Next Article in Journal
Pharmaceutical Applications of Metal Complexes and Derived Materials
Previous Article in Journal
Post-Synthesis Modulation of the Physicochemical Properties of Green-Synthesized Iron Oxide Nanoparticles with Tween 80 to Enhance Their Antibacterial Activity and Biocompatibility
Previous Article in Special Issue
Integrating Graphene Oxide and Mesenchymal Stem Cells in 3D-Printed Systems for Drug Delivery and Tissue Regeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in 3D-Printed Drug Delivery and Screening Platforms for Bone Disease Therapy

by
Iosif-Aliodor Timofticiuc
1,2,†,
Alex-Gabriel Grigore
2,†,
Elena-Teodora Tomescu
1,
Teona-Maria Vlaicu
1,
Serban Dragosloveanu
3,4,
Andreea-Elena Scheau
5,
Ana Caruntu
6,7,
Christiana Diana Maria Dragosloveanu
8,9,*,
Ioana Anca Badarau
10,
Andreea Cristiana Didilescu
11,
Constantin Caruntu
10,12 and
Cristian Scheau
10,5
1
Faculty of Medicine, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
2
“Foisor” Clinical Hospital of Orthopedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
3
Department of Orthopedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
4
Department of Orthopedics, “Foisor” Clinical Hospital of Orthopedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
5
Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
6
Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
7
Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
8
Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
9
Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
10
Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
11
Department of Embryology and Microbiology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
12
Department of Dermatology, “Prof. N.C. Paulescu,” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2025, 17(11), 1372; https://doi.org/10.3390/pharmaceutics17111372
Submission received: 29 September 2025 / Revised: 18 October 2025 / Accepted: 21 October 2025 / Published: 23 October 2025

Abstract

Bone diseases such as osteomyelitis, osteosarcoma, and osteoarthritis, as well as conditions caused by metabolic imbalances, including osteoporosis, require more efficient and optimized therapies. Systemic drug administration entails major disadvantages like cytotoxicity and adverse reactions, which can lead to serious complications or death. Therefore, local drug administration alternatives are currently under investigation for different pharmacological therapies. New vectors were created to improve control over administration, and 3D-printed and patient-specific drug delivery systems have been tested, revealing great potential. Moreover, 3D-printed platforms that mimic human tissues for drug testing are innovative solutions emerging for the pharmaceutical industry. Situated between in vitro and in vivo testing on human patients, they offer the advantage of reproducing functional architecture, providing results that are closer to those encountered in clinical trials performed on patients. In our article, we present the two categories of 3D systems, from the perspective of main drug groups (antibiotics, anticancer, and anti-inflammatory) as well as other categories, alongside their advantages, limitations, and their adaptations to 3D printing technologies. This article also highlights the technological drawbacks encountered in both delivery and screening systems, as well as the printing methods and materials used, including their physical and biological properties.

Graphical Abstract

1. Introduction

Three-dimensional (3D) printing has emerged in recent decades as one of the most promising and potent tools to fabricate complex, personalized products on demand with relative ease and low costs [1]. The technology has been widely used and implemented in a multitude of sectors, including automotive, construction, aerospace, medical, and many other industries [2,3,4]. In the pharmaceutical sector, 3D printing has enabled the development of personalized drug delivery systems, such as patient-specific tablets with controlled release profiles [5,6]. It also facilitates the rapid prototyping of medical devices and tissue engineering scaffolds [7]. Furthermore, this technology supports the fabrication of advanced screening platforms for disease modulation and targeted drug response, allowing for high-throughput, physiologically relevant testing [8,9,10,11]. Its precise control over formulation architecture further enhances the study of drug release kinetics and biological responses in preclinical models.
Due to these advantages of 3D printing, this technology has been especially significant in the field of orthopedics and craniofacial surgery, particularly in bone pathologies where traditional models may not sufficiently capture the complexity or biology of bone tissue architecture [12]. Bone-specific diseases, including osteoporosis, osteoarthritis, and bone cancers, pose unique therapeutic and modeling challenges to the development of therapeutics, drugs, and biologics because of the complexities of their pathophysiology and the need for a targeted, localized treatment approach [13,14,15]. Typically, traditional techniques do not allow for drugs to be effectively delivered to the bone or lack the ability to replicate the native bone microenvironment for testing. 3D printing has opened doors for designing and fabricating bone-mimetic scaffolds, as well as in vitro models of bone diseases that would exhibit structural and mechanical properties of native bone [16,17]. The ability to control the architecture and composition of a material enhances the application of 3D printing in studying more physiologically relevant conditions, including bone regeneration processes, drug release characteristics, and cellular behavior.
Regarding bone diseases, drug delivery systems are defined as specialized formulations or devices designed to bring and release therapeutic agents to the affected tissue [18]. The use of traditional systemic drug delivery frequently results in insufficient drug concentrations at the bone site with reduced efficacy, and an increased risk for systemic toxicity, especially when drug efficacy is limited by extensive bone damage [19]. In this case, being able to deliver an appropriate concentration locally at the site of disease is fundamental for achieving the desired bone concentrations, while limiting systemic drug levels and unintended side effects [20,21,22]. There are numerous examples of materials used to create effective bone drug delivery systems, including, but not limited to, biodegradable polymers (e.g., PLGA, PCL) [23,24], lipids (i.e., phospholipids in liposomes) [21,25], inorganic agents (e.g., silica, calcium phosphate) [26,27], and hydrogels [28]. Each has a range of benefits, including controlled release, targeting, and biocompatibility in therapeutic applications. Additionally, extensive research is conducted on using advanced materials such as metal-organic frameworks (MOFs) [29] and bioactive ceramics [30] for specialized applications like cancer therapy or for sustaining bone regeneration. Drug screening platforms, however, may be defined as in vitro or ex vivo models that mimic the tissue microenvironment and are useful in the assessment of the efficacy and safety of potential treatments [31,32]. Traditional models may be physiologically irrelevant due to the system‘s simplistic architecture and failure to allow dynamic interactions [33]. Advanced 3D bone mimetic platforms are attempting to provide structural, mechanical, and cellular complexity similar to native bone, in order to permit more predictive in vitro testing [34]. These systems are oriented towards addressing the limitations of traditional models, which did not allow for close cell–cell and cell–matrix interactions while also enabling high-throughput screening and real-time monitoring [33,35].
To address the current challenges in treating bone disease, new drug development approaches have emerged. Unlike conventional manufacturing methods, such as compression molding, granulation, and coating technologies [36,37,38], which lack the flexibility to produce unique dosage forms or release therapeutics in a complex profile, 3D printing offers a wide range of additive manufacturing technologies [39]. The American Society for Testing and Materials (ASTM) categorizes additive manufacturing into seven process types [40], of which we identified four main classes most frequently used in drug delivery systems and drug screening platforms for bone diseases: Material Extrusion (ME), Vat Photopolymerization (VPP), Powder Bed Fusion (PBF), and Binder Jetting (BJ) (Figure 1).
One of the more common processes of AM is Material Extrusion (ME) [41], in which a continuous filamentous material is extruded from a heated nozzle and deposited layer-by-layer to create three-dimensional objects [42]. The most common subtype of ME processing is Fused Filament Fabrication (FFF) [43], also known as Fused Deposition Modelling (FDM), which has been used for the fabrication of non-woven bone scaffolds using various thermoplastic materials (e.g., PCL, PLA, and PLGA) [44,45,46]. While the process parameters influence the mechanical properties in the final structure, [47] layer thickness, filament orientation, and inter-layer adhesion involvement lead to different mechanical properties in these scaffolds, and their final physical properties may differ from the biologically-inspired scaffold material [47]. In addition, extrusion bioprinting is another method of Material Extrusion that uses pneumatic or displacement-driven systems to extrude bioinks that contain living cells and growth factors at physiologically relevant temperatures, producing high-fidelity, multi-material soft and hard tissue scaffolds, including articular and osteochondral constructs [43].
Vat Photopolymerization (VPP) is another additive manufacturing (AM) process that can create spatial objects with enhanced precision and complex geometries. This technique is based on curing a light-sensitive resin layer-by-layer with a UV light source [48,49]. A well-known 3D printing technique from the VPP group is stereolithography (SLA), which was the first 3D printing technology to be commercialized in the late 90s and continues to be improved by advancements in optical components for better fidelity [49,50]. The resins used in VPP usually consist of monomers (that can be methacrylated for enhanced mechanical characteristics), photoinitiators, and additives, which polymerize into a thermally and mechanically stable construct [48,51].
The other technique in this group is Digital Light Processing (DLP), which utilizes a digital projector to cure an entire layer of resin at once, thus enhancing print speed and scalability [50,52,53,54,55,56]. In Powder Bed Fusion (PBF), a laser or electron beam selectively melts and fuses powdered materials, typically metal, but can also include polymers, ceramics, or composites, layer by layer [57,58,59]. The unsintered powder supports the part being manufactured [60,61]. PBF methods are limited to medium or smaller parts and require post-processing, for example, powder removal, sintering, and finishing [58,59,60]. Other PBF methods, such as SLS, SLM, DMLS, and EBM, offer flexibility to the materials and structural complexity [57,58,59,60,61,62,63,64]. Moreover, Binder Jetting (BJ) uses a print head to selectively deposit a liquid binder (with a volume around 5–15%) onto a powdered bed, bonding the materials under mechanical pressure without heat and enabling processing at room temperature [65,66,67]. This involves the use of water-based or solvent-based inks that can be used for sensitive materials like hydrogels and biologics in tissue engineering and drug delivery applications [67]. Moreover, as in PBF technology, unsintered powder acts as an internal support material, meaning that additional supportive structures needed in AM processes are eliminated, enabling complex geometries [68,69,70]. Although BJ is limited in terms of lower resolution (20 to 100 μm) and post-processing requirements and potential porosity, this technology can represent an alternative if complex constructs with enhanced fidelity are desired [65,66,67,69,70,71,72]. The purpose of this review is to analyze all the possibilities of locally delivering drugs or testing new medications or doses through 3D-printed models or platforms made of biocompatible materials. Although an exhaustive search was conducted for this work in this area, no studies were found on humans; instead, only in vitro or in vivo studies (using animal models) were identified. Given that the initial conceptualization included the expectation of finding results from human studies, it is worth noting that research in this field has not yet reached that stage. Thus, the present review is focused on the clinical translation of the methods discovered in vitro and on animal models, but also on how they can represent an alternative to the current drug testing technology, and in some situations, how they can completely eliminate the need for testing on human patients.

2. Methods

A literature search was conducted in the following electronic databases: PubMed and Scopus. The search was conducted in July 2025, using specific keywords, Medical Subject Headings (MeSH) terms, Boolean operators (AND”, “OR”, and “NOT” to eliminate literature reviews), and specific search field tags.
The search was composed of three search queries. The first search query was focused on finding any bone disease by using the structure “osteo*”, which resulted in osteosarcoma, osteomyelitis, osteoporosis, etc. Moreover, in the first query, other terms for chronic diseases with can affect the bones were added by using the Boolean operator “OR”: rickets, Paget’s disease, achondroplasia, fibrous dysplasia, cleidocranial dysplasia, spondylitis, Ewing, chondrosarcoma, and myeloma. The second query was focused on 3D printing or 3D bioprinting and other synonyms such as “additive manufacturing”, “printing”, “three-dimensional printing”, or “3D biofabrication”. The third search query was used to restrain the search area for drug delivery platforms or drug screening models (“drug delivery platform” [TiAb] OR “drug” [TiAb] OR “drug delivery system” [TiAb] OR “drug screening” [TiAb] OR “drug delivery” [TiAb] OR “disease modeling” [TiAb]).
The search was not restricted to any timeline. Articles were included only if available in full-text, in English, and if they were original research. Research was excluded based on title and abstract if it did not involve 3D printing or 3D bioprinting, if 3D-printed constructs were used only as surgical guides or medical accessories, or if the implant was not entirely 3D-printed. Any review articles, editorials, case reports, book chapters, meta-analyses, or systematic reviews were excluded.

3. 3D-Printed Drug Delivery Systems

3.1. Antibiotics

Osteomyelitis is a critical disease with a high risk of amputation. Even in the most usual manifestations, the occurring critical bone defects exceed the body’s ability to self-heal [73]. The development of local delivery systems was encouraged by the risks and variable efficiency of systemic antibiotherapy administration. In the last 20 years, the most used delivery material for antibiotic release has been polymethyl methacrylate (PMMA) [74].
However, the lack of control on the PMMA release behavior resulted in many aggravated cases of chronic osteomyelitis or even sepsis [75,76]. To overcome this challenge, 3D printing technologies were used to create a customizable, specific, and controllable drug delivery system for a variety of antibiotics (Figure 2). Advantages and challenges of unique scaffolds are discussed below, offering a broad view of the current research in patient-specific 3D printing-based antibiotherapy.

3.1.1. Vancomycin

Vancomycin is a frequently used antibiotic in the field of orthopedics, particularly for severe implant-related infections caused by resistant gram-positive bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) [77]. This antibiotic binds to a highly specific peptide terminal (D-Ala-D-Ala dipeptide) of the peptidoglycan precursors. By inhibiting this terminal, vancomycin stops the crosslinking of peptidoglycans and ultimately inhibits the bacterial wall synthesis [78]. Vancomycin dosing is complex, so therapeutic drug monitoring is essential, especially in patients who are in critical condition [79]. A good control of the release can minimize the risk of toxicity or adverse reactions. Furthermore, given the extensive pharmacokinetics of vancomycin in adult and pediatric populations, an individualized and highly specific dosing protocol is considered the gold standard [80,81].
If a severe infection or a local trauma occurs, the vascularization of the affected area is deficient [82,83]. In these cases, systemic administration of vancomycin would be highly inefficient, and achieving high concentrations of the antibiotic in the affected zone is improbable. Therefore, it is necessary to develop more efficient methods for local and controlled antibiotic administration systems. Various non-3D-printed scaffolds made of hydrogels or polymers, such as gelatin or tricalcium phosphates, were tested as vancomycin delivery systems [84]. However, for these standard scaffolds, the degradability rates, pore sizes, and other mechanical aspects (viscosity, strength, and elasticity modulus) are not controllable. Because of this, the drug release rate can be inconstant; a low degradability rate would directly affect the release, and a lack of control on the mechanical aspects could result in collapsed structures inside the scaffold.
In an in vitro study, a 3D bioprinting gelatin methacrylated (GelMA) scaffold for vancomycin delivery was tested. To solve the high degradability rate of the hydrogel, which would have rapidly released the entire quantity of the drug, zinc-based metal-organic (ZIF-8) nanoparticles were added. ZIF-8 nanoparticles provide supplementary crosslinking points during the biomaterials polymerization process, which enhances the overall rigidity [85]. As a result, the mechanical strength increases, and the early degradation is prevented [86]. Furthermore, it was discovered that at an optimal concentration of ZIF-8 of approximately 3 mg/mL, the nanoparticles exhibit high cytocompatibility and promote osteogenesis by stimulating RUNX2 and OPN, key regulators for osteogenic stem cell differentiation [86]. Therefore, a hydrogel-based scaffold with osteogenic properties and enhanced mechanical characteristics was printed, resulting in a tissue-engineered construct for bone regeneration that is optimal for vancomycin release and simultaneously exhibits antimicrobial activity. While vancomycin-loaded GelMA/ZIF-8 scaffold presents enhanced osteogenesis and antimicrobial properties, in vivo activity and long-term cytotoxicity remain to be studied.
Vancomycin-loaded hydrogels were also tested in vivo, in white mice models, for treating osteomyelitis. The main challenge when inserting a scaffold inside animal models is that the control of pH is lost, as opposed to in vitro testing. For this, the hydrogel (chitosan) was cross-linked with glutaraldehyde, thus ensuring a pH and temperature-responsive scaffold [87]. By the dual incorporation of the chitosan (chitosan loaded with vancomycin and chitosan-laden microspheres), a controlled release of the drug is ensured while also promoting osteogenesis. Moreover, by including the microspheres, a “shield” is created that protects the osteoblast from bacterial toxins, while vancomycin provides antimicrobial activity [87]. However, glutaraldehyde is a crosslinking agent that is considered toxic and carcinogenic. Therefore, an adaptation or substitute that ensures the cross-linking of the hydrogel needs to be identified, especially if the scaffolds are meant to be used in human patients [88,89,90]. One good example of an alternative agent is genipin, a plant-derived substance. It was shown that genipin, used as a cross-linking agent for chitosan, can enhance mechanical integrity and cytocompatibility, thus making it a better candidate for a chitosan-based drug delivery system for clinical use [91].
Chitosan can also be mixed with polymers for enhanced bioactive properties and osteogenesis. For example, in vitro studies revealed that constructs made of PMMA/PCL (polycaprolactone) and chitosan presented a plateau release of vancomycin after 24 h, while also stimulating human bone marrow stem cells [92]. PMMA and PCL are used for their capacity to create optimal pore sizes for better stimulation of the surrounding cells, and the chitosan, as mentioned earlier, is a perfect carrier for antibiotics, especially vancomycin. The rugosity of the polymer scaffold managed to offer a good adhesion to the chitosan coating, which offered a controlled release without premature degradation [92]. Good release of vancomycin and enhanced antimicrobial activity, in vitro, were also obtained with the following 3D-printed composites: PCL and silica [93], methylcellulose and hydroxyapatite (HA) [94], and PLA (polylactic acid)/HA and chitosan [95].
Metals can also be used for a drug delivery system for vancomycin. In an in vitro study, Titanium-Tantalum-Niobium-Zirconium (TTNZ), a metal alloy, was mixed with hydrogels (TTNZ-Chitosan-Hyaluronic Acid) and loaded with the antibiotic [96]. By testing different concentrations of vancomycin, the optimal concentration for antimicrobial activity and cell adhesion was 5% wt; furthermore, it was proven that the hydrogel loaded with vancomycin, when loaded in the porous structure of TTNZ, led to a slower but sustained release rate [96]. This represents a promising result from a clinical perspective, especially if a long prophylactic treatment is desired, rather than a critical infection treatment, where a high burst of drug release with a shorter period of plateau would be required. Metal-based drug delivery systems for vancomycin were also tested in vivo on New Zealand white rabbit models, where another titanium alloy was used (Ti6Al4V) [97]. Besides the already proven antimicrobial efficiency of these locally administering systems, the porous structure of this metal also promoted bone formation in the femoral defect in rabbits [97]. A strong rationale for choosing metal-based delivery systems, rather than hydrogels or polymers, is due to the ability to withstand very high loads [98]. If a custom-made full-prosthesis that also acts as an antimicrobial system is required, titanium can be a good choice.
A variety of 3D-printed patient-specific scaffolds were designed, with all existing categories of materials from the medical 3D printing industry (biopolymers, hydrogels, ceramics, and metal). All materials presented local efficiency and long drug release periods; some also acted as tissue-engineered scaffolds, where new bone formation was promoted. It is therefore the duty of the medical teams and future research to select optimal and specific mixes for the medical needs and requirements of each patient. The major disadvantages of systemic administration disappear when utilizing these solutions; therefore, an improvement in the patient’s life and a decrease in the risk associated with antibiotic resistance or the occurrence of systemic adverse reactions may be expected.

3.1.2. Ciprofloxacin (CFX)

Ciprofloxacin is a second-generation antibiotic of the fluoroquinolone group (1-cyclopropyl-6-fluoro-4-oxo-7-(piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid) that presents a broad antimicrobial effect, and is used in severe infections of various regions such as the respiratory tract, skin, and bone [99]. The main action mechanism against bacteria is via the inhibition of DNA gyrase, but unlike other antibiotics from the same class, it is more efficient against gram-negative bacteria, such as Pseudomonas aeruginosa, a known candidate found in foreign body implants, frequently used in orthopedic surgeries [100,101].
Many in vitro studies suggest that CFX is highly effective in treating osteomyelitis [102]. Moreover, many derivatives of CFX have shown great results in achieving anti-tumoral or anti-oxidation properties, which support the antimicrobial activity and healing [99,103]. Despite its great therapeutic advantages, oral administration can be accompanied by adverse effects with a great impact on post-orthopedic interventions recovery, such as tendinopathy, neuropathy, altered muscular metabolism, or even tendon rupture, along with increased toxicity for some patients [104,105]. As such, a local form of delivery would facilitate the partial bypass of the side effects, and an optimal solution to these challenges stands in personalized, 3D-printed drug delivery systems.
In an in vitro study, a 3D-printed thermopolymer scaffold was designed and loaded with CFX to test its effects in treating osteomyelitis [106]. To ensure a good delivery rate, the thermopolymer mix (biocompatible poly(ethylene oxide terephtalate)/poly(butylene terephtalate)-PEOT/PBT) was chosen accordingly, for its capability of ensuring a mechanically stable interconnected pore network, with a high percentage of pores, a key characteristic for controlled degradation and release of the drug [106]. When tested on human mesenchymal stromal cells, there was no relevant cytotoxicity present, and the release of CFX was sustained for one month after a burst phase in the first 24 h [106]. A stable and controllable delivery rate was, therefore, obtained [106]. Moreover, the polymers included in the scaffold enhanced the regenerative capabilities of the bone. If translated for in vivo animal/human tests, the scaffold might potentially heal large bone defects while ensuring a stable degradation rate for optimal drug release, thus combining targeted antimicrobial activity with bone regeneration.
PEOT/PBT is an elastomeric thermoplastic mix with important properties, such as high elasticity, viscosity, and wettability [107]. The crystal structures incorporated in its structure grant it mechanical integrity and stability, which, alongside the good degradability in an aqueous environment, make it a strong candidate for use in drug delivery [107]. However, many aspects need to be strictly controlled in order to achieve the desired construct, especially if the translation into clinical practice is desired. Moreover, thermoplastics are known mainly for their physical, rather than their biological and physiological properties [107]. In this context, other materials may be more appropriate for in vivo animal/human implementation. One example is silk fibroin hydrogel, a functional, bioactive material that can be transformed into various 3D bioprintable inks with increased strength, self-healing, adhesive, and conductive properties for sustained drug release or any combination of properties that is desired, depending on the mechanical or bioactive needs [108].
In this spirit, a team of researchers tested a 3D-printed silk fibroin hydrogel loaded with CFX-laden silica microparticles, in vitro, on a mouse pre-osteoblast cell line (MC3T3-E1) [109]. Alongside the antimicrobial effects, the integration of CFX in microparticles improved the control of drug release, lowering the burst phase time to 4 h compared to 24 h when using thermoplastic scaffolds [109]. Moreover, a very high cumulative release rate of approximately 50% was obtained after 72 h, proving that incorporating the CFX in particles is also effectively limiting scaffold-associated infections through its high release rate [109]. The innovative feature of this study is the synergistic combination of bone healing and bone inductive properties of the hydrogel scaffold with CFX’s antimicrobial activity. The increased surface rugosity and enhanced osteoinductive and osteoconductive properties of silk fibroin can play a key role when integrated in large bone defects caused by bone infections in human patients [109].
CFX can also be integrated into 3D-printed drug delivery systems in combination with other drugs, if more pharmacological effects are needed. Mixing it with other antibiotics for effect-compounding, such as gentamicin, or with anti-inflammatory drugs, such as dexamethasone, are just some examples [106,110]. Many other materials can be adapted for testing different delivery rates or other dynamics of the CFX. One good example is poly-caprolactone (PCL), a biocompatible and bioactive polymer with broad applications in 3D-printed scaffolds used in orthopedics, both in vitro and in vivo (animal models–white rats/rabbits) [110,111]. Using these approaches, high-precision, customizable, and locally effective systems can be obtained, allowing medical teams to tailor the antimicrobial treatment to each patient’s needs, aiming for effective recovery and healing, and reduced cytotoxicity and side effects.

