; Rompen, E.; Maalouf, E.; Brugnera Junior, A.; Brugnera, A.P.; Vescovi, P.; Zeinoun, T. A Randomized Comparative Clinical Study to Evaluate the Longevity of Esthetic Results of Gingival Melanin Depigmentation Treatment Using Different Laser Wavelengths (Diode, CO2, and Er: YAG). Photobiomodul Photomed. Laser Surg. 2020, 38, 167–173. [Google Scholar] [CrossRef] [PubMed]
  63. Sul, Y.T.; Byon, E.; Wennerberg, A. Surface characteristics of electrochemically oxidized implant and acid-etched implant: Surface chemistry, morphology, pore configurations, oxide thickness, crystal structure, and roughness. Int. J. Oral Maxillofac. Implant. 2008, 23, 631–640. [Google Scholar]
  64. Marin, C.; Granato, R.; Suzuki, M.; Gil, J.N.; Janal, M.N.; Coelho, P.G. Histomorphologic and histomorphometric evaluation of various endosseous implant healing chamber configurations at early implantation times: A study in dogs. Clin. Oral Implant. Res. 2010, 21, 577–583. [Google Scholar] [CrossRef] [PubMed]
  65. Bosshardt, D.D.; Chappuis, V.; Buser, D. Osseointegration of titanium, titanium alloy and zirconia dental implant: Current knowledge and open questions. Periodontology 2000 2017, 73, 22–40. [Google Scholar] [CrossRef] [PubMed]
  66. Jemat, A.; Ghazali, M.J.; Razali, M.; Otsuka, Y. Surface Modifications and Their Effects on Titanium Dental Implant. BioMed Res. Int. 2015, 2015, 791725. [Google Scholar] [CrossRef] [PubMed]
  67. Rupp, F.; Liang, L.; Geis-Gerstorfer, J.; Scheideler, L.; Hüttig, F. Surface characteristics of dental implant: A review. Dent. Mater. Off. Publ. Acad. Dent. Mater. 2018, 34, 40–57. [Google Scholar] [CrossRef] [PubMed]
  68. Tang, H.; Li, Y.; Ma, J.; Zhang, X.; Li, B.; Liu, S.; Zhang, K. Improvement of Biological and Mechanical Properties of Titanium Surface by Anodic Oxidation. Bio-Med. Mater. Eng. 2016, 27, 485–494. [Google Scholar] [CrossRef] [PubMed]
  69. Husain, F.; Gupta, S.; Sood, S.; Bhaskar, N.; Jain, A. To evaluate the effect of anodized dental implant surface on cumulative implant survival and success. A systematic review and meta-analysis. J. Indian Soc. Periodontol. 2022, 26, 525–532. [Google Scholar] [PubMed]
  70. Villaça-Carvalho, M.F.L.; de Araújo, J.C.R.; Beraldo, J.M.; Prado, R.F.D.; Moraes, M.E.L.; Manhães Junior, L.R.C.; Codaro, E.N.; Acciari, H.A.; Machado, J.P.B.; Regone, N.N.; et al. Bioactivity of an Experimental Dental Implant with Anodized Surface. J. Funct. Biomater. 2021, 12, 39. [Google Scholar] [CrossRef] [PubMed]
  71. Yeo, I.S. Reality of dental implant surface modification: A short literature review. Open Biomed. Eng. J. 2014, 8, 114–119. [Google Scholar] [CrossRef] [PubMed]
  72. Kim, K.H.; Ramaswamy, N. Electrochemical surface modification of titanium in dentistry. Dent. Mater. J. 2009, 28, 20–36. [Google Scholar] [CrossRef] [PubMed]
  73. Roguska, A.; Belcarz, A.; Zalewska, J.; Hołdyński, M.; Andrzejczuk, M.; Pisarek, M.; Ginalska, G. Metal TiO Nanotube Layers for the Treatment of Dental Implant Infections. ACS Appl. Mater. Interfaces 2018, 10, 17089–17099. [Google Scholar] [CrossRef] [PubMed]
  74. Alves-Rezende, M.C.R.; Capalbo, L.C.; De Oliveira Limírio, J.P.J.; Capalbo, B.C.; Limírio, P.H.J.O.; Rosa, J.L. The role of TiO2 nanotube surface on osseointegration of titanium implant: Biomechanical and histological study in rats. Microsc. Res. Tech. 2020, 83, 817–823. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, J.; Qian, S.; Liu, X.; Xu, L.; Miao, X.; Xu, Z.; Cao, L.; Wang, H.; Jiang, X. M2 macrophages contribute to osteogenesis and angiogenesis on nanotubular TiO2 surfaces. J. Mater. Chem. B 2017, 5, 3364–3376. [Google Scholar] [CrossRef] [PubMed]
