Next Article in Journal
Galactosed and Reduction-Responsive Nanoparticles Assembled from Trimethylchitosan–Camptothecin Conjugates for Enhanced Hepatocellular Carcinoma Therapy
Next Article in Special Issue
Compounding Tailored Veterinary Chewable Tablets Close to the Point-of-Care by Means of 3D Printing
Previous Article in Journal
Placenta-Derived Mesenchymal-like Adherent Stromal Cells as an Effective Cell Therapy for Cocaine Addiction in a Rat Model
Previous Article in Special Issue
Development and Validation of a Novel Tool for Assessing the Environmental Impact of 3D Printing Technologies: A Pharmaceutical Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Evolution of the 3D-Printed Drug Delivery Systems: A Review

1
Healthcare Industry Institute, University of Debrecen, Nagyerdei körút 98, H-4032 Debrecen, Hungary
2
Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, H-4032 Debrecen, Hungary
*
Author to whom correspondence should be addressed.
Submission received: 11 May 2022 / Revised: 15 June 2022 / Accepted: 18 June 2022 / Published: 21 June 2022
(This article belongs to the Special Issue The Evolution of Pharmaceutical Three-Dimensional Printing)

Abstract

:
Since the appearance of the 3D printing in the 1980s it has revolutionized many research fields including the pharmaceutical industry. The main goal is to manufacture complex, personalized products in a low-cost manufacturing process on-demand. In the last few decades, 3D printing has attracted the attention of numerous research groups for the manufacturing of different drug delivery systems. Since the 2015 approval of the first 3D-printed drug product, the number of publications has multiplied. In our review, we focused on summarizing the evolution of the produced drug delivery systems in the last 20 years and especially in the last 5 years. The drug delivery systems are sub-grouped into tablets, capsules, orodispersible films, implants, transdermal delivery systems, microneedles, vaginal drug delivery systems, and micro- and nanoscale dosage forms. Our classification may provide guidance for researchers to more easily examine the publications and to find further research directions.

Graphical Abstract

1. Introduction

Three-dimensional (3D) printing was developed more than 30 years ago to manufacture 3D objects based on a digital design. This layer-by-layer process enables a fast and cheap design cycle for the preparation of personalized medication [1]. The term 3D printing was coined as an umbrella term and encompasses a number of processes, and in many reviews the main types were described in detail [2,3,4,5]. Three-dimensional printing gave the means to the manufacture of a high-quality product within minutes in an easy manufacturing cycle. This on-demand manufacturing was time and material saving. Not to mention the fact that 3D printers could conquer the traditional manufacturing regime of ‘one size fits all’ [6]. As 3D printing was based on a computer-aided design (CAD), it provided the ability to quickly create and produce a flexible and innovative product [7]. Personalized medication carried the opportunity to create drug delivery systems for patient’s requirements. Furthermore, 3D printing gained access to the creation of unique dosage forms and achieving more complex drug release profiles [8]. The image could be made to meet the patient’s individual needs regarding their age, weight, organ function, and severity of disease [4]. The application of 3D printing technology might be an alternative way to construct effective, customized active pharmaceutical ingredient (API) combinations for the patient immediately [9]. The 3D printing technique opened up the opportunity for the development of tailored single and multi-drug products at the point-of-care [10].
In recent years, many comprehensive publications have been presented on the different designed drug dosage forms. As Moulton et al. highlighted, this kind of process created the opportunity for the manufacturing of controlled and modified release of the APIs, enabled the delivery of poorly water soluble drugs, increased drug stability, and reduced the used API amount without compromising the efficacy [11]. In 2018, two distinct research groups summarized the recent achievements in the manufacturing of pharmaceuticals but as a rapidly developing area the achievements vary from year to year [12,13]. Mohapatra et al. gathered together the newest publications in recent years and grouped the research based on the type of 3D printing [14].
The available reviews mostly focus on one or multiple drug dosage forms manufactured by one type of 3D printing technology. Cunha-Filho et al. discussed the fabricated drug delivery systems by fused deposition modeling (FDM) 3D printing [15]. While Gueche et al. summed up the oral dosage forms created by selective laser sintering (SLS) [16] and Wang et al. described the stereolithographic (SLA) constructed oral dosage forms [17]. Inkjet printing of pharmaceuticals was summarized by Dali et al. [18]. In more and more research, three-dimensional bioprinting was used which is a new era of 3D printing technologies where researchers aim to build living tissue models [19].
During the last decades multiple research groups were established to fabricate drug delivery systems. FabRx Ltd. is one of the most innovative start-up companies, which is a biotech company, designed to produce 3D-printed medication [20]. Regarding the diverse drug dosage forms, different reviews summarized the achievements. For example buccal patches were analyzed in the work of Shirvan et al. [21], implants in the work of Domsta et al. [22], oral dosage forms in the work of Khatri et al. [23], and transdermal delivery systems (TTS) in the work of Economidou et al. [24].
The aim of this work was to provide a comprehensive image on how the manufacturing of the different drug delivery systems started and where the experiments are headed now. The chosen drug delivery systems were divided into subgroups based on the type of the drug delivery system and the tables summarized the most important research in the last 20 years in chronological order.