3.1.3. Gentamicin

Gentamicin is an aminoglycoside widely used in the treatment of pseudomonal osseous infections and acts by binding to the 30S ribosomal unit of the bacteria, inhibiting protein synthesis [112]. Gentamicin is also frequently used in the treatment of orthopedic implant-related infections [113]. Regarding its safety, literature studies frequently report nephrotoxicity in cases of systemic administration, which imposes the need for an alternate route [114]. A large number of locally controlled delivery systems were clinically used to achieve high concentrations of antibiotics with low systemic absorption. The most intensively studied is the loading of gentamicin in a biopolymer, usually PMMA, that will be molded in a specific shape desired by the medical team for treating complex bone defects [113].
However, some studies conducted on standard medical biopolymers suggest that if the drug remains fixed inside the implant, usually due to a low degradability rate, the antimicrobial effect is minimized, and due to the surface roughness, bacterial colonization is facilitated, thus generating a high risk of sepsis or amputation [115]. Given that, other methods of drug delivery and various types of materials were tested. With the emerging field of 3D printing in medical applications, personalized and highly controlled release systems, such as specific porosities, mechanical properties, and degradability rates, were newly designed or optimized and tested in vitro, which could play an important role in future clinical practice [116,117,118].
Besides the infection cases caused by an insufficient release rate of antibiotics from PMMA-cement, many studies reported localized thermal necrosis due to the high exothermic polymerization when the cement was used in vivo on human patients [119,120,121]. By 3D printing PMMA in specific morphologies, there is no longer a need to manually shape the cements intraoperatively. Thus, the risk of thermal necrosis is eliminated because all the heat-generating reactions have already taken place preoperatively [118]. In an in vitro comparative study, PLA and PMMA scaffolds were designed and loaded with various antibiotics such as gentamicin, nitrofurantoin, and tobramycin [118]. The most effective system was the PMMA scaffold, which allowed for a controlled release rate of gentamicin and tobramycin; however, in the case of nitrofurantoin, only low effectiveness was achieved. The study proved that optimization of PMMA delivery shapes can be achieved using 3D printing with beads, disks, or layered filaments, and various antibiotics can be released without the disadvantages of the standard techniques [118]. Similar to PMMA, PLA scaffolds were loaded with the same antibiotics. While gentamicin release was not as clinically relevant as PMMA, with antimicrobial activity performing optimally only at a concentration 2.5 times higher than PMMA scaffolds, PLA managed to perform in releasing nitrofurantoin [118].
The main cause of the different behavior between the two similar biopolymers could reside in the extrusion temperatures. The range of temperatures for extruding PLA is around 175–275 °C, while for PMMA, it must be lower than 240 °C [122,123]. Therefore, another highly relevant aspect when loading antibiotics into polymers that are later extruded is the melting point of each antibiotic. For example, tobramycin, which has a melting point of around 200 °C, could only be printed optimally with PMMA scaffolds, because PLA requires higher temperatures for increased precision [124]. This can also explain the need for a higher concentration of gentamicin when released from PLA due to thermal inactivation of the antibiotic compared to PMMA. However, compared to PMMA, which already has clinical applications, the use of PLA for drug delivery should be further studied in vivo on animal models to gain a deeper understanding of how physiological microenvironments affect the release behavior.
Gentamicin can be used in combination with other drugs for immunosuppressed cancer patients. Methotrexate, an antifolate drug, is used in the treatment of various cancers, but similar to gentamicin, it can exhibit nephrotoxicity [125]. In severe cases of renal dysfunction, the systemic administration of these two drugs would be imprudent. However, when released controllably from a local scaffold, the risk of systemic absorption is substantially lower. In an in vitro study, the controlled release of gentamicin and methotrexate from a PLA-based 3D-printed scaffold was researched. For optimal release of the drugs, different printing temperatures were used compared to previous studies. A lower quality of the scaffold was preferred; thus, lower temperatures were tested, 175 °C for the antibiotic and 160 °C for methotrexate [117]. The 3D-printed scaffolds presented good release that inactivated the in vitro culture of osteosarcoma cells loaded with E. coli vitroids [117].
Other studies tested the same combination of drugs and demonstrated that an optimal release for 30 days can also be achieved if other materials are used, such as a composite PCL-HA [116]. The advantage of this composite is that the ceramic (HA) can be easily dispersed inside the PCL pores, which are usually formed at high temperatures, thus directly influencing the cellular activity and the drug release in a more controlled manner [126]. This type of polymer-based scaffold, which frequently requires high temperatures, is optimal for the release due to the feasibility of pore formation, a key element in the degradation of the scaffold that ensures the drug elution. However, to adapt this type of construct to a temperature-sensitive drug, some adjustments to the mixture need to be further researched. One example in this direction could be the addition of hydrogels to the mixtures; due to their properties of transitioning from liquid to gel states with temperature increase, lower printing temperatures for the entire composite can be achieved [127,128,129,130].

3.1.4. Tetracyclines

Tetracyclines represent an extensive class of antibiotics, with broad antimicrobial activity on both Gram-positive and Gram-negative bacteria [131]. Tetracycline, a first-generation representative, presents limitations regarding pharmacological bioavailability and pharmacokinetics [131]. However, the advantage of using tetracycline in orthopedic applications lies in its capacity to generate an osteoconductive interface. Therefore, it minimizes the bacterial colonization on the implant surface while also promoting osseointegration [132]. Moreover, some studies suggest that tetracycline can inhibit some matrix metalloproteinases, key enzymes for soft tissue regeneration, and can serve as a healing agent for tendinitis or articular defects [133]. Several customized 3D-printed drug delivery systems were created for delivering tetracycline.
A team of researchers developed a new material mix for a fused-deposition material (FDM) 3D printer [134]. By mixing thermoplastic polyurethane and polyvinyl alcohol, they created nanoporous filaments that presented good degradability and biocompatibility [134]. Using these novel filaments for the construction of the delivery scaffold, a drug system for tetracycline capable of both drug release and absorption was created. The limitation of this study is the short period of 3 days in which the release was tested; however, it was shown that the scaffold did not interfere with the cells (human primary fibroblast) life or functionality [134]. Moreover, the drug release remained stable and controllable. The mechanism of pore formation is based on the capacity of the PVA to dissolve in water [135,136,137]; thus, a sponge-like structure is created. The pore formation is not as optimal as in other materials, such as PLA or PCL, which could affect the mechanical integrity [138,139,140,141,142]. On the other hand, creating a polyurethane sponge-like structure offers absorptive properties to the structures, characteristics that are not often possible with other materials [143]. One of the reasons could be due to the elastomeric properties of the thermoplastic polyurethane (TPU); the pores formed by the elution of PVA expand when hydrated in a liquid environment, promoting absorption [144]. This creates the advantage of absorbing any drug like a sponge, without preloading the drug into the materials before printing. Another advantage is the use of FDM for this system, TPU and PVA being considered easy-printable materials [145,146,147]. Even if the study was performed in vitro, the translation to in vivo animal/human testing is feasible given the technology used, the material availability, and the rare characteristics of drug-loading through sponge-like absorption.
Even though first-generation tetracycline presents many advantages and applications, frequent bacterial resistance mechanisms develop with this drug, such as drug efflux pumps [148]. Therefore, efforts were made to develop other generations of tetracyclines. Doxycycline and minocycline are two relevant representatives of the second-generation tetracyclines, and they present enhanced tissue penetration due to the increased lipophilicity, as well as more feasible dosing protocols [149]. Composite materials were created for enhanced properties and tested in vivo on white rabbits with tibial defects. In this case, the loaded drug was doxycycline, and hydrogel was added to the mix for optimal release rate [150]. Moreover, to accomplish a bone defect repair, ceramics (hydroxyapatite) were added, which are known for their great osteoinductive and osteoconductive properties [111]. Photopolymers (PCLs) were also added for optimal pore formation [150]. The scaffold was tested in a wet state, and a higher release rate was observed compared with previous studies; more precisely, the release was controlled for 28 days in vivo, and after only 15 days, a 100% cumulative release was obtained [150]. As mentioned earlier, second-generation tetracyclines are more liposoluble, so this could stand as an argument for a higher release in the wet-state, purely based on the molecular properties and interactions between two immiscible media.
Improving cell adhesion can increase antimicrobial activity and cell proliferation for rapid healing [151,152]. This can be achieved by adding collagen to the composite mixture and was already demonstrated by a group of researchers who tested a minocycline 3D-printed drug delivery system in vitro on human bone marrow stromal cells [153]. The collagen addition did not affect the minocycline release dynamics. Tetracyclines not only exhibit antimicrobial activity like other antibiotics presented in this section but can also promote bone formation and healing. Although it frequently generates resistance mechanisms in bacteria, this could represent an interesting alternative, mainly due to its unique release mechanism. Translation in clinical use remains to be further studied.

3.1.5. Other Antibiotics

Other strategies in designing release systems were also studied. Some cases of chronic osteomyelitis require combined therapy strategies [151,154]. Systems able to sustain a combined and synergic release were created. It was observed that the morphology and structure of the scaffolds play an important role in the tandem drug release capabilities. Therefore, a four-circle concentric system was designed to deliver levofloxacin in combination with tobramycin, and thus the plateau period of release was prolonged to 60 days compared to previous studies where a porous scaffold maintained the release for 30 days [155]. The advantage of using 3D printing for these systems is the possibility to adapt the position, porosity, dimensions, and other mechanical properties for each circle [155]. In conclusion, a complex system is created in which each component can be loaded with a different drug, and control over each release mechanism is established, so that the synergistic effect is optimal.
3D-printed local systems were also designed for patients with osteoarticular tuberculosis. In this case, given that the treatment to kill Mycobacterium tuberculosis is a long-term therapy, local administration is essential to avoid systemic side effects [156]. Considering the potential of tissue-engineered scaffolds for the local administration of rifampicin, tricalcium phosphate was added alongside the drug-loaded microspheres to offer absorbent properties to the scaffold, while also promoting the healing of bone defects by enhancing the activity of osteogenic genes ALP, OCN, and BSP [157].
In the case of semi-synthetic antibiotics, such as roxithromycin, the adaptation for 3D printing is challenging. The main factor is represented by the solubility properties of the drug and also of the scaffold. In a study where PCL, a substance with low water absorption and hydrophobicity, was used for roxithromycin release, PEG was added to increase the hydrophilicity for a more sustained release and to enhance the bioactive properties [158]. Even though a constant release was achieved, a drug burst was observed in the first hours in vitro. In the cases where a more sustained release is desired from the first moments of activity, adding hydrogels in a multilayered system can represent the solution to limit the initial burst phase [159]. As long as all physical properties are taken into consideration (viscosity, pore sizes, elasticity, solubility, wettability, etc.), adaptations for good printability to various commercially available antibiotics can be achieved. Numerous “background” mechanisms are of great importance when designing a drug delivery system, the most relevant being cross-linkings, pore formation, degradability, and cytocompatibility, among many others.

3.2. Anticancer

Osteosarcoma mainly affects young people and is characterized by rapid invasion and metastasis [160]. The anticancer drug branch is constantly improving its therapy strategies, but the main challenge remains achieving a constant and sufficient drug release that can optimally kill tumor cells [161]. Similar to antibiotics, 3D-printed drug delivery systems can lead to high efficiency in treating osteosarcoma. Moreover, the base principles of 3D printing systems loaded with anticancer drugs do not differ from antibiotic-laden platforms in regard to the 3D printing technologies, materials, release mechanism, or other technical aspects. Given that, only relevant anticancer drugs will be discussed in this section, and the major challenges for each specific drug will be highlighted, pointing out the differences from other drugs in the adaptation for 3D printability.

3.2.1. Cisplatin

Cisplatin is the first choice as an anti-osteosarcoma drug and was tested in vivo on white mouse models [162,163]. Titanium alloy was used to create the load-bearing structure, and through electron beam melting 3D printing technology, a highly porous scaffold was produced [162]. However, highly porous titanium scaffolds are not characterized by absorption or release properties [164]. Therefore, inside the pores, a hydrogel structure (PLGA-PEG-PLGA) was created and loaded with cisplatin. Consequently, a dual system was created, as the hydrogel composite is capable of optimal drug release and has bone formation properties, while the titanium evenly distributes the drug-loaded hydrogel, preventing the whole system from collapsing [162]. Compared to antibiotics, the release balance is more challenging in this case; excessive cisplatin will increase the cytotoxicity, and a low concentration could generate resistant osteosarcoma cells [165]. Moreover, the affected zone could simultaneously be infected with resistant bacteria. The combination of drugs can be achieved, but the release is more challenging [166]. In the case of cisplatin, drug screening models for synergistic effects should be designed.

3.2.2. Doxorubicin

Doxorubicin is a general anticancer drug effective on a broad range of malignancies. However, the systemic administration of this drug can cause cardiotoxicity and myelosuppression [167]. Many categories of the previously presented materials were already designed in a 3D-printed drug delivery system for doxorubicin–metals: titanium alloys [168], Fe3O4 nanoparticles [169]; hydrogels: GelMA [170], cellulose [171]; ceramics: hydroxyapatite [172]. The major challenge when adapting doxorubicin to 3D printers and local delivery is the low hydrophilicity [173]. Due to low solubility, the water particles cannot penetrate the scaffold to ensure the drug elution [174]. This property directly affects the drug kinetics; thus, for good results, encapsulation is frequently needed [175]. Encapsulating the drug in liposomes or other micellar systems can modify its physical properties and transform its crystalline phase into a more amorphous state, which favors interaction with water [176,177].
Another solution stands in adding hydrophilic additives, and examples in this direction are the addition of clay particles or any bioresorbable materials, known for their capacity to increase the wettability of a scaffold [178,179]. Moreover, balancing the solubility can bypass the premature unloading or burst phase [167]. Generally, for a successful release, the solubility of the released item needs to be higher than that in the media, and when hydrophilicity is significantly increased, burst phases can occur [180]. Even though low solubility is contradictory to the burst phase for doxorubicin, it was shown that an important quantity of doxorubicin gets adsorbed to the surface of some materials, like PLGA, and gets released extensively when in contact with an aqueous medium [181]. The solution to that could be to ensure better homogeneity of the scaffold and to limit the external adsorption of the drug to the 3D-printed scaffold.

3.2.3. Zoledronate

Zoledronate is a drug used mainly for treating osteoporosis, but in the last decade, its anticancer properties drew a lot of attention as an innovative treatment option in osteosarcoma [182]. By inhibiting critical mechanisms for tumor progression, such as angiogenesis and invasion, preclinical studies suggest that it can play an important role in treating bone malignancies, while also repairing the bone defects caused by the cancerous cells [183]. Given its combined effects, it quickly became a great candidate for a tissue-engineered drug delivery system. In an in vitro study, where zoledronate was included in a 3D-printed PLLA scaffold, it was proven that the drug release is pH-responsive [184]. In the acidic media group, the zoledronate release was around 21%, and it sustained for several weeks, compared to the neutral pH group, with a very low rate of release. This property, combined with the powerful ability to inhibit the osteoclast activity, highlights the capacity of zoledronate to act only at the affected area, given that the osteoclast surrounding is acidic [184]. Studies that assessed the zoledronate local drug release in vivo on animal models exist only for loaded coatings [185]. To our knowledge, there are no reported in vivo studies with biocompatible materials laden with zoledronate that perform release by scaffold degradability.
Two main challenges occur when 3D printing with zoledronate. The first concern is the thermal stability of zoledronate, especially for extrusion-based 3D printing technologies that usually use high temperatures [186]. Studies on thermal stability for zoledronate suggest that small alterations appear at around 100–150 °C, and more critical processes for its structure start to occur above 180–190 °C [187]. For most of the materials compatible with extrusion-based 3D printers, the optimal printing temperature is above 200 °C [188]. As a solution for this issue, 3D-bioprinting in combination with different hydrogels with tailored properties, a method already described previously for thermal-sensitive antibiotics, can be used. Moreover, it was observed that due to the high polarity of the drug, it can interact with the polymers and affect its solubility and printability [189], which will directly impair the drug release and scaffold morphology, especially regarding pore sizes and homogeneity. These aspects were observed in in vitro studies or in literature studies on zoledronate and were not yet approached by the researchers in their attempt to adapt zoledronate to 3D printers. Therefore, thermal stability and polarity remain to be studied in further research.

3.3. Anti-Inflammatory and Other Drugs

Recent authors have studied synergistic effects of different mixes with antibiotics and anticancer drugs, attempting to directly solve the most serious bone diseases. Among the groups of drugs added as secondary products to better treat the aforementioned diseases, adaptations have also been made to create 3D drug delivery systems for anti-osteoporotic drugs, anti-inflammatory drugs, or even natural compounds with an antioxidant role. A frequently used anti-inflammatory drug is dexamethasone, a synthetic corticoid that also acts as an immunosuppressant and decongestant [190]. Dexamethasone is very sensitive to UV light, which can greatly affect its structure and pharmacological activity. Thus, when UV-based 3D printing technologies are used, higher concentrations of photopolymers need to be used for the scaffold compared to antibiotics or anticancer drugs. Therefore, scaffolds for dexamethasone delivery are usually made of polymers that protect the drug from UV denaturation, such as PTMC (poly (trimethylene carbonate)) [191] or PCL [192]. Other anti-inflammatory drugs that can be incorporated in 3D-printable systems made of hydrogels or polymers are ibuprofen and aspirin [193,194,195].
In some of the antimicrobial or anticancerous 3D-printed drug delivery systems, an antiosteoporotic drug or bone growth factor can be added to regenerate the affected bone area while also resolving the main cause of the defect. The approach should consider whether the drug has low solubility, therefore requiring encapsulation, or if the drug is thermosensitive, so then hydrogel can be added, or if the scaffold has low mechanical strength, so then metals or ZIF-8 can be used. Using this method, alendronate, bone-morphogenetic protein-2, and D3 vitamin were also included in systems for improved bioactive effects and healing in bone defects caused by infections, cancer, or osteoporosis [196,197,198,199,200,201]. Most of them were tested in vitro, and they did not show any different results when compared with the 3D-printed systems that were loaded with only one substance, either an antibiotic or an anticancer drug.
The same challenges regarding printability can appear when natural compounds are integrated into 3D-printable mixes. However, even though adapting them is highly challenging due to their complex bioactive properties, some studies presented successful results, but further research is required. As examples, we mention cucurbitacin, a natural compound that promotes angiogenesis, bone formation, and anti-cancer effects (integration into PLGA/TCP was successful with a controlled release rate of 1.70% each day) [202]. Curcumin, with in vivo osseointegration and in vitro chemoprevention, was successfully integrated in a TCP scaffold alongside allicin, another natural compound with antimicrobial activity, obtaining a controlled release for 30 days at a cumulative rate of 22% [203]. Using TCP 3D-printed scaffolds, other antioxidant natural compounds such as gingerol and allicin were integrated and locally released [204,205].
The many existing 3D printing technologies offer broad possibilities for material adaptations in order to sustain different releases of drugs, proteins, growth factors, or other substances. As long as good control over the solubility, printing temperatures, degradability rates, mechanical characteristics, and chemical properties can be achieved, successful studies could be done in future research.

4. 3D-Printed Drug Screening Models

In the pharmaceutical field, there is a constant need to produce new drugs with more targeted effects and fewer adverse reactions. Moreover, drugs already in use must constantly be improved, especially regarding their pharmacokinetics and pharmacodynamics [206]. The pharmaceutical industry classically uses two methods to verify the properties of drugs, as well as to verify their entire behavior when in contact with living elements [207]. Animal models and 2D cell cultures have been the central approach for drug testing for the majority of drugs [208,209]. Classic 2D cultures consist of a layer of cells attached to a hard surface, onto which different concentrations of various drugs can be administered. Through their interaction with cells, the degree of toxicity can be observed, and the method is reproducible and relatively easy to perform as a first-stage research in drug testing [210,211]. However, cell–cell and cell–microenvironment interactions under the drug influence are impossible to test with these 2D models. Moreover, there is a lack of architectural mimicry of tissues; thus, results regarding the drug activity can be mistaken [212,213].
Comparatively, animal models can provide the complexity of tissue activity and interactions and can play a key role in preclinical drug testing. However, in many cases, there are differences in the cellular metabolism in humans compared to animal models [214]. This difference can lead to mistaken results regarding physiological activity and cytotoxicity of a given drug. Moreover, animal models are not a feasible choice at present, given the costs, the long period necessary for experiments, and ethical considerations [207]. 3D printing technologies have emerged even in the pharmacological drug testing area as a solution to the drawbacks of classical methods. Multicellular, multilayered, and patient-specific models can be created using 3D printing [215]. Moreover, through 3D bioprinting, cells can also be printed homogeneously and in a controlled manner [216].
3D printing drug screening models aims to replace the need for conventional laboratory drug tests or animal models (Figure 3). Moreover, by generating complex 3D architectures, a more faithful model of the human physiological morphologies can be achieved. In particular, 3D-printed models and/or other organoid models could be suitable for mechanistic drug screening, cell–matrix interaction studies, and human cell-based pharmacodynamic testing, where human cellular responses and tissue-specific microenvironments are critical. The major advantage is that the 3D model closely mimics animal or human physiology, is customizable, and can accurately mimic a tissue. Furthermore, it has the advantage of being reproducible and easy to modify. The same model can be printed at any time to test multiple drugs under the same conditions. Also, certain parameters can be modified at any time to see how changing them can influence pharmacokinetics. These systems can model disease progression, local drug delivery, and bone remodeling dynamics and have the potential to yield more accurate results than 2D cultures.
However, animal models remain essential for the assessment of systemic pharmacokinetics (PK), biodistribution, and whole-body toxicity, which require the integrated physiology of multiple organs and metabolic systems. Thus, although 3D bioprinted bone models used in osteoporosis, osteoarthritis, and osteosarcoma studies can effectively replace early-stage screening and reduce animal use. Currently, 3D models as complementary preclinical tools can bring major advantages to drug studies and decrease the time required for their testing. In order to become complete replacements for in vivo testing, future studies should attempt to develop interconnected systems that also mimic the complexity and multifactoriality of biological communication between multiple organ systems of the body. In orthopedics, 3D models were printed for three main groups of bone disease: osteoporosis, osteoarthritis, and osteosarcoma.

4.1. 3D-Printed Drug Screening Models for Osteoporosis/Bone Defects

The first step to creating a drug testing platform for bone diseases is to mimic perfectly the internal porous structure. For this aspect, extensive research was performed on all categories of materials, and porous bone-like structures were successfully created using ceramics (hydroxyapatite, calcium triphosphate), hydrogels, and especially photopolymers (PLA, PCL) [217]. Another key aspect is achieving mechanical behaviors of the scaffold similar to those of natural bone. Even though materials, like metals and ceramics, are known for their bearing-loading properties, precisely controlling the exact values of different physical parameters is challenging [218,219]. A solution to achieve high control of the mechanical characteristics lies in 3D printing technologies based on the water-in-oil concept. With these, high internal phase emulsions are used as inks.
These inks are characterized by concentrated emulsion systems where the dispersed phase fractions go over 74% of the emulsion volume [220,221]. A team of researchers used this approach and created a PCL-PVA scaffold (PVA was used as dispersed phase) that was loaded with the osteoblast MG63 cell line [222]. The scaffold’s pores were similar to those of natural bone, but the main advantage is that by adjusting the emulsion composition, different mechanical properties can be achieved. While certain emulsion percentages were not tested, this PCL-PVA scaffold can perfectly mimic the human bone and can be adjusted to each patient’s mechanical profile. By adding the osteoblast, the pores were populated, and a human-like bone microenvironment was synthesized for in vitro studies. As reported in this study, various drugs can be tested on this platform, especially in terms of cytotoxicity and how the extracellular matrix can be modified under the activity of a certain drug [222]. Similar results were obtained by 3D bioprinting a bone organ model of graphene oxide composite laden with human mesenchymal stem cells instead of osteoblasts [223].
Interesting results were also obtained using 3D bioprinters for scaffold-free models [224]. Scaffold-free 3D-printed models do not require foundation biomaterials, because the cells rely on their interaction to create their mechanical support [225,226]. Thus, a more native-like model can be achieved. This approach was used by a team of researchers to specifically test drugs for osteogenesis control on differentiated bone marrow-derived mesenchymal stem cells [224]. While the effects on experimental drugs for osteogenesis inhibition (PD98059 and U0126) were studied, the emphasis was on drugs that promote osteogenesis (Icariin and Purmorphamine). It was shown that purmorphamine, a purine derivative that promotes osteogenesis by Hedgehog pathway signaling, can increase bone formation similar to BMP4 (bone morphogenetic protein 4) [224]. Icariin has also been tested and considered an efficient anti-osteoporotic drug, given the in vitro results.
To conclude, there are already good results in creating organ models that mimic the human bone. Due to the enhanced control offered by the 3D printing technologies and the possibilities to add cells in a precise manner, models that accurately mimic the bioactivity and morphologies of the bone can serve as drug screening platforms. Therefore, the classical testing platforms or the use of animal models can safely be replaced as a preclinical phase component before testing on humans.

