Next Article in Journal
Developing a Novel Enamel Adhesive with Amorphous Calcium Phosphate and Silver Nanoparticles to Prevent Demineralization during Orthodontic Treatment
Next Article in Special Issue
Conditional Mitigation of Dental-Composite Material-Induced Cytotoxicity by Increasing the Cure Time
Previous Article in Journal
Bioactive Synthetic Polymer-Based Polyelectrolyte LbL Coating Assembly on Surface Treated AZ31-Mg Alloys
Previous Article in Special Issue
Effect of Different Cavity Disinfectants on Adhesion to Dentin of Permanent Teeth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Three-Dimensional Impression of Biomaterials for Alveolar Graft: Scoping Review

by
Inês Francisco
1,2,3,
Ângela Basílio
1,
Madalena Prata Ribeiro
1,
Catarina Nunes
1,
Raquel Travassos
1,
Filipa Marques
1,
Flávia Pereira
1,
Anabela Baptista Paula
1,2,3,4,5,6,
Eunice Carrilho
2,3,4,5,6,*,
Carlos Miguel Marto
2,3,4,5,6,7 and
Francisco Vale
1,2,3
1
Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
2
Coimbra Institute for Clinical and Biomedical Research (ICBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
3
Laboratory for Evidence-Based Sciences and Precision Dentistry, University of Coimbra, 3000-075 Coimbra, Portugal
4
Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-075 Coimbra, Portugal
5
Clinical Academic Center of Coimbra (CACC), 3030-370 Coimbra, Portugal
6
Institute of Integrated Clinical Practice, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
7
Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
J. Funct. Biomater. 2023, 14(2), 76; https://doi.org/10.3390/jfb14020076
Submission received: 21 December 2022 / Revised: 22 January 2023 / Accepted: 26 January 2023 / Published: 29 January 2023
(This article belongs to the Special Issue Feature Papers in Dental Biomaterials)

Abstract

:
Craniofacial bone defects are one of the biggest clinical challenges in regenerative medicine, with secondary autologous bone grafting being the gold-standard technique. The development of new three-dimensional matrices intends to overcome the disadvantages of the gold-standard method. The aim of this paper is to put forth an in-depth review regarding the clinical efficiency of available 3D printed biomaterials for the correction of alveolar bone defects. A survey was carried out using the following databases: PubMed via Medline, Cochrane Library, Scopus, Web of Science, EMBASE, and gray literature. The inclusion criteria applied were the following: in vitro, in vivo, ex vivo, and clinical studies; and studies that assessed bone regeneration resorting to 3D printed biomaterials. The risk of bias of the in vitro and in vivo studies was performed using the guidelines for the reporting of pre-clinical studies on dental materials by Faggion Jr and the SYRCLE risk of bias tool, respectively. In total, 92 publications were included in the final sample. The most reported three-dimensional biomaterials were the PCL matrix, β-TCP matrix, and hydroxyapatite matrix. These biomaterials can be combined with different polymers and bioactive molecules such as rBMP-2. Most of the included studies had a high risk of bias. Despite the advances in the research on new three-dimensionally printed biomaterials in bone regeneration, the existing results are not sufficient to justify the application of these biomaterials in routine clinical practice.

1. Introduction

Craniofacial defects can originate from an array of etiological factors including congenital malformations, trauma, infection, rejection or implant failure, infection of bone graft, osteomyelitis, or surgical removal of tumors [1,2,3]. The craniofacial bone can also be impacted by systemic conditions such as osteodegenerative illnesses such as osteoporosis and arthritis, other impactful conditions include osteogenesis imperfecta and bone fibrous dysplasia [4]. All these conditions will compromise functional aspects such as phonation, mastication, and swallowing, which in turn affect the patient’s quality of life [5,6]. The two most common craniofacial bone defects are cancer of the head and neck and cleft lip and palate (CLP) [5,6,7,8,9]. CLP is a multifactorial pathology with several genetic and epigenetic factors as well as environmental factors such as geographical location, socioeconomical factors, and race [10,11]. In an attempt to minimize anomalies resulting from CLP, multidisciplinary treatment is initiated from birth and carries on into adulthood in order to achieve optimal results [12].
During the mixed dentition stage, individuals with CLP may require a secondary alveolar bone graft. During this period, this approach can result in relevant improvements such as closure of oronasal fistulae, stabilization of the two maxillary segments, and enhanced support of the alar base, which, in turn, will improve nasal and labial symmetry [13,14]. The secondary alveolar bone graft was introduced by Boyne and Sands in 1972 and it is currently regarded as the gold standard with the iliac crest being the most frequently chosen donor location [13]. In order to assert the proper timing to perform this procedure, the upper canine should have two thirds of its root developed which usually occurs between the ages of 9 and 11 [13].
The autologous bone graft can present with a variety of setbacks including limited amount of grafted bone, immune response risks, procedure time, and heavy costs. Additionally, a year after the procedure, bone reabsorption will happen in 40% of cases creating the need for re-intervention [15,16]. The main donor sites of autologous bone in craniomaxillofacial surgery are iliac crest graft and calvarial graft, but intraoral graft is also a possibility [17]. Currently, regenerative medicine has been established as a viable alternative in treatment of bone defects including CLP [18,19,20,21]. This approach can modulate the bone regeneration process and inflammation and enhance the healing process. Various biomaterials have been developed with the intent of overcoming the limitations of conventional bone grafts [22], such as heterologous or homologous bone graft [23,24]. These substituting materials can be used on their own or combined with an autologous bone graft and/or matrices. The most recognized tissue regeneration approach in the literature in the treatment of alveolar bone defects is bone morphogenetic protein 2 [25,26]. This approach provides comparable outcomes concerning bone volume, filling, and height to the gold standard technique with the iliac crest bone graft [26].
The matrices (Figure 1) are a subtract that allow for cell differentiation and proliferation. Their biocompatibility, biodegradability, osteoconduction, and mechanical properties are characteristics which can influence the success rate of the bone regeneration process [27].
These matrices can be three-dimensional (3D) printed enhancing its adaptation to the bone defect. With the use of 3D technologies, these matrices can be created and adapted according to the specific needs of each patient by changing their internal and external structures whilst using different materials [27,28].
The most commonly used matrices in bone defect treatment are bioceramic and are usually made out of hydroxyapatite (HA) or β-tricalcicum-phosphate (β-TCP). These materials are highly biocompatible and with osteoinductive abilities while also promoting rapid bone formation [29]. Despite a general increase of interest regarding 3D printed biomaterials in recent years, a comprehensive study regarding the general effectiveness of these biomaterials is lacking. To clarify this, we conducted a scoping review to assess the effectiveness of 3D printed biomaterials in the treatment of alveolar defects, which would be helpful for readership since it synthesizes what we know and the best future clinical approach in a single paper. Moreover, this knowledge will allow sustaining the realization of new future clinical studies. The aim of this paper is to put forth an in-depth review regarding the clinical efficiency of available 3D printed biomaterials for the correction of alveolar bone defects.

2. Materials and Methods

2.1. Study Research and Selection Strategy

Literature research was conducted on the PubMed data base via Medline, Cochrane Library, Web of Science Core Collection, EMBASE, and in gray literature. The last search was done, independently, on the 15th of August 2022 by two researchers.
A combination of Medical Subject Headings (Mesh) along with free text words were used in each of the databases (Appendix A). The following language filters were used: Portuguese, English, Spanish, and French. No filters were used regarding date of publication.
Two researchers initially scrutinized the articles independently by title and abstract. Subsequently, the articles were evaluated according to their full integral text; if doubts arose regarding the inclusion of a certain article, a third researcher was consulted.
The considered studies had to comply with the following inclusion criteria: in vitro, in vivo, ex vivo, and clinical studies; and studies that assessed bone regeneration resorting to 3D printed biomaterials. The exclusion criteria applied were as follows: non-clinical studies and every other type of research (editorials, academic books, and reports); case reports or descriptive studies; duplicated studies; studies with incomplete data; and studies that merely reported on the characterization of a new biomaterial without reporting on bone regeneration rates.

2.2. Data Extraction

After the eligibility process, the articles were sorted into different categories according to the type of study: in vitro, in vivo, ex vivo, or clinical. From each selected article, the following information was extracted: authors, date of publication, study design, experimental and control group, evaluation time, bone regeneration assessment method, results, and main conclusions.

2.3. Risk of Bias

The bias risk of the in vitro studies was obtained using the Faggion Jr. norms for pre-clinical studies regarding dental materials [30]. For the in vivo studies, the bias risk tool from the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) was used.

3. Results

3.1. Study Selection

The initial search, performed on the previously mentioned databases, gathered 792 studies. After removing duplicates, 604 studies were scrutinized according to title and abstract. Afterwards, all references deemed irrelevant for this systematic review were excluded, resulting in 123 potentially relevant studies. Given that 31 articles did not report bone regeneration rates, only 92 references were included in the final sample. The identification, screening and eligibility process is summarized in the flow chart (Figure 2).

3.2. Characteristics of the Included Studies

3.2.1. In Vitro Studies

Fifty-one articles analyzed the properties of biomaterials in vitro. The year of publication ranged from 2015 to 2022, with the exception of one study conducted in 2006 [31]. The most commonly used biomaterial in the control group was PCL matrix, followed by β-TCP and PLLA. Osteogenic activity through alkaline phosphatase was the most widely used method to assess bone regeneration, having been described in 26 articles. Seventy- two studies evaluated bone regeneration through the expression of osteogenesis-related genes. Only one study [32] reported the release rate of growth factors. On the other hand, one study [33] evaluated the porosity of the matrix and found that the presence of nanotubes is associated with more favorable results for osteogenesis when compared to larger pores. Table 1 summarizes the results of the in vitro studies included in this systematic review.

3.2.2. In Vivo Studies

In vivo bone regeneration was evaluated in 75 articles, published between 2015 and 2022, in various animal species, such as New Zealand rabbits, beagle dogs, and rat models. The number of animals used in each study ranged from 3 to 120, with seven articles not reporting the sample size [30,31,82,83,84,85].
The most commonly used biomaterial in the control group was β-TCP matrix, followed by PCL matrix. Regarding the evaluation method, microcomputerized tomography was the most used followed by histology. Other methods used were real-time polymerase chain reaction [42,86] and immunohistochemistry [87].
The most refracted matrices were PCL, β-TCP, and HA. In seven articles, the matrix of the experimental group contained bone morphogenetic protein-2 (BMP-2) [28,42,45,49,88,89]. Bone regeneration was superior in all experimental groups, with the exception of three articles [90,91,92], which found similar values between the control and experimental group. Regarding secondary outcomes, Van Hede et al. [73] analyzed matrix geometry, and found that the gyroid geometry results in better outcomes when compared to the orthogonal one. Chang et al. [43] found that combining HA matrix with an oxidized RGD peptide in a high stiffness matrix may be advantageous for maxillofacial regeneration when compared to low stiffness matrices.
Table 2 summarizes the results of the in vivo studies included in the present systematic review.

3.3. Synthesis of Quantitative Evidence

In the various studies evaluated, many different biomaterials are described. The most referenced biomaterial was β-tricalcium phosphate (β -TCP), used in 16 in vitro studies and 27 in vivo studies. The second most referenced biomaterial was polycaprolactone (PCL), mentioned in 16 in vitro studies and 20 in vivo studies. Hydroxyapatite (HA) was the third most used biomaterial, in 7 in vitro and 16 in vivo studies. There are other biomaterials/biomolecules that were used in more than 3 studies, namely: decellularized extracellular matrix (dECM), human recombinant bone protein type 2 (RhBMP-2), collagen, polylactic acid (PLLA), polylactic acid-co-glycolic acid (PLGA), calcium sulfate (SC), and different types of hydrogel (e.g., bone-derived extracellular matrix, β-TCP, cell-laden, nanocomposite, MicroRNA). All other biomaterials are mentioned only in a few studies, generating a multitude of results, which makes them difficult to analyze, and, consequently, to draw conclusions (Table 3).
The most used evaluation method was different in in vitro and in vivo studies. In the first ones, the most frequent methods were the following: determination of osteogenesis-related gene expression by qRT-PCR (27 studies), and the evaluation of alkaline phosphatase activity, a mineralization precursor protein, by p-nitrophenol assay (9 studies), and by a staining assay with the AKT assay kit (7 studies). In in vivo studies, radiological methods such as micro-CT (57 studies) and histological methods (56 studies) are the most used (Table 4).
The most used 3D printing technique mentioned in both types of studies is extrusion-based 3D printing (23 in vitro studies and 27 in vivo studies). However, there are other techniques used simultaneously in in vitro and in vivo studies, namely: fused deposition modeling (6 and 10, respectively), stereolithography (2 and 7, respectively), and laser sintering technique (3 in both). Other techniques are used, but only occasionally in 1 or 2 studies (Table 5).

3.4. Risk of Bias

The risk of bias of the in vitro and in vivo studies is summarized in Table 6 and Table 7, respectively. Regarding in vitro studies, none described the methodology to implementation sample. All in vivo studies also lacked information regarding sample allocation, allocation randomization process methodology, implementation, and protocol. All but three of the articles disclose information regarding study financing.
Regarding in vivo studies, most of the studies have serious methodological flaws, leaving out pivotal information such as sequence generation, allocation concealment, and blinding. Only six studies specify investigator blindness as a factor during outcome assessment. Lastly, seven other studies report no additional bias sources.

4. Discussion

The aim of the present systematic review was to report the current state of the art regarding the clinical efficiency of available 3D printed biomaterials for the correction of alveolar bone defects. Although the quantitative analysis of the results could not be executed due to the heterogeneity of the studies, the qualitative analysis allowed for a better understanding and evaluation of the published studies.
The conventional technique requires an autologous graft of cancellous bone and is considered the gold standard [13]. However, with the limited offer of donor bone as well as the bone reabsorption rate due to its adaptability to the defect site, a re-intervention may be necessary [15,16]. In an attempt to diminish these limitations, studies have been carried out in order to explore different approaches that can accelerate bone formation, reduce bone reabsorption and improve soft tissue scarring. 3D printed biomaterials can be specifically made to adapt to the bone defect site; this has led to an increase in studies regarding this topic over the last five years [27,28].
Out of the 75 in vivo studies included, 17 evaluated the efficiency of the PCL matrix [32,35,37,50,51,52,53,54,55,62,63,65,66,73,77,81,120]. This biomaterial is the most well reviewed biomaterial in literature due to its high biocompatibility, durability and subsequent extensive use [37]. Despite its low degradation rate, the PCL matrix is limited in terms of cellular adhesion and osteogenic differentiation, several authors [32,35,49,50,53,62] have suggested combining it with different polymers [37] and bioactive molecules such as rBMP-2, that promote proliferation and differentiation of mesenchymal stem cells into osteoblasts resulting in bone formation. Nonetheless, a recently published umbrella review regarding the efficiency of current approaches in regeneration of bone defects in non-syndromic patients with cleft palate concluded that rBMP2 seems to provide results similar to the iliac crest bone graft in terms of bone volume and vertical dimension [121]. Another limitation of the PCL matrix is its low hydrophilia [52], which can be amended when the matrix is combined with a hydrophilic material such as β -TCP [35,66,77] or polydopamine [37]. With the addition of graphene, the PCL matrix increases its capacity to induce the secretion of growth factors that boost angiogenesis [56].
The β-TCP matrix was reportedly used in 12 in vitro and 30 in vivo studies. This calcium phosphate bioceramic presents ideal biocompatibility and osteoconductivity [36,64,85]. In addition to those characteristics, the β-TCP matrix also contains components similar to the bone tissue apatite along with a good balance between reabsorption and degradation during bone formation. Despite all these attributes, the osteogenic abilities of this biomaterial showed subpar results when used in large bone defects [35,48,64] and thus falling short when compared to the autologous bone graft [70].
The hydroxyapatite matrix is one of the most referenced bioceramics in in vivo studies. When combined with β-TCP this matrix becomes highly biocompatible and with a great osteointegration rate [88,90,123]. However, more studies are required in order to fully understand the macro-design that can optimize bone regeneration [90]. Since the bone formation process involves the immune system, this can be modulated by biomaterials such as esphingosine-1-phosphate (S1O) which has been linked to the β-TCP matrix. This sphingolipid has been shown to increase the expression of genes related to osteogenesis, such as osteoporin (OPN), transcribing factor 2 related to a runt (RUNX2), and osteocalcin (OCN) [36]. In addition to this, the combination of β-TCP with strontium oxide (SrO), sillica (SiO2), magnesium (MgO), and zinc (ZnO) also proved to be effective in bone regeneration due to alterations in the physical and mechanical properties of the matrix [48].
Regarding PRF, this biomaterial can improve the reconstruction of the alveolar cleft. It is prepared from centrifuged autologous blood formed by a fibrin matrix that contains platelets, white blood cells, growth factors and cytokines. These factors may promote the uniqueness and differentiation pathways of osteoblasts, endothelial cells, chondrocytes, and various sources of fibroblasts, stimulating the regenerative capacity of the periosteum. Furthermore, the fibrous structure of PRF acts as a three-dimensional fibrin scaffold for cell migration [16]. In this way, PRF can be used with a bone substitute, allowing wound sealing, homeostasis, bone union, and graft stability [16]. In contrast, BMP-2 is usually applied in alloplastic bone grafts or scaffolding and is an effective inducer of bone and cartilaginous formation. Its application avoids the limitation of autologous bone grafts, which may be related to the shorter operative and hospitalization time. However, it has some adverse effects, such as nasal stenosis and localized edema at the graft site [26].
Another promising candidate for bone regeneration is the pure Zn L-PBF porous scaffold [74]. It presented relatively adjusted deterioration rates and mechanical strength for bone implants. Furthermore, they also showed well in vitro cytocompatibility with MC3T3-E1 cells and osteogenic capacity for hBMMSCs. The in vivo implantation results showed that pure Zn scaffolds have potential for applications in large bone defects with osteogenic properties [74].
Additionally, the microstructure of the matrices such as porosity, pore size, and structure play a very important role in cell viability and bone growth [115]. In contrast to traditional methods, the development of three-dimensional printing allows for the control of the microstructure. Therefore, a wide variety of materials and techniques are available to optimize the matrix [124]. Shim et al. reported that porosity affects osteogenesis, with matrices with 30% porosity showing better osteogenic capacity than groups with 50% and 70% porosity [115]. Regarding pore size, the literature suggests that the ideal size should be between 400 to 600 μm [63,103,111]. Finally, the pore configuration should also be considered in terms of the dynamic stability of the matrix. Recently, matrices with hierarchical structures have been studied. Zhang et al. demonstrated that tantalum matrices with hierarchical structures exhibited excellent hydrophilicity, biocompatibility, and osteogenic properties [33]. However, in the future, additional in vivo studies are required as to understand what structure the matrix should present in order to find a balance between cell viability and mechanical properties of the biomaterial, optimizing bone regeneration.
This systematic review presents some limitations that may alter the interpretation of the results, namely: (1) some of the included studies present a small sample size with only three animals; (2) the included studies present high risk of bias; (3) lack of evaluation of variables that interfere with bone regeneration, such as the position of the teeth in the bone graft, the width of the defect, the volume of grafted bone and the experience of the clinician; (4) absence of clinical studies; (5) heterogeneity of the studies in terms of matrix typology and follow-up used may difficult outcome assessment. Due to the heterogeneity in the methodology of the included studies, most of the studies selected in this systematic review were classified as having a high risk of bias, which may decrease the certainty of the results. According to the risk of bias analysis, the analyzed parameters with the highest risk of bias were sample allocation, allocation randomization process methodology, implementation, and protocol. These factors must be considered when figuring out the results of this review. The methodology of the several studies evaluated is very different and is not described enough, which makes their effective comparison impossible. Since there are numerous types of biomaterials/biomolecules and various combinations between them, future studies should define the most appropriate methodology, creating guidelines for its implementation and subsequent comparison.
In addition, future studies should be calibrated in order to use similar parameters and protocols, providing stronger evidence, focusing on the most described materials, namely β-tricalcium phosphate, polycaprolactone, hydroxyapatite with decellularized extracellular matrix (dECM), human recombinant bone protein type 2 (RhBMP-2), collagen, polylactic acid (PLLA), poly(lactic acid-co-glycolic acid (PLGA), and calcium sulfate (CS). Moreover, these promising materials should be evaluated and compared to each other in a single study in order to obtain more effective and clinically applicable conclusions. In the future, additional studies should be performed, more specifically blinded randomized studies with increased control of possible bias sources namely, the randomization process, concealment of the investigators of the experimental groups and description of the limitations of the studies. Moreover, the cost-effectiveness of the proposed new regenerative strategies should be evaluated, as it plays a crucial role in clinical decision making in healthcare systems, especially public institutions.
Lastly, future systematic reviews focused on 3D biomaterials should include only the most referenced evaluation and printing techniques. Therefore, for in vitro systematic reviews, the authors should compare PCL, b-TCP, RhBMP-2, and HA biomaterials created by extrusion printing, fused deposition, stereolithography, or laser sintering techniques. The chosen evaluation methodology should be gene expression by qRT-PCR and alkaline phosphatase activity. On the other hand, for in vivo systematic reviews, the authors should analyze the same biomaterials and the same technique printing, but the evaluation methodology should be based on radiology imaging and histology.