  76. Mussano, F.; Genova, T.; Serra, F.G.; Carossa, M.; Munaron, L.; Carossa, S. Nano-Pore Size of Alumina Affects Osteoblastic Response. Int. J. Mol. Sci. 2018, 19, 528. [Google Scholar] [CrossRef] [PubMed]
  77. Ding, X.; Zhou, L.; Wang, J.; Zhao, Q.; Lin, X.; Gao, Y.; Li, S.; Wu, J.; Rong, M.; Guo, Z.; et al. The effects of hierarchical micro/nanosurfaces decorated with TiO2 nanotubes on the bioactivity of titanium implant in vitro and in vivo. Int. J. Nanomed. 2015, 10, 6955–6973. [Google Scholar]
  78. Wagenberg, B.; Froum, S.J. Long-Term Bone Stability around 312 Rough-Surfaced Immediately Placed Implant with 2–12-Year Follow-Up. Clin. Implant. Dent. Relat. Res. 2015, 17, 658–666. [Google Scholar] [CrossRef] [PubMed]
  79. Ribeiro, A.; Oliveira, F.; Boldrini, L.; Leite, P.; Falagan-Lotsch, P.; Linhares, A.; Zambuzzi, W.; Fragneaud, B.; Campos, A.; Gouvêa, C. Micro-arc oxidation as a tool to develop multifunctional calcium-rich surfaces for dental implant applications. Mater. Sci. Eng. C 2015, 54, 196–206. [Google Scholar] [CrossRef] [PubMed]
  80. Giordano, C.; Visai, L.; Pedeferri, M.P.; Chiesa, R.; Cigada, A. Antibacterial treatments on titanium for implantology. Biomed. Pharmacother. 2006, 60, 472. [Google Scholar] [CrossRef]
  81. Ding, M.; Shi, J.; Wang, W.; Li, D.; Tian, L. Early osseointegration of micro-arc oxidation coated titanium alloy implant containing Ag: A histomorphometric study. BMC Oral Health 2022, 22, 628. [Google Scholar] [CrossRef] [PubMed]
  82. Shimabukuro, M. Antibacterial Property and Biocompatibility of Silver, Copper, and Zinc in Titanium Dioxide Layers Incorporated by One-Step Micro-Arc Oxidation: A Review. Antibiotics 2020, 9, 716. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, L.; Cai, B.; Huang, Y.; Wang, J.; Zhu, C.; Shi, K.; Song, Y.; Feng, G.; Liu, L.; Zhang, L. Comparative Study on 3D Printed Ti6Al4V Scaffolds with Surface Modifications Using Hydrothermal Treatment and Microarc Oxidation to Enhance Osteogenic Activity. ACS Omega 2021, 6, 1465–1476. [Google Scholar] [CrossRef] [PubMed]
  84. Hu, J.; Li, H.; Wang, X.; Yang, L.; Chen, M.; Wang, R.; Qin, G.; Chen, D.-F.; Zhang, E. Effect of Ultrasonic Micro-Arc Oxidation on the Antibacterial Properties and Cell Biocompatibility of Ti-Cu Alloy for Biomedical Application. Mater. Sci. Eng. C 2020, 115, 110921. [Google Scholar] [CrossRef] [PubMed]
  85. Djošić, M.; Janković, A.; Mišković-Stanković, V. Electrophoretic deposition of biocompatible and bioactive hydroxyapatite-based coatings on titanium. Materials 2021, 14, 5391. [Google Scholar] [CrossRef] [PubMed]
  86. Seuss, S.; Boccaccini, A. Alternating Current Electrophoretic Deposition of Antibacterial Bioactive Glass-Chitosan Composite Coatings. Int. J. Mol. Sci. 2014, 15, 12231–12242. [Google Scholar] [CrossRef]
  87. Nuswantoro, N.F.; Manjas, M.; Suharti, N.; Juliadmi, D.; Fajri, H.; Tjong, D.H.; Affi, J.; Niinomi, M. Gunawarman. Hydroxyapatite Coating on Titanium Alloy TNTZ for Increasing Osseointegration and Reducing Inflammatory Response In Vivo on Rattus Norvegicus Wistar Rats. Ceram. Int. 2021, 47, 16094–16100. [Google Scholar] [CrossRef]
  88. Gaafar, M.S.; Yakout, S.M.; Barakat, Y.F.; Sharmoukh, W. Electrophoretic Deposition of Hydroxyapatite/Chitosan Nanocomposites: The Effect of Dispersing Agents on the Coating Properties. RSC Adv. 2022, 12, 27564–27581. [Google Scholar] [CrossRef]
  89. Fiołek, A.; Zimowski, S.; Kopia, A.; Łukaszczyk, A.; Moskalewicz, T. Electrophoretic Co-Deposition of Polyetheretherketone and Graphite Particles: Microstructure, Electrochemical Corrosion Resistance, and Coating Adhesion to a Titanium Alloy. Materials 2020, 13, 3251. [Google Scholar] [CrossRef] [PubMed]