4.2. 3D-Printed Drug Screening Models for Osteoarthritis

Another form of scaffold-free 3D bioprinting is creating spheroids instead of cell sheets, which was attempted for osteoporosis models. Using the same principle, by aspiration-assisted 3D bioprinting techniques, cells can be placed in certain positions to facilitate their interaction to create self-assembled spheroids [225]. Differently, they create the extracellular matrix on the surface, and the interactions between the tested drug and the scaffold are closer in results to how they would act on real tissues. As such, the drug will reach each cell in different concentrations, depending on its position compared to the interface of interaction [227,228]. Using this concept, a team of researchers created spheroids from amniotic fluid-derived mesenchymal stromal cells, and a chondrogenic-like tissue was formed [229]. Although not tested in vitro or in vivo, it could act as a drug screening platform for osteoarthritis drug in vitro testing for local activity, like sclerostin, cyclopamine, osteoprotegerin, etc. [230].
Advanced systems that can behave as native tissue environments were also designed and created. One example in this direction is bioreactors, which are platforms that allow for various models to be cultivated, such as spheroids, organ-on-chip, and other organ models. They can enable dynamic systems by providing control over the fluids added to it, or the nutrient and metabolic exchanges [231]. Moreover, mechanical stress can be tested by changing the bioreactor environment’s mechanical characteristics [232]. These systems can also be 3D-printed, and an osteochondral bioreactor with fluid control was designed and printed using photopolymer resins by stereolithography [232]. Given that the fidelity of the bioreactors is higher compared to static models, they can also stand as possible drug screening systems for osteoarthritis.

4.3. Others

Osteosarcoma models were also 3D-printed, in order to test various anticancer drugs, already mentioned in the drug delivery systems section, such as cisplatin and doxorubicin [170,233,234,235,236]. However, given that 3D-printed delivery systems already exist in this case, the screening platforms can be helpful in testing synergistic effects or if some changes are made to the pharmacological properties of the drug. As base principles, the bone model is created in the same manner, with the only difference being the cell lines that are added. According to the previously cited studies, good results were obtained for MG-63, human osteosarcoma U-2 OS and its cisplatin-resistant variant, and HeLa-human bone osteosarcoma epithelial cell line. Using the bone organ models, any drug can be tested, and it is up to the researchers to select the drugs, cell lines, and materials to use for optimal results. To conclude, 3D-printed drug screening models can play a role as an intermediate test platform between in vivo studies on humans and in vitro studies.
In the next table (Table 1), we have integrated the research conducted on 3D printing, aiming to create either drug delivery systems or drug screening platforms. For each system, the technology, the materials and their properties, the types of drugs and their bioactive properties are specified.

5. Future Perspectives and Discussion

The 3D-printable drug delivery and drug screening systems with effects on the main orthopedic diseases, such as osteoporosis, osteoarthritis, and osteosarcoma, were presented here. There is a great need for new and therapeutically optimized treatments for a wide range of diseases with secondary pathological effects on the bone. Among these, the most relevant categories are metabolic diseases, which can alter the bone remodeling metabolism, the mineral metabolism, and the intracellular homeostasis; endocrine diseases, such as parathyroid disorders with effects on bone mineral metabolism or even renal diseases, are also of importance [267,268,269]. Given the complexity of the pathophysiological mechanisms underlying the manifestations of these diseases, there is a need for 3D models designated for local administration and screening for drug optimization, without directly affecting humans or laboratory animals through classical methods. An overview of the available technology studies is provided in Table 2.
Gaucher disease is a genetic disorder characterized by a deficiency of glucocerebrosidase, which leads to the accumulation of glucocerebrosides at the intracellular level, in lysosomes of multiple organs such as the spleen, liver, and bone marrow [270,271]. Bone damage is one of the major complications and causes of death, since bone remodeling and vascularization are totally compromised [272,273]. These effects lead to the appearance of chronic inflammation with unbearable pain and multiple pathological effects, ranging from osteopenia to osteonecrosis [274]. The current treatment stands in the restoration of the missing enzyme to reduce its accumulation in the lysosomes [275]. Pain relief and slowing of bone damage were reported with the combined treatment of enzyme replacement therapy and substrate reduction therapy [276]. However, local therapies with drugs for bone repair could present a potentiating element in the treatment of Gaucher disease.
A team of researchers managed to build a bone organ model laden with Gaucher cells. By aspiration-assisted 3D bioprinting (technology already presented in the drug screening section), a free-form spheroidal model was designed [277]. To mimic the affected bone, the spheroids were produced by the self-assembling co-culture of human bone marrow-derived mesenchymal stem cells and peripheral blood mononuclear cells from patients with Gaucher, which were differentiated into bone cell lineages [277]. While no drugs were specifically tested on this model, it could easily act as a drug screening platform where bone-healing medication or Gaucher drugs can be tested, such as monoclonal antibodies or Sclerostin [278,279]. Similar results could be obtained following the same methods for other metabolic diseases.
3D-printed systems can also be created to test potentially cytotoxic drugs. Microfluidic chips and transparent systems can be built using polymers such as PLA and PMMA [280,281]. The microfluidic chips have the potential to create randomly curved channels in which drugs can be loaded and “travel” in contact with the channel walls, where human cells are present [242]. The use of transparent materials allows testing the drug activity through optical methods, such as optical pharmacokinetics systems [282]. This type of model was created with relatively low costs using an FDM printer, where staurosporine, an apoptosis-inducing drug, was tested for its cytotoxic effects against human mesenchymal stem cells [242].
Theoretically, the potential of these highly controllable and customizable systems is limitless. As long as full control over the mechanical and bioactive parts is maintained, spatial models that perfectly mimic all existing tissues and organs can be created. Moreover, by using stem cells or different cell precursors or progenitors, the pathophysiological mechanisms produced by the affected cells can also be mimicked. As long as the properties of the drugs are known in terms of thermal and mechanical sensitivity, any drug can be adapted to 3D printing. By integrating these two concepts, patient-specific disease models can be created to facilitate the identification of customizable treatments. However, some challenges and limitations remain. In vivo studies and clinical translation are insufficiently addressed. Ethical considerations, methodological procedures, manufacturing protocols, legal regulation, and long-term studies are yet to be studied. In contrast, devices that are 3D-printed must be in accordance with applicable quality systems and regulations on medical devices. The U.S. Food and Drug Administration (FDA) states that if products are made through additive manufacturing, this does not change the regulatory pathway and should not fall under a special protocol [283]. All resulting products must comply with applicable Chemistry, Manufacturing, and Control (CMC) standards [283].
Rather, the FDA’s guidance on additive manufacturing emphasizes technical considerations, material quality, print validation and post-processing quality, physical and mechanical characteristics, and biological considerations (e.g., cleaning, sterility, and biocompatibility) [283,284]. In our view, regarding biocompatible materials, each manufacturer is required to provide the necessary documents to prove the safety of their product, and regarding the safety of production, each institution dealing with the production of such 3D-printable devices must think about and submit for approval a workflow with the possibility of verification by an audit or other third parties.
It is important to note that patient-adapted devices require regulatory authorities to review the design process and adaptation processes for use on the patient, not necessarily the individual device. In our opinion, the regulatory paths to be adopted could be similar to those adopted for the approval of the first pharmacoprinted drug by the FDA in August 2015, SPRITAM®, that is, complying with the following indications: those from the Chemistry, Manufacturing, and Control (CMC), as well as the 21 CFR 200s and 300s regulations [283,285,286].
In a 2025 article on clinical trials for 3D-printed drugs, it is mentioned that the FDA in the USA and the MHRA (Medicines and Healthcare products Regulatory Agency) in the UK are developing quality assurance and validation strategies for 3D manufacturing [287]. In the European Union, the Quality Innovation Group (QIG) (2025–2027) has been established to prepare a document aimed at integrating additive manufacturing and decentralized production into existing regulations and to encourage early dialogue between innovators and regulators [288]. The QIG focuses on standards (ISO/ASTM 52900 series) and interdisciplinary regulatory committees. These initiatives aim to address current translational barriers, such as reproducibility, lack of validated reference materials, and lack of clear quality control protocols [289]. Major challenges persist in terms of reproducibility, in contrast with 2D models or animal models.
Furthermore, drug delivery systems must be adapted in order to act as implants. In this regard, they must function biomechanically, and a major disadvantage to their integrity over time is biodegradability. However, the degradability of the prosthesis ensures the release of drugs, a key element for such a system. Given this contrast, it is possible that the total replacement of implants with drug-releasing implants is not an optimal solution. However, combined techniques could be advantageous. Integrating the constructions necessary for resistance over time and for biomechanical efficiency with drug delivery systems could be an option. Another option could be the effort of limiting the degradability over time of the entire implant by forcing osteogenesis to restore the integrity of the piece after the total elimination of the drug.
3D printing presents challenges in terms of stability over time and under various conditions that occur during printing, both for the scaffold made of the chosen biocompatible material and for the drugs [290]. To ensure a clinical translation of a 3D-printed product, we must mention that sterilization is mandatory, but this process can deform the models or leave residues. In a comparative study of steam sterilization (temperatures above 105 °C) with hydrogen peroxide or ethylene oxide plasma of 3D-printed models, it was found that steam sterilization at a high temperature significantly deformed models made of thermosensitive materials such as polylactic acid (PLA) [291,292]. Moreover, it was shown that steam sterilization at low temperature (105 °C), plasma sterilization, and gas sterilization preserved the fidelity of the models. However, it is worth mentioning that steam sterilization generally does not work on porous or architecturally complex models, and plasma or gas sterilization is less common in terms of their applicability in healthcare units [291].
The stability of active substances, such as drugs integrated into 3D models, during printing is another key issue. Many drugs degrade at high temperatures or under UV radiation (such as that used in vat-photopolymerization-based printing techniques). A review of 3D printing of thermolabile drugs indicates that semi-solid state extrusion, DLP, Binder Jetting, and SLA allow printing of thermolabile drugs because they avoid high temperatures, although photopolymerization can lead to light-induced degradation [293].
If temperatures frequently exceed 120 °C, this can lead to drug degradation, altered bioactivity, and poor mechanical stability. Therefore, the printing of heat-sensitive proteins, peptides, or antibiotics should be performed using potentially hybrid techniques that combine low temperatures with limited use of radiation to cover a wider range of drugs [294,295]. Researchers should also assess leachables from photoinitiators or unreacted monomers and confirm that sterilization does not alter drug potency or scaffold properties. Stability studies and aseptic printing processes should be included as potential verification methods in future studies [296].
To present in this section the future potential of printing technologies, we must also mention the possibility of 4D printing. By introducing this term, we refer to the potential of 3D models to present properties over time, such as changes in degradation behavior over time and as a function of temperature. In this sense, a review exemplifies the fact that scaffolds with osteogenic differentiation power can be generated that can change their shape and drug release capacities over time, which may have greater efficiency than the classic 3D models that were presented in this review [297]. We should also mention the potential that AI could have both in generating printing algorithms specific to certain materials or drugs, as well as by using AI in the future, potentially to generate 3D designs without human intervention [298].
From an ethical perspective, 3D printing should always be chosen over the unjustified use of animal models, but given that research in this field is still in its early stages, in vivo validation will still be required. There are no methodologies, protocols, or regulations that prove that 3D printing can completely replace the use of animal models. If future tests demonstrate the effectiveness of 3D printing in drug testing and the benefits will be significantly superior to classical methods, 3D printing might represent a new standard. However, to create the most realistic models, viable human cells from generic cultures or from the specific patient must be used directly. This may lead to issues regarding donor ethical considerations and, therefore, to limitations of using human cells on other patients. The lack of long-term studies cannot indicate whether excessive degradability could produce serious effects on the patients in whom these systems are used. Moreover, from a legal point of view, a policy has not yet been created that would assume responsibility for those who produce the 3D model, the hospital in which they would be used, or the manufacturer of the elements used. In the clinic, researchers should implement, as transparently as possible, the use of informed consent to clarify the use of cells for in vitro or in vivo studies (in the case of bioprinting). Anonymization should comply with data protection regulations, such as the GDPR (General Data Protection Regulation), ensuring that patient identities cannot be easily identified [299]. Standards, such as the Guidelines for Human Biobanks and Databases for Genetic Research, ISO 20387 (Biobanking), recommend clear consent materials, coding systems that separate personal identifiers from samples, and governance structures that control access [300]. Researchers should also consider return policies and data sharing agreements. New regulations are emerging that must be approved both for the pharmaceutical side, especially if combinations of drugs are used, but also for the implementation of scaffolds in patients.

6. Conclusions

3D printing has significant potential in creating personalized drug screening or local delivery systems at the site of the disease. The studies reviewed in this paper present optimistic in vitro and in vivo results for the administration of anticancer, antimicrobial, or bone reconstruction treatments, in a controlled manner. However, translation to patients remains to be further studied, especially from the perspective of reproducibility, regulatory, and also their long-term functionality. The development of new printing techniques and new materials has led drug testing to a possible new standard, from which it can further progress in the direction of personalized treatments in bone therapeutics (Figure 4).

Author Contributions

Conceptualization, I.-A.T., A.-G.G., S.D. and C.S.; methodology, S.D., A.-E.S., I.A.B. and C.C.; validation, A.C., I.A.B., A.C.D., C.C. and C.S.; formal analysis, I.-A.T., A.-G.G., A.-E.S. and C.D.M.D.; investigation, E.-T.T., T.-M.V. and C.D.M.D.; data curation, E.-T.T., T.-M.V., A.C. and C.D.M.D. writing—original draft preparation, I.-A.T., A.-G.G., E.-T.T., T.-M.V., S.D., A.-E.S., A.C., C.D.M.D., I.A.B., A.C.D., C.C. and C.S.; writing—review and editing, A.C., I.A.B., A.C.D., C.C. and C.S.; visualization, I.-A.T. and A.-G.G.; supervision, C.C. and C.S. All authors have read and agreed to the published version of the manuscript.