5. Conclusions

The most reported three-dimensional biomaterials were the PCL matrix, β-TCP matrix, and hydroxyapatite matrix. Despite the advances in the research on new three-dimensionally printed biomaterials in bone regeneration, the existing results are not sufficient to justify the application of these biomaterials in routine clinical practice.

Author Contributions

Conceptualization, I.F. and F.V.; methodology, E.C., C.M.M. and A.B.P.; validation, C.N. and R.T.; formal analysis, F.M. and F.P.; investigation, Â.B. and I.F.; data curation, C.N. and R.T.; writing, Â.B. and M.P.R.; writing—review and editing, I.F., C.M.M. and A.B.P.; visualization, E.C., F.M. and F.P.; supervision, I.F. and F.V.; project administration, F.V. and E.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

DatabaseSearch Phrase
Pubmed via Medline and Cochrane Library(“Printing, Three-Dimensional” [Mesh] OR “Printing, Three Dimensional” OR “Printings, Three-Dimensional” OR “Three-Dimensional Printings” OR “3-Dimensional Printing*” OR “3 Dimensional Printing*” OR “Printing, 3-Dimensional” OR “Printings, 3-Dimensional” OR “3-D Printing*” OR “3 D Printing*” OR “Printing, 3-D” OR “Printings, 3-D” OR “Three-Dimensional Printing” OR “Three Dimensional Printing” OR “3D Printing*” OR “Printing, 3D” OR “Printings, 3D”) AND (“Bone Regeneration”[Mesh] OR “Bone Regenerations*” OR “Regeneration, Bone” OR “Regenerations, Bone” OR Osteoconduction OR “Alveolar Bone Grafting”[Mesh] OR “alveolar bone grafting*” OR “Alveolar Cleft Grafting” OR “bone graft*” OR “Bone Substitutes”[Mesh] OR “bone substitute*” OR “Replacement Material, Bone” OR “Replacement Materials, Bone” OR “Materials, Bone Replacement” OR “Substitute, Bone” OR “Substitutes, Bone” OR “Bone Replacement Material*” OR “Material, Bone Replacement” ) AND (Dentistry[Mesh] OR dentistry OR oral* OR orofacial OR dental* OR maxillofacial OR “Surgery, Oral”[Mesh] OR “surgery, oral” OR “Maxillofacial Surgery” OR “Surgery, Maxillofacial” OR “Oral Surgery” OR “Cleft Palate”[Mesh] OR “cleft palate*” OR “Palate, Cleft” OR “Palates, Cleft” OR “Cleft Palate, Isolated”)
Web of Science Core Collection (WOS)TS = (“Print*, Three Dimensional” OR “Three-Dimensional Print*” OR “3-Dimensional Print*” OR “3 Dimensional Print*” OR “Print*, 3-Dimensional” OR “3-D Print*” OR “3D Print*” OR “Print*, 3-D” OR “ Print*, 3D”) AND TS = ( “Regenerati*, Bone” OR “Bone Regenerati*” OR osteoconduction OR “Alveolar Bone Graft*” OR “alveolar cleft grafting“ OR “bone graft*” OR “Replacement Material*, Bone” OR “Material*, Bone Replacement” OR “Substitute*, Bone” OR “Bone Replacement Material*” OR “ Material, Bone Replacement” OR “bone substitute*”) AND TS = (dent* OR oral* OR orofacial OR maxillofacial OR “Surgery, Oral” OR “oral surgery”)
EMBASE(‘printing, three dimensional’/exp OR ‘printing, three dimensional’ OR ‘printings, three-dimensional’ OR ‘three-dimensional printings’ OR ‘3-dimensional printing*’ OR ‘3 dimensional printing*’ OR ‘printing, 3-dimensional’ OR ‘printings, 3-dimensional’ OR ‘3-d printing*’ OR ‘3 d printing*’ OR ‘printing, 3-d’ OR ‘printings, 3-d’ OR ‘three-dimensional printing’/exp OR ‘three-dimensional printing’ OR ‘three dimensional printing’/exp OR ‘three dimensional printing’ OR ‘3d printing*’ OR ‘printing, 3d’ OR ‘printings, 3d’) AND (‘bone regeneration’/exp OR ‘bone regeneration’ OR ‘regeneration, bone’/exp OR ‘regeneration, bone’ OR ‘regenerations, bone’ OR ’osteoconduction’/exp OR osteoconduction OR ‘alveolar bone grafting’/exp OR ‘alveolar bone grafting’ OR ‘alveolar cleft grafting’ OR ‘bone graft*’ OR ‘bone graft’/exp OR ‘bone graft’ OR ‘bone transplantation’/exp OR ‘bone transplantation’ OR ‘bone prosthesis’/exp OR ‘bone prosthesis’ OR ‘bone substitute*’ OR ‘replacement material, bone’ OR ‘replacement materials, bone’ OR ‘materials, bone replacement’ OR ‘substitute, bone’ OR ‘substitutes, bone’ OR ‘bone replacement material*’ OR ‘material, bone replacement’) AND (dentistry OR ‘dentistry’/exp OR ‘dentistry’ OR oral OR orofacial OR ‘dental’/exp OR dental OR maxillofacial OR ‘oral surgery’/exp OR ‘oral surgery’)