2. The 3D Printing of Drug Delivery Systems

2.1. Tablets

The first publication of a 3D-printed tablet dates back to 1996 when solid samples were created with a desktop printer from PCL and PEO polymers containing blue and yellow dyes. Based on the results, complex drug delivery regimes could be created with this technique, such as the release of multiple drugs or multiphasic release of a single drug. This study demonstrated several simple examples of such devices and several construction methods that could be used to control the release of the drugs [25].
In the early 3D printing articles, droplet binding was used for the manufacturing of the samples when the used binder was not necessarily polymer but some other auxiliary material, e.g., Eudragit® or mannitol. The authors concluded that with this method adequate oral dosage forms can be manufactured which exhibit erosion or diffusion release mechanisms [26,27]. At the beginning of the research, the most important question for the authors was the type of the chosen additive manufacturing process, the used printing parameters, immediate- or delayed release tablet manufacturing, and first- or zero-order kinetic profile manufacturing [28].
Gbureck et al. used a unique technology for the manufacturing of the drug delivery system. Firstly, they created the sample with a 3D bioceramic powder printing process and then the used antibiotics were adsorbed during a week to fabricate the tablets [29]. Yu et al. produced an acetaminophen containing matrix tablet using a desktop 3D printer. The middle drug-containing regions of the tablets were formed by depositing the binder liquid containing release-modulation materials onto the automatically spread powder layers (Figure 1) [30]. Two years later, the same investigators decided to construct a drug delivery system from the same API, polymer, and printing technique but in this case the layers were not designed horizontally, yet vertically, to provide a different dissolution mechanism [31].
Guaifenesin-containing controlled release bilayer tablets were constructed with extrusion-based desktop 3D printing. The samples were formulated to demonstrate the production of relatively complex formulations that could mimic the release profile of a commercially available tablet [32].
Even though the wide range experiments started in the 2010s, the Food and Drug Administration agency (FDA) granted the approval of Spritam® in 2015. This 3D-printed tablet was used for the treatment of epileptic seizures. This was the first and still only approved 3D-printed drug delivery system. For the manufacturing, a so-called ZipDose Technology was used [33].
Goyanes et al. published four articles in 2015 about the manufacturing of tablets for distinct purposes with the use of FDM 3D printing and PVA filament. In one of the articles, the commercially produced PVA filaments were loaded with 5- and 4-amino salicylic acid in an ethanolic drug solution. A final drug loading of 0.06% w/w and 0.25% w/w was achieved for the APIs, respectively. This filament was created with unsimilar infill percentages in a nonidentical pattern (Figure 2). The dissolution tests showed that the release profiles depended on the used infill percentage and the used API itself [34].
A polypill was designed by a RegenHU extrusion-based 3D printer which contained distinct fillable ink cartridges for the production of semi-solid API-containing materials. In this research, three different API—nifedipine, captopril, and glipizide—containing inks were manufactured by HPMC. The three APIs could be found in three diverse compartments. From the nifedipine and the glipizide-containing formulations, the drug was released by diffusion and from the captopril formulation by osmosis. The schematic image of the samples is shown in Figure 3 [36].
In the same year, a newer design was applied which is also a polypill but contained five different APIs: amino salicylic acid (ASA) and hydrochlorothiazide (HCT) in the bottom layer, and atenolol, ramipril, and pravastatin in the middle area in three separated regions. The manufacturing was described in the previous article. The ASA and HCT formulation were an immediate release compartment, while the others were controlled with a cellulose acetate membrane to provide extended release. The graphical scheme is shown in Figure 4 [37].
Metolose® (a special cellulose ester) and PLA disks were created and co-extruded with nitrofurantoin as a model API. The research showed that the rheological properties depended on the amount of the undissolved particles and, as in the case of modified release tablets, the amount of the cellulose derivative affected the dissolution time from the fabricated filament [38].
In the work of Okwuosa et al., a 10% API-containing filament was produced by hot-melt extrusion. As an API dipyridamole or theophylline, as a polymer PVP, and as an excipient plasticizer was used. These constructed filaments were then FDM 3D printed. The novelty of this work is that, for the extrusion, thermostable filler (talc) was used which enabled lower temperature printing around 110 °C and the stability of the used APIs was not affected [39].
In the same year, Sadia et al. decided to use a pharmaceutical-grade non-melting filler (TCP) through the hot-melt extrusion to allow a consistent flow from the nozzle of the printer. This novel approach meant the addition of 20–50% non-melting component to the filament and four model drugs were incorporated separately. This process aimed at the fabrication of well-defined caplets. In the case of 5-ASA and prednisolone, 93% of the drug contents remained intact in the tablet but a significant drop in captopril content was observed due to thermal degradation. This procedure made the manufacturing of personalized immediate release tablets easy [40]. In Figure 5. the most important research results can be seen between 1996–2016.
Acosta-Vélez reported the production of a biocompatible photocurable pharmaceutical polymer for inkjet 3D printing that was suitable for the manufacturing of hydrophilic active pharmaceutical ingredients. More specifically, hyaluronic acid was functionalized with norbornene moieties. This conjugate in the presence of poly(ethylene) glycol dithiol, Eosin Y, and a visible light source underwent a polymerization reaction. The manufactured bioink was loaded with ropinirole HCL and dispensed through a piezoelectric nozzle onto a blank preform tablet, and then polymerized. The study confirmed the potential of inkjet printing for the rapid production of tablets through the deposition of a photocurable bioink designed for hydrophilic APIs [43].
Beck et al. combined two important, innovative fields: additive manufacturing and nanotechnology. The researchers’ idea was to first create their own filament with a channeling agent with hot-melt extrusion then FDM print it and finally load the previously fabricated channels with nanocapsules by soaking. The researchers believed that this method could improve the delivery of the drugs [44].
Chai et al. planned an intragastric floating tablet. For the manufacturing, first domperidone was hot-melt extruded with hydroxypropyl cellulose and then the produced filament was FDM 3D printed. Based on the authors’ findings, the sample with a hollow structure was successfully fabricated and the buoyancy of tablets was closely related to their densities. Due to the rigid shells produced by the melting deposition, HPC polymer chains dissociated slowly [45].
An oral dual-compartmental dosage unit was designed for the treatment of tuberculosis (Figure 6). The aim of the research was to physically isolate and modulate the release profile of an anti-tuberculosis drug combination because rifampicin and isoniazid negatively interact with each other upon simultaneous release in an acidic environment. The samples were fabricated in two steps; first, 3D printing of the outer structure, followed by hot-melt extrusion of the two different drug-containing filaments. This way, the two APIs were separated and resulted in modified release and an effective retardation, based on the authors’ findings [46].
A DuoTablet was designed, which meant that for the first time glipizide was hot-melt extruded with PVA and then the drug-loaded filament was printed and formed a double-chamber device composed of a tablet embedded within a larger tablet [47].
Gyroid lattice printlets were designed containing paracetamol with SLS technology. The novel structure was able to modulate the drug release from all four polymers. This work was the first to demonstrate the feasibility of using SLS to achieve customized drug release properties of several polymers, and avoided the alteration of the formulation composition [48].