Funding

Publication of this paper was supported by the University of Medicine and Pharmacy Carol Davila, through the institutional program Publish not Perish.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wang, S.; Chen, X.; Han, X.; Hong, X.; Li, X.; Zhang, H.; Li, M.; Wang, Z.; Zheng, A. A Review of 3D Printing Technology in Pharmaceutics: Technology and Applications, Now and Future. Pharmaceutics 2023, 15, 416. [Google Scholar] [CrossRef]
  2. Lakkala, P.; Munnangi, S.R.; Bandari, S.; Repka, M. Additive manufacturing technologies with emphasis on stereolithography 3D printing in pharmaceutical and medical applications: A review. Int. J. Pharm. X 2023, 5, 100159. [Google Scholar] [CrossRef]
  3. Sarmah, P.; Gupta, K. Recent Advancements in Fabrication of Metal Matrix Composites: A Systematic Review. Materials 2024, 17, 4635. [Google Scholar] [CrossRef]
  4. Tuli, N.T.; Khatun, S.; Rashid, A.B. Unlocking the future of precision manufacturing: A comprehensive exploration of 3D printing with fiber-reinforced composites in aerospace, automotive, medical, and consumer industries. Heliyon 2024, 10, e27328. [Google Scholar] [CrossRef] [PubMed]
  5. Stanojević, G.; Medarević, D.; Adamov, I.; Pešić, N.; Kovačević, J.; Ibrić, S. Tailoring Atomoxetine Release Rate from DLP 3D-Printed Tablets Using Artificial Neural Networks: Influence of Tablet Thickness and Drug Loading. Molecules 2021, 26, 111. [Google Scholar] [CrossRef] [PubMed]
  6. Md. Shoaib, A.; Ayesha, A.; Iftikhar, A.; Sheikh, S.-u.-N. Pharmaceutical Product Development Exploiting 3D Printing Technology: Conventional to Novel Drug Delivery System. Curr. Pharm. Des. 2018, 24, 5029–5038. [Google Scholar] [CrossRef]
  7. Vaz, V.M.; Kumar, L. 3D Printing as a Promising Tool in Personalized Medicine. AAPS PharmSciTech 2021, 22, 49. [Google Scholar] [CrossRef]
  8. Vanderburgh, J.; Sterling, J.A.; Guelcher, S.A. 3D Printing of Tissue Engineered Constructs for In Vitro Modeling of Disease Progression and Drug Screening. Ann. Biomed. Eng. 2017, 45, 164–179. [Google Scholar] [CrossRef]
  9. Marei, I.; Abu Samaan, T.; Al-Quradaghi, M.A.; Farah, A.A.; Mahmud, S.H.; Ding, H.; Triggle, C.R. 3D Tissue-Engineered Vascular Drug Screening Platforms: Promise and Considerations. Front. Cardiovasc. Med. 2022, 9, 847554. [Google Scholar] [CrossRef]
  10. Steinberg, E.; Friedman, R.; Goldstein, Y.; Friedman, N.; Beharier, O.; Demma, J.A.; Zamir, G.; Hubert, A.; Benny, O. A fully 3D-printed versatile tumor-on-a-chip allows multi-drug screening and correlation with clinical outcomes for personalized medicine. Commun. Biol. 2023, 6, 1157. [Google Scholar] [CrossRef]
  11. Mazzocchi, A.; Soker, S.; Skardal, A. 3D bioprinting for high-throughput screening: Drug screening, disease modeling, and precision medicine applications. Appl. Phys. Rev. 2019, 6, 011302. [Google Scholar] [CrossRef]
  12. Thiruchandran, G.; Dean, O.; Alim, D.; Crawford, A.; Salim, O. Three-dimensional printing in orthopaedic surgery: A review of current and future applications. J. Orthop. 2025, 59, 22–26. [Google Scholar] [CrossRef] [PubMed]
  13. Bădilă, A.E.; Rădulescu, D.M.; Niculescu, A.G.; Grumezescu, A.M.; Rădulescu, M.; Rădulescu, A.R. Recent Advances in the Treatment of Bone Metastases and Primary Bone Tumors: An Up-to-Date Review. Cancers 2021, 13, 4229. [Google Scholar] [CrossRef] [PubMed]
  14. Emami, A.; Namdari, H.; Parvizpour, F.; Arabpour, Z. Challenges in osteoarthritis treatment. Tissue Cell 2023, 80, 101992. [Google Scholar] [CrossRef]
  15. Patel, D.; Wairkar, S. Bone regeneration in osteoporosis: Opportunities and challenges. Drug Deliv. Transl. Res. 2023, 13, 419–432. [Google Scholar] [CrossRef]
  16. Lee, S.Y.; Lee, J.W. Mimicking microbone tissue by 3-dimensional printing. Organoid 2024, 4, e4. [Google Scholar] [CrossRef]
  17. Kortam, S.; Lu, Z.; Zreiqat, H. Recent advances in drug delivery systems for osteosarcoma therapy and bone regeneration. Commun. Mater. 2024, 5, 168. [Google Scholar] [CrossRef]
  18. Nair, A.; Varghese, B.A.; Gopi, S.; Jacob, J. 11—Smart drug delivery systems of natural products for inflammation: From fundamentals to the clinic. In Inflammation and Natural Products; Gopi, S., Amalraj, A., Kunnumakkara, A., Thomas, S., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 217–238. [Google Scholar] [CrossRef]
  19. Codrea, C.I.; Fruth, V. Three-Dimensionally Printed Scaffolds and Drug Delivery Systems in Treatment of Osteoporosis. Biomimetics 2025, 10, 429. [Google Scholar] [CrossRef]
  20. Motiei, M.; Kashanian, S.; Lucia, L.A.; Khazaei, M. Intrinsic parameters for the synthesis and tuned properties of amphiphilic chitosan drug delivery nanocarriers. J. Control. Release 2017, 260, 213–225. [Google Scholar] [CrossRef]
  21. Mirchandani, Y.; Patravale, V.B.; S, B. Solid lipid nanoparticles for hydrophilic drugs. J. Control. Release 2021, 335, 457–464. [Google Scholar] [CrossRef]
  22. Borandeh, S.; van Bochove, B.; Teotia, A.; Seppälä, J. Polymeric drug delivery systems by additive manufacturing. Adv. Drug Deliv. Rev. 2021, 173, 349–373. [Google Scholar] [CrossRef] [PubMed]
  23. Farjaminejad, S.; Farjaminejad, R.; Hasani, M.; Shojaei, S. Innovative Drug Delivery Systems in Bone Regeneration: Benefits and Applications in Tissue Engineering. J. Bionic Eng. 2025, 22, 2286–2307. [Google Scholar] [CrossRef]
  24. Qiu, E.; Liu, F. PLGA-based drug delivery systems in treating bone tumors. Front. Bioeng. Biotechnol. 2023, 11, 1199343. [Google Scholar] [CrossRef] [PubMed]
  25. Mehta, M.; Bui, T.A.; Yang, X.; Aksoy, Y.; Goldys, E.M.; Deng, W. Lipid-Based Nanoparticles for Drug/Gene Delivery: An Overview of the Production Techniques and Difficulties Encountered in Their Industrial Development. ACS Mater. Au 2023, 3, 600–619. [Google Scholar] [CrossRef]
  26. Grini, M.I.; Benbayer, C.; Saidi-Besbes, S.; Elaissari, A. Advances in mesoporous silica nanoparticles as carriers for drug delivery and other biomedical applications. Microporous Mesoporous Mater. 2025, 391, 113603. [Google Scholar] [CrossRef]
  27. Qiu, C.; Wu, Y.; Guo, Q.; Shi, Q.; Zhang, J.; Meng, Y.; Xia, F.; Wang, J. Preparation and application of calcium phosphate nanocarriers in drug delivery. Mater. Today Bio 2022, 17, 100501. [Google Scholar] [CrossRef]
  28. Bai, L.; Tao, G.; Feng, M.; Xie, Y.; Cai, S.; Peng, S.; Xiao, J. Hydrogel Drug Delivery Systems for Bone Regeneration. Pharmaceutics 2023, 15, 1334. [Google Scholar] [CrossRef]
  29. Zhao, Y.; Xiong, Y.; Zhao, Y. Beyond Drug Delivery: Metal–Organic Framework-Derived Nanosystems for Bone Regeneration under Complicated Pathological Microenvironments. Acc. Mater. Res. 2024, 5, 1532–1543. [Google Scholar] [CrossRef]
  30. Borges, R.; Pelosine, A.M.; de Souza, A.C.S.; Machado, J.; Justo, G.Z.; Gamarra, L.F.; Marchi, J. Bioactive Glasses as Carriers of Cancer-Targeted Drugs: Challenges and Opportunities in Bone Cancer Treatment. Materials 2022, 15, 9082. [Google Scholar] [CrossRef]
  31. Ingber, D.E. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat. Rev. Genet. 2022, 23, 467–491. [Google Scholar] [CrossRef]
  32. Wang, H.; Tian, L.; Goldstein, A.; Liu, J.; Lo, H.-C.; Sheng, K.; Welte, T.; Wong, S.T.C.; Gugala, Z.; Stossi, F.; et al. Bone-in-culture array as a platform to model early-stage bone metastases and discover anti-metastasis therapies. Nat. Commun. 2017, 8, 15045. [Google Scholar] [CrossRef]
  33. Paek, K.; Kim, S.; Tak, S.; Kim, M.K.; Park, J.; Chung, S.; Park, T.H.; Kim, J.A. A high-throughput biomimetic bone-on-a-chip platform with artificial intelligence-assisted image analysis for osteoporosis drug testing. Bioeng. Transl. Med. 2023, 8, e10313. [Google Scholar] [CrossRef]
  34. Fois, M.G.; van Griensven, M.; Giselbrecht, S.; Habibović, P.; Truckenmüller, R.K.; Tahmasebi Birgani, Z.N. Mini-bones: Miniaturized bone in vitro models. Trends Biotechnol. 2024, 42, 910–928. [Google Scholar] [CrossRef] [PubMed]
  35. Tang, Q.; Li, X.; Lai, C.; Li, L.; Wu, H.; Wang, Y.; Shi, X. Fabrication of a hydroxyapatite-PDMS microfluidic chip for bone-related cell culture and drug screening. Bioact. Mater. 2021, 6, 169–178. [Google Scholar] [CrossRef] [PubMed]
  36. Natoli, D.; Levin, M.; Tsygan, L.; Liu, L. Chapter 33—Development, Optimization, and Scale-Up of Process Parameters: Tablet Compression. In Developing Solid Oral Dosage Forms, 2nd ed.; Qiu, Y., Chen, Y., Zhang, G.G.Z., Yu, L., Mantri, R.V., Eds.; Academic Press: Boston, MA, USA, 2017; pp. 917–951. [Google Scholar] [CrossRef]
  37. Shanmugam, S. Granulation techniques and technologies: Recent progresses. Bioimpacts 2015, 5, 55–63. [Google Scholar] [CrossRef] [PubMed]
  38. Porter, S.C. Chapter 27—Coating of pharmaceutical dosage forms. In Remington, 23rd ed.; Adejare, A., Ed.; Academic Press: Cambridge, MA, USA, 2021; pp. 551–564. [Google Scholar] [CrossRef]
  39. Jadhav, A.; Jadhav, V.S. A review on 3D printing: An additive manufacturing technology. Mater. Today Proc. 2022, 62, 2094–2099. [Google Scholar] [CrossRef]
  40. Gail Kim, R.; Abisado, M.; Villaverde, J.; Sampedro, G. A Survey of Image-Based Fault Monitoring in Additive Manufacturing: Recent Developments and Future Directions. Sensors 2023, 23, 6821. [Google Scholar] [CrossRef]
  41. Ngo, T.D.; Kashani, A.; Imbalzano, G.; Nguyen, K.T.Q.; Hui, D. Additive manufacturing (3D printing): A review of materials, methods, applications and challenges. Compos. Part B Eng. 2018, 143, 172–196. [Google Scholar] [CrossRef]
  42. Bottini, L.; Boschetto, A.; Veniali, F.; Gaudenzi, P. 7—Additive manufacturing for CubeSat structure fabrication. In Next Generation CubeSats and SmallSats; Branz, F., Cappelletti, C., Ricco, A.J., Hines, J.W., Eds.; Elsevier: Amsterdam, The Netherlands, 2023; pp. 153–180. [Google Scholar] [CrossRef]
  43. Chartrain, N.A.; Gilchrist, K.H.; Ho, V.B.; Klarmann, G.J. 3D bioprinting for the repair of articular cartilage and osteochondral tissue. Bioprinting 2022, 28, e00239. [Google Scholar] [CrossRef]
  44. Lan, W.; Huang, X.; Huang, D.; Wei, X.; Chen, W. Progress in 3D printing for bone tissue engineering: A review. J. Mater. Sci. 2022, 57, 12685–12709. [Google Scholar] [CrossRef]
  45. Sabahi, N.; Roohani, I.; Wang, C.H.; Li, X. Material extrusion 3D printing of bioactive smart scaffolds for bone tissue engineering. Addit. Manuf. 2025, 98, 104636. [Google Scholar] [CrossRef]
  46. Nugroho, W.T.; Dong, Y.; Pramanik, A.; Chithirai Pon Selvan, M.; Zhang, Z.; Ramakrishna, S. Additive manufacturing of re-entrant structures: Well-tailored structures, unique properties, modelling approaches and real applications. Addit. Manuf. 2023, 78, 103829. [Google Scholar] [CrossRef]
  47. Tymrak, B.M.; Kreiger, M.; Pearce, J. Mechanical Properties of Components Fabricated with Open-Source 3-D Printers Under Realistic Environmental Conditions. Mater. Des. 2014, 58, 242–246. [Google Scholar] [CrossRef]
  48. Lefay, C.; Guillaneuf, Y. Recyclable/degradable materials via the insertion of labile/cleavable bonds using a comonomer approach. Prog. Polym. Sci. 2023, 147, 101764. [Google Scholar] [CrossRef]
  49. Islam, A.; Rahman, M.Z. 13.18—Recent advances in additive manufacturing techniques: An in-depth review. In Comprehensive Materials Processing, 2nd ed.; Hashmi, S., Ed.; Elsevier: Oxford, UK, 2024; pp. 352–378. [Google Scholar] [CrossRef]
  50. Khanna, N.; Zadafiya, K.; Patel, T.; Kaynak, Y.; Rahman Rashid, R.A.; Vafadar, A. Review on machining of additively manufactured nickel and titanium alloys. J. Mater. Res. Technol. 2021, 15, 3192–3221. [Google Scholar] [CrossRef]
  51. Lai, H.; Peng, X.; Li, L.; Zhu, D.; Xiao, P. Novel monomers for photopolymer networks. Prog. Polym. Sci. 2022, 128, 101529. [Google Scholar] [CrossRef]
  52. Xing, F.; Xu, J.; Yu, P.; Zhou, Y.; Zhe, M.; Luo, R.; Liu, M.; Xiang, Z.; Duan, X.; Ritz, U. Recent advances in biofabrication strategies based on bioprinting for vascularized tissue repair and regeneration. Mater. Des. 2023, 229, 111885. [Google Scholar] [CrossRef]
  53. Huh, J.T.; Yoo, J.J.; Atala, A.; Lee, S.J. Chapter 74—Three-dimensional bioprinting for tissue engineering. In Principles of Tissue Engineering, 5th ed.; Lanza, R., Langer, R., Vacanti, J.P., Atala, A., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 1391–1415. [Google Scholar] [CrossRef]
  54. Budharaju, H.; Suresh, S.; Sekar, M.P.; De Vega, B.; Sethuraman, S.; Sundaramurthi, D.; Kalaskar, D.M. Ceramic materials for 3D printing of biomimetic bone scaffolds—Current state-of-the-art & future perspectives. Mater. Des. 2023, 231, 112064. [Google Scholar] [CrossRef]
  55. Dizon, J.R.C.; Espera, A.H.; Chen, Q.; Advincula, R.C. Mechanical characterization of 3D-printed polymers. Addit. Manuf. 2018, 20, 44–67. [Google Scholar] [CrossRef]
  56. Sun, Y.; Juncos Bombin, A.D.; Boyd, P.; Dunne, N.; McCarthy, H.O. Application of 3D printing & 3D bioprinting for promoting cutaneous wound regeneration. Bioprinting 2022, 28, e00230. [Google Scholar] [CrossRef]
  57. Yu, H.Z. Chapter 6—Beyond metals and alloys: Additive friction stir deposition of metal matrix composites. In Additive Friction Stir Deposition; Yu, H.Z., Ed.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 203–232. [Google Scholar] [CrossRef]
  58. Joharji, L.; Mishra, R.B.; Alam, F.; Tytov, S.; Al-Modaf, F.; El-Atab, N. 4D printing: A detailed review of materials, techniques, and applications. Microelectron. Eng. 2022, 265, 111874. [Google Scholar] [CrossRef]
  59. Mohammadi, H.; Ahmad, Z.; Petrů, M.; Mazlan, S.A.; Faizal Johari, M.A.; Hatami, H.; Rahimian Koloor, S.S. An insight from nature: Honeycomb pattern in advanced structural design for impact energy absorption. J. Mater. Res. Technol. 2023, 22, 2862–2887. [Google Scholar] [CrossRef]
  60. Shahriar, M.A.; Kobir, M.H.; Rahman, S.; Rahman, M.Z.; Saha, B. 13.03—Overview of additive manufacturing and applications of 3D printed composites. In Comprehensive Materials Processing, 2nd ed.; Hashmi, S., Ed.; Elsevier: Oxford, UK, 2024; pp. 58–76. [Google Scholar] [CrossRef]
  61. Tufail, A.; Schmidt, F.; Maqbool, M. 15—Three-dimensional printing of hydroxyapatite. In Handbook of Ionic Substituted Hydroxyapatites; Khan, A.S., Chaudhry, A.A., Eds.; Woodhead Publishing: Sawston, UK, 2020; pp. 355–381. [Google Scholar] [CrossRef]
  62. Sinha, P.; Mukhopadhyay, T. Programmable multi-physical mechanics of mechanical metamaterials. Mater. Sci. Eng. R Rep. 2023, 155, 100745. [Google Scholar] [CrossRef]
  63. Hansen, C.J.; Peterson, A.M.; Park, J.H. Chapter 23—3D printing. In Handbook of Thermoset Plastics, 4th ed.; Dodiuk, H., Ed.; William Andrew Publishing: Boston, MA, USA, 2022; pp. 1021–1043. [Google Scholar] [CrossRef]
  64. V, K.; Kumar, B.N.; Kumar, S.S.; M, V. Magnesium role in additive manufacturing of biomedical implants—Challenges and opportunities. Addit. Manuf. 2022, 55, 102802. [Google Scholar] [CrossRef]
  65. Karunakaran, R.; Ortgies, S.; Tamayol, A.; Bobaru, F.; Sealy, M.P. Additive manufacturing of magnesium alloys. Bioact. Mater. 2020, 5, 44–54. [Google Scholar] [CrossRef]
  66. Elliott, A.M.; Cramer, C.L.; Nandwana, P.; Chmielus, M.; Mostafaei, A. Binder Jet-Metals. In Encyclopedia of Materials: Metals and Alloys; Caballero, F.G., Ed.; Elsevier: Oxford, UK, 2022; pp. 120–133. [Google Scholar] [CrossRef]
  67. Szymczyk-Ziółkowska, P.; Łabowska, M.B.; Detyna, J.; Michalak, I.; Gruber, P. A review of fabrication polymer scaffolds for biomedical applications using additive manufacturing techniques. Biocybern. Biomed. Eng. 2020, 40, 624–638. [Google Scholar] [CrossRef]
  68. Zhang, X.; Wu, X.; Shi, J. Additive manufacturing of zirconia ceramics: A state-of-the-art review. J. Mater. Res. Technol. 2020, 9, 9029–9048. [Google Scholar] [CrossRef]
  69. Peng, B.; Xu, H.; Song, F.; Wen, P.; Tian, Y.; Zheng, Y. Additive manufacturing of porous magnesium alloys for biodegradable orthopedic implants: Process, design, and modification. J. Mater. Sci. Technol. 2024, 182, 79–110. [Google Scholar] [CrossRef]
  70. Pontes, A.J. Chapter 7—Designing for additive manufacturing. In Design and Manufacturing of Plastics Products; Pouzada, A.S., Ed.; William Andrew Publishing: Norwich, NY, USA, 2021; pp. 249–292. [Google Scholar] [CrossRef]
  71. Lee, J.Y.; An, J.; Chua, C.K. Fundamentals and applications of 3D printing for novel materials. Appl. Mater. Today 2017, 7, 120–133. [Google Scholar] [CrossRef]
  72. Lv, X.; Ye, F.; Cheng, L.; Fan, S.; Liu, Y. Binder jetting of ceramics: Powders, binders, printing parameters, equipment, and post-treatment. Ceram. Int. 2019, 45, 12609–12624. [Google Scholar] [CrossRef]
  73. Dinh, P.; Hutchinson, B.K.; Zalavras, C.; Stevanovic, M.V. Reconstruction of osteomyelitis defects. Semin. Plast. Surg. 2009, 23, 108–118. [Google Scholar] [CrossRef]
  74. Nandi, S.K.; Mukherjee, P.; Roy, S.; Kundu, B.; De, D.K.; Basu, D. Local antibiotic delivery systems for the treatment of osteomyelitis–A review. Mater. Sci. Eng. C 2009, 29, 2478–2485. [Google Scholar] [CrossRef]
  75. Hasan, R.; Schaner, K.; Schroeder, M.; Wohlers, A.; Shreffler, J.; Schaper, C.; Subramanian, H.; Brooks, A. Extended release combination antibiotic therapy from a bone void filling putty for treatment of osteomyelitis. Pharmaceutics 2019, 11, 592. [Google Scholar] [CrossRef] [PubMed]
  76. Trombetta, R.P.; Ninomiya, M.J.; El-Atawneh, I.M.; Knapp, E.K.; de Mesy Bentley, K.L.; Dunman, P.M.; Schwarz, E.M.; Kates, S.L.; Awad, H.A. Calcium phosphate spacers for the local delivery of sitafloxacin and rifampin to treat orthopedic infections: Efficacy and proof of concept in a mouse model of single-stage revision of device-associated osteomyelitis. Pharmaceutics 2019, 11, 94. [Google Scholar] [CrossRef] [PubMed]
  77. Rybak, M.J. The pharmacokinetic and pharmacodynamic properties of vancomycin. Clin. Infect. Dis. 2006, 42 (Suppl. S1), S35–S39. [Google Scholar] [CrossRef] [PubMed]
  78. Yang, Z.; Vorpagel, E.R.; Laskin, J. Experimental and theoretical studies of the structures and interactions of vancomycin antibiotics with cell wall analogues. J. Am. Chem. Soc. 2008, 130, 13013–13022. [Google Scholar] [CrossRef]
  79. Yamaga, C.; Bostick, D.L.; Tabak, Y.P.; Liu-Ferrara, A.; Morel, D.; Yu, K. 210. Vancomycin Infusion Frequency and Intensity: Analysis of Real-World Data Generated from Automated Infusion Devices. Open Forum Infect. Dis. 2020, 7, S108. [Google Scholar] [CrossRef]
  80. Lietman, P.S.; Schaad, U.B.; McCracken Jr, G.H.; Nelson, J.D. Clinical pharmacology and efficacy of vancomycin in pediatric patients. J. Pediatr. 1980, 96, 119–126. [Google Scholar] [CrossRef]
  81. Wang, H.; Huang, L.; Wang, J.; Ni, Y.; Zhu, Z.; Gao, P.; Hu, Y.; Zhang, L.; Yang, J.; Fang, L. Population pharmacokinetic study of vancomycin in Chinese pediatric patients with hematological malignancies. Pharmacother. J. Hum. Human. Pharmacol. Drug Ther. 2020, 40, 1201–1209. [Google Scholar] [CrossRef]
  82. Mavrogenis, A.F.; Panagopoulos, G.N.; Kokkalis, Z.T.; Koulouvaris, P.; Megaloikonomos, P.D.; Igoumenou, V.; Mantas, G.; Moulakakis, K.G.; Sfyroeras, G.S.; Lazaris, A. Vascular injury in orthopedic trauma. Orthopedics 2016, 39, 249–259. [Google Scholar] [CrossRef]
  83. Amon, M.; Laschke, M.W.; Harder, Y.; Vollmar, B.; Menger, M.D. Impact of severity of local soft-tissue trauma on long-term manifestation of microcirculatory and microlymphatic dysfunctions. J. Trauma. Acute Care Surg. 2006, 61, 924–932. [Google Scholar] [CrossRef] [PubMed]
  84. Zhou, J.; Fang, T.; Wang, Y.; Dong, J. The controlled release of vancomycin in gelatin/β-TCP composite scaffolds. J. Biomed. Mater. Res. Part A 2012, 100, 2295–2301. [Google Scholar] [CrossRef] [PubMed]
  85. Anvari Kohestani, A.; Pishbin, F. 3D printing of bone scaffolds based on alginate/gelatin hydrogel ink containing bioactive glass 45S5 and ZIF-8 nanoparticles with sustained drug-release capability. Adv. Eng. Mater. 2023, 25, 2300563. [Google Scholar] [CrossRef]
  86. Choi, C.E.; Chakraborty, A.; Adzija, H.; Shamiya, Y.; Hijazi, K.; Coyle, A.; Rizkalla, A.; Holdsworth, D.W.; Paul, A. Metal organic framework-incorporated three-dimensional (3D) bio-printable hydrogels to facilitate bone repair: Preparation and in vitro bioactivity analysis. Gels 2023, 9, 923. [Google Scholar] [CrossRef]
  87. Lin, M.; Stehle, Y.; Chen, L.; Yang, M.; Zeng, K.; Wang, C.; Zhang, R.; Zhang, H.; Yang, J.; Hu, D.; et al. A 3D-printed chitosan-based pH-responsive dual functional scaffold for osteomyelitis: Synergistic antibacterial and osteogenic treatment. Carbohydr. Polym. 2025, 366, 123866. [Google Scholar] [CrossRef]
  88. Lu, L.; Yuan, S.; Wang, J.; Shen, Y.; Deng, S.; Xie, L.; Yang, Q. The formation mechanism of hydrogels. Curr. Stem Cell Res. Ther. 2018, 13, 490–496. [Google Scholar] [CrossRef]
  89. Wang, Y.; Wu, Q.; Ren, B.; Muskhelishvili, L.; Davis, K.; Wynne, R.; Rua, D.; Cao, X. Subacute pulmonary toxicity of glutaraldehyde aerosols in a human in vitro airway tissue model. Int. J. Mol. Sci. 2022, 23, 12118. [Google Scholar] [CrossRef]
  90. Zeiger, E.; Gollapudi, B.; Spencer, P. Genetic toxicity and carcinogenicity studies of glutaraldehyde—A review. Mutat. Res./Rev. Mutat. Res. 2005, 589, 136–151. [Google Scholar] [CrossRef]
  91. Hafezi, F.; Scoutaris, N.; Douroumis, D.; Boateng, J. 3D printed chitosan dressing crosslinked with genipin for potential healing of chronic wounds. Int. J. Pharm. 2019, 560, 406–415. [Google Scholar] [CrossRef]
  92. López-González, I.; Hernández-Heredia, A.B.; Rodríguez-López, M.I.; Auñón-Calles, D.; Boudifa, M.; Gabaldón, J.A.; Meseguer-Olmo, L. Evaluation of the In Vitro Antimicrobial Efficacy against Staphylococcus aureus and epidermidis of a Novel 3D-Printed Degradable Drug Delivery System Based on Polycaprolactone/Chitosan/Vancomycin—Preclinical Study. Pharmaceutics 2023, 15, 1763. [Google Scholar] [CrossRef]
  93. Zhang, Q.; Zhou, X.; Du, H.; Ha, Y.; Xu, Y.; Ao, R.; He, C. Bifunctional Hydrogel-Integrated 3D Printed Scaffold for Repairing Infected Bone Defects. ACS Biomater. Sci. Eng. 2023, 9, 4583–4596. [Google Scholar] [CrossRef]
  94. Iglesias-Mejuto, A.; Magariños, B.; Ferreira-Gonçalves, T.; Starbird-Pérez, R.; Álvarez-Lorenzo, C.; Reis, C.P.; Ardao, I.; García-González, C.A. Vancomycin-loaded methylcellulose aerogel scaffolds for advanced bone tissue engineering. Carbohydr. Polym. 2024, 324, 121536. [Google Scholar] [CrossRef]
  95. Gao, X.; Xu, Z.; Li, S.; Cheng, L.; Xu, D.; Li, L.; Chen, L.; Xu, Y.; Liu, Z.; Liu, Y. Chitosan-vancomycin hydrogel incorporated bone repair scaffold based on staggered orthogonal structure: A viable dually controlled drug delivery system. RSC Adv. 2023, 13, 3759–3765. [Google Scholar] [CrossRef]
  96. Huang, H.; Wu, Z.; Yang, Z.; Fan, X.; Bai, S.; Luo, J.; Chen, M.; Xie, X. In vitroapplication of drug-loaded hydrogel combined with 3D-printed porous scaffolds. Biomed. Mater. 2022, 17, 065019. [Google Scholar] [CrossRef]
  97. Zhang, T.; Zhou, W.; Yang, W.; Bi, J.; Li, H.; Gao, X.; Zhang, B.; Shi, G.; Li, K.; Wei, Z.; et al. Vancomycin-encapsulated hydrogel loaded microarc-oxidized 3D-printed porous Ti6Al4V implant for infected bone defects: Reconstruction, anti-infection, and osseointegration. Bioact. Mater. 2024, 42, 18–31. [Google Scholar] [CrossRef]
  98. Zhang, B.; Pei, X.; Zhou, C.; Fan, Y.; Jiang, Q.; Ronca, A.; D’Amora, U.; Chen, Y.; Li, H.; Sun, Y. The biomimetic design and 3D printing of customized mechanical properties porous Ti6Al4V scaffold for load-bearing bone reconstruction. Mater. Des. 2018, 152, 30–39. [Google Scholar] [CrossRef]
  99. Zhang, G.-F.; Liu, X.; Zhang, S.; Pan, B.; Liu, M.-L. Ciprofloxacin derivatives and their antibacterial activities. Eur. J. Med. Chem. 2018, 146, 599–612. [Google Scholar] [CrossRef] [PubMed]
  100. Brouqui, P.; Rousseau, M.C.; Stein, A.; Drancourt, M.; Raoult, D. Treatment of Pseudomonas aeruginosa-infected orthopedic prostheses with ceftazidime-ciprofloxacin antibiotic combination. Antimicrob. Agents Chemother. 1995, 39, 2423–2425. [Google Scholar] [CrossRef] [PubMed]
  101. LeBel, M. Ciprofloxacin: Chemistry, mechanism of action, resistance, antimicrobial spectrum, pharmacokinetics, clinical trials, and adverse reactions. Pharmacother. J. Hum. Pharmacol. Drug Ther. 1988, 8, 3–30. [Google Scholar] [CrossRef]
  102. Wald, E.R. Risk factors for osteomyelitis. Am. J. Med. 1985, 78, 206–212. [Google Scholar] [CrossRef]
  103. Liu, Y.; Liang, A.; Li, X.; Ma, Z.; Zhang, D. Efficacy Evaluation of Ciprofloxacin-Loaded Poly (Trimethylene Carbonate) Implants in the Treatment of Chronic Osteomyelitis. Front. Bioeng. Biotechnol. 2022, 10, 864041. [Google Scholar] [CrossRef]
  104. Baggio, D.; Ananda-Rajah, M.R. Fluoroquinolone antibiotics and adverse events. Aust. Prescr. 2021, 44, 161–164. [Google Scholar] [CrossRef]
  105. Sterne, G.M.; Richardson, M.L.; Warren, B.H. Imaging findings in two cases of fluoroquinolone-induced Achilles tendinopathy. Radiol. Case Rep. 2006, 1, 87–91. [Google Scholar] [CrossRef]
  106. Cámara-Torres, M.; Duarte, S.; Sinha, R.; Egizabal, A.; Álvarez, N.; Bastianini, M.; Sisani, M.; Scopece, P.; Scatto, M.; Bonetto, A.; et al. 3D additive manufactured composite scaffolds with antibiotic-loaded lamellar fillers for bone infection prevention and tissue regeneration. Bioact. Mater. 2021, 6, 1073–1082. [Google Scholar] [CrossRef]
  107. Moroni, L.; De Wijn, J.; Van Blitterswijk, C. Three-dimensional fiber-deposited PEOT/PBT copolymer scaffolds for tissue engineering: Influence of porosity, molecular network mesh size, and swelling in aqueous media on dynamic mechanical properties. J. Biomed. Mater. Res. Part. A Off. J. Soc. Biomater. Jpn. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 2005, 75, 957–965. [Google Scholar] [CrossRef]
  108. Zheng, H.; Zuo, B. Functional silk fibroin hydrogels: Preparation, properties and applications. J. Mater. Chem. B 2021, 9, 1238–1258. [Google Scholar] [CrossRef] [PubMed]
  109. Ng, P.; Pinho, A.R.; Gomes, M.C.; Demidov, Y.; Krakor, E.; Grume, D.; Herb, M.; Le, K.; Mano, J.; Mathur, S. Fabrication of antibacterial, osteo-inductor 3D printed aerogel-based scaffolds by incorporation of drug laden hollow mesoporous silica microparticles into the self-assembled silk fibroin biopolymer. Macromol. Biosci. 2022, 22, 2100442. [Google Scholar] [CrossRef] [PubMed]
  110. Stepanova, M.; Averianov, I.; Gofman, I.; Shevchenko, N.; Rubinstein, A.; Egorova, T.; Trulioff, A.; Nashchekina, Y.; Kudryavtsev, I.; Demyanova, E. Drug loaded 3D-printed poly (ε-caprolactone) scaffolds for local antibacterial or anti-inflammatory treatment in bone regeneration. Polymers 2023, 15, 3957. [Google Scholar] [CrossRef]