References

  1. Robey, P.G. Cell Sources for Bone Regeneration: The Good, the Bad, and the Ugly (But Promising). Tissue Eng. Part B Rev. 2011, 17, 423–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bhumiratana, S.; Vunjak-Novakovic, G. Concise Review: Personalized Human Bone Grafts for Reconstructing Head and Face. Stem Cells Transl. Med. 2012, 1, 64–69. [Google Scholar] [CrossRef]
  3. Weir, M.D.; Xu, H.H.K. Culture Human Mesenchymal Stem Cells with Calcium Phosphate Cement Scaffolds for Bone Repair. J. Biomed. Mater. Res. 2010, 93B, 93–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Illich, D.J.; Demir, N.; Stojković, M.; Scheer, M.; Rothamel, D.; Neugebauer, J.; Hescheler, J.; Zöller, J.E. Concise Review: Induced Pluripotent Stem Cells and Lineage Reprogramming: Prospects for Bone Regeneration. Stem Cells 2011, 29, 555–563. [Google Scholar] [CrossRef] [PubMed]
  5. Pagni, G.; Kaigler, D.; Rasperini, G.; Avila-Ortiz, G.; Bartel, R.; Giannobile, W.V. Bone Repair Cells for Craniofacial Regeneration. Adv. Drug Deliv. Rev. 2012, 64, 1310–1319. [Google Scholar] [CrossRef] [Green Version]
  6. He, X.; Dziak, R.; Mao, K.; Genco, R.; Swihart, M.; Li, C.; Yang, S. Integration of a Novel Injectable Nano Calcium Sulfate/Alginate Scaffold and BMP2 Gene-Modified Mesenchymal Stem Cells for Bone Regeneration. Tissue Eng. Part A 2013, 19, 508–518. [Google Scholar] [CrossRef] [Green Version]
  7. Raposo-Amaral, C.E.; Kobayashi, G.S.; Almeida, A.B.; Bueno, D.F.; de Souza e Freitas, F.R.; Vulcano, L.C.; Passos-Bueno, M.R.; Alonso, N. Alveolar Osseous Defect in Rat for Cell Therapy: Preliminary Report. Acta Cir. Bras. 2010, 25, 313–317. [Google Scholar] [CrossRef] [Green Version]
  8. Yoshioka, M.; Tanimoto, K.; Tanne, Y.; Sumi, K.; Awada, T.; Oki, N.; Sugiyama, M.; Kato, Y.; Tanne, K. Bone Regeneration in Artificial Jaw Cleft by Use of Carbonated Hydroxyapatite Particles and Mesenchymal Stem Cells Derived from Iliac Bone. Int. J. Dent. 2012, 2012, 352510. [Google Scholar] [CrossRef]
  9. Garland, M.A.; Reynolds, K.; Zhou, C.J. Environmental Mechanisms of Orofacial Clefts. Birth Defects Res. 2020, 112, 1660–1698. [Google Scholar] [CrossRef]
  10. Calzolari, E.; Bianchi, F.; Rubini, M.; Ritvanen, A.; Neville, A.J. Epidemiology of Cleft Palate in Europe: Implications for Genetic Research. Cleft Palate-Craniofac. J. 2004, 41, 244–249. [Google Scholar] [CrossRef]
  11. Taib, B.G.; Taib, A.G.; Swift, A.C.; van Eeden, S. Cleft Lip and Palate: Diagnosis and Management. Br. J. Hosp. Med. 2015, 76, 584–591. [Google Scholar] [CrossRef] [PubMed]
  12. Stal, S.; Chebret, L.; McElroy, C. The Team Approach in the Management of Congenital and Acquired Deformities. Clin. Plast. Surg. 1998, 25, 485–491. [Google Scholar] [CrossRef]
  13. Francisco, I.N. Overview of Care in Cleft Lip and Palate for Orthodontic Treatment. Ph.D. Thesis, Universidade do Porto, Porto, Portugal, 2021. [Google Scholar]
  14. Graber, L.; Vanarsdall, R.L.; Vig, K.W.L. Orthodontics Current Principles & Techniques, 4th ed.; Elsevier Mosby: St. Louis, MO, USA, 2005. [Google Scholar]
  15. Rawashdeh, M.A.; Telfah, H. Secondary Alveolar Bone Grafting: The Dilemma of Donor Site Selection and Morbidity. Br. J. Oral Maxillofac. Surg. 2008, 46, 665–670. [Google Scholar] [CrossRef] [PubMed]
  16. Francisco, I.; Fernandes, M.H.; Vale, F. Platelet-Rich Fibrin in Bone Regenerative Strategies in Orthodontics: A Systematic Review. Materials 2020, 13, 1866. [Google Scholar] [CrossRef]
  17. Valentini, V.; Cassoni, A.; Marianetti, T.M.; Romano, F.; Terenzi, V.; Iannetti, G. Reconstruction of Craniofacial Bony Defects Using Autogenous Bone Grafts: A Retrospective Study on 233 Patients. J. Craniofac. Surg. 2007, 18, 953–958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Ahn, G.; Lee, J.-S.; Yun, W.-S.; Shim, J.-H.; Lee, U.-L. Cleft Alveolus Reconstruction Using a Three-Dimensional Printed Bioresorbable Scaffold with Human Bone Marrow Cells. J. Craniofac. Surg. 2018, 29, 1880–1883. [Google Scholar] [CrossRef] [PubMed]
  19. Pradel, W.; Lauer, G. Tissue-Engineered Bone Grafts for Osteoplasty in Patients with Cleft Alveolus. Ann. Anat.-Anat. Anz. 2012, 194, 545–548. [Google Scholar] [CrossRef] [PubMed]
  20. Luaces-Rey, R.; Arenaz-Bua, J.; Lopez-Cedrun-Cembranos, J.; Herrero-Patino, S.; Sironvalle-Soliva, S.; Iglesias-Candal, E.; Pombo-Castro, M. Is PRP Useful in Alveolar Cleft Reconstruction? Platelet-Rich Plasma in Secondary Alveoloplasty. Med. Oral 2010, 15, e619–e623. [Google Scholar] [CrossRef] [Green Version]
  21. Morad, G.; Kheiri, L.; Khojasteh, A. Dental Pulp Stem Cells for in Vivo Bone Regeneration: A Systematic Review of Literature. Arch. Oral Biol. 2013, 58, 1818–1827. [Google Scholar] [CrossRef]
  22. Zumarán, C.; Parra, M.; Olate, S.; Fernández, E.; Muñoz, F.; Haidar, Z. The 3 R’s for Platelet-Rich Fibrin: A “Super” Tri-Dimensional Biomaterial for Contemporary Naturally-Guided Oro-Maxillo-Facial Soft and Hard Tissue Repair, Reconstruction and Regeneration. Materials 2018, 11, 1293. [Google Scholar] [CrossRef]
  23. Albanese, M.; Zotti, F.; Lanaro, L.; Trojan, D.; Paolin, A.; Montagner, G.; Iannielli, A.; Rodella, L.F.; Nocini, P.F. Fresh-Frozen Homologous Bone in Sinus Lifting: Histological and Radiological Analysis. Minerva Stomatol. 2019, 68, 226–235. [Google Scholar] [CrossRef] [PubMed]
  24. Chisci, G.; Fredianelli, L. Therapeutic Efficacy of Bromelain in Alveolar Ridge Preservation. Antibiotics 2022, 11, 1542. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, C.; Pan, W.; Feng, C.; Su, Z.; Duan, Z.; Zheng, Q.; Hua, C.; Li, C. Grafting Materials for Alveolar Cleft Reconstruction: A Systematic Review and Best-Evidence Synthesis. Int. J. Oral Maxillofac. Surg. 2018, 47, 345–356. [Google Scholar] [CrossRef] [PubMed]
  26. Francisco, I.; Paula, A.B.; Oliveiros, B.; Fernandes, M.H.; Carrilho, E.; Marto, C.M.; Vale, F. Regenerative Strategies in Cleft Palate: An Umbrella Review. Bioengineering 2021, 8, 76. [Google Scholar] [CrossRef] [PubMed]
  27. Thrivikraman, G.; Athirasala, A.; Twohig, C.; Boda, S.K.; Bertassoni, L.E. Biomaterials for Craniofacial Bone Regeneration. Dent. Clin. N. Am. 2017, 61, 835–856. [Google Scholar] [CrossRef]
  28. Zhang, L.; Yang, G.; Johnson, B.N.; Jia, X. Three-Dimensional (3D) Printed Scaffold and Material Selection for Bone Repair. Acta Biomater. 2019, 84, 16–33. [Google Scholar] [CrossRef]
  29. Seitz, H.; Rieder, W.; Irsen, S.; Leukers, B.; Tille, C. Three-Dimensional Printing of Porous Ceramic Scaffolds for Bone Tissue Engineering. J. Biomed. Mater. Res. 2005, 74B, 782–788. [Google Scholar] [CrossRef]
  30. Faggion, C.M. Guidelines for Reporting Pre-Clinical In Vitro Studies on Dental Materials. J. Evid. Based Dent. Pract. 2012, 12, 182–189. [Google Scholar] [CrossRef]
  31. Weinand, C.; Pomerantseva, I.; Neville, C.M.; Gupta, R.; Weinberg, E.; Madisch, I.; Shapiro, F.; Abukawa, H.; Troulis, M.J.; Vacanti, J.P. Hydrogel-β-TCP Scaffolds and Stem Cells for Tissue Engineering Bone. Bone 2006, 38, 555–563. [Google Scholar] [CrossRef]
  32. Park, J.Y.; Shim, J.-H.; Choi, S.-A.; Jang, J.; Kim, M.; Lee, S.H.; Cho, D.-W. 3D Printing Technology to Control BMP-2 and VEGF Delivery Spatially and Temporally to Promote Large-Volume Bone Regeneration. J. Mater. Chem. B 2015, 3, 5415–5425. [Google Scholar] [CrossRef]
  33. Zhang, Z.; Li, Y.; He, P.; Liu, F.; Li, L.; Zhang, H.; Ji, P.; Yang, S. Nanotube-Decorated Hierarchical Tantalum Scaffold Promoted Early Osseointegration. Nanomed. Nanotechnol. Biol. Med. 2021, 35, 102390. [Google Scholar] [CrossRef] [PubMed]
  34. Alksne, M.; Kalvaityte, M.; Simoliunas, E.; Rinkunaite, I.; Gendviliene, I.; Locs, J.; Rutkunas, V.; Bukelskiene, V. In Vitro Comparison of 3D Printed Polylactic Acid/Hydroxyapatite and Polylactic Acid/Bioglass Composite Scaffolds: Insights into Materials for Bone Regeneration. J. Mech. Behav. Biomed. Mater. 2020, 104, 103641. [Google Scholar] [CrossRef] [PubMed]
  35. Bae, E.-B.; Park, K.-H.; Shim, J.-H.; Chung, H.-Y.; Choi, J.-W.; Lee, J.-J.; Kim, C.-H.; Jeon, H.-J.; Kang, S.-S.; Huh, J.-B. Efficacy of RhBMP-2 Loaded PCL/β -TCP/BdECM Scaffold Fabricated by 3D Printing Technology on Bone Regeneration. BioMed Res. Int. 2018, 2018, 2876135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cao, Y.; Xiao, L.; Cao, Y.; Nanda, A.; Xu, C.; Ye, Q. 3D Printed β-TCP Scaffold with Sphingosine 1-Phosphate Coating Promotes Osteogenesis and Inhibits Inflammation. Biochem. Biophys. Res. Commun. 2019, 512, 889–895. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, Y.-W.; Shen, Y.-F.; Ho, C.-C.; Yu, J.; Wu, Y.-H.A.; Wang, K.; Shih, C.-T.; Shie, M.-Y. Osteogenic and Angiogenic Potentials of the Cell-Laden Hydrogel/Mussel-Inspired Calcium Silicate Complex Hierarchical Porous Scaffold Fabricated by 3D Bioprinting. Mater. Sci. Eng. C 2018, 91, 679–687. [Google Scholar] [CrossRef]
  38. Chiu, Y.-C.; Shie, M.-Y.; Lin, Y.-H.; Lee, A.K.-X.; Chen, Y.-W. Effect of Strontium Substitution on the Physicochemical Properties and Bone Regeneration Potential of 3D Printed Calcium Silicate Scaffolds. IJMS 2019, 20, 2729. [Google Scholar] [CrossRef] [Green Version]
  39. Cooke, M.E.; Ramirez-GarciaLuna, J.L.; Rangel-Berridi, K.; Park, H.; Nazhat, S.N.; Weber, M.H.; Henderson, J.E.; Rosenzweig, D.H. 3D Printed Polyurethane Scaffolds for the Repair of Bone Defects. Front. Bioeng. Biotechnol. 2020, 8, 557215. [Google Scholar] [CrossRef]
  40. Dai, Q.; Li, Q.; Gao, H.; Yao, L.; Lin, Z.; Li, D.; Zhu, S.; Liu, C.; Yang, Z.; Wang, G.; et al. 3D Printing of Cu-Doped Bioactive Glass Composite Scaffolds Promotes Bone Regeneration through Activating the HIF-1α and TNF-α Pathway of HUVECs. Biomater. Sci. 2021, 9, 5519–5532. [Google Scholar] [CrossRef]
  41. Dubey, N.; Ferreira, J.A.; Malda, J.; Bhaduri, S.B.; Bottino, M.C. Extracellular Matrix/Amorphous Magnesium Phosphate Bioink for 3D Bioprinting of Craniomaxillofacial Bone Tissue. ACS Appl. Mater. Interfaces 2020, 12, 23752–23763. [Google Scholar] [CrossRef]
  42. Fahimipour, F.; Dashtimoghadam, E.; Mahdi Hasani-Sadrabadi, M.; Vargas, J.; Vashaee, D.; Lobner, D.C.; Jafarzadeh Kashi, T.S.; Ghasemzadeh, B.; Tayebi, L. Enhancing Cell Seeding and Osteogenesis of MSCs on 3D Printed Scaffolds through Injectable BMP2 Immobilized ECM-Mimetic Gel. Dent. Mater. 2019, 35, 990–1006. [Google Scholar] [CrossRef]
  43. Gómez-Cerezo, M.N.; Lozano, D.; Arcos, D.; Vallet-Regí, M.; Vaquette, C. The Effect of Biomimetic Mineralization of 3D-Printed Mesoporous Bioglass Scaffolds on Physical Properties and in Vitro Osteogenicity. Mater. Sci. Eng. C 2020, 109, 110572. [Google Scholar] [CrossRef] [PubMed]
  44. Han, L.; Guo, Y.; Jia, L.; Zhang, Q.; Sun, L.; Yang, Z.; Dai, Y.; Lou, Z.; Xia, Y. 3D Magnetic Nanocomposite Scaffolds Enhanced the Osteogenic Capacities of Rat Bone Mesenchymal Stem Cells in Vitro and in a Rat Calvarial Bone Defect Model by Promoting Cell Adhesion. J. Biomed. Mater. Res. 2021, 109, 1670–1680. [Google Scholar] [CrossRef]
  45. Huang, K.-H.; Wang, C.-Y.; Chen, C.-Y.; Hsu, T.-T.; Lin, C.-P. Incorporation of Calcium Sulfate Dihydrate into a Mesoporous Calcium Silicate/Poly-ε-Caprolactone Scaffold to Regulate the Release of Bone Morphogenetic Protein-2 and Accelerate Bone Regeneration. Biomedicines 2021, 9, 128. [Google Scholar] [CrossRef] [PubMed]
  46. Jeong, J.E.; Park, S.Y.; Shin, J.Y.; Seok, J.M.; Byun, J.H.; Oh, S.H.; Kim, W.D.; Lee, J.H.; Park, W.H.; Park, S.A. 3D Printing of Bone-Mimetic Scaffold Composed of Gelatin/Β-Tri-Calcium Phosphate for Bone Tissue Engineering. Macromol. Biosci. 2020, 20, 2000256. [Google Scholar] [CrossRef] [PubMed]
  47. Kao, C.-T.; Lin, C.-C.; Chen, Y.-W.; Yeh, C.-H.; Fang, H.-Y.; Shie, M.-Y. Poly(Dopamine) Coating of 3D Printed Poly(Lactic Acid) Scaffolds for Bone Tissue Engineering. Mater. Sci. Eng. C 2015, 56, 165–173. [Google Scholar] [CrossRef] [PubMed]
  48. Ke, D.; Tarafder, S.; Vahabzadeh, S.; Bose, S. Effects of MgO, ZnO, SrO, and SiO2 in Tricalcium Phosphate Scaffolds on in Vitro Gene Expression and in Vivo Osteogenesis. Mater. Sci. Eng. C 2019, 96, 10–19. [Google Scholar] [CrossRef] [PubMed]
  49. Kim, B.-S.; Yang, S.-S.; Kim, C.S. Incorporation of BMP-2 Nanoparticles on the Surface of a 3D-Printed Hydroxyapatite Scaffold Using an ε-Polycaprolactone Polymer Emulsion Coating Method for Bone Tissue Engineering. Colloids Surf. B Biointerfaces 2018, 170, 421–429. [Google Scholar] [CrossRef] [PubMed]
  50. Kim, J.A.; Yun, H.; Choi, Y.-A.; Kim, J.-E.; Choi, S.-Y.; Kwon, T.-G.; Kim, Y.K.; Kwon, T.-Y.; Bae, M.A.; Kim, N.J.; et al. Magnesium Phosphate Ceramics Incorporating a Novel Indene Compound Promote Osteoblast Differentiation in Vitro and Bone Regeneration in Vivo. Biomaterials 2018, 157, 51–61. [Google Scholar] [CrossRef] [PubMed]
  51. Lee, S.J.; Won, J.-E.; Han, C.; Yin, X.Y.; Kim, H.K.; Nah, H.; Kwon, I.K.; Min, B.-H.; Kim, C.-H.; Shin, Y.S.; et al. Development of a Three-Dimensionally Printed Scaffold Grafted with Bone Forming Peptide-1 for Enhanced Bone Regeneration with in Vitro and in Vivo Evaluations. J. Colloid Interface Sci. 2019, 539, 468–480. [Google Scholar] [CrossRef] [PubMed]
  52. Li, J.; Chen, M.; Wei, X.; Hao, Y.; Wang, J. Evaluation of 3D-Printed Polycaprolactone Scaffolds Coated with Freeze-Dried Platelet-Rich Plasma for Bone Regeneration. Materials 2017, 10, 831. [Google Scholar] [CrossRef]
  53. Li, Y.; Li, Q.; Li, H.; Xu, X.; Fu, X.; Pan, J.; Wang, H.; Fuh, J.Y.H.; Bai, Y.; Wei, S. An Effective Dual-factor Modified 3D-printed PCL Scaffold for Bone Defect Repair. J. Biomed. Mater. Res. 2020, 108, 2167–2179. [Google Scholar] [CrossRef] [PubMed]
  54. Lin, Y.; Umebayashi, M.; Abdallah, M.-N.; Dong, G.; Roskies, M.G.; Zhao, Y.F.; Murshed, M.; Zhang, Z.; Tran, S.D. Combination of Polyetherketoneketone Scaffold and Human Mesenchymal Stem Cells from Temporomandibular Joint Synovial Fluid Enhances Bone Regeneration. Sci. Rep. 2019, 9, 472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lin, Y.-H.; Chiu, Y.-C.; Shen, Y.-F.; Wu, Y.-H.A.; Shie, M.-Y. Bioactive Calcium Silicate/Poly-ε-Caprolactone Composite Scaffolds 3D Printed under Mild Conditions for Bone Tissue Engineering. J. Mater. Sci. Mater. Med. 2018, 29, 11. [Google Scholar] [CrossRef] [PubMed]
  56. Lin, Y.-H.; Chuang, T.-Y.; Chiang, W.-H.; Chen, I.-W.P.; Wang, K.; Shie, M.-Y.; Chen, Y.-W. The Synergistic Effects of Graphene-Contained 3D-Printed Calcium Silicate/Poly-ε-Caprolactone Scaffolds Promote FGFR-Induced Osteogenic/Angiogenic Differentiation of Mesenchymal Stem Cells. Mater. Sci. Eng. C 2019, 104, 109887. [Google Scholar] [CrossRef] [PubMed]
  57. Martin, V.; Ribeiro, I.A.; Alves, M.M.; Gonçalves, L.; Claudio, R.A.; Grenho, L.; Fernandes, M.H.; Gomes, P.; Santos, C.F.; Bettencourt, A.F. Engineering a Multifunctional 3D-Printed PLA-Collagen-Minocycline-NanoHydroxyapatite Scaffold with Combined Antimicrobial and Osteogenic Effects for Bone Regeneration. Mater. Sci. Eng. C 2019, 101, 15–26. [Google Scholar] [CrossRef] [PubMed]
  58. Mi, X.; Su, Z.; Fu, Y.; Li, S.; Mo, A. 3D Printing of Ti3C2-MXene-Incorporated Composite Scaffolds for Accelerated Bone Regeneration. Biomed. Mater. 2022, 17, 035002. [Google Scholar] [CrossRef]
  59. Miao, Y.; Shi, X.; Li, Q.; Hao, L.; Liu, L.; Liu, X.; Chen, Y.; Wang, Y. Engineering Natural Matrices with Black Phosphorus Nanosheets to Generate Multi-Functional Therapeutic Nanocomposite Hydrogels. Biomater. Sci. 2019, 7, 4046–4059. [Google Scholar] [CrossRef]
  60. Midha, S.; Kumar, S.; Sharma, A.; Kaur, K.; Shi, X.; Naruphontjirakul, P.; Jones, J.R.; Ghosh, S. Silk Fibroin-Bioactive Glass Based Advanced Biomaterials: Towards Patient-Specific Bone Grafts. Biomed. Mater. 2018, 13, 055012. [Google Scholar] [CrossRef]
  61. Pan, T.; Song, W.; Xin, H.; Yu, H.; Wang, H.; Ma, D.; Cao, X.; Wang, Y. MicroRNA-Activated Hydrogel Scaffold Generated by 3D Printing Accelerates Bone Regeneration. Bioact. Mater. 2022, 10, 1–14. [Google Scholar] [CrossRef]
  62. Park, S.A.; Lee, H.; Kim, S.; Kim, K.; Jo, D.; Park, S. Three-dimensionally Printed Polycaprolactone/Beta-tricalcium Phosphate Scaffold Was More Effective as an rhBMP-2 Carrier for New Bone Formation than Polycaprolactone Alone. J. Biomed. Mater. Res. 2021, 109, 840–848. [Google Scholar] [CrossRef]
  63. Ratheesh, G.; Shi, M.; Lau, P.; Xiao, Y.; Vaquette, C. Effect of Dual Pore Size Architecture on In Vitro Osteogenic Differentiation in Additively Manufactured Hierarchical Scaffolds. ACS Biomater. Sci. Eng. 2021, 7, 2615–2626. [Google Scholar] [CrossRef]
  64. Remy, M.T.; Akkouch, A.; He, L.; Eliason, S.; Sweat, M.E.; Krongbaramee, T.; Fei, F.; Qian, F.; Amendt, B.A.; Song, X.; et al. Rat Calvarial Bone Regeneration by 3D-Printed β-Tricalcium Phosphate Incorporating MicroRNA-200c. ACS Biomater. Sci. Eng. 2021, 7, 4521–4534. [Google Scholar] [CrossRef]
  65. Roh, H.-S.; Lee, C.-M.; Hwang, Y.-H.; Kook, M.-S.; Yang, S.-W.; Lee, D.; Kim, B.-H. Addition of MgO Nanoparticles and Plasma Surface Treatment of Three-Dimensional Printed Polycaprolactone/Hydroxyapatite Scaffolds for Improving Bone Regeneration. Mater. Sci. Eng. C 2017, 74, 525–535. [Google Scholar] [CrossRef]
  66. Shim, J.-H.; Won, J.-Y.; Park, J.-H.; Bae, J.-H.; Ahn, G.; Kim, C.-H.; Lim, D.-H.; Cho, D.-W.; Yun, W.-S.; Bae, E.-B.; et al. Effects of 3D-Printed Polycaprolactone/β-Tricalcium Phosphate Membranes on Guided Bone Regeneration. IJMS 2017, 18, 899. [Google Scholar] [CrossRef] [Green Version]
  67. Shuai, C.; Yang, W.; Feng, P.; Peng, S.; Pan, H. Accelerated Degradation of HAP/PLLA Bone Scaffold by PGA Blending Facilitates Bioactivity and Osteoconductivity. Bioact. Mater. 2021, 6, 490–502. [Google Scholar] [CrossRef]
  68. Tcacencu, I.; Rodrigues, N.; Alharbi, N.; Benning, M.; Toumpaniari, S.; Mancuso, E.; Marshall, M.; Bretcanu, O.; Birch, M.; McCaskie, A.; et al. Osseointegration of Porous Apatite-Wollastonite and Poly(Lactic Acid) Composite Structures Created Using 3D Printing Techniques. Mater. Sci. Eng. C 2018, 90, 1–7. [Google Scholar] [CrossRef]
  69. Tsai, C.-H.; Hung, C.-H.; Kuo, C.-N.; Chen, C.-Y.; Peng, Y.-N.; Shie, M.-Y. Improved Bioactivity of 3D Printed Porous Titanium Alloy Scaffold with Chitosan/Magnesium-Calcium Silicate Composite for Orthopaedic Applications. Materials 2019, 12, 203. [Google Scholar] [CrossRef] [Green Version]
  70. Umeyama, R.; Yamawaki, T.; Liu, D.; Kanazawa, S.; Takato, T.; Hoshi, K.; Hikita, A. Optimization of Culture Duration of Bone Marrow Cells before Transplantation with a β-Tricalcium Phosphate/Recombinant Collagen Peptide Hybrid Scaffold. Regen. Ther. 2020, 14, 284–295. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, P.; Yin, H.-M.; Li, X.; Liu, W.; Chu, Y.-X.; Wang, Y.; Wang, Y.; Xu, J.-Z.; Li, Z.-M.; Li, J.-H. Simultaneously Constructing Nanotopographical and Chemical Cues in 3D-Printed Polylactic Acid Scaffolds to Promote Bone Regeneration. Mater. Sci. Eng. C 2021, 118, 111457. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, S.; Li, R.; Xu, Y.; Xia, D.; Zhu, Y.; Yoon, J.; Gu, R.; Liu, X.; Zhao, W.; Zhao, X.; et al. Fabrication and Application of a 3D-Printed Poly-ε-Caprolactone Cage Scaffold for Bone Tissue Engineering. BioMed Res. Int. 2020, 2020, 1–12. [Google Scholar] [CrossRef]
  73. Wu, Y.-H.; Chiu, Y.-C.; Lin, Y.-H.; Ho, C.-C.; Shie, M.-Y.; Chen, Y.-W. 3D-Printed Bioactive Calcium Silicate/Poly-ε-Caprolactone Bioscaffolds Modified with Biomimetic Extracellular Matrices for Bone Regeneration. IJMS 2019, 20, 942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Xia, D.; Qin, Y.; Guo, H.; Wen, P.; Lin, H.; Voshage, M.; Schleifenbaum, J.H.; Cheng, Y.; Zheng, Y. Additively Manufactured Pure Zinc Porous Scaffolds for Critical-Sized Bone Defects of Rabbit Femur. Bioact. Mater. 2023, 19, 12–23. [Google Scholar] [CrossRef] [PubMed]
  75. Xu, Z.; Wang, N.; Liu, P.; Sun, Y.; Wang, Y.; Fei, F.; Zhang, S.; Zheng, J.; Han, B. Poly(Dopamine) Coating on 3D-Printed Poly-Lactic-Co-Glycolic Acid/β-Tricalcium Phosphate Scaffolds for Bone Tissue Engineering. Molecules 2019, 24, 4397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Xu, Z.; Wang, N.; Ma, Y.; Dai, H.; Han, B. Preparation and Study of 3D Printed Dipyridamole/β-Tricalcium Phosphate/ Polyvinyl Alcohol Composite Scaffolds in Bone Tissue Engineering. J. Drug Deliv. Sci. Technol. 2022, 68, 103053. [Google Scholar] [CrossRef]