  90. Pipattanachat, S.; Qin, J.; Rokaya, D.; Thanyasrisung, P.; Srimaneepong, V. Biofilm Inhibition and Bactericidal Activity of NiTi Alloy Coated with Graphene Oxide/Silver Nanoparticles via Electrophoretic Deposition. Sci. Rep. 2021, 11, 14008. [Google Scholar] [CrossRef]
  91. Juliadmi, D.; Nuswantoro, N.F.; Fajri, H.; Indriyani, I.Y.; Affi, J.; Manjas, M.; Suharti, N.; Tjong, D.H.; Niinomi, M.; Gunawarman, G. The Coating of Bovine-Source Hydroxyapatite on Titanium Alloy (Ti-6Al-4V ELI) Using Electrophoretic Deposition for Biomedical Application. Mater. Sci. Forum 2020, 1000, 97–106. [Google Scholar] [CrossRef]
  92. Danlei, Z.; Haoran, D.; Yuting, N.; Wenjie, F.; Muqi, J.; Ke, L.; Qingsong, W.; William, M.P.; Zhen, Z. Electrophoretic Deposition of Novel Semi-Permeable Coatings on 3D-Printed Ti-Nb Alloy Meshes for Guided Alveolar Bone Regeneration. Dent. Mater. 2022, 38, 431–443. [Google Scholar]
  93. Nicoli, L.G.; Oliveira, G.J.P.L.; Lopes, B.M.V.; Marcantonio, C.; Zandim-Barcelos, D.L.; Marcantonio, E., Jr. Survival/Success of Dental Implant with Acid-Etched Surfaces: A Retrospective Evaluation after 8 to 10 Years. Braz. Dent. J. 2017, 28, 330–336. [Google Scholar] [CrossRef] [PubMed]
  94. Hamouda, I.M.; Enan, E.T.; Al-Wakeel, E.E.; Yousef, M.K. Alkali and heat treatment of titanium implant material for bioactivity. Int. J. Oral Maxillofac. Implant. 2012, 27, 776–784. [Google Scholar]
  95. Cho, S. The Removal Torque of Titanium Screw Inserted in Rabbit Tibia Treated by Dual Acid Etching. Biomaterials 2003, 24, 3611–3617. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, X.; Poon, R.W.Y.; Kwok, S.C.H.; Chu, P.K.; Ding, C. Plasma Surface Modification of Titanium for Hard Tissue Replacements. Surf. Coat. Technol. 2004, 186, 227–233. [Google Scholar] [CrossRef]
  97. Chang, E.; Chang, W.J.; Wang, B.C.; Yang, C.Y. Plasma Spraying of Zirconia-Reinforced Hydroxyapatite Composite Coatings on Titanium: Part I: Phase, Microstructure and Bonding Strength. J. Mater. Sci. Mater. Med. 1997, 8, 193–200. [Google Scholar] [CrossRef]
  98. Yan, J.; Huang, W.; Kuang, H.; Wang, Q.; Li, B. The Effect of Etched 3D Printed Cu-Bearing Titanium Alloy on the Polarization of Macrophage. Front. Mater. 2022, 9, 941311. [Google Scholar] [CrossRef]
  99. Xie, H.; Shen, S.; Qian, M.; Zhang, F.; Chen, C.; Tay, F.R. Effects of Acid Treatment on Dental Zirconia: An In Vitro Study. PLoS ONE 2015, 10, e0136263. [Google Scholar] [CrossRef] [PubMed]
  100. Chosa, N.; Taira, M.; Saitoh, S.; Sato, N.; Araki, Y. Characterization of apatite formed on alkaline-heat-treated Ti. J. Dent. Res. 2004, 83, 465–469. [Google Scholar] [CrossRef] [PubMed]
  101. Nishio, K.; Neo, M.; Akiyama, H.; Nishiguchi, S.; Kim, H.M.; Kokubo, T.; Nakamura, T. The effect of alkali and heat-treated titanium and apatite-formed titanium on osteoblastic differentiation of bone marrow cells. J. Biomed. Mater. Res. 2000, 52, 652–661. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, Y.; Chen, Y.; Kou, H.C.; Yang, P.; Wang, Y.; Lu, T. Enhanced bone healing in porous Ti implanted rabbit combining bioactive modification and mechanical stimulation. J. Mech. Behav. Biomed. Mater. 2018, 86, 336–344. [Google Scholar] [CrossRef] [PubMed]
  103. Wittneben, J.G.; Joda, T.; Weber, H.P.; Brägger, U. Screw retained vs. cement retained implant-supported fixed dental prosthesis. Periodontology 2000 2017, 73, 141–151. [Google Scholar] [CrossRef] [PubMed]
  104. Hafezeqoran, A.; Koodaryan, R. Effect of Zirconia Dental Implant Surfaces on Bone Integration: A Systematic Review and Meta-Analysis. Biomed. Res. Int. 2017, 2017, 9246721. [Google Scholar] [CrossRef] [PubMed]