  111. Timofticiuc, I.A.; Călinescu, O.; Iftime, A.; Dragosloveanu, S.; Caruntu, A.; Scheau, A.E.; Badarau, I.A.; Didilescu, A.C.; Caruntu, C.; Scheau, C. Biomaterials Adapted to Vat Photopolymerization in 3D Printing: Characteristics and Medical Applications. J. Funct. Biomater. 2023, 15, 7. [Google Scholar] [CrossRef] [PubMed]
  112. Abdelghany, S.M.; Quinn, D.J.; Ingram, R.J.; Gilmore, B.F.; Donnelly, R.F.; Taggart, C.C.; Scott, C.J. Gentamicin-loaded nanoparticles show improved antimicrobial effects towards Pseudomonas aeruginosa infection. Int. J. Nanomed. 2012, 7, 4053–4063. [Google Scholar] [CrossRef]
  113. Witkowski, J.; Wnukiewicz, W.; Reichert, P. Polymers as Carriers of Gentamicin in Traumatology and Orthopedic Surgery—Current State of Knowledge. Polim. Med. 2016, 46, 101–104. [Google Scholar] [CrossRef] [PubMed]
  114. Hodiamont, C.J.; van den Broek, A.K.; de Vroom, S.L.; Prins, J.M.; Mathôt, R.A.A.; van Hest, R.M. Clinical Pharmacokinetics of Gentamicin in Various Patient Populations and Consequences for Optimal Dosing for Gram-Negative Infections: An Updated Review. Clin. Pharmacokinet. 2022, 61, 1075–1094. [Google Scholar] [CrossRef] [PubMed]
  115. Neut, D.; van de Belt, H.; Stokroos, I.; van Horn, J.R.; van der Mei, H.C.; Busscher, H.J. Biomaterial-associated infection of gentamicin-loaded PMMA beads in orthopaedic revision surgery. J. Antimicrob. Chemother. 2001, 47, 885–891. [Google Scholar] [CrossRef] [PubMed]
  116. Tappa, K.; Jammalamadaka, U.; Weisman, J.A.; Ballard, D.H.; Wolford, D.D.; Pascual-Garrido, C.; Wolford, L.M.; Woodard, P.K.; Mills, D.K. 3D Printing Custom Bioactive and Absorbable Surgical Screws, Pins, and Bone Plates for Localized Drug Delivery. J. Funct. Biomater. 2019, 10, 17. [Google Scholar] [CrossRef]
  117. Weisman, J.A.; Nicholson, J.C.; Tappa, K.; Jammalamadaka, U.; Wilson, C.G.; Mills, D.K. Antibiotic and chemotherapeutic enhanced three-dimensional printer filaments and constructs for biomedical applications. Int. J. Nanomed. 2015, 10, 357–370. [Google Scholar] [CrossRef]
  118. Mills, D.K.; Jammalamadaka, U.; Tappa, K.; Weisman, J. Studies on the cytocompatibility, mechanical and antimicrobial properties of 3D printed poly(methyl methacrylate) beads. Bioact. Mater. 2018, 3, 157–166. [Google Scholar] [CrossRef]
  119. Szoradi, G.T.; Feier, A.M.; Zuh, S.G.; Russu, O.M.; Pop, T.S. Polymethyl Methacrylate Bone Cement Polymerization Induced Thermal Necrosis at the Cement–Bone Interface: A Narrative Review. Appl. Sci. 2024, 14, 11651. [Google Scholar] [CrossRef]
  120. Gundapaneni, D.; Goswami, T. Thermal isotherms in PMMA and cell necrosis during total hip arthroplasty. J. Appl. Biomater. Funct. Mater. 2014, 12, 193–202. [Google Scholar] [CrossRef]
  121. Boner, V.; Kuhn, P.; Mendel, T.; Gisep, A. Temperature evaluation during PMMA screw augmentation in osteoporotic bone—An in vitro study about the risk of thermal necrosis in human femoral heads. J. Biomed. Mater. Res. Part B Appl. Biomater. Off. J. Soc. Biomater. Jpn. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 2009, 90, 842–848. [Google Scholar] [CrossRef]
  122. Niedźwiedź, M.J.; Gil, M.; Gargol, M.; Podkościelny, W.M.; Mergo, P. Determination of the optimal extrusion temperature of the PMMA optical fibers. Photonics Lett. Pol. 2019, 11, 7–9. [Google Scholar]
  123. Frunzaverde, D.; Cojocaru, V.; Ciubotariu, C.-R.; Miclosina, C.-O.; Ardeljan, D.D.; Ignat, E.F.; Marginean, G. The influence of the printing temperature and the filament color on the dimensional accuracy, tensile strength, and friction performance of FFF-printed PLA specimens. Polymers 2022, 14, 1978. [Google Scholar] [CrossRef] [PubMed]
  124. Pignatello, R.; Mangiafico, A.; Basile, L.; Ruozi, B.; Furneri, P.M. Amphiphilic ion pairs of tobramycin with lipoamino acids. Eur. J. Med. Chem. 2011, 46, 1665–1671. [Google Scholar] [CrossRef] [PubMed]
  125. Maane, M.; Xiu, F.; Bellstedt, P.; Kullak-Ublick, G.A.; Visentin, M. Characterization of ligand-induced thermal stability of the human organic cation transporter 2 (OCT2). Front. Pharmacol. 2023, 14, 1154213. [Google Scholar] [CrossRef]
  126. Biscaia, S.; Branquinho, M.V.; Alvites, R.D.; Fonseca, R.; Sousa, A.C.; Pedrosa, S.S.; Caseiro, A.R.; Guedes, F.; Patrício, T.; Viana, T. 3D printed poly (ε-caprolactone)/hydroxyapatite scaffolds for bone tissue engineering: A comparative study on a composite preparation by melt blending or solvent casting techniques and the influence of bioceramic content on scaffold properties. Int. J. Mol. Sci. 2022, 23, 2318. [Google Scholar] [CrossRef]
  127. Agrawal, A.; Hussain, C.M. 3D-Printed Hydrogel for Diverse Applications: A Review. Gels 2023, 9, 960. [Google Scholar] [CrossRef]
  128. Yu, X.; Wang, P.; Gao, J.; Fu, Y.; Wang, Q.; Chen, J.; Chen, S.; Ding, J. Wet 3D printing of biodegradable porous scaffolds to enable room-temperature deposition modeling of polymeric solutions for regeneration of articular cartilage. Biofabrication 2024, 16, 035007. [Google Scholar] [CrossRef]
  129. Cochis, A.; Bonetti, L.; Sorrentino, R.; Contessi Negrini, N.; Grassi, F.; Leigheb, M.; Rimondini, L.; Farè, S. 3D Printing of Thermo-Responsive Methylcellulose Hydrogels for Cell-Sheet Engineering. Materials 2018, 11, 579. [Google Scholar] [CrossRef]
  130. Montero, J.; Becerro, A.; Pardal-Peláez, B.; Quispe-López, N.; Blanco, J.F.; Gómez-Polo, C. Main 3D Manufacturing Techniques for Customized Bone Substitutes. A Systematic Review. Materials 2021, 14, 2524. [Google Scholar] [CrossRef]
  131. Nguyen, F.; Starosta, A.L.; Arenz, S.; Sohmen, D.; Dönhöfer, A.; Wilson, D.N. Tetracycline antibiotics and resistance mechanisms. Biol. Chem. 2014, 395, 559–575. [Google Scholar] [CrossRef]
  132. Martin, V.; Bettencourt, A.F.; Santos, C.; Fernandes, M.H.; Gomes, P.S. Unveiling the osteogenic potential of tetracyclines: A comparative study in human mesenchymal stem cells. Cells 2023, 12, 2244. [Google Scholar] [CrossRef]
  133. Pasternak, B.; Aspenberg, P. Metalloproteinases and their inhibitors—Diagnostic and therapeutic opportunities in orthopedics. Acta Orthop. 2009, 80, 693–703. [Google Scholar] [CrossRef] [PubMed]
  134. Ahangar, P.; Li, J.; Nkindi, L.S.; Mohammadrezaee, Z.; Cooke, M.E.; Martineau, P.A.; Weber, M.H.; Saade, E.; Nateghi, N.; Rosenzweig, D.H. A nanoporous 3D-printed scaffold for local antibiotic delivery. Micromachines 2023, 15, 83. [Google Scholar] [CrossRef] [PubMed]
  135. Li, Z.; Souza, L.R.d.; Litina, C.; Markaki, A.E.; Al-Tabbaa, A. Feasibility of using 3D printed polyvinyl alcohol (PVA) for creating self-healing vascular tunnels in cement system. Materials 2019, 12, 3872. [Google Scholar] [CrossRef] [PubMed]
  136. Karyappa, R.; Nagaraju, N.; Yamagishi, K.; Koh, X.Q.; Zhu, Q.; Hashimoto, M. 3D printing of polyvinyl alcohol hydrogels enabled by aqueous two-phase system. Mater. Horiz. 2024, 11, 2701–2717. [Google Scholar] [CrossRef]
  137. Duran, C.; Subbian, V.; Giovanetti, M.T.; Simkins, J.R.; Beyette Jr, F.R. Experimental desktop 3D printing using dual extrusion and water-soluble polyvinyl alcohol. Rapid Prototyp. J. 2015, 21, 528–534. [Google Scholar] [CrossRef]
  138. Zhou, X.; Zhou, G.; Junka, R.; Chang, N.; Anwar, A.; Wang, H.; Yu, X. Fabrication of polylactic acid (PLA)-based porous scaffold through the combination of traditional bio-fabrication and 3D printing technology for bone regeneration. Colloids Surf. B Biointerfaces 2021, 197, 111420. [Google Scholar] [CrossRef]
  139. Dussault, A.; Pitaru, A.A.; Weber, M.H.; Haglund, L.; Rosenzweig, D.H.; Villemure, I. Optimizing design parameters of PLA 3D-printed scaffolds for bone defect repair. Surgeries 2022, 3, 162–174. [Google Scholar] [CrossRef]
  140. Buj-Corral, I.; Bagheri, A.; Petit-Rojo, O. 3D printing of porous scaffolds with controlled porosity and pore size values. Materials 2018, 11, 1532. [Google Scholar] [CrossRef]
  141. Huber, F.; Vollmer, D.; Vinke, J.; Riedel, B.; Zankovic, S.; Schmal, H.; Seidenstuecker, M. Influence of 3D printing parameters on the mechanical stability of PCL scaffolds and the proliferation behavior of bone cells. Materials 2022, 15, 2091. [Google Scholar] [CrossRef]
  142. Temple, J.P.; Hutton, D.L.; Hung, B.P.; Huri, P.Y.; Cook, C.A.; Kondragunta, R.; Jia, X.; Grayson, W.L. Engineering anatomically shaped vascularized bone grafts with hASCs and 3D-printed PCL scaffolds. J. Biomed. Mater. Res. Part A 2014, 102, 4317–4325. [Google Scholar] [CrossRef]
  143. Chamundeswari, V.N.; Siang, L.Y.; Chuah, Y.J.; Tan, J.S.; Wang, D.-A.; Loo, S.C.J. Sustained releasing sponge-like 3D scaffolds for bone tissue engineering applications. Biomed. Mater. 2017, 13, 015019. [Google Scholar] [CrossRef]
  144. Ahad, N.A.; Rozali, F.Z.; Rosli, N.H.; Hanif, N.I.H.; Parimin, N. Oil and water absorption behavior of TPU/natural fibers composites. Solid. State Phenom. 2018, 280, 374–381. [Google Scholar] [CrossRef]
  145. Desai, S.M.; Sonawane, R.Y.; More, A.P. Thermoplastic polyurethane for three-dimensional printing applications: A review. Polym. Adv. Technol. 2023, 34, 2061–2082. [Google Scholar] [CrossRef]
  146. Wei, C.; Solanki, N.G.; Vasoya, J.M.; Shah, A.V.; Serajuddin, A.T. Development of 3D printed tablets by fused deposition modeling using polyvinyl alcohol as polymeric matrix for rapid drug release. J. Pharm. Sci. 2020, 109, 1558–1572. [Google Scholar] [CrossRef] [PubMed]
  147. Ng, N.; Haq, R.A.; Marwah, O.; Ho, F.; Adzila, S. Optimization of polyvinyl alcohol (PVA) support parameters for fused deposition modelling (FDM) by using design of experiments (DOE). Mater. Today Proc. 2022, 57, 1226–1234. [Google Scholar] [CrossRef]
  148. Poole, K. Efflux pumps as antimicrobial resistance mechanisms. Ann. Med. 2007, 39, 162–176. [Google Scholar] [CrossRef]
  149. Barza, M.; Brown, R.B.; Shanks, C.; Gamble, C.; Weinstein, L. Relation between lipophilicity and pharmacological behavior of minocycline, doxycycline, tetracycline, and oxytetracycline in dogs. Antimicrob. Agents Chemother. 1975, 8, 713–720. [Google Scholar] [CrossRef]
  150. El-Habashy, S.E.; El-Kamel, A.H.; Essawy, M.M.; Abdelfattah, E.A.; Eltaher, H.M. 3D printed bioinspired scaffolds integrating doxycycline nanoparticles: Customizable implants for in vivo osteoregeneration. Int. J. Pharm. 2021, 607, 121002. [Google Scholar] [CrossRef]
  151. Zeltz, C.; Gullberg, D. The integrin–collagen connection–a glue for tissue repair? J. Cell Sci. 2016, 129, 653–664. [Google Scholar] [CrossRef]
  152. Somaiah, C.; Kumar, A.; Mawrie, D.; Sharma, A.; Patil, S.D.; Bhattacharyya, J.; Swaminathan, R.; Jaganathan, B.G. Collagen promotes higher adhesion, survival and proliferation of mesenchymal stem cells. PLoS ONE 2015, 10, e0145068. [Google Scholar] [CrossRef]
  153. 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 Mater. Biol. Appl. 2019, 101, 15–26. [Google Scholar] [CrossRef]
  154. Hedström, S.Å. General and local antibiotic treatment of chronic osteomyelitis. Scand. J. Infect. Dis. 1969, 1, 175–180. [Google Scholar] [CrossRef]
  155. Wu, W.; Ye, C.; Zheng, Q.; Wu, G.; Cheng, Z. A therapeutic delivery system for chronic osteomyelitis via a multi-drug implant based on three-dimensional printing technology. J. Biomater. Appl. 2016, 31, 250–260. [Google Scholar] [CrossRef]
  156. Bansal, R.; Sharma, D.; Singh, R. Tuberculosis and its treatment: An overview. Mini Rev. Med. Chem. 2018, 18, 58–71. [Google Scholar] [CrossRef]
  157. Gong, D.; Ma, Y.; Yang, X.; Xie, W.; Shao, L.; Zhen, P. Study on cytotoxicity of three-dimensional printed β-tricalcium phosphate loaded poly (lactide-co-glycolide) anti-tuberculosis drug sustained release microspheres and its effect on osteogenic differentiation of bone marrow mesenchymal stem cells. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2018, 32, 1131–1136. [Google Scholar] [CrossRef]
  158. Bai, J.; Wang, H.; Gao, W.; Liang, F.; Wang, Z.; Zhou, Y.; Lan, X.; Chen, X.; Cai, N.; Huang, W.; et al. Melt electrohydrodynamic 3D printed poly (ε-caprolactone)/polyethylene glycol/roxithromycin scaffold as a potential anti-infective implant in bone repair. Int. J. Pharm. 2020, 576, 118941. [Google Scholar] [CrossRef]
  159. Li, J.; Mooney, D.J. Designing hydrogels for controlled drug delivery. Nat. Rev. Mater. 2016, 1, 16071. [Google Scholar] [CrossRef]
  160. Yoon, S.J.; Moon, Y.J.; Chun, H.J.; Yang, D.H. Doxorubicin· hydrochloride/cisplatin-loaded hydrogel/nanosized (2-hydroxypropyl)-beta-cyclodextrin local drug-delivery system for osteosarcoma treatment in vivo. Nanomaterials 2019, 9, 1652. [Google Scholar] [CrossRef]
  161. Chen, C.; Dong, J.; Chen, H.; Wang, X.; Mei, J.; Wang, L.; Xian, C.J. Preparation of adriamycin gelatin microsphere-loaded decellularized periosteum that is cytotoxic to human osteosarcoma cells. J. Cell. Physiol. 2019, 234, 10771–10781. [Google Scholar] [CrossRef]
  162. Jing, Z.; Ni, R.; Wang, J.; Lin, X.; Fan, D.; Wei, Q.; Zhang, T.; Zheng, Y.; Cai, H.; Liu, Z. Practical strategy to construct anti-osteosarcoma bone substitutes by loading cisplatin into 3D-printed titanium alloy implants using a thermosensitive hydrogel. Bioact. Mater. 2021, 6, 4542–4557. [Google Scholar] [CrossRef]
  163. Raj, P.; Lal, B.; Gadewar, M.; Singh, A.; Prashanth, G. Cisplatin and nano-particle formulations of cisplatin for cancer therapy: A Review. J. Pharm. Res. Int. 2022, 34, 34–49. [Google Scholar] [CrossRef]
  164. Cheng, X.Q.; Xu, W.; Shao, L.H.; Shen, H.Q.; Liu, H.W. Enhanced osseointegration and antimicrobial properties of 3D-Printed porous titanium alloys with copper-strontium doped calcium silicate coatings. J. Biomater. Appl. 2025, 39, 607–619. [Google Scholar] [CrossRef]
  165. Marchandet, L.; Lallier, M.; Charrier, C.; Baud’huin, M.; Ory, B.; Lamoureux, F. Mechanisms of resistance to conventional therapies for osteosarcoma. Cancers 2021, 13, 683. [Google Scholar] [CrossRef]
  166. Zhu, C.; He, M.; Sun, D.; Huang, Y.; Huang, L.; Du, M.; Wang, J.; Wang, J.; Li, Z.; Hu, B.; et al. 3D-Printed Multifunctional Polyetheretherketone Bone Scaffold for Multimodal Treatment of Osteosarcoma and Osteomyelitis. ACS Appl. Mater. Interfaces 2021, 13, 47327–47340. [Google Scholar] [CrossRef]
  167. Ahangar, P.; Akoury, E.; Ramirez Garcia Luna, A.S.; Nour, A.; Weber, M.H.; Rosenzweig, D.H. Nanoporous 3D-printed scaffolds for local doxorubicin delivery in bone metastases secondary to prostate cancer. Materials 2018, 11, 1485. [Google Scholar] [CrossRef]
  168. Ma, L.; Zhou, J.; Wu, Q.; Luo, G.; Zhao, M.; Zhong, G.; Zheng, Y.; Meng, X.; Cheng, S.; Zhang, Y. Multifunctional 3D-printed scaffolds eradiate orthotopic osteosarcoma and promote osteogenesis via microwave thermo-chemotherapy combined with immunotherapy. Biomaterials 2023, 301, 122236. [Google Scholar] [CrossRef]
  169. Zhang, J.; Zhao, S.; Zhu, M.; Zhu, Y.; Zhang, Y.; Liu, Z.; Zhang, C. 3D-printed magnetic Fe(3)O(4)/MBG/PCL composite scaffolds with multifunctionality of bone regeneration, local anticancer drug delivery and hyperthermia. J. Mater. Chem. B 2014, 2, 7583–7595. [Google Scholar] [CrossRef]
  170. Jaiswal, C.; Dey, S.; Prasad, J.; Gupta, R.; Agarwala, M.; Mandal, B.B. 3D bioprinted microfluidic based osteosarcoma-on-a chip model as a physiomimetic pre-clinical drug testing platform for anti-cancer drugs. Biomaterials 2025, 320, 123267. [Google Scholar] [CrossRef]
  171. Liu, X.; Hu, H.; Ma, J.; Wang, B. Mineralized cellulose nanofibers reinforced bioactive hydrogel remodels the osteogenic and angiogenic microenvironment for enhancing bone regeneration. Carbohydr. Polym. 2025, 357, 123480. [Google Scholar] [CrossRef]
  172. Wang, L.; Dai, Z.; Bi, J.; Chen, Y.; Wang, Z.; Sun, Z.; Ji, Z.; Wang, H.; Zhang, Y.; Wang, L.; et al. Polydopamine-functionalized calcium-deficient hydroxyapatite 3D-printed scaffold with sustained doxorubicin release for synergistic chemo-photothermal therapy of osteosarcoma and accelerated bone regeneration. Mater. Today Bio 2024, 29, 101253. [Google Scholar] [CrossRef]
  173. Yuan, Y.; Wang, W.; Wang, B.; Zhu, H.; Zhang, B.; Feng, M. Delivery of hydrophilic drug doxorubicin hydrochloride-targeted liver using apoAI as carrier. J. Drug Target. 2013, 21, 367–374. [Google Scholar] [CrossRef]
  174. Park, C.; Lee, J.H.; Jin, G.; Ngo, H.V.; Park, J.-B.; Tran, T.T.; Tran, P.H.; Lee, B.-J. Release kinetics of hydroxypropyl methylcellulose governing drug release and hydrodynamic changes of matrix tablet. Curr. Drug Deliv. 2022, 19, 520–533. [Google Scholar] [CrossRef]
  175. Heshmatnezhad, F.; Nazar, A.R.S.; Aghaei, H.; Varshosaz, J. Production of doxorubicin-loaded PCL nanoparticles through a flow-focusing microfluidic device: Encapsulation efficacy and drug release. Soft Matter 2021, 17, 10675–10682. [Google Scholar] [CrossRef] [PubMed]
  176. Wang, Y.; Tu, S.; Steffen, D.; Xiong, M. Iron complexation to histone deacetylase inhibitors SAHA and LAQ824 in PEGylated liposomes can considerably improve pharmacokinetics in rats. J. Pharm. Pharm. Sci. 2014, 17, 583–602. [Google Scholar] [CrossRef] [PubMed]
  177. Shen, H.; He, D.; Wang, S.; Ding, P.; Wang, J.; Ju, J. Preparation, characterization, and pharmacokinetics study of a novel genistein-loaded mixed micelles system. Drug Dev. Ind. Pharm. 2018, 44, 1536–1542. [Google Scholar] [CrossRef] [PubMed]
  178. Van Oss, C.; Giese, R. The hydrophilicity and hydrophobicity of clay minerals. Clays Clay Miner. 1995, 43, 474–477. [Google Scholar] [CrossRef]
  179. Huang, W.Y.; Hibino, T.; Suye, S.-i.; Fujita, S. Electrospun collagen core/poly-l-lactic acid shell nanofibers for prolonged release of hydrophilic drug. RSC Adv. 2021, 11, 5703–5711. [Google Scholar] [CrossRef]
  180. Yeo, Y.; Park, K. Control of encapsulation efficiency and initial burst in polymeric microparticle systems. Arch. Pharmacal Res. 2004, 27, 1–12. [Google Scholar] [CrossRef]
  181. Kovshova, T.; Osipova, N.; Alekseeva, A.; Malinovskaya, J.; Belov, A.; Budko, A.; Pavlova, G.; Maksimenko, O.; Nagpal, S.; Braner, S. Exploring the interplay between drug release and targeting of lipid-like polymer nanoparticles loaded with doxorubicin. Molecules 2021, 26, 831. [Google Scholar] [CrossRef]
  182. Terpos, E. Bisphosphonate anticancer activity in multiple myeloma. Anti-Cancer Agents Med. Chem. Anti-Cancer Agents 2012, 12, 123–128. [Google Scholar] [CrossRef]
  183. Burkinshaw, R.; Thorpe, H.; Coleman, R. Bisphosphonates and prevention of metastases: The AZURE study. Breast Cancer Online 2007, 10, e21. [Google Scholar] [CrossRef]
  184. Di, W.; Shuai, Y.; Bo, W.; Wei, T.; Jinpeng, H.; Qian, G.; Deng, Y. A bifunctional zoledronate sustained-release system in scaffold: Tumor therapy and bone repair. Colloids Surf. B Biointerfaces 2023, 222, 113064. [Google Scholar] [CrossRef]
  185. Ran, Z.; Wang, Y.; Li, J.; Xu, W.; Tan, J.; Cao, B.; Luo, D.; Ding, Y.; Wu, J.; Wang, L.; et al. 3D-printed biodegradable magnesium alloy scaffolds with zoledronic acid-loaded ceramic composite coating promote osteoporotic bone defect repair. Int. J. Bioprint 2023, 9, 769. [Google Scholar] [CrossRef] [PubMed]
  186. Chen, Y.; Traore, Y.L.; Walker, L.; Yang, S.; Ho, E.A. Fused deposition modeling three-dimensional printing of flexible polyurethane intravaginal rings with controlled tunable release profiles for multiple active drugs. Drug Deliv. Transl. Res. 2022, 12, 906–924. [Google Scholar] [CrossRef] [PubMed]
  187. Albu, P.; Budiul, M.; Mateescu, M.; Chiriac, V.; Vlase, G.; Vlase, T. Studies regarding the induced thermal degradation, kinetic analysis and possible interactions with various excipients of an osseointegration agent: Zoledronic acid. J. Therm. Anal. Calorim. 2017, 130, 403–411. [Google Scholar] [CrossRef]
  188. Pang, R.; Lai, M.K.; Ismail, K.I.; Yap, T.C. Characterization of the Dimensional Precision, Physical Bonding, and Tensile Performance of 3D-Printed PLA Parts with Different Printing Temperature. J. Manuf. Mater. Process. 2024, 8, 56. [Google Scholar] [CrossRef]
  189. Liu, Q.; Qiu, L.; Wang, Y.; Lv, G.; Liu, G.; Wang, S.; Lin, J. Solvent effect on molecular structure, IR spectra, thermodynamic properties and chemical stability of zoledronic acid: DFT study. J. Mol. Model. 2016, 22, 84. [Google Scholar] [CrossRef]
  190. Johnson, D.B.; Lopez, M.J.; Kelley, B. Dexamethasone. In StatPearls [Internet]; StatPearls Publishing: Orlando, FL, USA, 2023. [Google Scholar]
  191. Zhang, X.; Geven, M.A.; Wang, X.; Qin, L.; Grijpma, D.W.; Peijs, T.; Eglin, D.; Guillaume, O.; Gautrot, J.E. A drug eluting poly(trimethylene carbonate)/poly(lactic acid)-reinforced nanocomposite for the functional delivery of osteogenic molecules. Int. J. Nanomed. 2018, 13, 5701–5718. [Google Scholar] [CrossRef]
  192. Chen, C.H.; Dash, B.S.; Ting, W.C.; Chen, J.P. Bone Tissue Engineering with Adipose-Derived Stem Cells in Polycaprolactone/Graphene Oxide/Dexamethasone 3D-Printed Scaffolds. ACS Biomater. Sci. Eng. 2024, 10, 6425–6440. [Google Scholar] [CrossRef]
  193. Zhao, L.; Chen, S.; Xie, C.; Liang, Q.; Xu, D.; Chen, W.; Xiao, X. The fabrication of multifunctional sodium alginate scaffold incorporating ibuprofen-loaded modified PLLA microspheres based on cryogenic 3D printing. J. Biomater. Sci. Polym. Ed. 2022, 33, 1269–1288. [Google Scholar] [CrossRef]
  194. Wei, H.; Chen, W.; Chen, S.; Zhang, T.; Xiao, X. 3D printing of MOF-reinforced methacrylated gelatin scaffolds for bone regeneration. J. Biomater. Sci. Polym. Ed. 2024, 35, 443–462. [Google Scholar] [CrossRef]
  195. Li, Y.; Bai, Y.; Pan, J.; Wang, H.; Li, H.; Xu, X.; Fu, X.; Shi, R.; Luo, Z.; Li, Y.; et al. A hybrid 3D-printed aspirin-laden liposome composite scaffold for bone tissue engineering. J. Mater. Chem. B 2019, 7, 619–629. [Google Scholar] [CrossRef]
  196. Vu, A.A.; Bose, S. Vitamin D3 release from traditionally and additively manufactured tricalcium phosphate bone tissue engineering scaffolds. Ann. Biomed. Eng. 2020, 48, 1025–1033. [Google Scholar] [CrossRef]
  197. Bose, S.; Chaudhari, V.S.; Kushram, P. 3D printed scaffolds with quercetin and vitamin D3 nanocarriers: In vitro cellular evaluation. J. Biomed. Mater. Res. Part A 2024, 112, 2110–2123. [Google Scholar] [CrossRef]
  198. Deng, N.; Sun, J.; Li, Y.; Chen, L.; Chen, C.; Wu, Y.; Wang, Z.; Li, L. Experimental study of rhBMP-2 chitosan nano-sustained release carrier-loaded PLGA/nHA scaffolds to construct mandibular tissue-engineered bone. Arch. Oral. Biol. 2019, 102, 16–25. [Google Scholar] [CrossRef] [PubMed]
  199. Ma, R.; Su, Y.; Cao, R.; Wang, K.; Yang, P. Enhanced osteogenic activity and bone repair ability of PLGA/MBG scaffolds doped with ZIF-8 nanoparticles loaded with BMP-2. Int. J. Nanomed. 2023, 18, 5055–5072. [Google Scholar] [CrossRef] [PubMed]
  200. Huang, K.H.; Lin, Y.H.; Shie, M.Y.; Lin, C.P. Effects of bone morphogenic protein-2 loaded on the 3D-printed MesoCS scaffolds. J. Formos. Med. Assoc. 2018, 117, 879–887. [Google Scholar] [CrossRef] [PubMed]
  201. Liu, X.; Wei, Y.; Xuan, C.; Liu, L.; Lai, C.; Chai, M.; Zhang, Z.; Wang, L.; Shi, X. A Biomimetic Biphasic Osteochondral Scaffold with Layer-Specific Release of Stem Cell Differentiation Inducers for the Reconstruction of Osteochondral Defects. Adv. Healthc. Mater. 2020, 9, e2000076. [Google Scholar] [CrossRef]
  202. Cheng, W.X.; Liu, Y.Z.; Meng, X.B.; Zheng, Z.T.; Li, L.L.; Ke, L.Q.; Li, L.; Huang, C.S.; Zhu, G.Y.; Pan, H.D.; et al. PLGA/β-TCP composite scaffold incorporating cucurbitacin B promotes bone regeneration by inducing angiogenesis. J. Orthop. Transl. 2021, 31, 41–51. [Google Scholar] [CrossRef]
  203. Bhattacharjee, A.; Jo, Y.; Bose, S. In vivo and In vitro properties evaluation of curcumin loaded MgO doped 3D printed TCP scaffolds. J. Mater. Chem. B 2023, 11, 4725–4739. [Google Scholar] [CrossRef]
  204. Bose, S.; Banerjee, D.; Vu, A.A. Ginger and Garlic Extracts Enhance Osteogenesis in 3D Printed Calcium Phosphate Bone Scaffolds with Bimodal Pore Distribution. ACS Appl. Mater. Interfaces 2022, 14, 12964–12975. [Google Scholar] [CrossRef]
  205. Bose, S.; White, B.; Chaudhari, V.S. Gingerol encapsulated lipid nanoparticles on 3D-printed scaffolds for orthopedic and dental applications. Biomater. Adv. 2025, 172, 214194. [Google Scholar] [CrossRef]
  206. Hock, F.J.; Pugsley, M.K. Drug Discovery and Evaluation: Safety and Pharmacokinetic Assays; Springer: Berlin/Heidelberg, Germany, 2024. [Google Scholar]
  207. Singh, V.K.; Seed, T.M. How necessary are animal models for modern drug discovery? Expert. Opin. Drug Discov. 2021, 16, 1391–1397. [Google Scholar] [CrossRef] [PubMed]
  208. Wang, H.; Brown, P.C.; Chow, E.C.; Ewart, L.; Ferguson, S.S.; Fitzpatrick, S.; Freedman, B.S.; Guo, G.L.; Hedrich, W.; Heyward, S. 3D cell culture models: Drug pharmacokinetics, safety assessment, and regulatory consideration. Clin. Transl. Sci. 2021, 14, 1659–1680. [Google Scholar] [CrossRef] [PubMed]
  209. Foglizzo, V.; Cocco, E.; Marchiò, S. Advanced cellular models for preclinical drug testing: From 2D cultures to organ-on-a-chip technology. Cancers 2022, 14, 3692. [Google Scholar] [CrossRef] [PubMed]
  210. Badr-Eldin, S.M.; Aldawsari, H.M.; Kotta, S.; Deb, P.K.; Venugopala, K.N. Three-dimensional in vitro cell culture models for efficient drug discovery: Progress so far and future prospects. Pharmaceuticals 2022, 15, 926. [Google Scholar] [CrossRef]
  211. Li, S.; Yang, K.; Chen, X.; Zhu, X.; Zhou, H.; Li, P.; Chen, Y.; Jiang, Y.; Li, T.; Qin, X. Simultaneous 2D and 3D cell culture array for multicellular geometry, drug discovery and tumor microenvironment reconstruction. Biofabrication 2021, 13, 045013. [Google Scholar] [CrossRef]
  212. Xu, Y.; Pachnikova, G.; Wang, H.; Wu, Y.; Przybilla, D.; Schäfer, R.; Chen, Z.; Zhu, S.; Keilholz, U. IC50: An unsuitable measure for large-sized prostate cancer spheroids in drug sensitivity evaluation. Bosn. J. Basic. Med. Sci. 2022, 22, 580. [Google Scholar] [CrossRef]
  213. Abbas, Z.N.; Al-Saffar, A.Z.; Jasim, S.M.; Sulaiman, G.M. Comparative analysis between 2D and 3D colorectal cancer culture models for insights into cellular morphological and transcriptomic variations. Sci. Rep. 2023, 13, 18380. [Google Scholar] [CrossRef]
  214. Jota Baptista, C.V.; Faustino-Rocha, A.I.; Oliveira, P.A. Animal models in pharmacology: A brief history awarding the nobel prizes for physiology or medicine. Pharmacology 2021, 106, 356–368. [Google Scholar] [CrossRef]
  215. Admane, P.; Mane, S.; Vinchurkar, K. 3D Printing Technology in the Pharmaceutical Industry and Its Application in Drug Delivery in the Context of Personalized Medication. In Personalized Medicine-New Perspectives; IntechOpen: London, UK, 2024. [Google Scholar]