  77. Yun, S.; Choi, D.; Choi, D.-J.; Jin, S.; Yun, W.-S.; Huh, J.-B.; Shim, J.-H. Bone Fracture-Treatment Method: Fixing 3D-Printed Polycaprolactone Scaffolds with Hydrogel Type Bone-Derived Extracellular Matrix and β-Tricalcium Phosphate as an Osteogenic Promoter. IJMS 2021, 22, 9084. [Google Scholar] [CrossRef] [PubMed]
  78. Zamani, Y.; Amoabediny, G.; Mohammadi, J.; Zandieh-Doulabi, B.; Klein-Nulend, J.; Helder, M.N. Increased Osteogenic Potential of Pre-Osteoblasts on Three-Dimensional Printed Scaffolds Compared to Porous Scaffolds for Bone Regeneration. Iran. Biomed. J. 2021, 25, 78–87. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, Y.; Wang, C.; Fu, L.; Ye, S.; Wang, M.; Zhou, Y. Fabrication and Application of Novel Porous Scaffold in Situ-Loaded Graphene Oxide and Osteogenic Peptide by Cryogenic 3D Printing for Repairing Critical-Sized Bone Defect. Molecules 2019, 24, 1669. [Google Scholar] [CrossRef] [Green Version]
  80. Zhao, N.; Wang, Y.; Qin, L.; Guo, Z.; Li, D. Effect of Composition and Macropore Percentage on Mechanical and in Vitro Cell Proliferation and Differentiation Properties of 3D Printed HA/β-TCP Scaffolds. RSC Adv. 2017, 7, 43186–43196. [Google Scholar] [CrossRef] [Green Version]
  81. Zhong, L.; Chen, J.; Ma, Z.; Feng, H.; Chen, S.; Cai, H.; Xue, Y.; Pei, X.; Wang, J.; Wan, Q. 3D Printing of Metal–Organic Framework Incorporated Porous Scaffolds to Promote Osteogenic Differentiation and Bone Regeneration. Nanoscale 2020, 12, 24437–24449. [Google Scholar] [CrossRef]
  82. van Hout, W.M.M.T.; Mink van der Molen, A.B.; Breugem, C.C.; Koole, R.; Van Cann, E.M. Reconstruction of the Alveolar Cleft: Can Growth Factor-Aided Tissue Engineering Replace Autologous Bone Grafting? A Literature Review and Systematic Review of Results Obtained with Bone Morphogenetic Protein-2. Clin. Oral. Investig. 2011, 15, 297–303. [Google Scholar] [CrossRef]
  83. Khojasteh, A.; Kheiri, L.; Motamedian, S.R.; Nadjmi, N. Regenerative Medicine in the Treatment of Alveolar Cleft Defect: A Systematic Review of the Literature. J. Cranio-Maxillofac. Surg. 2015, 43, 1608–1613. [Google Scholar] [CrossRef] [PubMed]
  84. Kamal, M.; Ziyab, A.H.; Bartella, A.; Mitchell, D.; Al-Asfour, A.; Hölzle, F.; Kessler, P.; Lethaus, B. Volumetric Comparison of Autogenous Bone and Tissue-Engineered Bone Replacement Materials in Alveolar Cleft Repair: A Systematic Review and Meta-Analysis. Br. J. Oral Maxillofac. Surg. 2018, 56, 453–462. [Google Scholar] [CrossRef] [PubMed]
  85. Bose, S.; Sarkar, N.; Banerjee, D. Effects of PCL, PEG and PLGA Polymers on Curcumin Release from Calcium Phosphate Matrix for in Vitro and in Vivo Bone Regeneration. Mater. Today Chem. 2018, 8, 110–120. [Google Scholar] [CrossRef] [PubMed]
  86. Lee, J.S.; Park, T.H.; Ryu, J.Y.; Kim, D.K.; Oh, E.J.; Kim, H.M.; Shim, J.-H.; Yun, W.-S.; Huh, J.B.; Moon, S.H.; et al. Osteogenesis of 3D-Printed PCL/TCP/BdECM Scaffold Using Adipose-Derived Stem Cells Aggregates; An Experimental Study in the Canine Mandible. IJMS 2021, 22, 5409. [Google Scholar] [CrossRef] [PubMed]
  87. Lee, S.-H.; Lee, K.-G.; Hwang, J.-H.; Cho, Y.S.; Lee, K.-S.; Jeong, H.-J.; Park, S.-H.; Park, Y.; Cho, Y.-S.; Lee, B.-K. Evaluation of Mechanical Strength and Bone Regeneration Ability of 3D Printed Kagome-Structure Scaffold Using Rabbit Calvarial Defect Model. Mater. Sci. Eng. C 2019, 98, 949–959. [Google Scholar] [CrossRef]
  88. Ishack, S.; Mediero, A.; Wilder, T.; Ricci, J.L.; Cronstein, B.N. Bone Regeneration in Critical Bone Defects Using Three-Dimensionally Printed β-Tricalcium Phosphate/Hydroxyapatite Scaffolds Is Enhanced by Coating Scaffolds with Either Dipyridamole or BMP-2: AGENTS THAT STIMULATE A2A RECEPTORS FURTHER ENHANCE HA/Β-TCP SCAFFOLDS BONE REGENERATION. J. Biomed. Mater. Res. 2017, 105, 366–375. [Google Scholar] [CrossRef] [Green Version]
  89. Yun, J.; Heo, S.; Lee, M.; Lee, H. Evaluation of a Poly(Lactic-Acid) Scaffold Filled with Poly(Lactide-Co-Glycolide)/Hydroxyapatite Nanofibres for Reconstruction of a Segmental Bone Defect in a Canine Model. Vet. Med. 2019, 64, 531–538. [Google Scholar] [CrossRef] [Green Version]
  90. Le Guéhennec, L.; Van Hede, D.; Plougonven, E.; Nolens, G.; Verlée, B.; De Pauw, M.; Lambert, F. In Vitro and in Vivo Biocompatibility of Calcium-phosphate Scaffolds Three-dimensional Printed by Stereolithography for Bone Regeneration. J. Biomed. Mater. Res. 2020, 108, 412–425. [Google Scholar] [CrossRef]
  91. Ryu, J.-I.; Yang, B.-E.; Yi, S.-M.; Choi, H.-G.; On, S.-W.; Hong, S.-J.; Lim, H.-K.; Byun, S.-H. Bone Regeneration of a 3D-Printed Alloplastic and Particulate Xenogenic Graft with RhBMP-2. IJMS 2021, 22, 12518. [Google Scholar] [CrossRef]
  92. Won, J.-Y.; Park, C.-Y.; Bae, J.-H.; Ahn, G.; Kim, C.; Lim, D.-H.; Cho, D.-W.; Yun, W.-S.; Shim, J.-H.; Huh, J.-B. Evaluation of 3D Printed PCL/PLGA/ β -TCP versus Collagen Membranes for Guided Bone Regeneration in a Beagle Implant Model. Biomed. Mater. 2016, 11, 055013. [Google Scholar] [CrossRef]
  93. Bekisz, J.M.; Flores, R.L.; Witek, L.; Lopez, C.D.; Runyan, C.M.; Torroni, A.; Cronstein, B.N.; Coelho, P.G. Dipyridamole Enhances Osteogenesis of Three-Dimensionally Printed Bioactive Ceramic Scaffolds in Calvarial Defects. J. Cranio-Maxillofac. Surg. 2018, 46, 237–244. [Google Scholar] [CrossRef] [PubMed]
  94. Chang, P.-C.; Lin, Z.-J.; Luo, H.-T.; Tu, C.-C.; Tai, W.-C.; Chang, C.-H.; Chang, Y.-C. Degradable RGD-Functionalized 3D-Printed Scaffold Promotes Osteogenesis. J. Dent. Res. 2021, 100, 1109–1117. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, M.-C.; Chiu, H.-C.; Kuo, P.-J.; Chiang, C.-Y.; Fu, M.M.; Fu, E. Bone Formation with Functionalized 3D Printed Poly-ε-Caprolactone Scaffold with Plasma-Rich-Fibrin Implanted in Critical-Sized Calvaria Defect of Rat. J. Dent. Sci. 2021, 16, 1214–1221. [Google Scholar] [CrossRef] [PubMed]
  96. Diomede, F.; Gugliandolo, A.; Cardelli, P.; Merciaro, I.; Ettorre, V.; Traini, T.; Bedini, R.; Scionti, D.; Bramanti, A.; Nanci, A.; et al. Three-Dimensional Printed PLA Scaffold and Human Gingival Stem Cell-Derived Extracellular Vesicles: A New Tool for Bone Defect Repair. Stem Cell Res. Ther. 2018, 9, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. El-Habashy, S.E.; El-Kamel, A.H.; Essawy, M.M.; Abdelfattah, E.-Z.A.; Eltaher, H.M. Engineering 3D-Printed Core–Shell Hydrogel Scaffolds Reinforced with Hybrid Hydroxyapatite/Polycaprolactone Nanoparticles for in Vivo Bone Regeneration. Biomater. Sci. 2021, 9, 4019–4039. [Google Scholar] [CrossRef] [PubMed]
  98. Fama, C.; Kaye, G.J.; Flores, R.; Lopez, C.D.; Bekisz, J.M.; Torroni, A.; Tovar, N.; Coelho, P.G.; Witek, L. Three-Dimensionally-Printed Bioactive Ceramic Scaffolds: Construct Effects on Bone Regeneration. J. Craniofac. Surg. 2021, 32, 1177–1181. [Google Scholar] [CrossRef]
  99. He, M.; Hou, Y.; Zhu, C.; He, M.; Jiang, Y.; Feng, G.; Liu, L.; Li, Y.; Chen, C.; Zhang, L. 3D-Printing Biodegradable PU/PAAM/Gel Hydrogel Scaffold with High Flexibility and Self-Adaptibility to Irregular Defects for Nonload-Bearing Bone Regeneration. Bioconjug. Chem. 2021, 32, 1915–1925. [Google Scholar] [CrossRef]
  100. Jia, L.; Yang, Z.; Sun, L.; Zhang, Q.; Guo, Y.; Chen, Y.; Dai, Y.; Xia, Y. A Three-Dimensional-Printed SPION/PLGA Scaffold for Enhanced Palate-Bone Regeneration and Concurrent Alteration of the Oral Microbiota in Rats. Mater. Sci. Eng. C 2021, 126, 112173. [Google Scholar] [CrossRef]
  101. Johnson, Z.M.; Yuan, Y.; Li, X.; Jashashvili, T.; Jamieson, M.; Urata, M.; Chen, Y.; Chai, Y. Mesenchymal Stem Cells and Three-Dimensional-Osteoconductive Scaffold Regenerate Calvarial Bone in Critical Size Defects in Swine. Stem Cells Transl. Med. 2021, 10, 1170–1183. [Google Scholar] [CrossRef]
  102. Kim, J.-W.; Yang, B.-E.; Hong, S.-J.; Choi, H.-G.; Byeon, S.-J.; Lim, H.-K.; Chung, S.-M.; Lee, J.-H.; Byun, S.-H. Bone Regeneration Capability of 3D Printed Ceramic Scaffolds. IJMS 2020, 21, 4837. [Google Scholar] [CrossRef]
  103. Lee, D.J.; Kwon, J.; Kim, Y.; Wang, X.; Wu, T.; Lee, Y.; Kim, S.; Miguez, P.; Ko, C. Effect of Pore Size in Bone Regeneration Using Polydopamine-laced Hydroxyapatite Collagen Calcium Silicate Scaffolds Fabricated by 3D Mould Printing Technology. Orthod. Craniofac. Res. 2019, 22, 127–133. [Google Scholar] [CrossRef] [PubMed]
  104. Liang, T.; Wu, J.; Li, F.; Huang, Z.; Pi, Y.; Miao, G.; Ren, W.; Liu, T.; Jiang, Q.; Guo, L. Drug-loading Three-dimensional Scaffolds Based on Hydroxyapatite-sodium Alginate for Bone Regeneration. J. Biomed. Mater. Res. 2021, 109, 219–231. [Google Scholar] [CrossRef] [PubMed]
  105. Lim, H.-K.; Hong, S.-J.; Byeon, S.-J.; Chung, S.-M.; On, S.-W.; Yang, B.-E.; Lee, J.-H.; Byun, S.-H. 3D-Printed Ceramic Bone Scaffolds with Variable Pore Architectures. IJMS 2020, 21, 6942. [Google Scholar] [CrossRef]
  106. Liu, A.; Sun, M.; Yang, X.; Ma, C.; Liu, Y.; Yang, X.; Yan, S.; Gou, Z. Three-Dimensional Printing Akermanite Porous Scaffolds for Load-Bearing Bone Defect Repair: An Investigation of Osteogenic Capability and Mechanical Evolution. J. Biomater. Appl. 2016, 31, 650–660. [Google Scholar] [CrossRef]
  107. Lopez, C.D.; Diaz-Siso, J.R.; Witek, L.; Bekisz, J.M.; Gil, L.F.; Cronstein, B.N.; Flores, R.L.; Torroni, A.; Rodriguez, E.D.; Coelho, P.G. Dipyridamole Augments Three-Dimensionally Printed Bioactive Ceramic Scaffolds to Regenerate Craniofacial Bone: Plast. Reconstr. Surg. 2019, 143, 1408–1419. [Google Scholar] [CrossRef]
  108. Naudot, M.; Garcia Garcia, A.; Jankovsky, N.; Barre, A.; Zabijak, L.; Azdad, S.Z.; Collet, L.; Bedoui, F.; Hébraud, A.; Schlatter, G.; et al. The Combination of a Poly-caprolactone/Nano-hydroxyapatite Honeycomb Scaffold and Mesenchymal Stem Cells Promotes Bone Regeneration in Rat Calvarial Defects. J. Tissue Eng. Regen. Med. 2020, 14, 1570–1580. [Google Scholar] [CrossRef]
  109. Pae, H.; Kang, J.; Cha, J.; Lee, J.; Paik, J.; Jung, U.; Kim, B.; Choi, S. 3D-printed Polycaprolactone Scaffold Mixed with Β-tricalcium Phosphate as a Bone Regenerative Material in Rabbit Calvarial Defects. J. Biomed. Mater. Res. 2019, 107, 1254–1263. [Google Scholar] [CrossRef]
  110. Qiao, S.; Wu, D.; Li, Z.; Zhu, Y.; Zhan, F.; Lai, H.; Gu, Y. The Combination of Multi-Functional Ingredients-Loaded Hydrogels and Three-Dimensional Printed Porous Titanium Alloys for Infective Bone Defect Treatment. J. Tissue Eng. 2020, 11, 204173142096579. [Google Scholar] [CrossRef]
  111. Qin, H.; Wei, Y.; Han, J.; Jiang, X.; Yang, X.; Wu, Y.; Gou, Z.; Chen, L. 3D Printed Bioceramic Scaffolds: Adjusting Pore Dimension Is Beneficial for Mandibular Bone Defects Repair. J. Tissue Eng. Regen. Med. 2022, 16, 409–421. [Google Scholar] [CrossRef]
  112. Qin, Y.; Liu, A.; Guo, H.; Shen, Y.; Wen, P.; Lin, H.; Xia, D.; Voshage, M.; Tian, Y.; Zheng, Y. Additive Manufacturing of Zn-Mg Alloy Porous Scaffolds with Enhanced Osseointegration: In Vitro and in Vivo Studies. Acta Biomater. 2022, 145, 403–415. [Google Scholar] [CrossRef]
  113. Rogowska-Tylman, J.; Locs, J.; Salma, I.; Woźniak, B.; Pilmane, M.; Zalite, V.; Wojnarowicz, J.; Kędzierska-Sar, A.; Chudoba, T.; Szlązak, K.; et al. In Vivo and in Vitro Study of a Novel Nanohydroxyapatite Sonocoated Scaffolds for Enhanced Bone Regeneration. Mater. Sci. Eng. C 2019, 99, 669–684. [Google Scholar] [CrossRef] [PubMed]
  114. 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] [PubMed]
  115. Shim, J.-H.; Jeong, J.; Won, J.-Y.; Bae, J.-H.; Ahn, G.; Jeon, H.; Yun, W.-S.; Bae, E.-B.; Choi, J.-W.; Lee, S.-H.; et al. Porosity Effect of 3D-Printed Polycaprolactone Membranes on Calvarial Defect Model for Guided Bone Regeneration. Biomed. Mater. 2017, 13, 015014. [Google Scholar] [CrossRef] [PubMed]
  116. Tovar, N.; Witek, L.; Atria, P.; Sobieraj, M.; Bowers, M.; Lopez, C.D.; Cronstein, B.N.; Coelho, P.G. Form and Functional Repair of Long Bone Using 3D-printed Bioactive Scaffolds. J. Tissue Eng Regen. Med. 2018, 12, 1986–1999. [Google Scholar] [CrossRef]
  117. Tulyaganov, D.U.; Fiume, E.; Akbarov, A.; Ziyadullaeva, N.; Murtazaev, S.; Rahdar, A.; Massera, J.; Verné, E.; Baino, F. In Vivo Evaluation of 3D-Printed Silica-Based Bioactive Glass Scaffolds for Bone Regeneration. JFB 2022, 13, 74. [Google Scholar] [CrossRef]
  118. Ulbrich, L.M.; de Souza Balbinot, G.; Brotto, G.L.; Leitune, V.C.B.; Soares, R.M.D.; Collares, F.M.; Ponzoni, D. 3D Printing of Poly(Butylene Adipate-co-terephthalate) (PBAT)/Niobium Containing Bioactive Glasses (BAGNb) Scaffolds: Characterization of Composites, in Vitro Bioactivity, and in Vivo Bone Repair. J. Tissue Eng. Regen. Med. 2022, 16, 267–278. [Google Scholar] [CrossRef]
  119. Van Hede, D.; Liang, B.; Anania, S.; Barzegari, M.; Verlée, B.; Nolens, G.; Pirson, J.; Geris, L.; Lambert, F. 3D-Printed Synthetic Hydroxyapatite Scaffold With In Silico Optimized Macrostructure Enhances Bone Formation In Vivo. Adv. Funct Materials 2022, 32, 2105002. [Google Scholar] [CrossRef]
  120. Wang, M.M.; Flores, R.L.; Witek, L.; Torroni, A.; Ibrahim, A.; Wang, Z.; Liss, H.A.; Cronstein, B.N.; Lopez, C.D.; Maliha, S.G.; et al. Dipyridamole-Loaded 3D-Printed Bioceramic Scaffolds Stimulate Pediatric Bone Regeneration in Vivo without Disruption of Craniofacial Growth through Facial Maturity. Sci. Rep. 2019, 9, 18439. [Google Scholar] [CrossRef] [Green Version]
  121. Yu, L.; Yang, Y.; Zhang, B.; Bai, X.; Fei, Q.; Zhang, L. Rapid Human-derived IPSC Osteogenesis Combined with Three-dimensionally Printed Ti6Al4V Scaffolds for the Repair of Bone Defects. J. Cell. Physiol. 2020, 235, 9763–9772. [Google Scholar] [CrossRef]
  122. Zhang, W.; Feng, C.; Yang, G.; Li, G.; Ding, X.; Wang, S.; Dou, Y.; Zhang, Z.; Chang, J.; Wu, C.; et al. 3D-Printed Scaffolds with Synergistic Effect of Hollow-Pipe Structure and Bioactive Ions for Vascularized Bone Regeneration. Biomaterials 2017, 135, 85–95. [Google Scholar] [CrossRef]
  123. Bekisz, J.M.; Fryml, E.; Flores, R.L. A Review of Randomized Controlled Trials in Cleft and Craniofacial Surgery. J. Craniofac. Surg. 2018, 29, 293–301. [Google Scholar] [CrossRef] [PubMed]
  124. Ghayor, C.; Chen, T.-H.; Bhattacharya, I.; Özcan, M.; Weber, F.E. Microporosities in 3D-Printed Tricalcium-Phosphate-Based Bone Substitutes Enhance Osteoconduction and Affect Osteoclastic Resorption. IJMS 2020, 21, 9270. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Most used matrices in bone regeneration. HA—Hydroxyapatite; β-TCP—β-tricalcicum-phosphate; PLLA—Polylactic acid; PGA—Glycolic acid; PCL—Polycaprolactone.
Figure 1. Most used matrices in bone regeneration. HA—Hydroxyapatite; β-TCP—β-tricalcicum-phosphate; PLLA—Polylactic acid; PGA—Glycolic acid; PCL—Polycaprolactone.
Jfb 14 00076 g001
Figure 2. Flowchart.
Figure 2. Flowchart.
Jfb 14 00076 g002
Table 1. Characteristics of in vitro studies.
Table 1. Characteristics of in vitro studies.
Authors, YearControl GroupExperimental GroupCell CultureEvaluation TimeBone Regeneration Evaluation MethodPrinting TechniqueResultsConclusion
Alksne M. et al., 2020
[34]
PLLA scaffold -PLLA scaffold + HA 10%
-PLLA scaffold + BG
Rat dental pulp stem cells DPSCs1, 7, 10 daysALP activity evaluated by p-nitrophenol assay and osteogenesis-related gene expression quantified with qPCR Extrusion-based bioprintingThe scaffold with BG shows better osteoinductive properties than that with HAPLLA+BG scaffold is promising in bone regeneration
Bae E. et al., 2018
[35]
PCL/ β TCP scaffold-dECM/PCL/ β TCP scaffold
-dECM/PCL/β TCP/ rhBMP-2 scaffold
MC3T3-E1 cells (mouse preosteoblasts)1, 3, 5, 7, 14, 21 e 28 daysALP activity evaluated by p-nitro phenol assayExtrusion-based bioprintingThe dECM/PCL/β TCP/rhBMP-2 scafffold showed higher FA expression than the other scaffoldsdECM can be combined with rhBMP-2 to enhance bone regeneration
Cao Y. et al., 2019
[36]
β TCP scaffoldS1P coated β -TCP scaffoldRAW264.7 cells (macrophage cells) + BMSC cells (Rat bone marrow stromal cells)3 daysOsteogenic-related gene expression quantified by
qRT-PCR
3D-BioplotterS1P-coated β-TCP scaffold increased the expression of osteogenesis-related genesS1P-coated β-TCP scaffold promotes bone regeneration
Chen Y. et al., 2018
[37]
Cells cultured on the tissue culture plate without scaffold-PDASC/PCL scaffold
-PDASC/PCL/hydrogel scaffold
RFP-HUVEC cells + Wharton’s jelly mesenchymal stem cells (WJMSCs)1, 3, 7 daysOsteogenic-related protein secretion determined by an ELISAInkjet-based bioprintingPDASC/PCL/hydrogel scaffold showed higher expression of osteogenesis-related proteinsPDASC/PCL/hydrogel scaffold can be applied in bone regeneration
Chiu Y. et al., 2019 [38]SC scaffoldSrSC scaffoldMouse fibroblasts L929 cell line1, 3, 7 daysExpression levels of osteogenic-related proteins via western blot3D printingIncreased mineralization in the SrSC scaffoldSrSC scaffold is promising in bone regeneration
Cooke M. et al., 2020 [39] DPSCs without dexamethasone and β-glycerol-2-phosphate in a LayFomm scaffoldDPSCs with dexamethasone and β-glycerol-2-phosphate in a LayFomm scaffoldDental Pulp Stem Cells (DPSCs)21 daysHistological evaluation of the calcified matrix formedFused deposition modelingDPSCs with dexamethasone and β-glycerol-2-phosphate in a LayFomm scaffold are able to form mineralized matrixLayFomm is a promising scaffold for craniofacial bone regeneration
Dai Q. et al., 2021 [40]0Cu-BG-2Cu-BG
-5Cu-BG
-10Cu-BG
Mouse bone mesenchymal stem cells (BMSCs)1, 3, 7 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based hydrogel 3D printing-In the presence of Cu there is increased differentiation of stem cells
-The highest osteogenesis-related gene expression occurred in the group with 2Cu
Bioactive glass containing Cu promotes stem cell proliferation and regenerated bone tissue quality
Dubey N. et al., 2020 [41]Hydrogel scaffoldHydrogel scaffold with MgPDental pulp stem cells (DPSCs)7, 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Microvalve BioprintingThe scaffold with MP increased the expression of osteogenesis-related genesThe presence of MP in the scaffold can increase bone formation
Fahimipour F. et al., 2019 [42]βTCP/collagen/heparin scaffoldβTCP/collagen/heparin/ BMP-2 scaffoldMesenchymal stem cells (MSCs)7, 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingThe presence of BMP-2 led to an increased expression of osteogenesis-related genesThe β TCP/collagen/heparin/ BMP-2 scaffold is effective and should be explored for other bioactive molecules
Gómez-Cerezo M. et al., 2020 [43]BG/ PVA scaffold-BG/PVA-2d
-BG/PVA-30d
rBMSCs (femora marrow rats)3, 7 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based additive manufacturing methodThe BG/PVA-2d scaffold showed higher expression of genes related to osteogenesisImmersion of the BG/PVA scaffold in PBS improves the osteogenic properties of the scaffolf
Han L. et al., 2021 [44]PLGA scaffold without Fe coatingFe-coated PLGA scaffoldrBMSCs1, 2, 3, 7 e 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
3D printingFe-coated PLGA scaffold increased expression of osteogenesis-related genes3D scaffolds with nanocomposites enhance osteogenic differentiation of mesenchymal stem cells
Huang K. et al., 2021 [45]SC/ CS scaffoldSC/CS/BMP-2 scaffoldHuman dental pulp stem cells (hDPSCs)3 daysALP activity via western blotExtrusion-based bioprintingThe SC/CS/BMP-2 scaffold showed higher levels of osteogenic ALP activitySC/CS/BMP-2 scaffold is promising for bone regeneration
Jeong J. et al., 2020 [46]100% gelatin scaffoldGelatin and β-TCP scaffoldMC3T3-E1 preosteoblast cells7 daysALP activity evaluated by p-nitro phenol assayExtrusion-based bioprintingScaffolds with 60% β-TCP and 40% gelatin show the best cellular activityScaffolds with 60% β-TCP and 40% gelatin are a bone substitute with potential