  105. Sailer, I.; Karasan, D.; Todorovic, A.; Ligoutsikou, M.; Pjetursson, B.E. Prosthetic failures in dental implant therapy. Periodontology 2000 2022, 88, 130–144. [Google Scholar] [CrossRef] [PubMed]
  106. Barros Lucena, G.A.; Molon, R.S.; Moretti, A.J.; Shibli, J.A.; Rêgo, D.M. Evaluation of Microbial Contamination in the Inner Surface of Titanium Implant Before Healing Abutment Connection: A Prospective Clinical Trial. Int. J. Oral Maxillofac. Implant. 2018, 33, 853–862. [Google Scholar] [CrossRef] [PubMed]
  107. Kawakita, E.; Wang, Z.; Kato, T.; Inaba, T.; Kasai, Y. Basic research on a cylindrical implant made of shape-memory alloy for the treatment of long bone fracture. Open Orthop. J. 2012, 6, 239–244. [Google Scholar] [CrossRef] [PubMed]
  108. Planinić, D.; Dubravica, I.; Šarac, Z.; Poljak-Guberina, R.; Celebic, A.; Bago, I.; Cabov, T.; Peric, B. Comparison of different surgical procedures on the stability of dental implant in posterior maxilla: A randomized clinical study. J. Stomatol. Oral Maxillofac. Surg. 2021, 122, 487–493. [Google Scholar] [CrossRef] [PubMed]
  109. Fritz, M.E. Two-stage implant systems. Adv. Dent. Res. 1999, 13, 162–169. [Google Scholar] [CrossRef]
  110. Tavelli, L.; Barootchi, S.; Avila-Ortiz, G.; Urban, I.A.; Giannobile, W.V.; Wang, H.L. Peri-implant soft tissue phenotype modification and its impact on peri-implant health: A systematic review and network meta-analysis. J. Periodontol. 2021, 92, 21–44. [Google Scholar] [CrossRef] [PubMed]
  111. Zhang, X.; Wang, F. Perioperative care experience of 10 cases of two-stage non-embedded implanted dentures. Guangxi Med. 2002, 8, 1314–1315. [Google Scholar]
  112. Mously, H.A. Effect of Two Implant-supported Partial Overdenture Attachment Design on the Periodontal Health. J. Contemp. Dent. Pract. 2020, 21, 68–72. [Google Scholar] [CrossRef] [PubMed]
  113. Al-Johany, S.S.; Al Amri, M.D.; Alsaeed, S.; Alalola, B. Dental Implant Length and Diameter: A Proposed Classification Scheme. J. Prosthodont. 2016, 26, 252–260. [Google Scholar] [CrossRef] [PubMed]
  114. Raszewski, Z.; Nowakowska-Toporowska, A.; Nowakowska, D.; Więckiewicz, W. Update on Acrylic Resins Used in Dentistry. Mini Rev. Med. Chem. 2021, 21, 2130–2137. [Google Scholar] [CrossRef] [PubMed]
  115. Gad, M.M.; Al-Thobity, A.M. The impact of nanoparticles-modified repair resin on denture repairs: A systematic review. Jpn. Dent. Sci. Rev. 2021, 57, 46–53. [Google Scholar] [CrossRef] [PubMed]
  116. Ghayebloo, M.; Alizadeh, P. Effect of zirconia nanoparticles on ZrO2-Bearing Lithium-Silicate glass-ceramic composite obtained by spark plasma sintering. J. Mech. Behav. Biomed. Mater. 2020, 110, 103880. [Google Scholar] [CrossRef] [PubMed]
  117. Siddiqi, A.; Payne, A.G.T.; De Silva, R.K.; Duncan, W.J. Titanium allergy: Could it affect dental implant integration? Clin. Oral Implant. Res. 2011, 22, 673–680. [Google Scholar] [CrossRef] [PubMed]
  118. Chouirfa, H.; Bouloussa, H.; Migonney, V.; Falentin-Daudré, C. Review of titanium surface modification techniques and coatings for antibacterial applications. Acta Biomater. 2019, 83, 37–54. [Google Scholar]
  119. Joda, T.; Zarone, F.; Ferrari, M. The complete digital workflow in fixed prosthodontics: A systematic review. BMC Oral Health. 2017, 17, 124. [Google Scholar] [CrossRef] [PubMed]
  120. Wang, C.; Liu, J.; Shi, F.D.; Liu, T.; Lu, Q. Some important issues in the development of glass-ceramic dental materials are explored. Fiberglass 2022, 49, 1–6. [Google Scholar]