The goal of Hollander et al. was to study the printability of poly(dimethyl siloxane) (PDMS) with a semi-solid extrusion printer in combination with the UV-assisted crosslinking technology using UV-LED light to produce drug delivery systems. Samples with different pore sizes and API amount were prepared and contained prednisolone as a model drug. By altering the surface area/volume ratio, it was possible to create structures with different release rates. The study shows that this 3D printing technique in combination with UV-LED crosslinking was an applicable method and an interesting alternative for manufacturing controlled release devices containing temperature-susceptible drugs [49].
Kollamaram et al. aimed to fabricate low-melting and thermolabile drugs by reducing the FDM printing temperature. For this purpose, two immediate release polymers, Kollidon VA64 and Kollidon 12PF, were investigated and ramipril was used as the model low melting point drug (109 °C). The drug loaded filaments were extruded at 70 °C and contained 3 w/w% API, while the printing temperature was 90 °C. This work demonstrated that the selection and use of new excipients could make this technique suitable for drugs with lower melting temperatures [50].
Gastro-floating tablet were created with three kinds of infill percentage and prepared by hydroxypropyl methylcellulose (HPMC K4M) and hydroxypropyl methylcellulose (HPMC E15) as hydrophilic matrices and microcrystalline cellulose (MCC PH101) as extrusion molding agent (Figure 7). The study determined that floating could be maintained for up to eight hours with the combination of traditional pharmaceutical excipients and a modern technique [51].
Tablets with a novel design approach of caplets with perforated channels were fabricated by Sadia et al. to accelerate drug release from FDM 3D-printed samples (Figure 8). The experimental arrangement was to use different channel widths, lengths, and alignments. Based on the results, the parameters should be carefully considered in addition to surface area when optimizing drug release from samples. The incorporation of short channels could be adopted in the patterns of dosage forms built from polymeric filaments [52].
Scoutaris et al. created indomethacin (as a model drug) containing PEG filaments, then FDM 3D printed to construct chewable tablets. The shapes of the samples were variable (lion, heart, and teddy bear) for improved patient compliance in the case of children. This research also demonstrated that 3D printing could be effectively used for the manufacturing of personalized medication in the field of pediatrics [53].
In the same year a high—up to 60 w/w%—API-containing filaments were designed by TPU polymers and a prolonged release profile was achieved with a time period of 24 h. High drug loaded filaments were fabricated which were heat stable through the FDM 3D printing [54]. In Figure 9. the most important research results can be seen between 2017–2018.
Goyanes et al. aimed to produce a drug delivery system for a rare metabolic disorder called maple syrup urine disease, which required strict dietary restriction and oral supplementation of isoleucine. In the research, isoleucine containing printlets were constructed in six diverse flavors and four distinct API amounts with a special 3D printer (The Magic Candy Factory). The dissolution profile of the samples was adequate and the patients—with different preferences in terms of flavor and color—reported good acceptability of the formulations [55].
Öblom et al. designed isoniazid-containing filaments and then printed tablets with FDM printing technology. As a polymer, nonidentical cellulose based (such as HPMC, HPC, or Eudragit) filaments were fabricated with a constant content of 30 w/w% API. The effect of the used polymer, the size, and the infill percentage were investigated using the dissolution profile. Drug release characteristics could be altered by changing these critical printing parameters and allowing personalization of the tablets [56].
A polypill with SLA technique was manufactured from six different APIs: paracetamol, chloramphenicol, acetylsalicylic acid, naproxen, caffeine, and prednisolone, where the created structure was cylinder (Figure 10) or ring shaped. For the fabrication, a novel method was developed to fabricate multi-layered constructs with variable drug contents and shapes [57].
Nineteen semisolid formulations were prepared for a fractional factorial design. The variables were the amount of the API and unalike soluble and insoluble excipients. First, a Carbopol gel was made; then, with diclofenac sodium a semisolid pasta was created and then this special “ink” was 3D printed with a Bioplotter printer. The researchers found out that with the determination of critical process parameters a robust and consistent 3D printing method could be achieved [58].
Awad et al. decided to manufacture tablets by 3D printing technology with braille and moon patterns in various shapes for patients with visual impairment. Printlets with different shapes were fabricated to offer additional information, such as the medication indication or its dosing regimen. Despite the presence of the patterns, the printlets retained their original properties of a conventional tablet [59].
A special technique was used for the avoidance of high printing temperature in the case of FDM printing. A study was performed to develop novel core-shell gastroretentive floating pulsatile drug delivery systems using a hot-melt extrusion-paired FDM 3D printing and direct compression method. In the research, hydroxypropyl cellulose (HPC) and ethyl cellulose (EC)-based filaments were fabricated using hot-melt extrusion technology and were utilized as feedstock material for printing shells through the production. The directly compressed theophylline tablet was used as the core. The researchers fabricated a gastro-floating theophylline-containing drug delivery system [60].
An osmotically controlled dosage form was designed where the core contained the PVA, diltiazem, and osmogene. The 3D-printed core was covered by cellulose acetate to provide modified release. In the graphic, an imported hole and several linear cavities were shaped to achieve the controlled release profile [61].
Karavasili et al. created chocolate and corn syrup ink to print ibuprofen and paracetamol-containing dosage forms for children. The main concept was to fabricate a chewable tablet for pediatric use and to 3D print both hydrophilic and lipophilic APIs [62].
Melting solidification printing was used as a novel technique for the manufacture of oral solid dosage forms to avoid the use of solvents and high temperatures. This process was performed with a special ink—Gelucire® 50/13 (fatty polyethylene glycol esters)—which could be used to obtain a floating sustained release system with improved dissolution and absorption of drugs, for example, from ricobendazole, which had showed a low and erratic bioavailability [63].
Tsintavi et al. dedicated their work to partially coating tablets with a glyceride, namely Precirol ATO 5, using a semi-solid 3D printer as an approach for tuning the release of two APIs, the hydrophilic methyl-levodopa hydrochloride and the lipophilic acyclovir. The surface coating percentage, the number of coating layers, and the coated sides of the tablet controlled the release profile and diverse dissolution profiles were reached [64].
Multi-layered polyprintlets were produced from PEG 300, PEGDA, and four different antihypertensive drugs: irbesartan, atenolol, hydrochlorothiazide, and amlodipine by SLA 3D printing. The created drug delivery system could deliver a low-dose combination therapy, but an interaction occurred between PEGDA and amlodipine. This unexpected drug–polymer interaction had a serious impact because it highlighted the need to screen the biocompatibility properties of photoreactive monomers to ensure the safety and compatibility of drug-loaded oral dosage forms produced by SLA [65].
Dapagliflozin-containing self-nanoemulsifying tablets were manufactured by semisolid pressure-assisted microsyringe (PAM) extrusion-based 3D printing technique. This work combined two very investigated fields recently in pharmaceutic manufacturing: SNEDDS and 3D printing. For the manufacturing, a liquid and a solid phase were fabricated separately. First, the solid phase was melted then the liquid phase ingredients were added. This semi-solid syringe was transferred to the extruder syringe when 3D printing could take place. This manufacturing enabled the manufacturing of a special drug delivery system with the combination of two innovative research fields [66].