Kao C. et al., 2015 [47]PLLA scaffoldPLLA/PDA scaffoldHuman adipose-derived stem cells (hADSCs)3, 7 daysALP activity evaluated by p-nitro phenol assayStereolithographyALP activity was higher in the PLLA/PDA scaffoldPDA is a promising tool in bone regeneration
Ke, D. et al., 2018 [48]β TCP scaffoldβ-TCP, SrO, SiO2, MgO and ZnO scaffoldHuman preosteoblast cell line (hFOB 1.19)3, 9 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Fused deposition modelingThe β TCP/MgO and β TCP/SiO2 scaffolds demonstrated the highest expression of osteogenesis-related genesThe β TCP/MgO and β TCP/SiO2 scaffolds are promising for bone regeneration
Kim B. et al., 2018 [49]PCL scaffoldPCL + BMP-2 + HA scaffoldHuman bone marrow-derived mesenchymal stem cells (hMSCs)7 daysALP activity3D printingThe PCL+ BMP-2 + HA scaffold increased the activity of FAOsteogenic properties are superior in the PCL + BMP-2 + HA scaffold
Kim J. et al., 2017 [50]MgP ceramic scaffoldMgP/KR-34893 scaffoldHuman bone marrow-derived mesenchymal stem cells (hMSCs)1, 3, 5, 7 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingMgP/ KR-34893 scaffold increased the expression of osteogenesis-related genesAddition of KR-34893 promotes greater osteogenic differentiation
Lee S. et al., 2018 [51]PCL scaffoldPCL/BFP-1 scaffoldHuman tonsil-derived mesenchymal stem cells (hTMSCs)7, 14 daysALP activity evaluated by p-nitro phenol assayFused deposition modelingThe PCL/BFP-1 scaffold was shown to have the highest osteogenic efficacyThe PCL/BFP-1 scaffold is promising is efficient in bone regeneration
Li J. et al., 2017 [52]PCL scaffold-PCL and traditional PRP scaffold
-PCL/PRP scaffold freeze-dried
Human dental pulps DPSCs7, 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Fused deposition modelingThe freeze-dried PCL/PRP scaffold increased the expression of osteogenesis-related genesThe freeze-dried PCL/PRP scaffold promotes greater bone formation
Li Y. et al., 2019 [53]PCL scaffoldPCL/Asp@Lipo/BFP-1 scaffoldHuman mesenchymal stem cells (hMSCs)7, 14, 21 daysALP activity quantified by AKP assay kit3D printing, method not describedThe 3:7 Asp@Lipo/BFP-1 ratio was shown to have the highest osteogenic efficacyThis is a promising scaffold for craniofacial bone regeneration
Lin Y. et al., 2019 [54]Culture of hSF-MSCsPEEK scaffold with hSF-MSCsHuman mesenchymal stem cells (MSCs)1, 4, 7, 14, 21 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Laser sintering techniquehSF-MSCs proliferate in the PEEK scaffoldPEEK/ hSF-MSCs is a promising scaffold in bone regeneration
Lin YH. et al., 2017 [55]PCL scaffold-PCL/10%SC scaffold
-PCL/30%SC scaffold
-PCL/50%SC scaffold
Wharton’s Jelly mesenchymal stem cells (WJMSCs)7 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingPCL/50% scaffold induced higher expression of osteogenesis-related genesPCL/SC scaffold shows favorable osteoconductive properties and is a promising biomaterial for bone regeneration
Lin YH. et al., 2019 [56]Neat grapheneGCP scaffoldHuman Wharton’s Jelly mesenchymal stem cells (WJMSCs)3, 7 daysOsteogenesis-related gene expression via western blotExtrusion-based bioprintingGCP scaffold induced higher expression of osteogenesis-related proteinsGCP scaffold promotes osteogenesis
Martin V. et al., 2019 [57]PLLA/col scaffold-PLLA/col/MH scaffold
-PLLA/col/MH/HA scaffold
Human bone marrow-derived mesenchymal stem cells (hMSCs)5, 10, 15 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprinting-Incorporation of HA increased the expression of osteogenesis-related genes
-The combination of HA and MH resulted in increased osteogenic activity
PLLA/col/MH/HA scaffolds stimulates osteogenesis and has a therapeutic action against Staphylococcus aureus, which makes it promising in bone regeneration
Mi X. et al., 2022 [58]HA/Sodium alginate scaffoldHA/Sodium alginate/Ti3C2 MXeneBone mesenchymal stem cells (BMSCs)7, 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingThe experimental scaffold exhibited excellent biocompatibility, promoted cell proliferation and upregulated osteogenic gene expressionTi3C2 MXene composite 3D-printed scaffolds are promising for clinical bone defect treatment
Miao Y. et al., 2019 [59]Hidrogel scaffoldHidrogel scaffold with FPMesenchymal stem cells hMSCs7, 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Nanosheets via liquid phase stripping methodThe addition of FP increased the osteogenesis-related gene expressionHydrogel and FP scaffold may constitute a good strategy for bone regeneration
Midha S. et al., 2018 [60]Bioactive glass 45S5-Bioactive Silk Fibrin Glass with Strontium
-Strontium-free fibrin silk bioactive glass
TVA-BMSC cell line21 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingThe strontium group showed higher expression of osteogenesis-related genesSilk fibrin bioactive glass promising for bone formation
Pan T. et al., 2022 [61]Hydrogel scaffold combined with miRNAHydrogel scaffold with miRNA and 0.25;1;2.5% GTAMesenchymal stem cells hMSCs7, 14, 21, 28, 42 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingThe scaffold with 1% GTA presented the best characteristics for bone regenerationThe hydrogel/miRNA/1%GTA scaffold is promising for bone regeneration
Park J. et al., 2015 [32]PCL scaffoldPCL/VEGF/BMP-2 scaffoldHuman dental pulp stem cells (DPSCs)7, 14 daysGrowth Factor Release RateExtrusion-based bioprintingBone regeneration was superior in the scaffold with growth factorsScaffolds with growth factors are a promising alternative
Park S. et al., 2020 [62]PCL scaffoldPCL/ β TCP scaffoldMouse preosteoblast cell line MC3T3-E17 daysALP activity quantified by AKP assay kitSelective laser sinteringThe PCL/ β TCP scaffold showed higher ALPThe addition of β TCP to the PCL scaffold is advantageous for bone regeneration
Ratheesh G. et al., 2021 [63]FDM-manufactured PCL scaffoldPCL scaffold by FDM and MEWHuman joint tissue explant cells3, 7, 21 daysOsteogenesis-related gene expression quantified by
qRT-PCR
MEW and FDMThe PCL scaffold by FDM/MEW showed higher expression of genes related to osteogenesisMEW membrane promotes a more favorable environment for osteogenic differentiation
Remy M. et al., 2021 [64]β TCP/miRNA scaffoldβTCP/miRNA/collagen scaffoldPrimary human BMSCs (hBMSCs)7 daysOsteogenesis-related gene expression quantified by
qRT-PCR
StereolithographyThe β TCP/miRNA/collagen scaffold showed higher expression of osteogenesis-related genesThe β TCP/miRNA/collagen scaffold is promising in the treatment of bone defects
Roh H. et al., 2016 [65]PCL/HA scaffoldPCL/HA and MgO scaffoldPre-osteoblast (MC3T3-E1) cells1, 3 e 5 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingThe addition of MgO increased the osteogenesis-related gene expressionPCL/HA/MgO scaffold is promising for bone formation
Shim J. et al., 2017 [66]Collagen membrane-PCL scaffold
-PCL/ β-TCP scaffold
NIH3T3 (mouse fibroblasts) + MC3T3-E1 (mouse preosteoblasts1, 4, 7,
14 days
Proliferation rates of fibroblastsMultilayer membrane 3D printingOsteogenic differentiation was higher in the PCL/ β-TCP scaffoldThe PCL/ β-TCP scaffold shows good results in bone regeneration
Shuai C. et al., 2020 [67]HA/PLLA scaffoldHA/PLLA e PGA scaffoldMG-63 human osteoblast-like cells8 weeksFormation of mineralized matrixLaser-assisted bioprintingThe HA/PLLA/PGA scaffold has proven to be a suitable environment for cell cultureThe HA/PLLA/PGA scaffold is capable of bone and vascular formation
Tcacencu I. et al., 2018 [68]-SW ceramic glass-ceramic scaffold
-PLLA scaffold
AW/PLLA scaffoldBone marrow-derived stromal cells (BMSCs)7, 14 daysALP activity evaluated by p-nitro phenol assayIndirect 3D printing/fused filament fabricationThe AW scaffold showed higher activity of ALPAW scaffold has good osteoconductive properties
Tsai C. et al., 2019 [69]Ti scaffoldTi scaffold with Mg- SC and CH Human Wharton’s Jelly mesenchymal stem cells (WJMSCs)3, 7 daysALP activity quantified by AKP assay kitSelective laser meltingThe Ti/Mg-CS/CH scaffold increased the activity of ALPTi/Mg-CS/CH scaffold increases osteogenesis
Umeyama R. et al., 2020 [70]β TCP scaffoldβ TCP/RCP scaffoldBone marrow cells isolated from C57BL/6J mice4, 7, 14 daysOsteogenesis-related gene expression quantified by
qRT-PCR
3D printingThe β TCP/RCP showed higher Osteogenesis-related gene expressionThe addition of RCP is efficient in bone regeneration
Wang P. et al., 2021 [71]PLLA scaffold-Sodium hydroxide conditioned PLLA scaffold
-PlA scaffold with PDA conditioned with NaOH
Bone marrow stromal cells (BMSCs)7, 14 daysALP activity evaluated by p-nitro phenol assayFused deposition modellingThe PLLA scaffold with PDA conditioned with sodium hydroxide showed higher activity of ALPPLLA scaffold with PDA conditioned with sodium hydroxide is promising for bone formation
Wang S. et al., 2020 [72]PCL e Bio-Oss scaffold PCL/ Bio-Os/NaOH scaffoldHuman bone marrow-derived mesenchymal stem cells (hBMMSCs)7, 14 daysALP activity quantified by AKP assay kitFused deposition modelingThe PCL/ Bio-Oss/NaOH scaffold increased ALPPCL/ Bio-Oss/NaOH scaffold is promising for bone formation
Weinand C. et al., 2006 [31]β TCP scaffold β TCP/type I collagen in hydrogel scaffoldBone-marrow-derived differentiated mesenchymal stem cells (MSCs)6 weeksOsteogenesis-related gene expression quantified by
qRT-PCR
Inkjet-based bioprintingOsteogenesis-related gene expression was higher in β TCP/type I collagen scaffoldThe β TCP/type I collagen scaffold is promising for bone formation
Wu Y. et al., 2019 [73]SC and PCL scaffolddECM/SC/PCL scaffoldHuman Wharton’s Jelly mesenchymal stem cells (WJMSCs)6 h, 1 and 7 daysOsteogenesis-related gene expression quantified by
qRT-PCR
Extrusion-based bioprintingThe dECM/SC/PCL scaffold increased the expression of osteogenesis-related genesdECM/SC/PCL scaffold is promising for bone regeneration
Xia D. et al., [74]Zinc scaffold Pure zinc porous scaffold Mouse pre-osteogenic cells (MC3T3-E1 cell line)7, 14 daysOsteogenesis-related gene expression quantified by qRT-PCRLaser powder bed fusion technologyPure zinc porous scaffold showed higher expression of osteogenesis-related genesPure Zn porous scaffolds with customized structures represent a promising biodegradable solution for treating large bone defect
Xu Z. et al., 2019 [75]β TCP/PLGA scaffoldβ TCP/PLGA/PDA scaffoldMouse pre-osteogenic cells (MC3T3-E1 cell line)7, 14 daysALP activity quantified by AKP assay kitExtrusion-based bioprintingβ TCP/PLGA/PDA scaffold increased ALP activityThe addition of PDA promotes osteogenesis
Xu Z. et al., 2022 [76]β TCP/PVA scaffoldβ TCP/ PVA/ dipyridamole scaffoldMouse pre-osteogenic cells (MC3T3-E1 cell line)7, 14 daysALP activity quantified by ALP assay kitExtrusion-based bioprintingThe β TCP/ PVA/ dipyridamole scaffold increased ALPβ TCP/PVA/dipyridamole composite scaffolds have brilliant potential in new bone formation as a suitable alternative
Yun S. et al., 2021 [77]PCL scaffolddECM/β TCP/PCL scaffoldMG63 cells1, 3, 5, 7, 14 daysALP activity quantified by AKP assay kitExtrusion-based bioprintingThe dECM/ β TCP/PCL scaffold increased ALPThe dECM/β TCP/PCL scaffold was shown to have superior osteogenic potential
Zamani Y. et al., 2021 [78]β TCP/PLGA scaffold by solvent/leach technique3D printed β TCP/ PLGA scaffoldMC3T3-E1 pre-osteoblasts14 daysALP activity evaluated by p-nitro phenol assayExtrusion-based bioprintingThe β TCP/ PLGA 3D scaffold showed higher ALP activityThe β TCP/ PLGA 3D scaffold is more favorable for bone formation
Zhang Y. et al., 2019 [79]β TCP/PLGA scaffoldβ TCP/PLGA/OG/BMP-2 scaffoldrMSCs1, 4, 7 daysALP activity evaluated by p-nitro phenol assayExtrusion-based bioprintingβ TCP/ PLGA/ OG/ BMP-2 scaffold increased ALP activityβ TCP/PLGA/OG/BMP-2 is a promising scaffold for bone regeneration
Zhang Z. et al., 2021 [33]p-Ta scaffoldp-Ta-nt scaffoldMC3T3-E1 preosteoblasts7 daysOsteogenesis-related gene expression quantified by qRT-PCR3D printing laser melting systemTantalum scaffold with nanotubes showed higher expression of osteogenesis-related genesTantalum scaffold with nanotubes holds promise for bone formation
Zhao N. et al., 2017 [80]β TCP scaffold e HÁ scaffoldHA/β TCP scaffold with different HA compositions (0.20, 0.40, 0.60, 0.80 and 1.00)Bone mesenchymal stem cells (BMSCs)1, 4, 7 daysOsteogenesis-related gene expression quantified by qRT-PCR3D printing40% HA scaffold showed higher osteogenic capacityHA / β TCP scaffold is promising for bone formation
Zhong L. et al., 2020 [81]PCL scafold-PCL/DCPD scaffold
-PCL/DCPD and nanoZIF-8 scaffold
Bone mesenchymal stem cells (BMSCs)25 daysOsteogenesis-related gene expression quantified by qRT-PCRExtrusion-based bioprintingPCL/DCPD/nanoZIF-8 scaffold increased osteogenesis-related gene expressionThe PCL/DCPD/ nanoZIF-8 scaffold is a bone substitute with potential
3D—three dimensional, Asp@Lipo—aspirin loaded liposomes, AW—apatite-volastonite, BFP-1—bone forming peptide 1, BG—bioactive glass, BG/PVA-2d—bioactive glass/polyvinyl acid in phosphate-salt buffer 2 days, BG/PVA-30d—bioactive glass/polyvinyl acid in phosphate buffered saline 30 days, Bio-Oss—deproteinized bovine bone mineral, BMP-2—bone morphogenetic protein type-2, CH—chitosan, CS—calcium sulfate, Cu—copper, Cu (10Cu-BG) —bioactive glass with 15% copper, Cu (2Cu-BG) —bioactive glass with 7% copper, Cu (5Cu-BG)—bioactive glass with 10% copper, DCPD—calcium phosphate dihydrate, dECM—decellularized extracellular matrix, FA—alkaline phosphatase, Fe—iron, FDM—fusion and deposition method, FP—black phosphorus, GCP—calcium silicate with graphene/polycaprolactone, GTA—glutaraldehyde, HA—hydroxyapatite, hSF-MSCs—synovial mesenchymal stem cells, KR-34893—bioactive organic compound, MEW—melt electrospinning writing, MgO—magnesium oxide, MgP—magnesium phosphate, MH—minocycline, miRNA—microRNA, nanoZIF-8—nanoscale zeolitic imidazolate framework-8, NaOH—sodium hydroxide, nt—nanotubes, OG—graphene oxide, PCL—polycaprolactone, PDA—polydopamine, PDASC—polydopamine modified calcium silicate, PEEK—polyetheretherketone, PGA—polyglycolic acid, PLGA—poly(lactic acid-co-glycolic acid), PLLA—polylactic acid, PLLA/col—polylactic acid/collagen, PRP—platelet-rich plasma, p-Ta—porous tantalum, PVA—polyvinyl acid, PBS—phosphate-saline buffer, RCP—recombinant collagen peptide, rhBMP-2—human recombinant bone protein type 2, S1P—sphingosine-1-phosphate, SC—calcium silicate, SiO2—silica, SrO—strontium oxide, SrSC—calcium strontium silicate, Ti—titanium, VEGF—endothelial growth factor, ZnO—zinc oxide, β TCP—β-tricalcium phosphate.
Table 2. Characteristics of in vivo studies.
Table 2. Characteristics of in vivo studies.
Authors, YearSample Size (n)/Animal ModelControl GroupExperimental GroupEvaluation TimeBone Regeneration Evaluation MethodPrinting TechniqueResultsConclusion
Bae E. et al., 2018 [35]n = 28 male SD rats Group without scaffold (n = 7)-Group with scaffold PCL/β-TCP (n = 7)
-Group with scaffold dECM/ PCL/β-TCP (n = 7)
-Group with scaffold dECM/ PCL/β-TCP/rhBMP-2 (n = 7)
4 weeksμ-CT, histologyExtrusion-based 3D printingBone formation was significantly higher in the group with the dECM/ PCL/β-TCP/rhBMP-2 scaffold (43.32% ± 7.63)The dECM/PCL/β-TCP/rhBMP-2 scaffold promotes bone regeneration
Bekisz J. et al., 2018 [93]n = 10 defects in 5 Finn Dorset sheepsGroup with HA/ β-TCP/collagen scaffold (n = 5)Group with HA/ β-TCP/collagen/dipyridamole 100 μM scaffold (n = 5)3, 6 weeksμ-CT, histologyExtrusion-based 3D printingOsteogenesis was higher in the experimental group at 3 and 6 weeksDipyridamole significantly increases the capacity for bone regeneration
Bose S. et al., 2018 [85]Male SD rats Group with β-TCP scaffoldGroup with β-TCP/curcumin/PCL/PEG scaffold 6 weeksHistologyBinder jettingThe formation of mineralized bone, after 6 weeks, was higher in the experimental group (44.9%)The β-TCP/curcumin/PCL/PEG scaffold is an excellent candidate for bone regeneration
Chang P. et al., 2021 [94]n = male SD rats Group without scaffold (n = 6)-Group with HA scaffold (n = 6)
-Group with HA and nonoxidized RGD peptide with lower stiffness (n = 6)
-Group with HA scaffold and nonoxidized RGD peptide with osteoid-like stiffness (n = 6)
-Group with HA scaffold and oxidized RGD peptide with osteoid-like stiffness (n = 6)
7, 28 daysμ-CT, HistologyExtrusion-based 3D printing-Limited bone regeneration was observed in the group with HA scaffold and nonoxidized RGD peptide with osteoid-like stiffness
-There was greater bone formation at both time points in the group with HA scaffold and oxidized RGD peptide with osteoid-like stiffness
The combination of HA with oxidized RGD peptide in a osteoid-like stiffness scaffold may be beneficial for maxillofacial regeneration
Chen M. et al., 2021 [95]n = 32 male SD rats Group without scaffold (n = 8)-Group with PRF (n = 8)
-Group with PCL scaffold (n = 8)
-Group with PRF/PCL scaffold (n = 8)
4, 8 weeksμ-CT, histologyFused deposition modeling-More mineralization was observed in the groups with scaffold at 4 and 8 weeks
-The presence of PRF did not influence bone formation
The use of PCL scaffolds enhances bone formation
Chiu Y. et al., 2019 [38]New Zealand rabbitsGroup with SC scaffoldGroup with SrSC scaffold4, 8 weeksμ-CT, histology3D printingThere is more bone and vascular formation in the experimental group at 4 (26.3 ± 1.9%) and 8 weeks (45.7 ± 6.2%)SrSC scaffold enhances bone regeneration
Cooke M. et al., 2020 [39]n = 12 male SD ratsGroup without LayFomm scaffold (n = 6)Group with LayFomm scaffold (n = 6)6 weeksμ-CTFused deposition modeling-The mechanical properties of the scaffold are a limitation in large defects
-There is greater production of mineralized tissue in the group with LayFomm scaffold
LayFomm scaffold is promising in craniofacial regeneration
Dai Q. et al., 2021 [40]n = 40 defects in 20 male SD ratsDefects without scaffold-Defects with Gel/SF scaffold
-Defect with Gel/SF/0Cu-BG scaffold
-Defect with Gel/SF/2Cu-BG, Gel/SF/5Cu-BG and Gel/SF/10Cu-BG scaffold
4, 8 weeksμ-CT, histologyExtrusion-based hydrogel 3D printing-The group with the Gel/SF/2Cu-BG scaffold produced the largest number of blood vessels
-At 4 weeks, the Gel/SF/5Cu-BG scaffold presented the highest bone formation
-At 8 weeks, the Gel/SF/2Cu-BG scaffold presented the highest bone formation
The most effective scaffold for bone regeneration was Gel/SF/5Cu-BG
Diomede F. et al., 2018 [96]n = 24 male Wistar ratsGroup with PLLA scaffold (n = 4)-Group with PLLA scaffold and hGMSCs (n = 4)
-Group with PLLA/EV scaffold (n = 4)
-Group with PLLA/hGMSCs/EVs scaffold (n = 4)
-Group with PLLA/PEI-EVs scaffold (n = 4)
-Group with PLLA/EIP-EVs/hGMSCs scaffold (n = 4)
6 weeksμ-CTFused deposition modelingThe groups with the PLLA/PEI-EVs and PLLA/PEI-EVs/ hGMSCs scaffolds demonstrated greater bone regeneration and better osteogenic properties with 12.27% and 9.71% new bone formation, respectivelyPLLA scaffolds conjugated with PEI-EVs are promising in bone regeneration
Dubey N. et al., 2020 [41]n = 16 male Fisher 344 ratsGroup without scaffold (n = 4)-Group with PTFE (n = 4)
-Group with ECM scaffold (n = 4)
-Group with ECM/MgP scaffold (n = 4)
4, 8 weeksμ-CT, histologyMicrovalve 3D printing-The control group and the PTFE membrane group showed little bone formation
-In the group with the ECM/AMP scaffold, a greater bone density was observed at 4 and 8 weeks than in the other groups
The presence of MgP enhances bone regeneration and is promising for bone defect repair
El-Habashy S. et al., 2021 [97]n = 24 New Zealand rabbitsGrupo without scaffold (n = 6)-Group with polyvinyl acid scaffold (n = 6)
-Group with HA scaffold (n = 6)
-Group with HA/PCL scaffold (n = 6)
2, 6 weeksμ-CTExtrusion-based 3D printingThe HA/PCL scaffold showed better biocompatibility, osteoconduction and osteogenic properties at both time pointsHA/PCL scaffold is promising in bone defect repair
Fahimipour F. et al., 2019 [42]n = 15 male Fisher 344 rats-Group with β-TCP/collagen/heparin scaffold (n = 5)
-Group with β-TCP/collagen/BMP-2 scaffold (n = 5)
Group with β TCP/collagen/heparin/BMP-2 scaffold
(n = 5)
6 weeksHistology, qPCRInkjet-based 3D printingThe experimental group showed superior osteogenic differentiation and increased bone formationThe bioactive molecule BMP-2 increases scaffold efficiency in bone regeneration