  121. Chong, E.; Pelletier, M.H.; Mobbs, R.J.; Walsh, W.R. The design evolution of interbody cages in anterior cervical discectomy and fusion: A systematic review. BMC Musculoskelet. Disord. 2015, 16, 99. [Google Scholar] [CrossRef] [PubMed]
  122. Chung, I.; Lee, J.; Li, L.; Seol, Y.J.; Lee, Y.M.; Koo, K.T. A preclinical study comparing single-and double-root 3d-printed Ti-6Al-4V implant. Sci. Rep. 2023, 13, 862. [Google Scholar] [CrossRef] [PubMed]
  123. Bruno, V.; Berti, C.; Barausse, C.; Badino, M.; Gasparro, R.; Ippolito, D.R.; Felice, P. Clinical Relevance of Bone Density Values from CT Related to Dental Implant Stability: A Retrospective Study. BioMed Res. Int. 2018, 2018, 6758245. [Google Scholar] [CrossRef] [PubMed]
  124. Cucchi, A.; Vignudelli, E.; Franco, S.; Levrini, L.; Castellani, D.; Pagliani, L.; Rea, M.; Modena, C.; Sandri, G.; Longhi, C. Tapered, Double-Lead Threads Single Implants Placed in Fresh Extraction Sockets and Healed Sites of the Posterior Jaws: A Multicenter Randomized Controlled Trial with 1 to 3 Years of Follow-Up. Biomed. Res. Int. 2017, 2017, 8017175. [Google Scholar] [CrossRef] [PubMed]
  125. Maglione, M.; Bevilacqua, L.; Dotto, F.; Costantinides, F.; Lorusso, F.; Scarano, A. Observational Study on the Preparation of the Implant Site with Piezosurgery vs. Drill: Comparison between the Two Methods in terms of Postoperative Pain, Surgical Times, and Operational Advantages. Biomed. Res. Int. 2019, 2019, 8483658. [Google Scholar] [CrossRef]
  126. Fujiwara, S.; Kato, S.; Bengazi, F.; Urbizo Velez, J.; Tumedei, M.; Kotsu, M.; Botticelli, D. Healing at implant installed in osteotomies prepared either with a piezoelectric device or drills: An experimental study in dogs. Oral Maxillofac. Surg. 2021, 25, 65–73. [Google Scholar] [CrossRef] [PubMed]
  127. Clauser, T.; Lin, G.H.; Lee, E.; Del Fabbro, M.; Wang, H.L.; Testori, T. Risk of early implant failure in grafted and non-grafted sites: A systematic review and meta-analysis. Int. J. Oral Implantol. 2022, 15, 31–41. [Google Scholar]
  128. Munakata, M.; Kataoka, Y.; Yamaguchi, K.; Sanda, M. Risk Factors for Early Implant Failure and Selection of Bone Grafting Materials for Various Bone Augmentation Procedures: A Narrative Review. Bioengineering 2024, 11, 192. [Google Scholar] [CrossRef] [PubMed]
  129. French, D.; Grandin, H.M.; Ofec, R. Retrospective cohort study of 4591 dental implant: Analysis of risk indicators for bone loss and prevalence of peri-implant mucositis and peri-implantitis. J. Periodontol. 2019, 90, 691–700. [Google Scholar] [CrossRef] [PubMed]
  130. Laleman, I.; Lambert, F. Implant connection and abutment selection as a predisposing and/or precipitating factor for peri-implant diseases: A review. Clin. Implant. Dent. Relat. Res. 2023, 25, 723–733. [Google Scholar] [CrossRef] [PubMed]
  131. Sun, J.S.; Liu, K.C.; Hung, M.C.; Lin, H.Y.; Chuang, S.L.; Lin, P.J.; Chang, J.Z. A cross-sectional study for prevalence and risk factors of peri-implant marginal bone loss. J. Prosthet. Dent. 2023, in press. [Google Scholar] [CrossRef] [PubMed]
  132. Yao, J.; Tang, H.; Gao, X.L.; McGrath, C.; Mattheos, N. Patients’ expectations to dental implant: A systematic review of the literature. Health Qual. Life Outcomes 2014, 12, 153. [Google Scholar] [CrossRef] [PubMed]
  133. McCrea, S.J.J. An Analysis of Patient Perceptions and Expectations to Dental Implant: Is There a Significant Effect on Long-Term Satisfaction Levels. Int. J. Dent. 2017, 2017, 8230618. [Google Scholar] [CrossRef] [PubMed]
  134. Kashbour, W.A.; Rousseau, N.S.; Thomason, J.M.; Ellis, J.S. Provision of information on dental implant treatment: Patients’ thoughts and experiences. Clin. Oral Implant. Res. 2018, 29, 309–319. [Google Scholar] [CrossRef] [PubMed]