A special method was designed for the fabrication of tablets with customizable dosages, durations, and combinations of multiple drugs by FDM 3D printing technology. The method and the structure of the tablet was simple: first, a template was printed by FDM 3Dprinter; then, a solution was poured into a PDMS mold where solidification take place. Finally, the samples were covered with pre-printed white wax coatings. The tablets were customized by varying the amount of excipient used, the height of the tablet, and the number and amount of used APIs (paracetamol, phenylephrine HCl, and diphenhydramine HCl). Based on the authors’ findings, with the use of templates a high variety of tablets could be constructed [67].
In a study, photoabsorbers were used to improve the SLS printability of five different colorless drugs and distinct excipients with low glass transition temperature and low stability. The forming mechanism of amorphous and crystalline polymers was sintering and melting, respectively. Immediate-release tablets with a high drug loading of 90% and sustained-release tablets with tunable dissolution behavior were successfully prepared, suggesting that the SLS technique had great prospects in producing personalized oral preparations [40,68]. In Figure 11, the most important research results could be seen since 2019.
In recent decades, the number of publications on 3D-printed tablets has multiplied. The above-described research was highlighted because of their novelty in some way. Since it would be completely impossible to characterize all the innovative research, we tempted to summarize even more research in the two tables below. The studies were classified based on the publication year and then in alphabetical order of the author. Table 1 summarizes the publications between 1996 and 2016, and Table 2 from 2017 to present.
Some advantages and limitations should also be mentioned. One of the biggest advantages against the conventional tablets were the possibility of incorporating several drug substances into one product to produce a polypill, which is personalized regarding both the combination of drug substances and the doses. These tablets would benefit the drug treatments of several medical conditions and would improve adherence to medications. We had to mention that low printing efficiency was one of the major limitations [69]. The other limitations were the lower productivity, higher costs, and incapability of production and delivery on-demand compared to the conventional tablets [70]. In addition, the healthcare professionals expressed some concerns associated with medication safety and quality aspects, including dose accuracy, quality control, stability, shelf life of formulations, and the identification of drug products at hospital wards [71]. Another research group named poor surface quality and mechanical strength of the final object as a limiting factor [72]. The possibility to 3D print personalized medications not only at an industry or pharmacy setting, nor compounding or community, but also even at the patient’s home could revolutionize the healthcare system [70].
Table 1. The grouping of the manufactured tablets based on the publication year and then in alphabetical order between 1996 and 2016.
Table 1. The grouping of the manufactured tablets based on the publication year and then in alphabetical order between 1996 and 2016.
YearType of 3D PrintingType of PolymerType of APIArticle
1996desktop 3D printerPCL, PEOyellow and blue dyeWu et al. [25]
2000droplet bindingmethacrylate copolymerschlorpheniramineKatstra et al. [26]
droplet bindingmethacrylate copolymerschlorpheniramine, diclofenacRowe et al. [73]
2003droplet binding (TheriForm™ process)none (mannitol)captoprilLee et al. [27]
2006droplet binding (TheriForm™ process)Kollidon SR (80% polyvinyl acetate, 19% polyvinyl pyrrolidone)pseudoephedrineWang et al. [28]
2007bioceramic powder printingResomer RG502H (polylactide-polyglycolide 50:50)vancomycin, ofloxacin, and tetracyclineGbureck et al. [29]
powder binding desktop 3D machinePVPacetaminophenYu et al. [30]
2009powder binding desktop 3D machinePVP K30acetaminophenYu et al. [31]
2012SLSPCLprogesteroneSalmoria et al. [74]
2014FDMPVAfluoresceinGoyanes et al. [75]
Extrusion-based 3D printer (Fab@Home)PAAguaifenesinKhaled et al. [32]
2015—FDA approvedZipDoseunknownlevetiracetamAprecia Pharmaceuticals [33]
2015FDMPVAparacetamol, caffeineGoyanes et al. [35]
FDMPVAparacetamolGoyanes et al. [41]
FDMPVAbudesonideGoyanes et al. [42]
FDMPVA5- and 4- amino salicylic acidGoyanes et al. [34]
RegenHU 3D printerHPMCnifedipine, captopril, glipizideKhaled et al. [36]
RegenHU 3D printerHPMCASA, HCT, atenolol, pravastatin, captoprilKhaled et al. [37]
FDMEudragit RL100
and RS100, HPC
theophyllinePietrzak et al. [76]
FDMPVAprednisoloneSkowyra et al. [77]
2016FDMEudragit EPO, Soluplus and PVAfelodipineAlhijjaj et al. [78]
FDMPLA, HPMCnitrofurantoinBoetker et al. [38]
FDMPVAparacetamol, caffeineGoyanes et al. [79]
FDMPVPdipyridamole or theophyllineOkwuosa et al. [39]
FDMEudragit EPOtheophylline, 5-ASA, captopril, prednisoloneSadia et al. [40]
SLAPEGDA4-ASA, paracetamolWang et al. [17]
Table 2. The grouping of the manufactured tablets based on the publication year and then in alphabetical order between 2017 and 2021.
Table 2. The grouping of the manufactured tablets based on the publication year and then in alphabetical order between 2017 and 2021.
YearType of 3D PrintingType of PolymerType of APIArticle
2017inkjet printingPEGropiniroleAcosta-Vélez et al. [43]
FDMPCL, Eudragit RL 100nanocapsulesBeck et al. [44]
FDMHPCdomperidoneChai et al. [45]
inkjet printingPEGDAropiniroleClark et al. [80]
SLSKollicoat IRparacetamolFina et al. [81]
FDMPEO, PLArifampicin, isoniazidGenina et al. [46]
FDMHPMCASparacetamolGoyanes et al. [82]
FDMHECfood coloringGoyanes et al. [83]
FDMbeeswaxfenofibrateKyobula et al. [84]
FDMPVAglipizideLi et al. [47]
FDMPVPtheophyllineOkwuosa et al. [85]
FDMKollidon® VA64, Kollicoat® IR,
Affinsiol™15 cP and HPMCAS
haloperidolSolanki et al. [86]
FDMPVAcurcuminTagami et al. [87]
FDMPLAacetaminophenZhang et al. [88]
2018inkjet printing with piezoelectric nozzlePEG, PEGDAnaproxenAcosta-Vélez et al. [89]
FDMHPCtheophyllineArafat et al. [90]
FDMEudrgait EPOwarfarinArafat et al. [91]
SLSEudragit (L100-55 and RL)paracetamolFina et al. [48]
SLSHPMC E5, Kollidon VA64paracetamolFina et al. [92]
UV-assisted crosslinking technologyPDMSprednisoloneHollander et al. [49]
ZMorph®Kollicoat®, PLAaripiprazoleJamróz et al. [69]
RegenHU bioprinterPVP K25paracetamolKhaled et al. [93]
FDMKollidon VA64, Kollidon 12PFramiprilKollamaram et al. [50]
extrusion-based MAMIIHPMC K4M, HPMC E15, MCC PH101, PVPdipyridamoleLi et al. [51]
SLAPEGdaparacetamolRobles- Martinez et al. [94]
SLSHPMCparacetamolTrenfield et al. [95]
FDMPolyplasdone-XL®hydrochlorothiazideSadia et al. [52]
FDMPEGindomethacinScoutaris et al. [53]
FDMTPUtheophylline, metforminVerstraete et al. [54]
2019SLSKollidon® VA 64diclofenacBarakh Ali et al. [96]
direct single-screw powder extruder (FabRx)HPCitraconazoleGoyanes et al. [97]
specially adapted 3D printer (The Magic Candy Factory)pectinisoleucineGoyanes et al. [55]
FDMHPMCcarvedilolIlyés et al. [98]
FDMPEOtheophyllineIsreb et al. [99]
FDMEudragit® RS 100acetaminophenKrause et al. [100]
FDMHPMCAS, PEG 400pregabalinLamichhane et al. [101]
FDMCellulose based polymersisoniazidÖblom et al. [56]
SLAPEGdaparacetamol, chloramphenicol, acetylsalicylic acid, naproxen, caffeine, prednisoloneRobles-Martinez et al. [57]
FDMHPMCacyclovirShin et al. [102]
Bioplotter 3D printerPolyplasdonediclofenac sodiumZidan et al. [58]
pressure-assisted microsyringePVPginkgolideWen et al. [103]
FDMPVAparacetamolXu et al. [104]
2020SLSKollidon VA64ondansetronAllahham et al. [105]
SLSKollidon VA64paracetamolAwad et al. [59]
FDMHPMCtheophyllineCheng et al. [106]
semi-solid 3D extrusion printerHPCMlevetiracetamCui et al. [72]
FDMHPC, ECtheophyllineDumpa et al. [60]
FDMHPCcaffeineFanous et al. [107]
FDMPVAdiltiazemGioumouxouzis et al. [61]
SLSKollicoat® IRlopinavirHamed et al. [108]
FDMKollicoat® IR, PLA, PVAbicalutamideJamróz et al. [109]
DLP, SLS, SSE, FDMPVA, PEGDAplaceboJanuskaite et al. [110]
inkjet technology XYZprinting 3D Food Printer (Model 3C10A)chocolate, corn syrupibuprofen, paracetamolKaravasili et al. [62]
SLSMCCclindamycinMohamed et al. [111]
direct powder extrusionPEOtramadolOng et al. [112]
melting solidification printing processGelucire 50/13ricobendazoleReal et al. [63]
semi-solid 3D printerPrecirol ATO 5methyldopa, acyclovirTsintavi et al. [64]
FDMHPCcinnarizineVo et al. [113]
SLAPEG 300, PEGDAirbesartan, atenolol, hydrochlorothiazide, amlodipineWu et al. [65]
2021pressure-assisted microsyringePEG 400, PEG 6000dapagliflozinAlgahtani et al. [66]
direct powder extrusionKollidon VA64praziquantelBoniatti et al. [114]
SLSPVPAropiniroleDavis et al. [115]
SSEemulsion gelfenofibrateJohannesson et al. [116]
FDMPEG 1000paracetamol, phenylephrine HCl, diphenhydramine HClTan et al. [67]
FDMPCLindomethacin, theophyllineViidik et al. [117]
FDMPEGDAwarfarin sodiumXu et al. [118]
SLSPVAindomethacin, nifedipine, tinidazole, ibuprofen, metoprolol, paracetamol, diclofenac sodiumYang et al. [68]