Fama C. et al., 2020 [98]n = 14 defects in 7 rats-------Group with porous β-TCP scaffold
(n = 7)
-Group with non-porous β-TCP scaffold (n = 7)
8 weeksμ-CT, histology3D printed scaffolds -In the groups with the non-porous scaffold, greater bone formation was observed
-The porous scaffold exhibited greater soft tissue volume
Non-porous scaffold enhances bone regeneration
Guéhennec L. et al., 2019 [90]n = 12 male SD ratsGroup with HA scaffold (n = 6)Group with HA:60- β TCP:40 scaffold (n = 6)3, 6 monthsμ-CT, histologyStereolithographyThe groups showed similar amount of bone formed 3 and 6 months after interventionCalcium phosphate scaffolds have good osseointegration and biocompatibility and should be studied to achieve the ideal level of bone regeneration
Han L. et al., 2021 [44]n = 14 male SD ratsGroup without scaffold (n = 6)-Group with Fe-coated PLGA scaffold (n = 4)
-Group with PLGA scaffold without Fe coating (n = 4)
8 weeksμ-CT3D printing The amount of bone formed was higher in the Fe-coated scafold, followed by the uncoated scaffoldMagnetic scaffold promotes bone regeneration
He M. et al., 2021 [99]n = 12 female SD ratsGroup without scaffold (n = 4)Group with hydrogel scaffold with PPG-1.5 (n = 4)4 weeksHistologyExtrusion-based 3D printingIn the group with the PPG-1.5 scaffold, bone formation was higherPPG-1.5 scaffold provides good mechanical support for bone growth
Huang K. et al., 2021 [45]n = 6 male New Zealand rabbitsGroup with SC/CS scaffold (n = 3)Group with SC/CS/BMP-2 scaffold
(n = 3)
4 weeksμ-CT, histologyExtrusion-based 3D printingThe MS/CS/BMP-2 scaffold promoted greater vascular and bone growthThe MS/CS scaffold can act as a carrier for BMP-2 and is an ideal biomaterial for bone regeneration
Ishack S. et al., 2017 [88]n = 15 murine ratsGroup with HA/β-TCP scaffold (n = 5)-Group with HA/ β-TCP/dipyridamole scaffold (n = 5)
-Group with HA/ β-TCP/BMP-2 scaffold (n = 5)
2, 4, and 8 weeksμ-CT, histologyExtrusion-based 3D printingThe experimental groups demonstrated greater bone formation at 2, 4 and 8 (47.5 ± 5% for dipyridamole and 48.3 ± 4% for BMP-2) weeks compared to the control groupAddition of dipyridamole and BMP-2 to HA/ β-TCP scaffold promotes bone formation
Jeong J. et al., 2020 [47]n = 20 male SD rats
Group with 100% gelatin scaffold (n = 4)Group with gelatin scaffold (40%) and β-TCP (60%)4 weeksμ-CTExtrusion-based 3D printingThe scaffold with β-TCP induced significantly more bone formationThe presence of β-TCP provides a more favorable environment for bone formation
Jia L. et al., 2021 [100]n = 18 male SD ratsGroup without scaffold (n = 6)-Group with PLLA scaffold (n = 6)
-Group with PLLA scaffold and iron oxide (n = 6)
4 weeksμ-CTDirect ink writing techniqueIron oxide scaffold promoted bone formation and altered the composition of the oral microbiomIron oxide scaffold can be used to treat bone defects of the palate
Johnson Z. et al., 2021 [101]n = 6 yorkshire farm pigsGroup without scaffold (n = 3)Group with HA/ β-TCP scaffold (n = 3)8 weeksμ-CT, histologyStereolithographyBone regeneration was superior in the group with the HA/ β-TCP scaffoldHA/ β-TCP scaffold seems to be effective in bone regeneration
Ke D. et al., 2018 [48]n=12
rat distal femoral defects
Group with β TCP scaffoldGroup with β TCP, SiO2, and MgO scaffold8, 12, 16 weeksHistologyFused deposition modeling-At week 8, both groups had similar amounts of mineralized bone
-The experimental group presented greater bone formation at 12 and 16 weeks
The β TCP/Si/Mg scaffold significantly increased osteogenesis compared to the control group matrix, making it promising for bone regeneration
Kim J. et al., 2020 [102]n = 12 adult male beaglesGroup without scaffold (n = 4)-Group with β-TCP/ HA scaffold without synthetic polymer (n = 4)
-Group with β-TCP/ HA scaffold with synthetic polymer (n = 4)
4, 8 weeksHistology, imagiologiaStereolithographyThe group with the β-TCP/ HA scaffold without the synthetic polymer showed greater bone regeneration in both momentsThe β-TCP/ HA scaffold without the synthetic polymer can be used for bone regeneration
Kim J. et al., 2017 [50]n = 24 male SD ratsGroup without scaffold (n = 6)-Group with MgP scaffold (n = 6)
-Group with MgP scaffold and 5 μM of KR-34893 (n = 6)
-Group with MgP scaffold and 25 μM KR-34893 (n = 6)
4, 8 weeksμ-CT, histologyExtrusion-based 3D printing-The number of osteoclasts decreases in the presence of KR-34893
- Bone formation is higher in groups with scaffold containing KR-34893
The compound KR-34893 is gradually released from the scaffold, increasing bone volume
Lee D. et al., 2018 [103]n = 12 male SD rats------Group with HCCS-PDA scaffold and 250 μm pore size (n = 6)
Group with HCCS-PDA scaffold and 500 μm pores
(n = 6)
8 weeksμ-CT, histologyDigital light processing-type 3D printing system-Limited bone growth was observed in the group with the 250 μm pore scaffold
-The group with the 500 μm pore scaffold showed greater bone regeneration
The pore size of the HCCS-PDA scaffold that induces the most effective bone regeneration is 500 μm
Lee J. et al., 2021 [86]n = 10 beaglesGroup with PCL/ β-TCP/dECM scaffold
(n = 5)
Group with PCL/ β-TCP/bdECM scaffold + ADSC injection
(n = 5)
8 weeksμ-CT, histology, qPCRFused deposition modelingThe experimental group demonstrated greater expression of genes related to osteogenesis and osteoblastsInjection of stem cells derived from adipose tissue enhances ossification
Lee S. et al., 2019 [51]n = 12 Male New Zealand white rabbits Group with PCL scaffold (n = 3)-Group with PCLD scaffold (n = 3)
-Group with PCLDB100 scaffold (n = 3)
-Group with PCLDB1000 scaffold (n = 3)
8 weeksHistology, imagiologiaFused deposition modelingIn the group treated with PCLDB1000 scaffold, a higher rate of bone formation and number of blood vessels was observedPCLDB1000 scaffold is promising for bone regeneration
Lee SH. et al., 2019 [87]New Zealand rabbitsGroup with PCL scaffoldGroup with PCL kagome-structure scaffold4, 16 weeksμ-CT, histology, immunohistochemistryExtrusion-based 3D printingThe experimental group demonstrated bone formation at 4 and 16 weeksThe scaffold with kagome-structure can be applied in bone defect reconstruction
Liang T. et al., 2021 [104]n = 9 beaglesGroup without scaffold -Group with HA/SA scaffold
-Group with HA/SA/NG scaffold
-Group with HA/SA/CGRP scaffold
1, 2, and 3 monthsμ-CTMicro extrusion 3D printing -Greater bone growth was observed in the experimental groups at months 1, 2, and 3
-The groups with HA/SA/NG and HA/SA/CGRP scaffolds demonstrated greater osteogenic potential
-HA/SA scaffold is promising for bone regeneration
-NG and CGRP may lead to increased bone proliferation
Li J. et al., 2017 [52]n = 24 ratos machos SDGrupo com matriz PCL
(n = 8)
-Group with PCL matrix and traditional PRP (n = 8)
-PCL matrix/PRP freeze-dried (n= 8)
2, 4, 8, 12 weeksμ-CT, histologyFused deposition modeling-Addition of freeze-dried PRP to the PCL matrix promotes greater bone regenerationAddition of freeze-dried PRP to the PCL matrix promotes greater bone regeneration
Li Y. et al., 2019 [53]Male New Zealand rabbits Group without scaffold-Group with PCL/Asp@Lipo/BFP-1 scaffold
-Group with PCL/Asp@Lipo scaffold
-Group with PCL/BFP-1 scaffold
8 weeksHistology3D printing, method not describedThe group treated with PCL/Asp@Lipo/BFP-1 scaffold showed greater bone formation, followed by the group treated with PCL/BFP-1The hybrid scaffold PCL/Asp@Lipo/BFP-1 showed good osteogenic properties
Lim H. et al., 2020 [105]n = 12 male New Zealand rabbits-----Group with HA/TCP scaffols with pores 0.8; 1.0; 1.2; 1.4 mm4, 8 weeksμ-CTDigital light processing-At week 4, larger pores result in greater bone formation
-At week 8, there was no correlation between % bone formation and pore size
Pore size only influences bone regeneration in the initial phase
Lin YH. et al., 2019 [54]n = 10 female New Zealand rabbitsGroup without PEEK scaffold-Group with PEEK scaffold and hSF-MSCs in standard culture medium
-Group with PEEK scaffold + hSF-MSCs in osteogenic culture medium
-Group with PEEK scaffold
4, 12 weeksμ-CT, histologyLaser sintering techniqueThe largest volume of bone formed was observed in the group with PEEK scaffold + hSF-MSCs) in a standard culture medium at 4 and 12 weeksThe combination of PEEK scaffold + hSF-MSCs is effective in regenerating bone defects
Lin YH. et al., 2017 [56]n = 12 New Zealand rabbitsGroup with SC/PCL scaffold (n = 6)Group with graphene/SC/PCL scaffold in a 10/40/50 ratio (n = 6)4, 8 weeksμ-CT, histologyExtrusion-based 3D printingIn the experimental group, the volume of bone formed was significantly higher at 4 and 8 weeksPCL scaffolds containing graphene and calcium silicate are promising in bone regeneration
Liu A. et al., 2016 [106]n = 20 male New Zealand rabbitsGroup with β
TCP scaffold (n = 10)
Group with akermanite scaffold (n = 10)6, 12 weeksμ-CT, histologyExtrusion-based 3D printing-The percentage of bone formed at 6 and 12 weeks was significantly higher in the experimental group
-The βTCP scaffold exhibited low mechanical properties
Akermanite scaffold is promising in bone regeneration
Lopez C. et al., 2019 [107]n = 15 New Zealand rabbitsGroup with β
TCP scaffold (n = 5)
-Group with β-TCP and collagen scaffold (n = 5)
-Group with β-TCP and collagen and dipyridamole scaffold (n = 5)
8 weeksμ-CT, histologyExtrusion-based 3D printingIn the groups without dipyridamole, less bone growth and more residual scaffold was observed than in the group with dipyridamoleDipyridamole significantly increased the bone regenerative capacity of the bioceramic scaffold
Mi X. et al., 2022 [58]n = 36 male SD ratsGroup without scaffold (n = 12)-Group with HA/sodium alginate scaffold (n = 12)
-Group with HA/sodium alginate/Ti3C2 MXene scaffold (n = 12)
4, 8 weeksμ-CT, histologyExtrusion-based 3D printingThe group with the scaffold with Ti3C2 MXene promoted bone healing to a significantly greater degree than the other groupsThe Ti3C2 MXene composite 3D-printed scaffolds are promising for clinical bone defect treatment
Miao Y. et al., 2019 [59]Male Wistar rats-Group without scaffold
-Hydrogel scaffold group
Group with hydrogel scaffold and FP nanoparticles3, 6, and 9 weeksμ-CT, histologyNanosheets via liquid phase stripping method-The incorporation of FP promoted mineralization and reinforced the mechanical properties of the scaffold
-Bone regeneration in the experimental group was superior at 3, 6, and 9 weeks
The hydrogel/FP scaffold can be applied in bone regeneration
Naudot M. et al., 2020 [108]n = 22
male SD rats
Group with PCL scaffold (n = 11)Group with PCL/HA/ BM-MSCs scaffold (n = 11)2 monthsμ-CT, histologyElectrospinning and electrosprayingThe experimental group showed significantly higher bone formation over the two monthsThe combination of PCL scaffold with HA and BM-MSCs is promising for bone defect regeneration
Pan T. et al., 2022 [61]n = 20
BALB/c rats
-Group without scaffold (n = 4)
-Group with hydrogel scaffold combined with miRNA (n = 4)
-Group with hydrogel scaffold with miRNA and 0.25 GTA (n = 4)
-Group with hydrogel scaffold with miRNA and 1 GTA (n = 4)
-Group with hydrogel scaffold with miRNA and 2.5 GTA (n = 4)
2, 4, 8 weeksμ-CT, histologyExtrusion-based 3D printingBone regeneration was significantly higher in the groups with 1GTA and 2.5GTA at 2, 4 and 8 weeksThe presence of miRNA and GTA induces osteogenesis, making this scaffold promising for the area of bone regeneration
Park S. et al., 2020 [62]n = 8 defects in 4 male beagles Defects in a PCL scaffold (n = 2)-Defects with PCL/T50 scaffold (n = 2)
-Defects with PCL/T0/B2 scaffold (n = 2)
-Defects with PCL/T50/B2 scaffold (n = 2)
3 monthsμ-CTSelective laser sintering-The volume of bone formed in defects with the PCL/T50 scaffold was significantly higher than with the PCL scaffols
-In the scaffolds with rhBMP-2, bone regeneration was significantly higher
PCL/T50 scaffold is beneficial for transporting rhBMP-2 and regenerating bone in mandibular defects
Park J. et al., 2015 [32]n = 30 BALB/c-nu/nu Group with PCL scaffold (n = 10)-Group with PCL/BMP-2 scaffold (n = 10)
-Group with PCL/BMP-2/VEGF scaffold (n = 10)
4 weeksQuantification of osteogenic genes in dental pulp stem cellsExtrusion-based 3D printingBone regeneration was faster in the vascularized scaffoldVascularized scaffold is promising in bone regeneration
Pae H. et al., 2018 [109]n = 10 male New Zealand rabbitsGroup without scaffold-Group with PCL scaffold
-Group with PCL/10% β-TCP scaffold
-Group with PCL/10% β-TCP and collagen membrane
2, 8 weeksμ-CT3D printingBone formation was only observed in the scaffolds containing β-TCPAddition of β-TCP to the PCL scaffold increases osteoconductivity
Qiao S. et al., 2020 [110]n = 30 female New Zealand rabbitsGroup with Ti scaffold (n = 15)Group with Ti scaffold modified by hydrogel with medium concentrations of silver nanoparticles (n = 15)6, 12 weeksμ-CT, histology3D printingThe experimental group showed significantly higher bone regeneration at 6 and 12 weeksHydrogel-modified Ti scaffold with medium concentrations of silver nanoparticles is promising for treating bone defects
Qin H. et al., 2022 [111]n = 24 male New Zealand white rabbits-----Group with magnesium-substituted calcium scaffold with 480 μm pore size
-Group with magnesium-substituted calcium scaffold with 600 μm pore size
-Group with magnesium-substituted calcium scaffold with 720 μm pore size
2,4,8, 12 weeksμ-CT, histologyDigital light processingThere was a higher new bone ingrowth rate in the 600 μm group than the other two groups at 4–12 weeks post-implantationThe magnesium-substituted calcium scaffold with 600 μm pore size is promising to guide new bone ingrowth
Qin Y. et al., 2022 [112]n = 10 male New Zealand rabbitsGroup with pure Zn scaffolds (n = 10)Group with Zn-1Mg porous scaffolds (n = 10)6, 12 weekshistologyLaser powder bed fusionThe experimental group showed enhanced bone formation compared with pure Zn counterpartsZn-1Mg porous scaffolds presented promising results to fulfill customized requirements of biodegradable bone implants.
Remy M. et al., 2021 [64]n = 30 male SD rats-Group with β
TCP scaffold (n = 5)
-Group with β
TCP scaffold and collagen (n = 5)
-Group with β-TCP/collagen/empty vector (n = 5)
-Group with β-TCP/pDNA 5 μg miRNA 200c (n = 5)
-Group with β-TCP/collagen/pDNA 1 μg miRNA-200c (n = 5)
-Group with β-TCP/collagen/pDNA 5 μg miRNA-200c (n = 5)
4 weeksμ-CT, histologyStereolithography-The groups that contained miR-200c demonstrated greater bone formation
-Bone formation was higher in the scaffold containing βTCP/collagen/pDNA 5 μg miR-200c
Incorporation of miR increases scaffold efficacy in bone regeneration
Rogowska-Tylman J. et al., 2019 [113]n = 15 male rabbits-Group with β
TCP scaffold
-Group with PCL scaffold
-Group with β-TCP/ HA scaffold
-Group with PCL/HA scaffold
3 monthsμ-CT, histology, immunohistochemistryFoaming process/3D printingThe highest bone growth occurred in the group that had the β-TCP/ HA scaffold, followed by the group with the β
TCP
The addition of HA particles increases bone regeneration
Ryu J. et al., 2021 [91]n = 32 mandibular defects in male beagle dogs-Group without scaffold (n = 8)
-Group with Bio-Oss and rhBMP-2
(n = 12)
Group with HA scaffold/ β-TCP/ rhBMP-2 (n = 12)6, 12 weeksHistology, imagiologyStereolithographyThere was no significant difference between the Bio-Oss group and the experimental groupBone formation is not significantly different with HA scaffold/ β- TCP/ rhBMP-2 or with Bio-Oss particles and rhBMP-2
Seo Y. et al., 2022 [114]n = 40 bone defects in New Zealand White rabbitGroup without scaffold (n = 10)-Group with β-TCP/ HA scaffold with 0.8 mm pore diameter (n = 10)
-Group with β-TCP/ HA scaffold with 1 mm pore diameter (n = 10)
-Group with β-TCP/ HA scaffold with 1.2 mm pore diameter (n = 10)
2, 8 weeksμ-CT, histologyStereolithographyAmong the experimental groups, the 1.0- and 1.2-mm groups exhibited signifcantly larger areas of new bone compared with the 0.8-mm groupβ-TCP/ HA block substitutes with different pore diameter promoted faster bone regeneration than that in the natural healing group
Shim J. et al., 2017 [66]n = 3 male beagle dogsGroup with collagen membrane (n = 1)-Group with PCL scaffold
(n = 1)
-Group with PCL scaffold/ β-TCP
(n = 1)
8 weeksμ-CT, histologyMultilayer membrane 3D printingPCL/ β-TCP scaffold is more effective than PCL and than collagen membrane in terms of bone regenerationPCL/ β-TCP scaffold appears to be a more effective alternative to collagen membrane in bone regeneration
Shim J. et al., 2017 [115]n = 8 New Zealand rabbits Group without scaffold-Group with 30% porous PCL membrane
-Group with 50% porous PCL membrane
-Group with 70% porosity PCL membrane
4 weeksμ-CT, Histometric AnalysisExtrusion-based 3D printing-The group with the 30% porosity scaffold showed a higher level of bone formation compared to the experimental groups
-The control group obtained more bone formation than the scaffold with 50% porosity
-Bone formation was significantly higher in PCL membranes with low porosity
-The PCL membrane with 30% porosity is the most favorable for bone regeneration
Shuai C. et al., 2021 [67]n = 18 New Zealand rabbits Group without scaffold (n = 6)-Group with PLLA/PGA/HA scaffold (n = 6)
-Group with PLLA/HA scaffold (n = 6)
4, 8 weeksμ-CTLaser-assisted 3D printingThe PLLA/PGA/HA scaffold showed greater osteogenesis and vascularizationPLLA/PGA/HA scaffold is promising for bone regeneration
Tcacencu I. et al., 2018 [68]n = 15 male SD ratsGroup with PLLA scaffold (n = 3)-Group with glass-ceramic scaffold AW (n = 3)
-PLLA/AW scaffold Group
(n = 6)
12 weeksHistologyIndirect 3D printing/fused filament fabrication-No bone formation was observed in the control group
-The highest bone formation occurred in the group with the PLLA/AW scaffold
PLLA/AW scaffold is effective in bone regeneration
Tovar N. et al., 2018 [116]n = 14 New Zealand rabbits Group without scaffold (n = 4)Group with β-TCP scaffold (n = 10)8, 12, 24 weeksμ-CT, histologyExtrusion-based 3D printing-The control group showed limited bone growth
-In the experimental group, the amount of bone formed was greater at 12 and 24 weeks
The β-TCP scaffolds are biocompatible, resorbable and can regenerate bone
Tsai C. et al., 2019 [69]n = 12 New Zealand rabbits Group with titanium scaffold (n = 6)Group with titanium/Mg- CS and CH scaffold6 weeksHistologySelective laser meltingLess bone regeneration was observed in the control groupMineralization was higher in the experimental scaffold, which makes it promising for bone defect regeneration
Tulyaganov D. et al., 2022 [117]n = 16 male Chinchilla rabbitsGroup with glass powder (n = 8)Group with robocast glass scaffold (n = 8)3, 6 monthsHistologyExtrusion-based 3D printingThe scaffolds exhibited a clear osteogenic effect upon implantation and underwent gradual resorption followed by ossificationThe scaffold is promising in bone tissue engineering and show promise for potential translation to clinical assessment
Ulbrich L. et al., 2021 [118]n = 120 male Wistar rats-Group with empty bone defects
-Group with autogenous bone
-Group with Bio-Oss scaffold
-Group with PBAT scaffold
Group with PBAT/BG scaffold15, 30, 60 daysμ-CTFused deposition modelingPBAT/ BAGNb presented new bone formation comparable to controlsThe combination of PBAT and BAGNb may be an alternative to produce bioactive materials with controllable shapes and properties for bone regeneration treatments
Umeyama R. et al., 2020 [70]C57BL/6J male ratsGroup with β-TCP/RCP scaffoldGroup with β-TCP/RCP scaffold and bone marrow cells cultured in an osteogenic environment for 4, 7, and 14 days8 weeksHistology3D printingThe group with the scaffold whose cells had been cultured in an osteogenic environment for 7 days showed the highest osteogenic potentialBone marrow cells should be cultured in osteogenic medium for 7 days before integrating β-TCP/RCP scaffold