  216. Mihaylova, A.; Shopova, D.; Parahuleva, N.; Yaneva, A.; Bakova, D. (3D) bioprinting—Next dimension of the pharmaceutical sector. Pharmaceuticals 2024, 17, 797. [Google Scholar] [CrossRef] [PubMed]
  217. Bisht, B.; Hope, A.; Mukherjee, A.; Paul, M.K. Advances in the fabrication of scaffold and 3D printing of biomimetic bone graft. Ann. Biomed. Eng. 2021, 49, 1128–1150. [Google Scholar] [CrossRef] [PubMed]
  218. Carluccio, D.; Demir, A.; Bermingham, M.; Dargusch, M. Challenges and opportunities in the selective laser melting of biodegradable metals for load-bearing bone scaffold applications. Metall. Mater. Trans. A 2020, 51, 3311–3334. [Google Scholar] [CrossRef]
  219. Entezari, A.; Roohani-Esfahani, S.-I.; Zhang, Z.; Zreiqat, H.; Dunstan, C.R.; Li, Q. Fracture behaviors of ceramic tissue scaffolds for load bearing applications. Sci. Rep. 2016, 6, 28816. [Google Scholar] [CrossRef]
  220. Li, X.; Fan, L.; Li, J. Extrusion-based 3D printing of high internal phase emulsions stabilized by co-assembled β-cyclodextrin and chitosan. Food Hydrocoll. 2023, 134, 108036. [Google Scholar] [CrossRef]
  221. Wang, M.; Zhang, J.; Fan, L.; Li, J. Fabrication of fat-reduced water-in-oil emulsion and the application in 3D printing. Food Res. Int. 2023, 172, 113118. [Google Scholar] [CrossRef]
  222. Ghosh, S.; Yadav, A.; Rani, S.; Takkar, S.; Kulshreshtha, R.; Nandan, B.; Srivastava, R.K. 3D Printed Hierarchical Porous Poly(ε-caprolactone) Scaffolds from Pickering High Internal Phase Emulsion Templating. Langmuir 2023, 39, 1927–1946. [Google Scholar] [CrossRef]
  223. Zhang, J.; Griesbach, J.; Ganeyev, M.; Zehnder, A.K.; Zeng, P.; Schädli, G.N.; Leeuw, A.; Lai, Y.; Rubert, M.; Müller, R. Long-term mechanical loading is required for the formation of 3D bioprinted functional osteocyte bone organoids. Biofabrication 2022, 14, 035018. [Google Scholar] [CrossRef]
  224. Breathwaite, E.; Weaver, J.; Odanga, J.; Dela Pena-Ponce, M.; Lee, J.B. 3D Bioprinted Osteogenic Tissue Models for In Vitro Drug Screening. Molecules 2020, 25, 3442. [Google Scholar] [CrossRef]
  225. Ayan, B.; Heo, D.; Zhang, Z.; Dey, M.; Povilianskas, A.; Drapaca, C.; Ozbolat, I. Aspiration-assisted bioprinting for precise positioning of biologics. Sci. Adv. 2020, 6, eaaw5111. [Google Scholar] [CrossRef]
  226. Khoshnood, N.; Zamanian, A. A comprehensive review on scaffold-free bioinks for bioprinting. Bioprinting 2020, 19, e00088. [Google Scholar] [CrossRef]
  227. Friedrich, J.; Seidel, C.; Ebner, R.; Kunz-Schughart, L.A. Spheroid-based drug screen: Considerations and practical approach. Nat. Protoc. 2009, 4, 309–324. [Google Scholar] [CrossRef] [PubMed]
  228. Aguilar, I.N.; Olivos III, D.J.; Brinker, A.; Alvarez, M.B.; Smith, L.J.; Chu, T.-M.G.; Kacena, M.A.; Wagner, D.R. Scaffold-free bioprinting of mesenchymal stem cells using the Regenova printer: Spheroid characterization and osteogenic differentiation. Bioprinting 2019, 15, e00050. [Google Scholar] [CrossRef] [PubMed]
  229. Zuliani, C.C.; da Cunha, J.B.; de Souza, V.M.; de Andrade, K.C.; Moraes, Â.M.; Coimbra, I.B. Amniotic fluid MSCs for scaffold-free cartilage repair: Spheroid fusion and chondrogenic microtissue development. Future Sci. OA 2025, 11, 2476922. [Google Scholar] [CrossRef]
  230. Apostu, D.; Lucaciu, O.; Mester, A.; Oltean-Dan, D.; Baciut, M.; Baciut, G.; Bran, S.; Onisor, F.; Piciu, A.; Pasca, R.D. Systemic drugs with impact on osteoarthritis. Drug Metab. Rev. 2019, 51, 498–523. [Google Scholar] [CrossRef]
  231. Oliveira, M.B.; Neto, A.I.; Correia, C.R.; Rial-Hermida, M.I.; Alvarez-Lorenzo, C.; Mano, J.F. Superhydrophobic chips for cell spheroids high-throughput generation and drug screening. ACS Appl. Mater. Interfaces 2014, 6, 9488–9495. [Google Scholar] [CrossRef]
  232. Yan, Y.; Song, L.; Madinya, J.; Ma, T.; Li, Y. Derivation of cortical spheroids from human induced pluripotent stem cells in a suspension bioreactor. Tissue Eng. Part A 2018, 24, 418–431. [Google Scholar] [CrossRef]
  233. Han, W.; El Botty, R.; Montaudon, E.; Malaquin, L.; Deschaseaux, F.; Espagnolle, N.; Marangoni, E.; Cottu, P.; Zalcman, G.; Parrini, M.C.; et al. In vitro bone metastasis dwelling in a 3D bioengineered niche. Biomaterials 2021, 269, 120624. [Google Scholar] [CrossRef]
  234. Ji, X.; Bei, H.P.; Zhong, G.; Shao, H.; He, X.; Qian, X.; Zhang, Y.; Zhao, X. Premetastatic Niche Mimicking Bone-On-A-Chip: A Microfluidic Platform to Study Bone Metastasis in Cancer Patients. Small 2023, 19, e2207606. [Google Scholar] [CrossRef]
  235. Lu, Z.; Miao, X.; Zhang, C.; Sun, B.; Skardal, A.; Atala, A.; Ai, S.; Gong, J.; Hao, Y.; Zhao, J.; et al. An osteosarcoma-on-a-chip model for studying osteosarcoma matrix-cell interactions and drug responses. Bioact. Mater. 2024, 34, 1–16. [Google Scholar] [CrossRef]
  236. Pellegrini, E.; Desando, G.; Petretta, M.; Cellamare, A.; Cristalli, C.; Pasello, M.; Manara, M.C.; Grigolo, B.; Scotlandi, K. A 3D collagen-based bioprinted model to study osteosarcoma invasiveness and drug response. Polymers 2022, 14, 4070. [Google Scholar] [CrossRef]
  237. Adhami, M.; Dastidar, A.G.; Anjani, Q.K.; Detamornrat, U.; Tarrés, Q.; Delgado-Aguilar, M.; Acheson, J.G.; Manda, K.; Clarke, S.A.; Moreno-Castellanos, N.; et al. 3D-printing of dipyridamole/thermoplastic polyurethane materials for bone regeneration. Drug Deliv. Transl. Res. 2025, 15, 2467–2482. [Google Scholar] [CrossRef]
  238. Bharathi, R.; Harini, G.; Sankaranarayanan, A.; Shanmugavadivu, A.; Vairamani, M.; Selvamurugan, N. Nuciferine-loaded chitosan hydrogel-integrated 3D-printed polylactic acid scaffolds for bone tissue engineering: A combinatorial approach. Int. J. Biol. Macromol. 2023, 253, 127492. [Google Scholar] [CrossRef]
  239. Farto-Vaamonde, X.; Auriemma, G.; Aquino, R.P.; Concheiro, A.; Alvarez-Lorenzo, C. Post-manufacture loading of filaments and 3D printed PLA scaffolds with prednisolone and dexamethasone for tissue regeneration applications. Eur. J. Pharm. Biopharm. 2019, 141, 100–110. [Google Scholar] [CrossRef]
  240. Oryan, A.; Hassanajili, S.; Sahvieh, S. Zoledronate loaded polylactic acid/polycaprolactone/hydroxyapatite scaffold accelerates regeneration and led to enhance structural performance and functional ability of the radial bone defect in rat. Iran. J. Vet. Res. 2023, 24, 122–125. [Google Scholar] [CrossRef] [PubMed]
  241. Remy, M.T.; Upara, C.; Ding, Q.J.; Miszuk, J.M.; Sun, H.; Hong, L. MicroRNA-200c release from Gelatin-Coated 3D-Printed PCL scaffolds enhances bone regeneration. ACS Biomater. Sci. Eng. 2024, 10, 2337–2350. [Google Scholar] [CrossRef] [PubMed]
  242. Riester, O.; Laufer, S.; Deigner, H.-P. Direct 3D printed biocompatible microfluidics: Assessment of human mesenchymal stem cell differentiation and cytotoxic drug screening in a dynamic culture system. J. Nanobiotechnol. 2022, 20, 540. [Google Scholar] [CrossRef] [PubMed]
  243. Lipreri, M.V.; Totaro, M.T.; Raimondi, I.; Cortini, M.; Baldini, N.; Avnet, S. A customizable micropatterned platform for osteosarcoma spheroid generation, imaging, and drug screening. Biomater. Adv. 2025, 177, 214419. [Google Scholar] [CrossRef]
  244. Fan, S.; Tan, Y.; Yuan, X.; Liu, C.; Wu, X.; Dai, T.; Ni, S.; Wang, J.; Weng, Y.; Zhao, H. Regulation of the immune microenvironment by pioglitazone-loaded polylactic glycolic acid nanosphere composite scaffolds to promote vascularization and bone regeneration. J. Tissue Eng. 2024, 15, 20417314241231452. [Google Scholar] [CrossRef]
  245. Zhang, J.; Zhang, Y.; Chen, J.; Gong, W.; Tu, X. The Osteocyte with SB216763-Activated Canonical Wnt Signaling Constructs a Multifunctional 4D Intelligent Osteogenic Module. Biomolecules 2024, 14, 354. [Google Scholar] [CrossRef]
  246. Zha, Y.; Li, Y.; Lin, T.; Chen, J.; Zhang, S.; Wang, J. Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics 2021, 11, 397–409. [Google Scholar] [CrossRef]
  247. Tan, Y.; Fan, S.; Wu, X.; Liu, M.; Dai, T.; Liu, C.; Ni, S.; Wang, J.; Yuan, X.; Zhao, H. Fabrication of a three-dimensional printed gelatin/sodium alginate/nano-attapulgite composite polymer scaffold loaded with leonurine hydrochloride and its effects on osteogenesis and vascularization. Int. J. Biol. Macromol. 2023, 249, 126028. [Google Scholar] [CrossRef] [PubMed]
  248. Noory, P.; Farmani, A.R.; Ai, J.; Bahrami, N.; Bayat, M.; Ebrahimi-Barough, S.; Farzin, A.; Shojaie, S.; Hajmoradi, H.; Mohamadnia, A.; et al. Enhancing in vitro osteogenic differentiation of mesenchymal stem cells via sustained dexamethasone delivery in 3D-Printed hybrid scaffolds based on polycaprolactone-nanohydroxyapatite/alginate-gelatin for bone regeneration. J. Biol. Eng. 2025, 19, 48. [Google Scholar] [CrossRef] [PubMed]
  249. Gao, X.; Yang, J.; Liu, L.; Hu, Z.; Lin, R.; Tang, L.; Yu, M.; Chen, Z.; Gao, C.; Zhang, M.; et al. An electrostatic encapsulation strategy to motivate 3D-printed polyelectrolyte scaffolds for repair of osteoporotic bone defects. Bioact. Mater. 2025, 46, 1–20. [Google Scholar] [CrossRef] [PubMed]
  250. Rodríguez-González, R.; Delgado, J.; Delgado, L.M.; Pérez, R.A. Silica 3D printed scaffolds as pH stimuli-responsive drug release platform. Mater. Today Bio 2024, 28, 101187. [Google Scholar] [CrossRef]
  251. Zhang, W.; Shi, W.; Wu, S.; Kuss, M.; Jiang, X.; Untrauer, J.B.; Reid, S.P.; Duan, B. 3D printed composite scaffolds with dual small molecule delivery for mandibular bone regeneration. Biofabrication 2020, 12, 035020. [Google Scholar] [CrossRef]
  252. Wu, Y.; Woodbine, L.; Carr, A.M.; Pillai, A.R.; Nokhodchi, A.; Maniruzzaman, M. 3D Printed Calcium Phosphate Cement (CPC) Scaffolds for Anti-Cancer Drug Delivery. Pharmaceutics 2020, 12, 1077. [Google Scholar] [CrossRef]
  253. Xu, T.; Gao, S.; Yang, N.; Zhao, Q.; Zhang, Y.; Li, T.; Liu, Z.; Han, B. A personalized biomimetic dual-drug delivery system via controlled release of PTH(1-34) and simvastatin for in situ osteoporotic bone regeneration. Front. Bioeng. Biotechnol. 2024, 12, 1355019. [Google Scholar] [CrossRef]
  254. Wen, Y.T.; Dai, N.T.; Hsu, S.H. Biodegradable water-based polyurethane scaffolds with a sequential release function for cell-free cartilage tissue engineering. Acta Biomater. 2019, 88, 301–313. [Google Scholar] [CrossRef]
  255. Romero-Torrecilla, J.A.; de Anleo, M.E.-G.; Martínez-Ohárriz, C.; Ripalda-Cemboráin, P.; López-Martínez, T.; Abizanda, G.; Valdés-Fernández, J.; Prandota, J.; Muiños-López, E.; Garbayo, E. 3D-printed polycaprolactone scaffolds functionalized with poly (lactic-co-glycolic) acid microparticles enhance bone regeneration through tunable drug release. Acta Biomater. 2025, 198, 219–233. [Google Scholar] [CrossRef]
  256. 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. B Appl. Biomater. 2020, 108, 2167–2179. [Google Scholar] [CrossRef]
  257. Nichols, D.A.; Sondh, I.S.; Little, S.R.; Zunino, P.; Gottardi, R. Design and validation of an osteochondral bioreactor for the screening of treatments for osteoarthritis. Biomed. Microdevices 2018, 20, 18. [Google Scholar] [CrossRef] [PubMed]
  258. Han, X.; Sun, M.; Chen, B.; Saiding, Q.; Zhang, J.; Song, H.; Deng, L.; Wang, P.; Gong, W.; Cui, W. Lotus seedpod-inspired internal vascularized 3D printed scaffold for bone tissue repair. Bioact. Mater. 2021, 6, 1639–1652. [Google Scholar] [CrossRef] [PubMed]
  259. Mo, X.; Meng, K.; Li, Z.; Lan, S.; Ren, Z.; Fu, X.; Li, C.; Sun, T.; Xie, D.; Zhang, Z.; et al. An Integrated Microcurrent Delivery System Facilitates Human Parathyroid Hormone Delivery for Enhancing Osteoanabolic Effect. Small Methods 2025, 9, e2401144. [Google Scholar] [CrossRef] [PubMed]
  260. Pektas, H.K.; Demidov, Y.; Ahvan, A.; Abie, N.; Georgieva, V.S.; Chen, S.; Farè, S.; Brachvogel, B.; Mathur, S.; Maleki, H. MXene-Integrated Silk Fibroin-Based Self-Assembly-Driven 3D-Printed Theragenerative Scaffolds for Remotely Photothermal Anti-Osteosarcoma Ablation and Bone Regeneration. ACS Mater. Au 2023, 3, 711–726. [Google Scholar] [CrossRef]
  261. Bubpamala, T.; Promoppatum, P.; Pholpabu, P. Drug-Releasing Tannic Acid-Mediated Adhesive PEG Hydrogel for Porous Titanium Implants. ACS Omega 2024, 9, 887–895. [Google Scholar] [CrossRef]
  262. Kwon, B.J.; Seon, G.M.; Lee, M.H.; Koo, M.A.; Kim, M.S.; Kim, D.; Han, J.J.; Kim, D.; Kim, J.; Park, J.C. Locally delivered ethyl-2,5-dihydroxybenzoate using 3D printed bone implant for promotion of bone regeneration in a osteoporotic animal model. Eur. Cell Mater. 2018, 35, 1–12. [Google Scholar] [CrossRef]
  263. Fan, D.; Zhang, C.; Wang, H.; Wei, Q.; Cai, H.; Wei, F.; Bian, Z.; Liu, W.; Wang, X.; Liu, Z. Fabrication of a composite 3D-printed titanium alloy combined with controlled in situ drug release to prevent osteosarcoma recurrence. Mater. Today Bio 2023, 20, 100683. [Google Scholar] [CrossRef]
  264. Jing, Z.; Yuan, W.; Wang, J.; Ni, R.; Qin, Y.; Mao, Z.; Wei, F.; Song, C.; Zheng, Y.; Cai, H.; et al. Simvastatin/hydrogel-loaded 3D-printed titanium alloy scaffolds suppress osteosarcoma via TF/NOX2-associated ferroptosis while repairing bone defects. Bioact. Mater. 2024, 33, 223–241. [Google Scholar] [CrossRef]
  265. Bose, S.; Sarkar, N.; Majumdar, U. Micelle encapsulated curcumin and piperine-laden 3D printed calcium phosphate scaffolds enhance in vitro biological properties. Colloids Surf. B Biointerfaces 2023, 231, 113563. [Google Scholar] [CrossRef]
  266. Vu, A.A.; Kushram, P.; Bose, S. Effects of Vitamin A (Retinol) Release from Calcium Phosphate Matrices and Porous 3D Printed Scaffolds on Bone Cell Proliferation and Maturation. ACS Appl. Bio Mater. 2022, 5, 1120–1129. [Google Scholar] [CrossRef] [PubMed]
  267. Onyiriuka, A. Mineral and bone disorders secondary to chronic kidney disease (renal osteodystrophy). Sri Lanka J. Diabetes Endocrinol. Metab. 2015, 5, 91. [Google Scholar] [CrossRef]
  268. Evaggelou, E.; Lambrou, G.I. Acid-base disorders and the impact on metabolic bone disease in hemodialysis patients. J. Res. Pract. Musculoskelet. Syst. 2021, 5, 65–70. [Google Scholar] [CrossRef]
  269. Prasad, N.; Hamosh, A.; Sponseller, P. Orthopaedic Manifestations of Inborn Errors of Metabolism. J. Bone Jt. Surg. Rev. 2021, 9, e20. [Google Scholar] [CrossRef] [PubMed]
  270. Marcucci, G.; Zimran, A.; Bembi, B.; Kanis, J.; Reginster, J.-Y.; Rizzoli, R.; Cooper, C.; Brandi, M.L. Gaucher disease and bone manifestations. Calcif. Tissue Int. 2014, 95, 477–494. [Google Scholar] [CrossRef]
  271. Rosenbloom, B.E.; Weinreb, N.J. Gaucher disease: A comprehensive review. Crit. Rev. Oncog. 2013, 18, 163–175. [Google Scholar] [CrossRef]
  272. Mucci, J.M.; Rozenfeld, P. Pathogenesis of bone alterations in Gaucher disease: The role of immune system. J. Immunol. Res. 2015, 2015, 192761. [Google Scholar] [CrossRef]
  273. Soloveva, A.; Yatsyk, G.; Ponomarev, R.; Lukina, K.; Kostina, I.; Mamonov, V.; Lukina, E. Reversible and irreversible radiological signs of bone involvement in type I Gaucher disease. Russ. J. Hematol. Transfusiol. 2019, 64, 49–59. [Google Scholar] [CrossRef]
  274. Wenstrup, R.; Roca-Espiau, M.; Weinreb, N.; Bembi, B. Skeletal aspects of Gaucher disease: A review. Br. J. Radiol. 2002, 75, A2–A12. [Google Scholar] [CrossRef]
  275. Barranger, J.; O’Rourke, E. Lessons learned from the development of enzyme therapy for Gaucher disease. J. Inherit. Metab. Dis. 2001, 24, 89–96. [Google Scholar] [CrossRef]
  276. Amato, D.; Patterson, M.A. Combined miglustat and enzyme replacement therapy in two patients with type 1 Gaucher disease: Two case reports. J. Med. Case Rep. 2018, 12, 19. [Google Scholar] [CrossRef]
  277. Banerjee, D.; Ivanova, M.M.; Celik, N.; Kim, M.H.; Derman, I.D.; Limgala, R.P.; Ozbolat, I.T.; Goker-Alpan, O. Biofabrication of anin-vitrobone model for Gaucher disease. Biofabrication 2023, 15, 045023. [Google Scholar] [CrossRef] [PubMed]
  278. Barneveld, R.A.; Tegelaers, F.P.; Ginns, E.I.; Visser, P.; Laanen, E.A.; Brady, R.O.; Galjaard, H.; Barranger, J.A.; Reuser, A.J.; Tager, J.M. Monoclonal antibodies against human β-glucocerebrosidase. Eur. J. Biochem. 1983, 134, 585–589. [Google Scholar] [CrossRef] [PubMed]
  279. Ivanova, M.M.; Dao, J.; Kasaci, N.; Friedman, A.; Noll, L.; Goker-Alpan, O. Wnt signaling pathway inhibitors, sclerostin and DKK-1, correlate with pain and bone pathology in patients with Gaucher disease. Front. Endocrinol. 2022, 13, 1029130. [Google Scholar] [CrossRef] [PubMed]
  280. Duong, L.H.; Chen, P.-C. A novel solvent bonding method for creating a 3D, nonplanar, and hybrid PLA/PMMA microfluidic chip. In Proceedings of the 2018 IEEE 13th Annual International Conference on Nano/Micro Engineered and Molecular Systems (NEMS), Singapore, 1–4 April 2018; pp. 177–180. [Google Scholar]
  281. Lynh, H.D.; Pin-Chuan, C. Novel solvent bonding method for creation of a three-dimensional, non-planar, hybrid PLA/PMMA microfluidic chip. Sens. Actuators A Phys. 2018, 280, 350–358. [Google Scholar] [CrossRef]
  282. Reif, R.; Wang, M.; Joshi, S.; A’Amar, O.; Bigio, I.J. Optical method for real-time monitoring of drug concentrations facilitates the development of novel methods for drug delivery to brain tissue. J. Biomed. Opt. 2007, 12, 034036. [Google Scholar] [CrossRef]
  283. Di Prima, M.; Coburn, J.; Hwang, D.; Kelly, J.; Khairuzzaman, A.; Ricles, L. Additively manufactured medical products—The FDA perspective. 3D Print. Med. 2016, 2, 1. [Google Scholar] [CrossRef]
  284. Beitler, B.G.; Abraham, P.F.; Glennon, A.R.; Tommasini, S.M.; Lattanza, L.L.; Morris, J.M.; Wiznia, D.H. Interpretation of regulatory factors for 3D printing at hospitals and medical centers, or at the point of care. 3D Print. Med. 2022, 8, 7. [Google Scholar] [CrossRef]
  285. Food and Drug Administration, USA, Highlights of Prescribing Information—Spritam. 2015. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/207958s000lbl.pdf (accessed on 15 October 2025).
  286. Velagaleti, R.; Burns, P.K.; Gill, M.; Prothro, J. Impact of current good manufacturing practices and emission regulations and guidances on the discharge of pharmaceutical chemicals into the environment from manufacturing, use, and disposal. Environ. Health Perspect. 2002, 110, 213–220. [Google Scholar] [CrossRef]
  287. Parramon-Teixido, C.J.; Rodríguez-Pombo, L.; Basit, A.W.; Worsley, A.; Cañete-Ramírez, C.; Alvarez-Lorenzo, C.; Cabañas-Poy, M.J.; Goyanes, A. A framework for conducting clinical trials involving 3D printing of medicines at the point-of-care. Drug Deliv. Transl. Res. 2025, 15, 3078–3097. [Google Scholar] [CrossRef]
  288. European Medicines Agency. Quality Innovation Group. 2024. Available online: https://www.ema.europa.eu/en/committees/working-parties-other-groups/chmp-working-parties-other-groups/quality-innovation-group (accessed on 15 October 2025).
  289. Alexander, A.E.; Wake, N.; Chepelev, L.; Brantner, P.; Ryan, J.; Wang, K.C. A guideline for 3D printing terminology in biomedical research utilizing ISO/ASTM standards. 3D Print. Med. 2021, 7, 8. [Google Scholar] [CrossRef] [PubMed]
  290. Török, G.; Gombocz, P.; Bognár, E.; Nagy, P.; Dinya, E.; Kispélyi, B.; Hermann, P. Effects of disinfection and sterilization on the dimensional changes and mechanical properties of 3D printed surgical guides for implant therapy—Pilot study. BMC Oral. Health 2020, 20, 19. [Google Scholar] [CrossRef] [PubMed]
  291. Rynio, P.; Galant, K.; Wójcik, Ł.; Grygorcewicz, B.; Kazimierczak, A.; Falkowski, A.; Gutowski, P.; Dołęgowska, B.; Kawa, M. Effects of Sterilization Methods on Different 3D Printable Materials for Templates of Physician-Modified Aortic Stent Grafts Used in Vascular Surgery-A Preliminary Study. Int. J. Mol. Sci. 2022, 23, 3539. [Google Scholar] [CrossRef] [PubMed]
  292. Pérez Davila, S.; González Rodríguez, L.; Chiussi, S.; Serra, J.; González, P. How to Sterilize Polylactic Acid Based Medical Devices? Polymers 2021, 13, 2115. [Google Scholar] [CrossRef]
  293. Abdella, S.; Youssef, S.H.; Afinjuomo, F.; Song, Y.; Fouladian, P.; Upton, R.; Garg, S. 3D Printing of Thermo-Sensitive Drugs. Pharmaceutics 2021, 13, 1524. [Google Scholar] [CrossRef]
  294. Peng, H.; Han, B.; Tong, T.; Jin, X.; Peng, Y.; Guo, M.; Li, B.; Ding, J.; Kong, Q.; Wang, Q. 3D printing processes in precise drug delivery for personalized medicine. Biofabrication 2024, 16, 032001. [Google Scholar] [CrossRef]
  295. Dumpa, N.; Butreddy, A.; Wang, H.; Komanduri, N.; Bandari, S.; Repka, M.A. 3D printing in personalized drug delivery: An overview of hot-melt extrusion-based fused deposition modeling. Int. J. Pharm. 2021, 600, 120501. [Google Scholar] [CrossRef]
  296. Rengarajan, V.; Clyde, A.; Pontsler, J.; Valiente, J.; Peel, A.; Huang, Y. Assessing Leachable Cytotoxicity of 3D-Printed Polymers and Facile Detoxification Methods. 3D Print. Addit. Manuf. 2023, 10, 1110–1121. [Google Scholar] [CrossRef]
  297. Gharehdaghi, N.; Nokhbatolfoghahaei, H.; Khojasteh, A. 4D printing of smart scaffolds for bone regeneration: A systematic review. Biomed. Mater. 2024, 20, 012003. [Google Scholar] [CrossRef]
  298. Serrano, D.R.; Luciano, F.C.; Anaya, B.J.; Ongoren, B.; Kara, A.; Molina, G.; Ramirez, B.I.; Sánchez-Guirales, S.A.; Simon, J.A.; Tomietto, G.; et al. Artificial Intelligence (AI) Applications in Drug Discovery and Drug Delivery: Revolutionizing Personalized Medicine. Pharmaceutics 2024, 16, 1328. [Google Scholar] [CrossRef]
  299. Pettersson, A.B.V.; Ballardini, R.M.; Mimler, M.; Li, P.; Salmi, M.; Minssen, T.; Gibson, I.; Mäkitie, A. Core Legal Challenges for Medical 3D Printing in the EU. Healthcare 2024, 12, 1114. [Google Scholar] [CrossRef]
  300. Annaratone, L.; De Palma, G.; Bonizzi, G.; Sapino, A.; Botti, G.; Berrino, E.; Mannelli, C.; Arcella, P.; Di Martino, S.; Steffan, A.; et al. Basic principles of biobanking: From biological samples to precision medicine for patients. Virchows Arch. 2021, 479, 233–246. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the four major types of additive manufacturing (AM) technologies commonly used in pharmaceutical applications in bone disease modeling: Material Extrusion (ME), Vat Photopolymerization (VPP), Powder Bed Fusion (PBF), and Binder Jetting (BJ). Each technology is illustrated with its respective solidification and material deposition mechanisms. Also illustrated are hybrid/emerging AM systems, representing combinations of multiple printing modalities and advanced fabrication approaches. Created in BioRender. Timofticiuc, I. (2025) https://BioRender.com/5lvi4qg. Date of last access–5 September 2025.
Figure 1. Schematic representation of the four major types of additive manufacturing (AM) technologies commonly used in pharmaceutical applications in bone disease modeling: Material Extrusion (ME), Vat Photopolymerization (VPP), Powder Bed Fusion (PBF), and Binder Jetting (BJ). Each technology is illustrated with its respective solidification and material deposition mechanisms. Also illustrated are hybrid/emerging AM systems, representing combinations of multiple printing modalities and advanced fabrication approaches. Created in BioRender. Timofticiuc, I. (2025) https://BioRender.com/5lvi4qg. Date of last access–5 September 2025.
Pharmaceutics 17 01372 g001
Figure 2. Diagram depicting evaluation pathways of 3D-printed drug-loaded constructs. Each construct produced by a different additive manufacturing technology is examined in an in vitro/in vivo setting for properties such as biocompatibility, pharmacokinetics, and therapeutic efficacy. These constructs can allow a sustained drug release upon implantation and exhibit good compatibility with bone tissue for localized and effective treatment of diseased bone. Created in BioRender. Timofticiuc, I. (2025). https://BioRender.com/pacx9kq. Date of last access–5 September 2025.
Figure 2. Diagram depicting evaluation pathways of 3D-printed drug-loaded constructs. Each construct produced by a different additive manufacturing technology is examined in an in vitro/in vivo setting for properties such as biocompatibility, pharmacokinetics, and therapeutic efficacy. These constructs can allow a sustained drug release upon implantation and exhibit good compatibility with bone tissue for localized and effective treatment of diseased bone. Created in BioRender. Timofticiuc, I. (2025). https://BioRender.com/pacx9kq. Date of last access–5 September 2025.
Pharmaceutics 17 01372 g002
Figure 3. Illustration of drug screening workflow for bone-related diseases such as osteoarthritis, osteoporosis, osteomyelitis, and osteosarcoma. Diseased cells are extracted from patients and 3D bioprinted alongside biocompatible materials, resulting in platforms closely mimicking the bone microenvironment. These customized models are then used to test and modulate responses to various therapeutic candidates, enabling high-throughput, physiologically relevant, and patient-specific drug evaluation. Created in BioRender. Timofticiuc, I. (2025) https://BioRender.com/bwgn10n. Date of last access–5 September 2025.
Figure 3. Illustration of drug screening workflow for bone-related diseases such as osteoarthritis, osteoporosis, osteomyelitis, and osteosarcoma. Diseased cells are extracted from patients and 3D bioprinted alongside biocompatible materials, resulting in platforms closely mimicking the bone microenvironment. These customized models are then used to test and modulate responses to various therapeutic candidates, enabling high-throughput, physiologically relevant, and patient-specific drug evaluation. Created in BioRender. Timofticiuc, I. (2025) https://BioRender.com/bwgn10n. Date of last access–5 September 2025.
Pharmaceutics 17 01372 g003
Figure 4. Current landscape, challenges, and future roadmap for 3D-printed bone therapeutics. Created in BioRender. Timofticiuc, I. (2025) https://BioRender.com/7vq75i5. Date of last access–15 October 2025.
Figure 4. Current landscape, challenges, and future roadmap for 3D-printed bone therapeutics. Created in BioRender. Timofticiuc, I. (2025) https://BioRender.com/7vq75i5. Date of last access–15 October 2025.
Pharmaceutics 17 01372 g004
Table 1. This table highlights the main results of research conducted on drug delivery and screening systems using 3D printing technologies. The table is categorized by the main technologies, and for each one, the materials from which the scaffold was made, the drug released, and a summary of the properties of the entire scaffold (both drug and material properties) are presented. It is also mentioned whether the study is performed in vitro or in vivo (no in vivo studies on human patients were found).