  135. Albrektsson, T.; Chrcanovic, B.; Jacobsson, M.; Wennerberg, A. Osseointegration of implant—A biological and clinical overview. JSM Dent. Surg. 2017, 2, 1022–1028. [Google Scholar]
  136. Amengual-Peñafiel, L.; Córdova, L.A.; Constanza Jara-Sepúlveda, M.; Brañes-Aroca, M.; Marchesani-Carrasco, F.; Cartes-Velásquez, R. Osteoimmunology drives dental implant osseointegration: A new paradigm for implant dentistry. Jpn. Dent. Sci. Rev. 2021, 57, 12–19. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, X.; Li, Y.; Feng, Y.; Cheng, H.; Li, D. The role of macrophages in osseointegration of dental implant: An experimental study in vivo. J. Biomed. Mater. Res. Part A 2020, 108, 2206–2216. [Google Scholar] [CrossRef] [PubMed]
  138. Vasconcelos, D.M.; Santos, S.G.; Lamghari, M.; Barbosa, M.A. The two faces of metal ions: From implant rejection to tissue repair/regeneration. Biomaterials 2016, 84, 262–275. [Google Scholar] [CrossRef]
  139. Saravi, B.; Vollmer, A.; Lang, G.; Adolphs, N.; Li, Z.; Giers, V.; Stoll, P. Impact of renin-angiotensin system inhibitors and beta-blockers on dental implant stability. Int. J. Implant. Dent. 2021, 7, 31. [Google Scholar] [CrossRef] [PubMed]
  140. Alvarez, C.; Monasterio, G.; Cavalla, F.; Córdova, L.A.; Hernández, M.; Heymann, D.; Garlet, G.P.; Sorsa, T.; Pärnänen, P.; Lee, H.M.; et al. Osteoimmunology of Oral and Maxillofacial Diseases: Translational Applications Based on Biological Mechanisms. Front. Immunol. 2019, 10, 1664. [Google Scholar] [CrossRef] [PubMed]
  141. Lin, H.; Sohn, J.; Shen, H.; Langhans, M.T.; Tuan, R.S. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials 2019, 203, 96–110. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Comparison of healthy tooth structure and dental implant structure. Natural teeth include natural crowns, gums, and fibrous bone; dental implants include artificial crowns, abutments, and the implant. Created through Adobe Photoshop (PS) CS 8.0 drawing software.
Figure 1. Comparison of healthy tooth structure and dental implant structure. Natural teeth include natural crowns, gums, and fibrous bone; dental implants include artificial crowns, abutments, and the implant. Created through Adobe Photoshop (PS) CS 8.0 drawing software.
Coatings 14 00781 g001
Figure 2. The processing flow of SLM [13].
Figure 2. The processing flow of SLM [13].
Coatings 14 00781 g002
Figure 3. The processing flow of EBM [18].
Figure 3. The processing flow of EBM [18].
Coatings 14 00781 g003
Figure 4. The processing flow of SLS [24].
Figure 4. The processing flow of SLS [24].
Coatings 14 00781 g004
Figure 5. Implant modeling with three types of implant: one-stage implant, two-stage non-embedded implant, and two-stage embedded implant. Created through Adobe Photoshop (PS) CS 8.0 drawing software.
Figure 5. Implant modeling with three types of implant: one-stage implant, two-stage non-embedded implant, and two-stage embedded implant. Created through Adobe Photoshop (PS) CS 8.0 drawing software.
Coatings 14 00781 g005
Figure 6. Illustration of the potential role of beta-blockers and RAS inhibitors in peri-implant bone remodeling processes. Created through Adobe Photoshop (PS) CS 8.0 drawing software. AP-1, activator protein 1; ARB, angiotensin II type 1 receptor blocker; RANKL, receptor activator of nuclear factor-kappa β ligand; OPG, osteoprotegerin; ATF-4, activating transcription factor 4; ROS, reactive oxygen species; NF-xβ, nuclear factor-kappa β; ACE, angiotensin-converting enzyme; Ang, angiotensin; AT1R, angiotensin II type 1 receptor; AT2R, angiotensin II type 2 receptor.