Van hede D. et al., 2021 [119]
n = 16
Wistar male rats
-CaP matrix with orthogonal geometry
-CAP matrix + Bio-Oss
Group with CaP matrix with gyroid geometry4, 8 weeksμ-CTStereolithographyIn the group with the gyroid scaffold, greater bone formation was observed at 4 and 8 weeksGyroid geometry is promising for bone regeneration
Wang M. et al., 2019 [120]n = 16 New Zealand rabbits Group with autologous bone graft
(n = 8)
Group with β-TCP scaffold and dipyridamole (n = 8)24 weeksHistology3D printingThe group with the experimental scaffold demonstrated greater bone regenerationThe β-TCP and dipyridamole scaffold is promising in bone defect regeneration
Wang P. et al., 2021 [71]n = 72 SD female rats Group with PLLA scaffold (n = 8)-Group with PLLA scaffold conditioned with sodium hydroxide (n = 8)
-Group with PLLA scaffold with PDA conditioned with sodium hydroxide (n = 8)
4, 8 weeksμ-CT, histologyFused deposition modelingBone formation at weeks 4 and 8 was higher in the group with the scaffold with PDA, followed by the PLLA scaffold conditioned with sodium hydroxideThe presence of PDA increases osteogenesis in the scaffold
Wang S. et al., 2020 [72]n = 12 female BALB/c miceGroup with PCL/Bio-Oss scaffold (n = 6)Group with PCL/Bio-Oss/NaOH scaffold (n = 6)8 weeksHistologyFused deposition modeling
In the group with the PCL/Bio-Oss/NaOH scaffold, a greater bone formation was observedNaOH treatment increased the hydrophilicity of the scaffold by increasing the osteogenic properties
Won J. et al., 2016 [92]n = 3 male beagle dogsGroup with collagen membraneGroup with PCL/PLGA/β-TCP and Bio-Oss scaffold8 weeksμ-CT, histologyExtrusion-based 3D printing-Bone formation was similar in both groups
-The scaffold of the experimental group showed better mechanical properties
The PCL/PLGA/β-TCP scaffold promotes bone regeneration levels similar to collagen membrane, but has better mechanical properties
Wu Y. et al., 2019 [73]Wistar ratsGroup with SC/PCL scaffoldGroup with dECM/SC/PCL scaffold4 weeksμ-CTExtrusion-based 3D printingBone regeneration was superior in the dECM/SC/PCL groupDecellularization combined with 3D scaffolds can be applied in bone regeneration
Xia D. et al., 2022 [74]n = 15 New Zealand rabbitsGroup with zinc scaffold Group with pure zinc porous scaffold4, 12, 24 weeksμ-CTLaser powder bed fusion technologyBone regeneration was superior in the group with pure zinc porous scaffoldPure Zn porous scaffolds with customized structures represent a promising biodegradable solution for treating large bone defect
Xu Z. et al., 2019 [75]n = 6 BALB/c miceGroup without scaffold-Group with PLGA/ β -TCP scaffold
-Group with PLGA scaffold/ β -TCP/1 mg polydopamine
-Group with PLGA scaffold / β -TCP/2 mg polydopamine
2, 6 weeksμ-CT, histologyExtrusion-based 3D printingThe higher the PDA concentration, the greater the bone regeneration at 2 and 6 weeksThe addition of PDA allows for good results, and has a lot of potential in bone regeneration
Yu L. et al., 2020 [121]n = 18 SD ratsGroup with Ti scaffold-Group with Ti and MSC scaffold
-Group with Ti scaffold and RA
8 weeksμ-CT, histology3D printing-In the control group, bone formation was almost null
-The greatest bone regeneration occurred in the group with RA
The combination of pluripotent stem cells and Ti scaffolds with RA can be used to repair bone defects
Yun J. et al., 2019 [89]n = 12 beagles Group without scaffold-Group with PLLA/PLGA/HA scaffold
-Group with PLLA/PLGA/HA/BMP-2 scaffold
20 weeksμ-CT, histology, imagiologyExtrusion-based 3D printing-The PLLA/PLGA/HA scaffold is biodegradable and was replaced by bone
-Bone regeneration was significantly higher in the group with BMP-2
Bone defects can be successfully treated with PLLA/PLGA/HA/BMP-2
Yun S. et al., 2021 [77]n = 27 SD ratsGroup without scaffold (n = 3)-Group with dECM scaffold (n = 8)
-Group with β TCP scaffold (n = 8)
-Group with dECM/ β TCP scaffold (n = 8)
4 weeksμ-CT, histologyExtrusion-based 3D printingThe group with the dECM/ β TCP scaffold showed greater bone formationThe dECM/ β TCP scaffold has ideal osteogenic potential to treat bone defects
Zhang W. et al., 2017 [122]n = 38 male New Zealand rabbitsGroup with β-TCP scaffold
(n = 12)
-Group with BRT scaffold
(n = 12)
-Group with BRT-H scaffold (n = 14)
4, 12 weeksμ-CTExtrusion-based 3D printingThe group with the BRT-H scaffold promoted significantly more bone regenerationBRT-H scaffold is promising in the repair of large bone defects
Zhang Y. et al., 2019 [79]n = 24 male Wistar ratsGroup without scaffold (n = 6)-Group with β TCP/ PLGA/ OG /BMP- 2 (n = 6)
-Group with β TCP/ PLGA/OG (n = 6)
-Group with β TCP/ PLGA
(n = 6)
4, 12 weeksμ-CT, histologyExtrusion-based 3D printingIn the group with β TCP/ PLGA/OG/BMP- 2 the highest bone formation was observed, followed by the group with β TCP/PLGA/OG and β TCP/PLGABMP-2 peptide and OG are favorable for bone growth and enhance bone regeneration, making PTG/P scaffold promising in the repair of bone defects
Zhang Z. et al., 2021 [33]n = 12 New Zealand rabbitsGroup with p-Ta scaffold (n = 6)Group with p-Ta-nt scaffold (n = 6)2 weekshistology3D printing laser melting systemBone formation was significantly higher in the experimental groupTantalum matrices with nanotubes show promise in bone regeneration
Zhong L. et al., 2020 [81]n = 24 male SD ratsGroup without scaffold (n = 6)-Group with PCL scaffold (n = 6)
-Group with PCL/DCPD scaffold (n = 6)
-Group with PCL/DCPD scaffold/ nanoZIF-8 (n = 6)
12 weeksμ-CTExtrusion-based 3D printingThe group with the PCL/DCPD/nanoZIF-8 scaffold induced significantly more bone formation NanoZIF-8 has great potential in treating bone defects
ADSCs—adipose tissue derived stem cells, Asp@Lipo—aspirin loaded liposomes, AW—apatite/volastonite, BFP-1—bone forming peptide 1, Bio-Oss—deproteinized bovine bone minerals, BM-MSCs—bone marrow derived mesenchymal stem cells, BMP-2—bone morphogenetic protein-2, BRT—β tricalcium phosphate, silicon, magnesium, and calcium, BRT-H—β tricalcium phosphate, silicon, magnesium, and calcium with hollow pipe structure, CaP—calcium phosphate, CGRP—hydroxyapatite/sodium alginate/calcitonin gene-related peptide, CH—chitosan, CS—calcium sulfate, DCPD—calcium phosphate dihydrate, dECM—decellularized extracellularized matrix, dECM—decellularized extracellular matrix, ECM—natural-like extracellular matrix, ETG—sodium hydroxide-conditioned polylactic acid, EV—extracellular vesicle, FP—black phosphorus, Gel/SF—gelatin/silk fibrin, Gel/SF/0Cu-BG—silk gelatin/fibrin and bioactive glass, Gel/SF/10Cu-BG—silk gelatin/fibrin/bioactive glass and 15% copper, Gel/SF/2Cu-BG—silk gelatin/fibrin/bioactive glass and 7% copper, Gel/SF/5Cu-BG—silk gelatin/fibrin/bioactive glass and 10% copper, GTA—glutaraldehyde, HA—hydroxyapatite, HCCS-PDA—calcium silicate and hydroxyapatite collagen with polydopamine binding, hGMSCs—human gum mesenchymal stem cells, hSF-MSCs—synovial mesenchymal stem cells, KR-34893—bioactive organic compound, LayFomm—polyvinyl acid + polyurethane, mg—milligram, Mg- CS—calcium silicate, MgO—magnesium oxide, MgP—magnesium phosphate, miRNA—microRNA, MSC—mesenchymal stem cells, NG—naringin, OG—graphene oxide, PBAT—poly(butylene adipate-co-terephthalate), PCL—polycaprolactone, PCL/T0/B2—polycaprolactone/human recombinant bone protein type 2, PCL/T50—ratio 1:1 polycaprolactone / β tricalcium phosphate, PCL/T50/B2—polycaprolactone/β tricalcium phosphate/human recombinant bone protein type 2, PCLD—dopamine-immersed polycaprolactone, PCLDB100—dopamine-immersed polycaprolactone and BFP-1 at 100 ug/mL, PCLDB1000—dopamine-immersed polycaprolactone and BFP-1 at 1000 ug/mL, pDNA—DNA plasmid, PEEK—polyetherketone, PEG—polyethylene glycol, PEI-EVs—polylactic acid/extracellular vesicle with polyethyleneimine, PGA—polyglycolic acid, PLGA—poly(lactic acid-co-glycolic acid), PPG-1. 5—polyacrylamide, polyurethane, PRF—platelet-rich fibrin, PRP—platelet-rich plasma, p-Ta-nt—tantalum with nanotubes, PTFE—polytetrafluoroethylene, PTG—polylactic acid with polydopamine conditioned with sodium hydroxide, qPCR—real-time polymerase chain reaction, RA—retinoic acid, RCP—recombinant collagen peptide, rhBMP-2—human recombinant bone protein type 2, SA—sodium alginate, SC—calcium silicate, SD—Sprague Dawley, SiO2—silica, SrSC—calcium strontium silicate, Ti—titanium, β-TCP—β-tricalcium phosphate, μ-CT—microcomputed tomography.
Table 3. Biomaterials described in the included studies (in vitro and in vivo).
Table 3. Biomaterials described in the included studies (in vitro and in vivo).
In Vitro StudiesIn Vivo Studies
Biomaterialsβ-TCP1627
PCL1620
HA716
PLLA76
CS46
Collagen45
PLGA45
dECM35
Hydrogel53
MgP22
Zn-1Mg04
BG31
PDA30
MgO21
HCCS-PDA12
Ti11
PVA20
OG11
p-Ta-nt11
nanoZIF-811
DCPD11
Layform11
Sodium alginate11
Gelatin11
SiO11
PEEK11
PGA11
AW11
Gel/SF01
CaP matrix01
Robocast glass01
PEI-EVs01
PTFE01
Polyvinyl acid01
PEG01
PCLD01
SA01
Graphene01
Akermanite01
Ti3C2 MXene11
FP nanoparticles01
PBAT01
Polydopamine01
BRT01
GCP10
Bioactive Silk Fibrin Glass10
BiomoleculesRhBMP-267
Dipyridamole14
PRF03
hSF-MSCs12
miRNA21
NaOH21
Curcumin01
RGD01
Asp@Lipo11
BFP-111
RCP11
VEGF11
Heparin01
ADSCs01
NG01
CGRP01
BM-MSCs01
pDNA01
DPSCs10
Dexamethasona10
Glycerol10
KR-3489310
PRP10
Table 4. Analysis of evaluation methods in in vitro and in vivo studies.
Table 4. Analysis of evaluation methods in in vitro and in vivo studies.
Study Typeµ-CTHistologyqRT-PCR (Osteogenesis-Related Gene Expression)p-Nitrophenol Assay
(ALP Activity)
AKT Assay Kit
(ALP Activity)
ImagiologyWestern-Blot
(Expression Levels of Osteogenic-Related Proteins)
ImunohistochemistryWestern-Blot
(ALP Activity)
ELISA
(Osteogenic-Related Protein Secretion)
In vitro01279702011
In vivo575630040200
Table 5. Analysis of biomaterials 3D printing techniques in in vitro and in vivo studies.
Table 5. Analysis of biomaterials 3D printing techniques in in vitro and in vivo studies.
Study TypeExtrusion Based BioprintingFused Deposition Modeling3D Printing (No Specific Method)StereolithograhyLaser Sintering TechniqueDigital Light Processing Type 3D Printing SystemSelective Laser meltingLaser Powder Bed FusionInkjet-Based BioprintingMicrovalve BioprintingExtrusion-Based HydrogelNanosheets via Liquid Phase Stripping MethodMultilayer Membrane 3D PrintingIndirect 3D Printing/Fused Filament FabricationBinder JettingDirect Ink Writing TechniqueMicro Extrusion Foaming Process/3D PrintingElectrospinning and Electrospraying3D Printed Scaffolds
In vitro236623121211111000000
In vivo2710873322111111111111
Table 6. Risk of bias of in vitro studies.
Table 6. Risk of bias of in vitro studies.
Structured SummaryScientific Background and Explanation of Rationale Specific Objectives and/or HypothesesIntervention for Each GroupOutcomeSample SizeRandom AllocationAllocation Concealment MechanismImplementationBlindingStatistical MethodsOutcomes and EstimationLimitationsFundingProtocol
Alksne M. et al., 2020 [34]YYYYYYNNNNYYYYY
Bae E. et al., 2018 [35]YYYYYNYYNYYYYYN
Cao Y. et al., 2019 [36]YYYYYNNNNNYYNYN
Chen Y. et al., 2018 [37] YYYYYNNNNNYYNYN
Chiu Y. et al., 2019 [38]YYYYYNNNNNYYNYN
Cooke M. et al., 2020 [39]YYYYYNNNNNYYYYN
Dai Q. et al., 2021 [40]YYYYYNNNNNYYNYN
Dubey N. et al., 2020 [41]YYYYYNYNNNYYYNN
Fahimipour F. et al., 2019 [42]YYYYYNNNNNYYYYN
Gómez-Cerezo M. et al., 2020 [43]YYYYYNNNNNYYNYN
Han L. et al., 2021 [44]YYYYYNYYNNYYYYN
Huang K. et al., 2021 [45]YYYYYNNNNNYYYYY
Jeong J. et al., 2020 [46]YYYYYNNNNNYYNYN
Kao C. et al., 2015 [47]YYYYYNNNNNYYNYN
Ke, D. et al., 2018 [48]YYYYYNNNNNYYNYN
Kim B. et al., 2018 [49]YYYYYNNNNYYYNYN
Kim J. et al., 2017 [50]YYYYYNNNNNYYNYN
Lee S. et al., 2018 [51]YYYYYYYYNNYYNYN
Li J. et al., 2017 [52]YYYYYNYNNNYYYYY
Li Y. et al., 2019 [53]YYYYYNNNNNYYNYY
Lin Y. et al., 2019 [54]YYYYYNNNNNYYNNY
Lin YH. et al., 2017 [55]YYYYYNNNNNYYNSN
Lin YH. et al., 2019 [56]YYYYYNNNNNYYNYY
Martin V. et al., 2019 [57]YYNYYNNNNNYYNYN
Mi X. et al., 2022 [58]YYYYYNYNNNYYYYN
Miao Y. et al., 2019 [59]YYYYYNNNNNYYNYN
Midha S. et al., 2018 [60]YYYYYNNNNNYYYYN
Pan T. et al., 2022 [61]YYYYYNYNNNYYYYN
Park J. et al., 2015 [32]YYYYYNYNNNYYNYN
Park S. et al., 2020 [62]YYYYYNNNNNYYNYY
Ratheesh. G. et al., 2021 [63]YYYYYNNNNNYYYYN
Remy M. et al., 2021 [64]YYYYYNNNNNYYYSN
Roh H. et al., 2016 [65]YYYYYNYNNNYYNYN
Shim J. et al 2017 [115]YYYYYNYNNNYNNYN
Shuai C. et al., 2020 [67]YYYYYNNNNNYYNYN
Tcacencu I. et al., 2018 [68]YYYYYNYNNNYYNYN
Tsai C. et al., 2019 [69]YYYYYNNNNNYYNYN
Umeyama R. et al., 2020 [70]YYYYYNNNNNYYNYN
Wang P. et al., 2021 [71]YYYYYNYNNNYYNYN
Wang S. et al., 2020 [72]YYYYYNNNNNYYYYN
Weinand C. et al., 2006 [31]YYYYYNNNNNYNYNN
Wu Y. et al., 2019 [73]YYYYYNNNNNYNNYN
Xia D. et al., 2022 [74]YYYYYNNNNNYYYYY
Xu Z. et al., 2019 [75]YYYYYNNNNNYNNYN
Xu Z. et al., 2022 [76]YYYYYNYNNNYYNYN
Yun S. et al., 2021 [77]YYYYYNYNNNYNYYN
Zamani Y. et al., 2021 [78]YYYYYNYNNNYYNYN
Zhang Y. et al., 2019 [79]YYYYYNYNNNYYNYN
Zhang Z. et al., 2021 [33]YYYYYNYNNNYYYYN
Zhong L. et al., 2020 [81]YYYYYNYNNNYYYYN
Zhao N. et al., 2017 [80]YYYYYNNNNNYNYYN
Y—Yes; N—No.
Table 7. Risk of bias of in vivo studies.
Table 7. Risk of bias of in vivo studies.
Sequence GenerationBaseline CharacteristicsAllocation ConcealmentRandom HousingBlindingRandom Outcome AssessmentBlindingIncomplete Outcome Data Selective Outcome ReportingOther Sources of Bias
Bae E. et al., 2018 [35]NYNYNYYYYY
Bekisz J. et al., 2018 [93]NYNNNYNYYY
Bose S. et al., 2018 [85]NYNUNYNYYY
Chang P. et al., 2021 [94]NYNNNYNYYY
Chen M. et al., 2021 [95]NYNNNYNYYY
Chiu Y. et al., 2019 [38]NYNNNYNYYY
Cooke M. et al., 2020 [39]NYNUNYNYYY
Dai Q. et al., 2021 [40]NYNNNYNYYY
Diomede F. et al., 2018 [96]NYNYNYNYYY
Dubey N. et al., 2020 [41]NYNNNYNYYY
El-Habashy S. et al., 2021 [97]NYNNNYNYYY
Fahimipour F. et al., 2019 [42]NYNYNYNYYY
Fama C. et al., 2020 [98]UNNNNNNNNY
Guéhennec L. et al., 2019 [90]NYNNNYNYYN
Han L. et al., 2021 [44]NYNNNYNYYY
He M. et al., 2021 [99]NYNNNYNYYY
Huang K. et al., 2021 [45] NYNNNYNYYY
Ishack S. et al., 2017 [88]NYNNNYNYYY
Jeong J. et al., 2020 [46]NYNNNYNYYY
Jia L. et al., 2021 [100]NYNYNYNYYY
Johnson Z. et al., 2021 [101]NYNNNYNYYY
Ke D. et al., 2018 [48]NYNYNYNYYY
Kim J. et al., 2020 [102]NYNNNYNYYY
Kim J. et al., 2017 [50]NYNNNYNYYY
Lee D. et al., 2018 [103]NYNNNYNYYY
Lee J. et al., 2021 [86]NYNNNYNYYY
Lee S. et al., 2019 [51]NYNNNYNYYY
Lee SH. et al., 2019 [87]NYNNNYNYYY
Liang T. et al., 2021 [104]NYNYNYNYYY
Li J. et al., 2017 [52]NYNNNYNYYY
Li Y. et al., 2019 [53]NYNNNYNYYY
Lim H. et al., 2020 [105]NYNNNYNYYY
Lin YH. et al., 2019 [56]NYNNNYNYYY
Lin YH. et al., 2017 [55]NYNNNYNYYY
Liu A. et al., 2016 [106]NYNNNYNYYY
Lopez C. et al., 2019 [107]NYNNNYYYYY
Mi X. et al., 2022 [58]NYNNNYNYYY
Miao Y. et al., 2019 [59]NNNNNYNYYY
Naudot M. et al., 2020 [108]NYNUNYNYYY
Pan T. et al., 2022 [61]NYNNNYNYYY
Park S. et al., 2020 [62]NYNUNYNYYY
Park J. et al., 2015 [32]NYNNNYNYYY
Pae H. et al., 2018 [109]NYNYNYNYYY
Qiao S. et al., 2020 [110]NYNNNYNYYY
Qin H. et al., 2022 [111]NYNYNYNYYN
Qin Y. et al., 2022 [112]NYNYNYNYYY
Remy M. et al., 2021 [64]NYNNNYNYYY
Rogowska-Tylman J. et al., 2019 [113]NYNNNNNNYY
Ryu J. et al., 2021 [91]NYNNNYYYYN
Seo Y. et al., 2022 [114]NYNYNYNYYY
Shim J. et al., 2017 [115]NYNNNYNYYY
Shim J. et al., 2018 [66]NYNYNYYYYN
Shuai C. et al., 2021 [67]NYNNNYNYYY
Tcacencu I. et al., 2018 [68]NYNNNYNYYY
Tovar N. et al., 2018 [116]NYNNNYNYYN
Tsai C. et al., 2019 [69]NNNNNYNYYY
Tulyaganov D. et al., 2022 [117]NYNYNYNYYY
Ulbrich L. et al., 2021 [118]YYNYNYNYYY
Umeyama R. et al., 2020 [70]NUNNNYNYYN
Van hede D. et al., 2021 [119]NNNNNYNYYN
Wang M. et al., 2019 [120]NYNNNYYYYY
Wang P. et al., 2021 [71]NYNNNYNYYY
Wang S. et al., 2020 [72]NYNUNYNYYY
Won J. et al., 2016 [92]NYNNNYYYYY
Wu Y. et al., 2019 [73]NNNYNYNYYY
Xia D. et al., 2022 [74]NYNYNYNYYY
Xu Z. et al., 2019 [76]NNNNNYNYYY
Yu L. et al., 2020 [121]NNNNNYNYYY
Yun J. et al., 2019 [89]NYNNNYNYYY
Yun S. et al., 2021 [77]NNNYNYNYYY
Zhang W. et al., 2017 [122]NYNNNYNYYY
Zhang Y. et al., 2019 [79]NYNNNYNYYY
Zhang Z. et al., 2021 [33]NNNNNYNYYY
Zhong L. et al., 2020 [81]NYNNNYNYYY
Y—Yes; N—No; U—unclear.
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