Table 1. This table highlights the main results of research conducted on drug delivery and screening systems using 3D printing technologies. The table is categorized by the main technologies, and for each one, the materials from which the scaffold was made, the drug released, and a summary of the properties of the entire scaffold (both drug and material properties) are presented. It is also mentioned whether the study is performed in vitro or in vivo (no in vivo studies on human patients were found).
Broad 3D Printing TechnologySpecific 3D Printing TechnologyDrugsScaffold Material3D-Printer Commercial NameDiseases Targeted + Delivery/ScreeningBiological and Mechanical PropertiesRelease ProfileIn VitroIn VivoReferences
Material ExtrusionFDM (Fused Deposition Modeling)Dipyridamole (DIP)Thermoplastic polyurethane (TPU)Flashforge
Creator Pro (Flashforge, China)
-Bone defects
-Drug delivery system
-Shore hardness–80A
-Moderate flexibility
-Resistance to deformation
-Non-degradable, good biocompatibility
-Osseointegration
-High cell viability and proliferation at DIP 10% w/w
-High opacity
-Compression modulus of 10 MPa
-Slow, sustained over 30 days
-10% DIP, the release was ~90% (2888 µg), vs. 5% DIP, where the release was ~3% (58 µg)
-No burst
MC3T3-E1 (murine pre-osteoblasts)Not tested[237]
TetracyclineTPU + PVA porous filaments (PORO-LAY LAY-FOMM 60; LAY-FELT)Flashforge
Creator Pro (Flashforge, China)
-Bone defects
-Drug delivery system
-High porosity
-Release and absorption properties
-High cell viability
Not specifiedStaphylococcus aureus (cat# 470179-208), Pseudomonas fragi (cat# 470179-090); human ACL fibroblasts (from three patients)Not tested[134]
Nuciferine (NF)PLA/CS-NF porous scaffold (PLA with chitosan-nuciferine)Ultimaker 3-Bone defects
-Drug delivery system
-Low molecular weight
-75–85% deacetylation
-Average pore size: 100–325 µm
-Osteoblast differentiation
-Burst day 1, then sustained to 21 days;
-68.5–71.8% cumulative by day 21 (dose-dependent 60–100 μM)
Mouse MSCs (C3H10T1/2)Not tested[238]
Prednisolone; DexamethasonePLA (Polylactic Acid)Regemat3D (Spain)-Bone defects
-Drug delivery system
-Osteoinductivity-Loaded filament scaffolds: <10% in first 2 weeks (sustained)
-Post-print loaded: ~50% burst in first 6 h
Murine fibroblasts (CCL-163, ATCC); human bone marrow MSCs (hMSC; ATCC PCS-500-012)Not tested[239]
MinocyclinePLA scaffold coated with type-I collagen +
citrate-hydroxyapatite nanoparticles (HA NPs)
BQ Hephestos i3 (Prusa-style)-Osteomyelitis
-Drug delivery system
-Compressive strength: ≈13 MPa
-High Young’s modulus values
-Low plasticity
-Cell adhesion
-Burst within first hour, then gradual 4–24 hStaphylococcus aureus; human bone-marrow-derived stromal cells (hMSCs)Not tested[153]
Gentamicin; Tobramycin; NitrofurantoinPMMA bone cement (Orthoset 3) or PLA pellets, silicone-oil coatedMakerBot Replicator (first gen)-Osteomyelitis
-Drug delivery system
Not mentionedNot specifiedEscherichia coli and Staphylococcus aureusNot tested[118]
ZoledronatePLA/PCL + HA NPs (indirect: printed + freeze-dried)ZMorph (indirect printing + freeze-drying)-Bone defect
-Drug delivery system
-Pore diameter ≈ 141.01 ± 48.25 µm
-Wall thickness ≈ 27.51 ± 2.94 µm
-Porosity ≈ 69–71%
-Compressive modulus ≈ 0.6339 ± 0.03995 MPa
-Weight loss ≈ 2 ± 2.5% at 35 days;
-Slow degradation
-Structural stability
Not specifiedNot testedForty adult males (Wistar rats)[240]
No drug (Plasmid DNA encoding microRNA-200c)PCL scaffold coated with collagen/gelatin sponges Regemat 3D + Facilan PCL 100-Bone defect
-Drug delivery system
-Pore size 450 µm;
-Porosity 55.75%;
-Gelatin coating thickness ≈ 17.52 µm;
-Basic pH gelatin slowed the release and improved regeneration
-Collagen/acidic gelatin: rapid over 72 h
-Basic gelatin: slowed, sustained over 72 h
-PCL + polymer coats mimic sponge controls
-Burst vs. sustained, depending on polymer
Human preosteoblasts, embryonic palatal mesenchymal (HEPM; ATCC)12-week-old male Sprague–Dawley rats (N per group = 3–7)[241]
Any drugPLA/PMMA/PC microfluidic chipsUltimaker 3-Osteosarcoma model;
-Drug-screening platform
Not mentionedNo specific drug testedSaOS-2 osteoblasts; human MSCs (hMSCs)Not mentioned[242]
DoxorubicinPLACreality Ender 3 Pro-Osteosarcoma model
-Drug-screening platform
-The surface microstructure allowed the optimal development of spheroids and their immobilizationNot specifiedOsteosarcoma lines 143B, MG63 (ATCC)Not tested[243]
3D bioprinting Icariin; Purmorphamine; PD98059; U0126Scaffold-free-spheroids
(bone marrow-derived mesenchymal stem cells (BM-MSC))
Regenova® (Cyfuse)-Osteoporosis model
-Drug-screening platform
-Icariin and Purmorphamine are osteoinductive drugs
-Osteogenesis promoters
-PD98059; U0126 are osteogenesis inhibitors
Not specifiedBone marrow MSCs at passage 4 (LifeNet Health)Not tested[224]
VancomycinMethylcellulose + nanohydroxyapatite (aerogel ink)Cellink BIO X (BIOX)-Osteomyelitis
-Drug delivery system
-Bone repair and antimicrobial activity
-Optimal porosity
-Textural stability after 7 months
-Burst first 8 h, then sustained to 72 hMouse embryo fibroblasts (NIH/3T3); MC3T3-E1 pre-osteoblastsArtemia salina eggs; fresh fertilized hens’ eggs; tests in triplicate[94]
Pioglitazone (PIO)nano-attapulgite/PVA/gelatinNot specified-Bone defect
-Drug delivery system
-High viscosity
-Osteogenic differentiation
-PIO clear absorbance: 238 nm
-Minimal cytotoxicity
-PLGA nanospheres reduced burst; sustained to day 9 (with PNs), total ~32.5% ± 1.5% on day 9
-Without PNs ~41% ± 2.5% at day 5
RAW 264.7 (mouse macrophage); mouse BMSCs (CRL-12424, ATCC); HUVECsSprague–Dawley rats, 3-month-old (N = 30)[244]
SB216763PCLBioprinter DOME-PCI3D-Osteoporosis model
-Drug-screening platform
-Induces angiogenesis
-Osteogenic differentiation (Wnt signaling)
-High cell survival rate (91% after 7 days)
Not specifiedHUVECs; murine ST2 stromal cell lineNot tested[245]
Growth factor (VEGF)PCL modified with amine groups (CP05)3D printer (Allcct, China)-Bone defect
-Drug-screening platform
-PCL molecular weight: 50 kDa
-Printing temperature: 180 °C
-PCL scaffold was optimal for the porous structure of the bone, in which new vessels (due to VEGF) started to form
-Controllable release (qualitative)Rat BMSCs (rBMSCs)Male Sprague–Dawley rats (N = 40)[246]
Leonurine hydrochlorideGelatin/alginate + nano-attapulgiteRegenHU 3D Discovery (microvalve)-Bone defects with vascular impairment
-Drug delivery system
-Good biocompatibility
-Bone mineralization
-Increased blood vessel formation
-Tube formation and cellular migration (key elements for angiogenesis)
-Explosive within 72 h, then stabilized
-~18% over 17 days (nanostructured scaffold)
BMSCs; HUVECsMale Wistar rats, 8–10 weeks (N = 50).[247]
Dexamethasone (DEX); Amphotericin B; GentamicinPCL + nano-HA + alginate/gelatin hydrogelTwo-nozzle pneumatic printer-Osteomyelitis
-Drug delivery system
-PCL-nHA high compressive modulus: 46.37 ± 2.58 MPa
-PCL-nHA low compressive strength 4.51 ± 0.47 MPa
-Dexamethasone microparticles (MPs-DEX) -spherical: 0.24–5.58 µm
-Negative zeta potential of the drug microparticles
-Free MPs-DEX: 55.8% day-1 burst, ~90.4% by day 30.
-MPs-DEX entrapped: ~7.0% day-1 then sustained to ~55.8% by day 30
Human endometrial MSCs (hEnMSCs).Not tested[248]
Doxycycline (in DX/HAp/PCL NPs)Gelatin + PVA + hyaluronic acid; integrated with DX/HAp/PCL NPsRobota (dual extrusion)-Osteomyelitis
-Drug delivery system
-Osteoconduction;
-Bioresorption and immune tolerance;
-Pore sizes (90.4 ± 3.9 and 196.6 ± 38.8 µm);
-Wet-state scaffolds: 100% by 15 days
-NPs alone: slower (to 24–28 days)
-Biphasic 24-day profiles with method-dependent differences
Not testedNew Zealand white rabbits (N = 30)[150]
Growth factor (BMP-2)PCL + mesoporous calcium silicate NPsBioScaffolder 3.1 (GeSiM)-Orofacial bone repair
-Drug delivery system
-BMP-2 did not affect the mechanical integrity of the PCL scaffold
-Porosity: 65%
-Pore sizes: 542 ± 13 µm
-Good cell viability and adhesion
-Burst day 1
-Sustained, day 14 amount ~4× day 1
Human Wharton’s Jelly MSCs (WJMSCs)Not tested[200]
BMP-2 (in ZIF-8 NPs)PLGA + MBG + ZIF-8EnvisionTEC FDM bioprinter-Bone defects
-Drug delivery system
-Compressive strength: ≈2.9 MPa
-Increased mechanical strength due to the addition of ZIF-8
-Osteoinductive activity
-Slow;
-~40% by 72 h, ~57% by 174 h, continuing thereafter
MC3T3-E1Sprague–Dawley rats (N = 12).[199]
Salvianolic acid BAlginate/ε-polylysineBioScaffolder 3.1 (GeSiM)-Bone defects
-Drug delivery system
-Angiogenesis promoter
-Suppressed osteoclast activity
-+146% bone mass
-~60% cumulative by day 80C3H10 cellsFemale Sprague–Dawley rats, 10 weeks (N = 32)[249]
Ibuprofen (IBU)GelMA + ZIF-8 nanoparticlesNot specified-Bone defects
-Drug delivery system
Not mentionedNot specifiedMC3T3-E1Not tested[194]
Model protein (Cytochrome C) Sol–gel Ca-doped silica (TEOS/CaCl2 inks)Cellink BIO X (mechanical extrusion)-Bone regeneration
-Drug delivery system
-Calcium increased the pore size (from 1.66 nm to 9.10 nm)
-Brittle behavior at 120 N
-Adsorption of Cytochrome C of 84% after 5 days
-pH-controlled release
-By increasing the calcium concentration, larger drug molecules can be released
-pH-dependent
-No release at pH 7.5
-Initial burst (24 h) at pH ≤ 5.5, then fast to 72 h
-Continued up to 28 d (21 days at pH 5.5).
Rat MSCs (rMSCs)Not tested[250]
Resveratrol (RVS); Strontium ranelate (SrRn)PCL/hydrogelNot stated-Craniomaxillofacial bone defects
-Drug delivery system
-Mandibular bone formation after 8 weeks in rats’ mandibular defects
-Enhanced angiogenesis
-Osteoclast inhibition
SrRn: >70% day 1 burst, then sustained ≥21 days
-RVS: <30% over 21 days
-Scaffolds are structurally stable over 21 days.
Mouse MSCs; human osteoclasts (from PB monocytes); HUVECsSprague–Dawley rats: bone defect only (N = 5), scaffolds only (N = 6), scaffolds + RVS + SrRn (N = 6)[251]
5-FluorouracilCalcium phosphate cement (CPC)Bioprinter + Innotere-Osteosarcoma
-Drug delivery system
-Poor solubility of the anticancer drug
-Shape fidelity and increased ultimate tensile strength
-Complete release within 2 h for all coated scaffoldsHeLa; HEK293T cell linesNot tested[252]
Parathormone (1-34)(PTH1-34); SimvastatinGelMA + PLASunp Biomaker Pro (Sunp Biotech)-Osteoporosis
-Drug delivery system
-Mechanical strength similar to natural bone
-Stable biodegradation
-Porosity rates were around 50%
-Compressive modulus of GelMA-PLA: ≈40 MPa
-Water absorption of GelMA-PLA: ≈255%
-Simvastatin: 6.29% initial, then gradual to day 19
-PTH1-34 minimal initial, cumulative peak ~day 40 (PLGA microspheres mediated)
MC3T3-E1.12-week-old female SD rats, sham (N = 3) and OVX (N = 21)[253]
Water-based extrusion printing(general drug-releasing)PCL HIPE Pickering emulsions (PCL + PVA dispersed phase)Not specified-Osteosarcoma model
-Drug screening platform
-Good cellular adhesion
-Optimal pore morphology that resembles the natural bone
Not specifiedMG63 osteoblastsNot tested[222]
SDF-1; Y27632PU (Polyurethane) dispersion + collagenNot specified-Osteoarthritis
-Drug delivery system
-Promotion of cartilage proliferation
-Cell free scaffold
-Stromal cell-derived factor attracted mesenchymal stem cells from the surroundings, which helped in cartilage development
-Y27632 burst at 12 h increases with loading (12–30%)
-Then, ~0.10–0.19%/h
-Scaffold releases reach effective ~10 µM by 48–62 h
Human bone marrow MSCs (hMSCs)Adult male New Zealand white rabbits; independent triplicate runs[254]
Melt-extrusion Ciprofloxacin (CFX); Gentamicincopolymer PEOT/PBT (poly(ethylene oxide terephthalate)/poly(butylene terephthalate))Not specified-Osteomyelitis
-Drug delivery system
-Interconnected pore network
-High porosity
-Load-bearing mechanics
-CFX/MgAl: sustained ~1 month
-GTM/ZrP: burst <24 h, then slow
-Kinetics: Stage I (modified Freundlich), Stage II diffusion-limited; incomplete total release due to polymer diffusion limits
Staphylococcus epidermidis; Pseudomonas aeruginosa; human MSCs (hMSCs)Not tested[106]
Micro-extrusion CiprofloxacinMethacrylated silk fibroin (SF-MA) + HMSC-MA silica capsules (polymer–silica aerogel)Micro-extrusion system (name not specified)-Osteomyelitis
-Drug delivery system
-Large bacteriostatic rings
-Osteoinductive
-Osteogenic
-Burst first 4 h; ~35% by 72 h (most), up to ~50% with higher HMSC content due to high loading (95%) and mesoporesStaphylococcus aureus (ATCC 29213); Escherichia coli (K-12 DH5α); MC3T3-E1; human MSCsNot tested[109]
Melt Electrowriting (MEW) + FDM (hybrid)—(BMP-2 in PLGA MicroPs as cargo)PCL mesh + PLGA microparticles (InductOs BMP-2 formulation)MEW (outer) + FDM (core)-Critical-sized bone defects
-Drug delivery system
-PLGA allowed the controlled release of rhBMP-2
-Limited ectopic bone growth
-15-day study
-Most rhBMP-2 released within 24 h
-High-dose MPs ~73% vs. low-dose ~69%;
-Amidation did not alter MPs’ kinetics
Human periosteum-derived MSCs (hPMSCs)Female Sprague–Dawley rats, 8–12 weeks; groups ≥6 animals; randomized double-blind; doses: 0.2–1.2 µg rhBMP-2[255]
Melt Electrohydrodynamic PrintingRoxithromycinPCL/PEGCustom system-Osteomyelitis
-Drug delivery system
-The addition of PEG increased the hydrophilicity of the scaffold
-Increased wettability
-Increased cellular proliferation
->70% within 6 h (burst), then slow to 24 h
-PEG increased early release (0–15% PEG: 72–88% at 6 h; 76–91% at 24 h)
Escherichia coli; Staphylococcus aureus; MG63 cellsNot tested[158]
Modified/Hybrid/Custom extrusionAspirinPCLCustom system-Bone defect
-Drug delivery system
Not mentioned-Sustained releaseHuman MSCs (hMSCs)Not tested[256]
Vat PhotopolymerizationSLA (Stereolithography)CisplatinDS-3000 biocompatible resin (DWS)DWS 028J+, Italy-Osteosarcoma model
-Drug screening model
-Osteosarcoma cells were taken from a patient with residual cancerous cells (HBCx-66)
-Pore sizes: 100–500 µm
-Young’s modulus: ≈ 2.45 GPa
-Initial burst, then sustained >15 days
-Release coupled to PLGA-PEG-PLGA hydrogel degradation within 3D-printed implant
Bone marrow MSCs; breast cancer PDX cells; osteosarcoma (143B, HOS, MG63)5-week-old female BALB/c nude mice; 5 groups, N = 5/group[233]
Bioreactor to test candidate drugs Somos WaterShed XC11122 resin; agarose porous medium; GelMA hydrogel scaffold3D Systems Viper si2-Osteoarthritis model
-Drug screening platform
-Fluid velocity inside the bioreactor is tunable to test the drug’s elution
-Hydraulic resistance
Not specifiedNot testedNot tested[257]
DLP (Digital Light Processing)VancomycinGelMA with Zn-MOFCELLINK Lumen XTM DLP bioprinter-Osteomyelitis
-Drug delivery system
-Compressive modulus 52.14 ± 19.42 kPa
-If ZIF-8 nanoparticles are added, the compressive modulus increases to 128.13 ± 19.46 kPa
-Enhanced cell adhesion, migration, and growth
-Sustained over 48 h
-GelMA ~93% and GelMA/ZIF-8 ~88% cumulative
Escherichia coli DH5α; human adipose-derived stem cells (hASCs)Not tested[86]
Deferoxamine (DPO) liposomesTCP (tricalcium phosphate) + GelMAAUTOCERA-M (Beijing, China)-Bone defect
-Drug delivery system
-GelMA microspheres diameter: 200 ± 30 µm
-Porosity 64 ± 5%
-Polydispersity index 0.19 ± 0.03
-Bioceramic compressive strength: 2.2 ± 0.2 MPa
-Suitable for non-load-bearing applications
-Pure DFO liposomes: 67.6% in 6 h
-Scaffold-loaded DFO: ~36% at 6 h; ~69% by 7 days
-Total in vitro 48 h for pure liposomes
Rat MC3T3-E1Rats divided into control, β-TCP, and TGL groups (N = 10 each; total N = 30).[258]
High-res PhotopolymerizationHuman parathyroid hormone (1-34 PTH)XS-BIO resin; silver-coated syringe tip & steel counter-electrodes (electrodes as metals)Miro P213 (Xingsheng)-Osteoporosis
-Drug delivery system
-No fracture in the microchannels at compressive loads of 40 N
-No deformation for 20 s at forces of 20 N
Not specifiedHUVECsFemale Sprague–Dawley rats (N = 30); OVX (N = 25) and sham (N = 5)[259]
Photo-crosslinking-assisted printingSorafenibMethacrylated silk fibroin (SF-MA) + cetyltrimethylammonium bromide (CTAB); Ti3C2Tₓ MXenePhoto-crosslinking SF-MA gel setup-Osteosarcoma model
-Drug screening platform
-Controlled pore sizes
-MXene increased the photothermal conversion
-Laser irradiation was used to increase the scaffolds’ temperature and to favor the release of sorafenib
-pH- and laser-responsive
-At pH 7: 76% at 5 h; pH 4.3: 18% at 5 h
-Acidic + laser: 35% at 5 h (vs 18% without)
MC3T3-E1; MG-63Not tested[260]
Powder Bed Fusion (PBF)SLS (Selective Laser Sintering)ZoledronatePLLA + mesoporous silicon (SBA15NH2)Home-made SLS system-Osteosarcoma
-Drug delivery system
-In the acidic pH, the release is approximately two times higher, a key aspect for localized treatment of osteosarcoma or osteoporosis-Initial burst phase release
-Fast first 24 h, then slow
-Acidic pH: 16.7% (24 h) + 21.7% later
-Neutral pH: 9.6% (24 h) + 11.7% later
MG-63 (osteosarcoma); RAW264.7 (mouse macrophage/osteoclast precursor)Not tested[184]
SLM (Selective Laser Melting)Doxorubicin;
indoleamine 2,3-dioxygenase inhibitor
Ti6Al4V + Zn(NO3)2·6H2O; Metal-organic-framework (MW) compositeConcept Laser M2-Osteosarcoma
-Drug delivery system
-High biocompatibility
-Corrosion resistance
-Bone-like elastic modulus
-Microwave thermal sensitivity
-Acidic pH 5.5 + MW: 61.6% at 24 h; acidic no MW: 39.5%
-Neutral pH 7.4: 27.2% (no MW) or 33.9% (with MW)
Murine OS K7M2; rBMSCsBALB/c mice (male, 4–5 weeks) and male SD rats (numbers not specified)[168]
Strontium ranelateTannic Acid (TA) coated with PEG hydrogelTRUMPF TruPrint 1000-Osteoporosis model
-Drug screening model
-The PEG coating increased the titanium crosslinkability
-Optimal biodegradability
-The release was diffusion-based, and it was slower than in other materials, given that the tannic acid scaffold is denser
-TA 20% → ~90% release in 2 days
-TA 80% → ~50% release in 15 days
L929 mouse fibroblastsNot tested[261]
Ethyl-2,5-DihydroxibenzoateTitanium coated with PLGAWinforsys Metasys250-Osteoporosis
-Drug delivery system
-Inhibition of bone resorption
-Promoter of bone formation
-PLGA coating further stimulated the bone formation
Not specifiedMC3T3-E1 mouse pre-osteoblastsFemale Sprague–Dawley rats (N = 20; 3 months old)[262]
Zoledronic acid (ZA)JDBM Mg-Nd-Zn-Zr alloy + ceramic coatingZRapid Tech SLM150-Osteoporosis
-Drug delivery systems
-Porosity: 80%
-Pore-diameter: 600 µm
-The ceramic coating reduced the biodegradability to ≈5 mg/day
-Corrosion resistance
-Slow, controllable
-~8% in first 3 days
-~54% by 30 days (release driven by coating degradation)
Rat BMSCs (rBMSCs); mouse BMMs (bone-marrow macrophages)Female Sprague–Dawley rats, 3-month-old (N = 60)[185]
EBM (Electron Beam Melting)PaclitaxelTitanium + PEG Q10plus, GE, SUA-Osteosarcoma
-Drug delivery system
-In the acidic environment, the osteosarcoma cells were successfully killed-pH-responsive: ~11% at 72 h (pH 7.4)
-~85% at 72 h (pH 6.5)
-Stable at 7.4, fast intracellular release at 6.5
-The releases were controlled for over 1 month
143B, HOS, MG63; SaOS2, U2OSBALB/c nude mice (N = 30), 4 groups, n = 6/group[263]
CisplatinTi6Al4V + PLGA-PEG-PLGA hydrogelArcam S12 (Molndal/Gothenburg, Sweden)-Osteosarcoma
-Drug delivery system
-Low elastic modulus
-Low stiffness
-Good osteoinduction
-Large drug-loading due to the porous structure
-Initial burst then sustained >15 days
-Release matched hydrogel degradation
-Some hydrogel detachment observed
Osteosarcoma 143B, HOS, MG63BALB/c nude mice, 5 groups (N = 5/group)[162]
SimvastatinPorous Ti6Al4V (3DTi) + hydrogelArcam S12 (Molndal/Gothenburg, Sweden)-Bone defect (osteosarcoma)
-Drug delivery system
-In vitro and in vivo (rabbit condyles)
-The tumor volume reduced by 59–77% without any other damage
-The release was sustained for 18 days
-Anti-osteosarcoma and osteogenic effects
-Continuous release up to 18 days aligned with hydrogel degradation143B (human OS); L929 (mouse fibroblasts)Six-week-old female BALB/c nude mice; 3 groups (n = 6 each).[264]
Binder JettingBinder Jet 3D PrintingCurcumin loaded with Magnesiumβ-TCP (β-tricalcium phosphate)ExOne (Irwin, PA)-Osteosarcoma
-Drug delivery system
-In vivo osseointegration;
-In vitro chemoprevention;
-High cell viability;
-30 days: pH 7.4 ~22% (TCP) vs. ~17% (Mg-TCP); pH 5.0 ~30% (TCP) vs. ~23% (Mg-TCP)
-Weibull fits R2 > 0.9
THP1-derived osteoclasts; hFOB; MG-63; S. aureus; P. aeruginosaSprague–Dawley rats (supplier Envigo, numbers not specified)[203]
Gingerol; Allicinβ-TCPExOne-Osteoporosis
-Drug delivery system
-Porosity: 50%
-Antimicrobial activity
-Ginger promoted osteoblast proliferation by over 50%
-Reduced osteoclast activity
-Porous core with dense exterior with 10 MPa compressive strength
-Bone healing after 10 weeks
-Allicin sustained ~2 days (35–45%); gingerol release higher with PEG 50:50 vs. 65:35Human fetal osteoblasts (hFOB); osteoclasts (THP1-derived)Sprague–Dawley rats (N = 20)[204]
Quercetin; Vitamin D3 (lipid NPs)β-TCPExOne-Osteosarcoma
-Drug delivery system
-Compressive strength around 10 MPa;
-Osteoblast proliferation
-Osteoclast inhibition
-Antimicrobial activity against Staphylococcus aureus and Pseudomonas aeruginosa
-Biphasic; method- and pH-dependent; up to ~60 days
-Power-law kinetics (R2 > 0.99)
-↑ Acidic pH release
-Above 80% release rate at pH 7.4
-Controlled release for 60 days
hFOB; THP-1-derived osteoclasts; MG-63; P. aeruginosa; S. aureusNot tested[197]
Curcumin + Piperine (coated with Carvacrol)β-TCPProMetal® (ExOne)-Osteosarcoma
-Drug delivery system
-Compressive strength of 12.3 MPa
-Pore diameter: 400 µm
-Biphasic; method- and pH dependent;
-Up to ~60 days Power-law kinetics (R2 > 0.99)
-↑ Acidic pH release
-In combination with piperine, the curcumin availability increases to over 2000%
-94% antibacterial activity
-Cumulative release of 80% for curcumin and 90% piperine
-Controlled release for 60 days
hFOB (passage 6); MG-63 (passage 6); Staphylococcus aureusNot tested[265]
Gingerolβ-TCPExOne Innovent+-Osteosarcoma
-Drug delivery system
-Antioxidant properties
-Tumor necrosis factor suppression
-No cytotoxicity
-In a very acidic environment (pH 5.0), there is an initial burst release of 16% compared to 12% in pH 7.4hFOB (ATCC CRL-11372); MG-63; Staphylococcus aureusNot tested[205]
Retinol (vitamin A)β-TCP scaffold with PCL/PEG coatingExOne powder-bed printer-Bone defect
-Drug delivery scaffold
-Bulk density: ≈1.0 g/cm−3
-Porosity: 50%
-Compressive strength: 5.1 ± 1.1 MPa
-Full release by 1 day at pH 5
-~50% plateau by 7 days at pH 7.4; Weibull fits reported;
hFOB; THP-1-derived osteoclastsNot tested[266]
Vitamin D3β-TCP scaffold with PCL/PEG coatingExOne -Bone defect
-Drug delivery scaffold
-Pore diameter: 350 µm
-Porosity: 48.9 ± 2.54%
-Compressive strength: 7.0 ± 0.66 MPa
-Relative density: 32.2 ± 1.23%
-Osteoblast proliferation was enhanced by 64% compared to traditional osteoinductive methods
-3D scaffolds > porogen scaffolds
-After 60 days in PBS: 3D-printed model released ~47.5% vs. porogen ~37.5%
hFOB Not tested[196]
Sitafloxacin; Rifampinα-TCP/HA; PLGA coatingModified ZPrinter 450 (3D Systems)-Osteomyelitis
-Drug delivery system
-Reduced bacterial colonization
-Improved mechanical robustness
-The PLGA coating increased the Young’s modulus by three times, and the maximum stress by 4.4 times
->95% in 12 h (burst). PLGA-coated: biphasic
-Initial burst then ~zero-order for 2 weeks (dose-dependent)
Not testedFemale Balb/cJ mice (13–15 weeks)[76]
Table 2. Comparative summary table on 3D printing technology, release profile, and biocompatibility for each major drug class addressed in the manuscript.
Table 2. Comparative summary table on 3D printing technology, release profile, and biocompatibility for each major drug class addressed in the manuscript.
Drug ClassMain 3D Printing TechniquesRelease ProfileBiocompatibility/Biological ResponseReferences
Antibiotics
  • FDM (PLA, PCL, PMMA)
  • Bioprinting hydrogels (GelMA, methylcellulose/nHAp)
  • DLP
  • Binder Jetting (CaP);
  • Melt-extrusion (PEOT/PBT)
  • Micro-extrusion (SF-MA)
  • MEW + FDM hybrid
  • Melt-electrohydrodynamic PCL/PEG
  • Frequent early burst (often 30–95% within 6–48 h for some systems), then sustained release typically to ~30 days
  • Some systems longer (e.g., ciprofloxacin in PEOT/PBT sustained ≥1 month)
  • MOF/hydrogel could potentially increase release profiles
  • Overall biocompatible carriers (PLA/PCL/GelMA/CaP/Ti)
  • Strong antimicrobial activity, reduced colonization, preserved cell viability, and bone integration
  • PMMA curing can generate heat
  • High local rifampin can be cytotoxic if overdosed (not at reported loads)
[76,86,94,106,109,118,134,150,153,248]
Anti-inflammatory
  • FDM (PLA)
  • Bioprinting (PCL/alginate/gelatin with drug microparticles)
  • Modified/custom extrusion (PCL + liposomes)
  • Surface-loaded parts can show fast burst (e.g., ~50% dexamethasone in 6 h), while embedded/encapsulated systems show tempered burst and sustained release ~2–4 weeks (e.g., DEX MPs to day 30)
  • Cytocompatible
  • Osteoinductivity reported for some constructs
  • Degradation stable and tunable via composite design
[194,239,248,256]
Anticancer
  • SLA/DLP
  • SLS/SLM/EBM (Ti alloys and ceramics)
  • Binder Jetting (β-TCP)
  • Photo-crosslinking-assisted printing
  • Tumor-on-chip/microfluidic platforms
  • Highly tunable: from ultra-fast (e.g., 5-FU complete in ~2 h) to prolonged/pH- or stimulus-responsive (e.g., paclitaxel >1 month at pH 7.4; faster at pH 6.5; cisplatin sustained >15 days; DOX release boosted by microwave/acidic pH)
  • Overall span ≈ 2 h to ≥60 days depending on drug/carrier
  • Effective local tumor suppression with minimal systemic exposure
  • Maintained osteogenesis/osseointegration of scaffolds
  • Responsive systems (pH/thermal/photothermal) allow on-demand dosing
[162,168,184,185,233,240,243,252,259,263]
Other drugs and growth factors
  • FDM
  • Bioprinting (alginate/gelatin, PVA, GelMA)
  • SLS/SLM
  • Binder Jetting
  • MEW hybrid
  • Wide spectrum: from fast burst (e.g., VEGF, leonurine early 72 h burst) to very prolonged: BMP-2 (sustained 2+ weeks; ZIF-8 composite ~57% by 174 h and continuing), Salvianolic acid B (~60% by day 80), vitamins/natural products like Vitamin D3 and Gingerol sustained up to 60 days, Dipyridamole sustained 30 days (no burst)
  • Strong osteogenesis and angiogenesis
  • Enhanced osteoblast proliferation and osseointegration
  • Osteoclast inhibition (e.g., Strontium ranelate); generally biocompatible with occasional initial bursts governed by coating/encapsulation
[196,197,199,200,204,205,249,259,261,265,266]
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