Figure 6. Illustration of the potential role of beta-blockers and RAS inhibitors in peri-implant bone remodeling processes. Created through Adobe Photoshop (PS) CS 8.0 drawing software. AP-1, activator protein 1; ARB, angiotensin II type 1 receptor blocker; RANKL, receptor activator of nuclear factor-kappa β ligand; OPG, osteoprotegerin; ATF-4, activating transcription factor 4; ROS, reactive oxygen species; NF-xβ, nuclear factor-kappa β; ACE, angiotensin-converting enzyme; Ang, angiotensin; AT1R, angiotensin II type 1 receptor; AT2R, angiotensin II type 2 receptor.
Coatings 14 00781 g006
Table 1. Studies on the different 3D printing methods.
Table 1. Studies on the different 3D printing methods.
Study3D Printing MethodResultsReferences
Osteogenesis of 3D-Printed porous Ti-6Al-4V implant with different pore sizes.SLMSLM was able to fabricate porous Ti6Al4V implants with proper mechanical properties analogous to human bone.[26]
Micro/nano topography of selective laser melting titanium inhibits osteoclastogenesis via mediation of macrophage polarization.SLMBoth in vivo and in vitro studies revealed that the SLA- and SAH-treated SLM-Ti implants significantly inhibited peri-implant osteoclast activity.[27]
Micro-/nano-topography of selective laser melting titanium enhances adhesion and proliferation.SLMCreating appropriate micro-/nano-topographies affected cell behavior and increased the stability of the SLM-Ti.[28]
Osteoinduction of porous Ti implants with a channel structure fabricated by selective laser melting.SLMThe RP-based SLM technique was very effective for investigating the influence of pore size on osteoinduction.[29]
A biomimetic hierarchical structure on selective laser melting titanium with enhanced hydrophobic surface.SLMThe SLM-Ti had a superhydrophilic surface, and the contact angles quickly reduced to zero upon complete wetting.[30]
Improved osseointegration of 3D-printed Ti-6Al-4V implants with a micro/nano surface topography.EBMAfter acid etching and anodic oxidation, the hydrophilicity and bioactivity of EBM-Ti were improved.[31]
Osteoconductivity of bioactive Ti-6Al-4V implants with lattice-shaped interconnected large pores fabricated by electron beam melting.EBMEBM-Ti with NaOH, CaCl2, heating, and water treatment, with lattice-like pores, with superior mechanical properties and biological activity.[32]
Electron beam-melted, free-form-fabricated titanium alloy implants: Material surface characterization and early bone response in rabbits.EBMCompared with machined Ti, EBM-Ti implants had higher surface roughness, thicker surface oxides, and better osteogenic activity.[33]
Mechanical properties of selective laser sintering pure titanium and Ti-6Al-4V, and its anisotropy.SLSThe SLS process with Ti-6Al-4V powder had great performance for the fabrication of dental prostheses.[34]
In vitro and vivo comparisons of the porous Ti-6Al-4V alloys fabricated by the selective laser melting technique and sintering technique.SLSMicrostructure and mechanical properties of the SLS porous Ti-6Al-4V were more similar to the cancellous bone, without obvious stress shielding.[35]
3D laser-printed porous Ti-6Al-4V dental implants for compromised bone support.SLSIn micro-CT analysis, new bone formation and osseointegration within the SLS-ITRI implants were observed.[36]
Table 2. Comparison of the advantages and disadvantages of different implantation methods and their use.
Table 2. Comparison of the advantages and disadvantages of different implantation methods and their use.
By Structure ClassificationOne-Stage ImplantTwo-Stage Non-Embedded ImplantTwo-Stage Embedded Implant
Planting patternsThe appropriate position is selected in the alveolar bone, the implant is placed in the hole, and the screw is closed; 7–10 days later the stitches are removed.Non-invasive surgery (gum surgery): The implant is directly inserted into the alveolar bone and mounted on a healing abutment. The implant is directly or indirectly exposed in the mouth.First surgery: To ensure that the bone tissue has a long enough time to heal, we place the implant in the alveolar bone and bury it completely under the soft tissue, without exposing it to the mouth.
Second procedure: After the bone has healed, the abutment is attached to the implant, the gums are cut, and the abutment is connected to the implant using the implant’s center screw.
MeritsStrong function: Can restore tooth function, chewing function is better than traditional dentures.
No grinding: Artificial root restoration, without grinding healthy teeth next to it.
High comfort level: No foreign body sensation, and conducive to maintaining oral hygiene.
The implant soft tissue and bone tissue have the same healing time, which is more favorable for healing soft tissue.
The implant and periodontal tissue heal well, with improved initial stability.
The need for secondary surgery shortens the dental implant time and reduces secondary trauma to the oral cavity.
The non-submersible implant has a higher joint plane and only one joint, thus providing a more favorable biological width and gingival margin height.
High stability: Higher implant stability due to adequate healing time between implant and bone tissue.
Difficult to infection: The risk of infection is reduced because the implant is completely embedded under the soft tissue.