2.2. Capsules

The first 3D-printed capsular devices were manufactured in 2015 by Melocchi et al. For the manufacturing, hydroxypropyl cellulose-containing filaments were created by hot-melt extrusion and then the filament was 3D printed. The manufactured samples were swellable erodible capsules for oral pulsatile drug release [119].
Fused deposition modeling and inkjet printing were used to fabricate capsules from different polymer formulations. The capsules were formed by three parts: two hollow parts which had a cylindrical closed end and a rounded open end; the middle part acted like a joint and a partition (Figure 12). The hollow parts differed in geometry and wall-thickness. The samples were filled with model APIs and the results showed that the device was able to successfully release the model APIs in pulses within 2 h [120].
A research group combined the versatility of 3D printing capsules with controlled geometry and the drug release properties of nanocellulose hydrogel to accurately modulate its drug release properties. As a novel method, the capsules were filled with a drug dispersion composed of model compounds and anionic cellulose nanofiber hydrogel. The main benefits of this device were that the release could be modulated simply by modulating the inner geometry of the PLA capsule and as the API did not undergo heating a wide range of APIs could be used. e.g., proteins and liposomes [121].
As it could be seen. capsules were investigated by a few research groups because only hard-shell capsules can be manufactured by 3D printing. The advantage of the manufacturing by 3D printing, more or less the same as in the case of the tablets as personalized drug delivery systems, is that it could be made with flexible on-demand doses with better health outcomes. As a limitation, a research group mentioned the API stability and the low amount of pharmaceutical grade polymeric carriers [119]. The published research on the created capsules can be found in Table 3.