Francisco, I.; Basílio, Â.; Ribeiro, M.P.; Nunes, C.; Travassos, R.; Marques, F.; Pereira, F.; Paula, A.B.; Carrilho, E.; Marto, C.M.; et al. Three-Dimensional Impression of Biomaterials for Alveolar Graft: Scoping Review. J. Funct. Biomater. 2023, 14, 76. https://doi.org/10.3390/jfb14020076

AMA Style

Francisco I, Basílio Â, Ribeiro MP, Nunes C, Travassos R, Marques F, Pereira F, Paula AB, Carrilho E, Marto CM, et al. Three-Dimensional Impression of Biomaterials for Alveolar Graft: Scoping Review. Journal of Functional Biomaterials. 2023; 14(2):76. https://doi.org/10.3390/jfb14020076

Chicago/Turabian Style

Francisco, Inês, Ângela Basílio, Madalena Prata Ribeiro, Catarina Nunes, Raquel Travassos, Filipa Marques, Flávia Pereira, Anabela Baptista Paula, Eunice Carrilho, Carlos Miguel Marto, and et al. 2023. "Three-Dimensional Impression of Biomaterials for Alveolar Graft: Scoping Review" Journal of Functional Biomaterials 14, no. 2: 76. https://doi.org/10.3390/jfb14020076

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