Timofticiuc, I.-A.; Grigore, A.-G.; Tomescu, E.-T.; Vlaicu, T.-M.; Dragosloveanu, S.; Scheau, A.-E.; Caruntu, A.; Dragosloveanu, C.D.M.; Badarau, I.A.; Didilescu, A.C.; et al. Advances in 3D-Printed Drug Delivery and Screening Platforms for Bone Disease Therapy. Pharmaceutics 2025, 17, 1372. https://doi.org/10.3390/pharmaceutics17111372

AMA Style

Timofticiuc I-A, Grigore A-G, Tomescu E-T, Vlaicu T-M, Dragosloveanu S, Scheau A-E, Caruntu A, Dragosloveanu CDM, Badarau IA, Didilescu AC, et al. Advances in 3D-Printed Drug Delivery and Screening Platforms for Bone Disease Therapy. Pharmaceutics. 2025; 17(11):1372. https://doi.org/10.3390/pharmaceutics17111372

Chicago/Turabian Style

Timofticiuc, Iosif-Aliodor, Alex-Gabriel Grigore, Elena-Teodora Tomescu, Teona-Maria Vlaicu, Serban Dragosloveanu, Andreea-Elena Scheau, Ana Caruntu, Christiana Diana Maria Dragosloveanu, Ioana Anca Badarau, Andreea Cristiana Didilescu, and et al. 2025. "Advances in 3D-Printed Drug Delivery and Screening Platforms for Bone Disease Therapy" Pharmaceutics 17, no. 11: 1372. https://doi.org/10.3390/pharmaceutics17111372

APA Style

Timofticiuc, I.-A., Grigore, A.-G., Tomescu, E.-T., Vlaicu, T.-M., Dragosloveanu, S., Scheau, A.-E., Caruntu, A., Dragosloveanu, C. D. M., Badarau, I. A., Didilescu, A. C., Caruntu, C., & Scheau, C. (2025). Advances in 3D-Printed Drug Delivery and Screening Platforms for Bone Disease Therapy. Pharmaceutics, 17(11), 1372. https://doi.org/10.3390/pharmaceutics17111372

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