2.3. Orodispersible Films

The first 3D-printed oral film was printed by thermal inkjet printing where the used API (salbutamol sulfate) was dissolved in the aqueous solution, the ink cartridges were filled with this solution, and it was printed onto a commercial potato starch film. The authors concluded that this process was suitable for the manufacturing of aqueous drug solutions into thin polymer films but the viscosity and API stability had to be controlled [125].
In another work, the aim was to evaluate the applicability of orodispersible films (ODFs), porous copy paper sheets, and water impermeable transparency films (TFs) which contained rasagiline mesylate (RM) as a low dose active pharmaceutical ingredient. Flexible doses of the API were obtained by printing several subsequent layers on top of the already printed ones, using an off-the-shelf consumer thermal inkjet (TIJ) printer [126].
A research group manufactured the drug dosage form with a special 3D printing method which incorporated two different methods: piezoelectric- and solenoid valve-based inkjet printing technologies to allow the dispensing of an extensive range of fluids. The research demonstrated the opportunity to 3D print a wide range of formulations for the patient needs. The fabrication avoided the risk of drug degradation by ink heating and of substrate damage (by contact printing) and the manufacturing scheme avoided the emergence of defects [127].
Vakili et al. used inkjet printing to create orodispersible films, which contained propranolol hydrochloride. The drug delivery systems were designed with escalating doses of propranolol hydrochloride on three different substrates and three unalike area sizes were used through the 3D printing with thermal inkjet printing technology. A thin sweetener coating layer of saccharin was successfully included in the final dosage form to increase the patient compliance among pediatric patients [128].
Aripiprazole-containing orodispersible films were fabricated with FDM technology from PVA by Jamróz et al. The aripiprazole in the sample is fully amorphous due to the two-step hot-melt extrusion process (filament fabrication and 3D printing) and the high concentration of PVA polymer helped to maintain the amorphous form [129].
In a study, benzydamine hydrochloride and HEC were used for the manufacturing of a printing dispersion. For the 3D printing, a modified FDM technique was used in which the FDM extruder was replaced by linear syringe pump. This method could be implemented into compounding practice of pharmacies allowing preparation of ODFs for the patients in small batches, and could eliminate preliminary test printing [130].
A research group aimed to investigate semisolid extrusion 3D printing for production of warfarin-containing orodispersible films. The applied 3D printing method was unique because a one-step-process utilized disposable syringes hindering the printing material to be in contact with the printing equipment. The method was successfully utilized to produce transparent, smooth and thin, yet flexible and strong orodispersible films containing therapeutic doses of warfarin. The authors found it to be a potential method for on-demand compounding right at the bedside [131].
With the 3D printing technique, the limitations of the conventional manufacturing process could be eliminated. In addition, a precise amount of API could be printed, and it was feasible to print fixed API combinations. These attributes made the oromucosal films especially interesting for the administration of potent APIs, for example, for the treatment of cardiovascular disorders, schizophrenia, or migraine. A huge advantage of the printing technique was the possibility to integrate safety features in the form of QR codes with the dosage form. However, some challenges such as increased dosing remained even with the use of 3D printing [132]. Some other limitations were the dose inaccuracy and the liquid formulation by artificial instruments based on a research group [133]. The published research on the fabricated orodispersible films can be found in Table 4.

2.4. Implants

Levofloxacin-containing PLA implants were designed with inkjet printing. The manufactured samples were 10 mm in width and rounded. In this research, a complex release profile was demonstrated in the 100-day monitoring period when one pulse of release appeared from the 5th to 25th day, and another pulse began at the 50th day and ended at the 80th day, with a lag time of 25 days between the two pulses, wherein a steady state of release was observed at about 5 µg/mL [141].
Rifampicin and isoniazid-containing multi-layered concentric cylindrical implants were fabricated against tuberculosis. The multi-layered concentric cylinder was divided into four layers from the center to the periphery and the APIs were distributed individually into the different layers in a specific sequence of isoniazid–rifampicin–isoniazid–rifampicin. The dissolution tests proved that the API liberation takes place orderly from the outside to the center and the peak concentrations were between 8 and 12 days. In this study, a programmed release multi-drug implant with a complex construction was fabricated by 3D printing [142].
A research group prepared dexamethasone-containing tailored drug delivery platforms where two distinct designs—structure A: rolled and sealed; structure B: layer-by-layer—were extrusion printed. As the API liberation was continuous for more than 4 months, these samples could be used as implants [143].
Genina et al. manufactured intrauterine device and subcutaneous rods from ethylene vinyl acetate (EVA) copolymer with FDM printing. The samples were containing indomethacin as a model API and with the device the drug dissolution was over 30 days. A long-acting 3D-printed implantable system was built [144].
In a study, levofloxacin and tobramycin-containing implants were fabricated for the treatment of osteomyelitis. A multi-layered concentric cylinder construction was created by powder-based inkjet printing. A sustained and programmed drug delivery system was provided [145].
In a study, the effect of the used polymers on the drug release profile of quinine was examined as a model drug. The used polymers were Eudragit® RS, PCL, PLLA, and EC and affected the dissolution profile of the samples. The fastest relative drug release was observed from PCL where the dissolved API amount was approximately 76% in 51 days and the lowest from Eudragit RS and EC with less than 5% of quinine release in 78 and 100 days, respectively [146].
Qamar et al. manufactured an implantable mesh for the treatment of hernia. PP and PVA meshes were produced with distinct pore size, shape, and thread thickness. The meshes were filled with ciprofloxacin for the management of hernia. Based on the research, animals implanted with ciprofloxacin HCl loaded meshes exhibited fewer fluctuations in body temperature and faster wound healing [147].
The purpose of a study was to demonstrate the applicability of 3D printing methods for the fabrication of patient-specific fixation implants that allow localized drug delivery. The 3D printing was used to fabricate gentamicin and methotrexate loaded fixation devices, including screws, pins, and bone plates [148].
In a study, PLLA samples were printed using a special Zcorp Zprinter 650 then immersed into the solution of various anticancer drugs such as cisplatin, ifosfamid, methotrexate, or doxorubicin and finally dried. The proposed 3D-printed drug delivery system could simultaneously realize individual local chemotherapy, multi-drug delivery, long-term sustainable drug release, and non-reoperation in osteosarcoma treatment [149].
Ciprofloxacin containing PLA implants were fabricated with the combination of semi-solid extrusion and fused-deposition modeling for the treatment of bone infections. The authors found this method more adequate than the conventional method for manufacturing [150].
The 3D printing technologies have promising potential for the manufacture of sophisticated drug implants and patient-specific macro-porosity implants with personalized drug release behavior. In general, the currently commercially available implants lack the personalization of the treatment and consideration of several issues such as anatomical differences, age, genders, and disease condition [151]. A research group determined that 3D-printed implants could display complex drug release patterns compared to conventionally fabricated drug implants [141]. The manufacturing of 3D printing provided the possibility to produce smaller, less invasive, and more site-specific implants compared to the conventional ones [152]. Even though the clinical data—to support the routine use of the 3D-printed customized implants—are currently limited, the patient-specific implants may gain more attention and popularity in the prophylaxis and treatment of diseases such as complicated bone infections and bone tuberculosis in the near future when the digital and manufacturing technology advances further [150].
Some published research on the produced implants can be found in Table 5.