Good bone integration: The closer bonding between the implant and the bone tissue is conducive to long-term stability.
Good long-term results: The long-term results are usually better because the connection between the implant and the abutment is better.
High success rate: Due to the above advantages, the success rate of two-stage embedded implants is usually higher.
ShortcomingsLong operation cycle: It usually takes 3 to 6 months between primary and secondary operations to facilitate the full integration of the implant and the alveolar bone.
Postoperative discomfort: After the first phase of the surgery, the mouth may be swollen and painful.
As implants are directly exposed to the oral environment, they may be more susceptible to infection by the oral bacteria.
During healing, the implant may be affected by the bite force, thus affecting its stability.
Two operations are required: Compared to a one-stage implant, the two-stage embedded implant requires two operations, increasing the risk and complexity of the procedure.
Long healing time: Due to the need to wait for bone tissue and implant healing, the entire treatment process is longer, usually taking several months.
Higher cost: The cost of two-stage embedded implants is usually higher due to the need for two surgeries and longer treatment times.
Table 3. Proposed classification scheme for dental implants based on length.
Table 3. Proposed classification scheme for dental implants based on length.
TermUltra-ShortShortStandardLong
Measurements≤6 mm>6 mm to <10 mm10 mm to <13 mm≥13 mm
Comments6 mm or lessFrom more than 6 mm to less than 10 mmFrom 10 mm to less than 13 mm13 mm or more
Table 4. Proposed classification scheme for dental implants based on diameter.
Table 4. Proposed classification scheme for dental implants based on diameter.
TermUltra-NarrowNarrowStandardWide
Measurements<3.0 mm≥3.0 mm to ≤3.75 mm≥3.75 mm to ≤5 mm≥5 mm
CommentsLess than 3.0 mmFrom 3.0 mm to less than 3.75 mmFrom 3.75 mm to less than 5 mm5.0 mm or more
Table 5. Advantages and disadvantages of different types of dental implant.
Table 5. Advantages and disadvantages of different types of dental implant.
Dental Implant MaterialsAdvantagesDisadvantages
PMMA-ZrO biomimetic nanocompositesStrong weather resistance
Wide variety
Non-toxic
Environmental protection
Good overall mechanical properties
Tensile properties
The fracture toughness and curvature strength are much higher than those of traditional dental ceramics
Poor wear resistance
High cost
Poor heat resistance
Zirconia–glass ceramic composite materialHigh density
High hardness
Low thermal expansion coefficient
High melting point
Good corrosion resistance
Good insulation
Biocompatibility
Easy to damage
Affects the service life
The preparation process requirements are high
High price
Titanium (chemical)Light weight
High strength
High-temperature resistance (to be used at higher temperatures for a long time)
Low-temperature resistance (to maintain good ductility and toughness)
Non-toxic
The new dental restorative material titanium has good biocompatibility and strong strength
The tribological properties of the natural teeth are very good
May cause allergies
High raw material cost
High weight
Low hardness
Difficult to process
Easy to oxidize
Expensive
Difficult to refine
Glass ceramicsExcellent lithium silicate glass ceramics
High bending strength
Transparency
High biocompatibility
Repair versatility
Comfort to wear
Easy to damage
Affects the service life
The preparation process requirements are high
High price
Resin matrix compositeA series of resin composites with uniform size dispersion
Low water absorption
Good biocompatibility was prepared for modified alumina nanoparticles
Poor durability
Unstable color
Easy color
Silicon nitrideGood antibacterial properties
No biotoxicity
Good biocompatibility
Mechanical properties
Good osseoconjugation
High mechanical strength
High fracture rate
Low elasticity
Heat shock
Low wear resistance
Can cause a foreign body reaction
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

Gao, J.; Pan, Y.; Gao, Y.; Pang, H.; Sun, H.; Cheng, L.; Liu, J. Research Progress on the Preparation Process and Material Structure of 3D-Printed Dental Implants and Their Clinical Applications. Coatings 2024, 14, 781. https://doi.org/10.3390/coatings14070781

AMA Style

Gao J, Pan Y, Gao Y, Pang H, Sun H, Cheng L, Liu J. Research Progress on the Preparation Process and Material Structure of 3D-Printed Dental Implants and Their Clinical Applications. Coatings. 2024; 14(7):781. https://doi.org/10.3390/coatings14070781

Chicago/Turabian Style

Gao, Jingjing, Yang Pan, Yuting Gao, Hanyu Pang, Haichuan Sun, Lijia Cheng, and Juan Liu. 2024. "Research Progress on the Preparation Process and Material Structure of 3D-Printed Dental Implants and Their Clinical Applications" Coatings 14, no. 7: 781. https://doi.org/10.3390/coatings14070781

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

Article Metrics

Back to TopTop