2.5. TTS

Anti-acne drug loaded masks/patches were fabricated by Goyanes et al., but as this system provides transdermal delivery, we decided to subgroup the research here. In the research, salicylic-acid-containing filaments were used for the FDM 3D printing but the API showed significant thermal degradation. The manufacturing by SLA contained a higher amount of drug and showed no drug degradation, so the researchers found this method more adequate [160].
Yi et al. manufactured a 3D-printed biodegradable patch with a versatile shape and incorporated a high drug concentration for the achievement of a controlled drug release profile. The patches composed of poly(lactide-co-glycolide), polycaprolactone, and 5-fluorouracil were the antitumor agent. With the use of 3D printing technology, the geometry of the patch and the drug release kinetics could be manipulated. The patches were flexible, and released the drug over four weeks with minimized side effects [161].
A research group developed an electro hydrodynamic (EHD) printing technique to fabricate antibiotic-containing patches using polycaprolactone (PCL), polyvinyl pyrrolidone (PVP), and their composite system (PVP-PCL). Drug loaded 3D patches possessed perfectly aligned fibers giving rise to fibrous strut orientation, variable inter-strut pore size, and controlled film width (via layering). The used polymer type and the printed patch void size affected the dissolution profiles [162].
If we compared the advantages and the limitations, we found that the conventional manufacturing process involves solvent evaporation and multiple steps which were often time consuming in comparison with the 3D printing method. In case of the TTSs, the adhesion was a critical point and the skin could be undulating and curved, especially on the nose or head, which the conventional TTSs could not take into consideration [151]. The combination of 3D scanning and 3D printing had the potential to create personalized drug loaded devices, adapted in shape and size to individual patients [160]. The 3D printing of differently shaped patches demonstrated the ability to manipulate the drug release by altering the surface area which allowed precise control of the liberation amount and placement of the API, high efficacy, and minimized systemic toxicity [161]. A few published studies on the created TTSs can be found in Table 6.

2.6. Microneedles

In the early research of Ovsianikov et al., a placebo microneedle was developed by femtosecond laser two photon polymerization 3D printing technology [164]. In 2013, an amphotericin B containing microneedle was created by the combination of visible light dynamic mask micro stereolithography, micro molding, and piezoelectric inkjet printing. Based on the results, the printing process was found to be a scalable approach that could be used to incorporate pharmacologic agents, even with complex solubility profiles, within microneedles [165]. The same researchers fabricated miconazole-containing microneedles with the same technology but with a special polymer called Gantrez® AN 169 BF (poly(methyl vinyl ether-co-maleic anhydride)). The manufactured sample had potential use in transdermal treatment of cutaneous fungal infections [166].
A dacarbazine-containing drug delivery system was produced which could be used for the therapy of skin cancer locally. For the manufacturing, a special 3D printing process was used named as multi-material micro stereolithography (μSL). First, the microneedle array was built and then the API was added with blending due to the crosslinking effect of the polymer (Figure 13) [167].
Insulin polymeric layers on metal microneedles were constructed by Ross et al. The dissolution profiles showed rapid insulin release rates in the first 20 min, suggesting that solid-state insulin delivery via microneedles was feasible [168].
Lim et al. used 3D printing for manufacturing a dual-function microneedle array on personalized curved surfaces (microneedle splint) for drug delivery and splinting of the affected finger. Sufficient penetration efficiency was achieved, and the final microneedle splint showed biocompatibility. A significantly higher amount of diclofenac permeated through the skin with the use of the microneedle splint as compared to intact skin. The fabricated microneedle splint could thus be a potential new approach to treat trigger finger via personalized splinting without affecting normal hand function [169].
Two research groups produced insulin-releasing microneedles from Dental SG with the use of inkjet printing and the SLA technique. In the case of the research of Pere et al., the API was released rapidly within 30 min [170]. Economidou et al. reached better glucose levels and hypoglycemia control [171].
One of the biggest advantages of 3D printing was that the technique allowed the incorporation of pharmacologic agents even with a complex solubility profile into the microneedles for patient needs [165]. The manufactured microneedles provided accurate dosing which also maintained the mechanical strength in comparison with the existing microneedles, e.g., made of metal. Furthermore, microneedles that have extremely fine tips in nano scale allowed good penetration of skin and potentially cell targeted delivery [172]. A few studies on the 3D-printed microneedles can be found in Table 7.

2.7. Vaginal Drug Delivery Systems

Even though this subsection consists of numerous drug delivery systems, we would like to discuss them together for easier accessibility.
The article of Genina et al. was already discussed in the section of implants because two distinct type of drug delivery systems were fabricated: intrauterine device (IUD) and subcutaneous rods [144].
Tappa et al. designed estrogen and progesterone-containing PCL filaments and FDM printed into different structures: mesh, IUD, and subdermal implant. As the FDM printing with PCL was possible at lower temperatures (around 130 °C), the API remained stable based on the TG/DSC results. This was the first study to offer data on hormone-loaded 3D-printed constructs [176].
Bioadhesive vaginal films were produced by Varan et al., where the used APIs were paclitaxel and cidofovir, which had antiviral efficacy and used for the treatment of cervical cancer locally. The basic idea was to increase the solubility of the paclitaxel with a cyclodextrin complex while cidofovir was encapsulated in PCL nanoparticles to manufacture an ink [177].
Our research group fabricated a vaginal ring as a carrier system for the treatment of bacterial vaginosis. For the manufacturing, FDM 3D printing was used and then these pre-printed samples were easily filled with the jellified APIs (vaginal gels) depending on the patient’s need. (Figure 14). This article focused on avoiding the loss and decomposition of API through the printing process and manufacturing a drug delivery system, which could be directly and rapidly printed at the bedside or in the pharmacy as individualized medication [178].
The currently available approaches did not consider the specific anatomy of each patient, the patient’s medical conditions, age, and gender specificity which limited the effective therapy whereas every patient is unique and should require different doses of drugs and hormones [179]. In addition, other factors such as variability in drug absorption related with menstrual cycle, menopause, and pregnancy were different among patients [180]. Therefore, patient-specific systems are needed to provide personalized shape, size, and tailored drug release to improve the efficacy and to increase compliance. The 3D-printed delivery system in this regard can provide personalized geometry, snug fit, increase efficacy, improve patient compliance, and prevent the post-surgical complications [151].
Some research on the manufacturing of vaginal drug delivery systems can be found in Table 8. The research was classified based on the publication year and then in alphabetical order. In the first column, the type of the drug delivery system is described.

2.8. Micro and Nanoscale Dosage Forms

Another area that concerns the researchers is the possible manufacturing of micro and nanoscale drug delivery systems. In a study, a poorly soluble model drug was used to construct a 10% folic acid containing nanosuspension and the printing was performed on an inkjet-based microdosing dispenser head. In the research, the authors found this method adequate for the incorporation of poorly soluble drugs to increase the oral absorption [183].
Scoutaris et al. produced poorly soluble felodipine and PVP-containing solution for inkjet printing. Based on the authors’ research, inkjet printing could be used to prepare this novel drug dosage form consisting of micro-sized dried deposits from sprayed picolitre droplets containing a drug on a substrate. The novelty of the work was that a scalable dosage form could be produced whereby many droplets could be produced to achieve a dissolution profile equivalent to conventional bulk dosage formulations [184].
In another study, rifampicin and PLGA-containing inks were fabricated for micro-pattern printing on a glass or titanium carrier. This research combines special ink formulations in microscale range and the author’s idea was to use these micro-patterns in orthopedic surgeries [185].
Paclitaxel-containing PLGA inks were created for piezoelectric inkjet printing where four different shapes were printed. Microparticles with diverse geometries exhibited non-similar drug release rates mainly due to nonidentical surface areas [186].
Some studies on the manufacturing of micro and nanoscale systems can be found in Table 9. The research was classified based on the publication year and then in alphabetical order. In the first column, the type of the drug delivery system was described.

2.9. Other

A few other interesting but not mandatory ‘drug delivery systems’ are discussed in this section. Kitson et al. used ibuprofen with a modified Prusa FDM 3D printer where the API was deposited in a pre-printed vessel. In this case, the API could be directly printed to a drug delivery system, e.g., tablet or capsule, to receive a ready-to-use drug dosage form [187].
Long et al. 3D-printed a chitosan-pectin hydrogel. To the gel, lidocaine hydrochloride was incorporated as a potential wound dressing candidate. The scaffolds were printed using an extrusion-based 3D printer using a mechanical positive displacement dispensing system followed by lyophilization. The research confirmed the possible manufacturing of hydrogels with 3D printing [188].
Seoane-Viaño et al. fabricated tacrolimus-containing rectal suppositories with semi-solid extrusion-based 3D printing. For the printing, Gelucire 44/14 and coconut oil were used. The designed drug dosage form could contribute to a long-lasting effect on the treatment of colitis [189,190].

3. Future Perspective

Three-dimensional printing enables the manufacture of personalized drug delivery systems. With the use of this modern technique, cost-effectiveness, simplified production, complex formulations, and increased opportunities for collaboration can be achieved. The high flexibility provides the production of a multitude of drug products with tailored release profiles and designs [20]. Additionally, 3D printing has to challenge some limitations in the future. As 3D printing right now cannot compete with the speed of the conventional drug manufacturing method, in the future the improvement in the printing speed and resolution has to be adjusted which can also reduce the energy consumption and cost as a recent, huge concern. The printed product dimension accuracy and scale size (e.g., micro or nanoscale) must be improved also. As 3D printing technology is limited in terms of materials, the manufacturing of novel polymers, inks, etc., are still required and necessary for the proper manufacturing [191]. Even though the FDA approved Spritam® in 2015, the used ZipDose® technology was similar to traditional powder compaction that could help the process of approval. At present, there are no fixed guidelines for the regulation of 3D-printed pharmaceutical dosage forms, so the researchers have to face regulatory concerns [151]. Another interesting area is bioprinting where living cells, tissues, or organs can be printed and exploratory studies of bioprinting in pharmaceutics have shown promising applications of this technique, for example, in the field of target identification and validation or in vitro efficacy assessment [19].

4. Conclusions

In conclusion, 3D printing represents a very interesting and modern technique in the field of drug manufacturing which is about to revolutionize the health industry. Three-dimensional printing is a layer-by-layer, automated process, which enables the manufacturing of complex, personalized products on-demand. In the last decade, the number of publications multiplied year-by-year. The FDA approved the first and only 3D-printed drug in 2015, which supported the commercial feasibility of this technology. In the last two decades, dozens of research groups aimed to manufacture different drug dosage forms, e.g., tablets, capsules, implants, even rectal suppositories by 3D printing to improve the safety, efficacy, and tolerability of medicines and provide individualized therapy for those most in need.

Author Contributions

I.B. conceived the overall project, and wrote the conclusion part. Z.U. wrote the first part of the introduction and P.F. wrote the second half of the introduction. P.A. wrote Section 2 and was responsible for drawing the figures. All authors have read and agreed to the published version of the manuscript.

Funding

The publication is supported by the GINOP-2.3.4-15-2020-00008 project. The project is co-financed by the European Union and the European Regional Development Fund. The research was supported by the Thematic Excellence Programme (TKP2020-IKA-04) of the Ministry for Innovation and Technology in Hungary. Project no. TKP2021-EGA-18 has been implemented with the support provided from the National Research, Development and Innovation Fund of Hungary, financed under the TKP2021-EGA funding scheme.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Norman, J.; Madurawe, R.D.; Moore, C.M.V.; Khan, M.A.; Khairuzzaman, A. A new chapter in pharmaceutical manufacturing: 3D-printed drug products. Adv. Drug Deliv. Rev. 2017, 108, 39–50. [Google Scholar] [CrossRef] [PubMed]
  2. Stansbury, J.W.; Idacavage, M.J. 3D printing with polymers: Challenges among expanding options and opportunities. Dent. Mater. 2016, 32, 54–64. [Google Scholar] [CrossRef] [PubMed]
  3. 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]
  4. Alhnan, M.A.; Okwuosa, T.C.; Sadia, M.; Wan, K.W.; Ahmed, W.; Arafat, B. Emergence of 3D Printed Dosage Forms: Opportunities and Challenges. Pharm. Res. 2016, 33, 1817–1832. [Google Scholar] [CrossRef] [PubMed]
  5. Singh, S.; Ramakrishna, S.; Singh, R. Material issues in additive manufacturing: A review. J. Manuf. Process. 2017, 25, 185–200. [Google Scholar] [CrossRef]
  6. Elkasabgy, N.A.; Mahmoud, A.A.; Maged, A. 3D printing: An appealing route for customized drug delivery systems. Int. J. Pharm. 2020, 588, 119732. [Google Scholar] [CrossRef]
  7. Okafor-Muo, O.L.; Hassanin, H.; Kayyali, R.; ElShaer, A. 3D Printing of Solid Oral Dosage Forms: Numerous Challenges with Unique Opportunities. J. Pharm. Sci. 2020, 109, 3535–3550. [Google Scholar] [CrossRef]
  8. Aquino, R.P.; Barile, S.; Grasso, A.; Saviano, M. Envisioning smart and sustainable healthcare: 3D Printing technologies for personalized medication. Futures 2018, 103, 35–50. [Google Scholar] [CrossRef]
  9. Ursan, I.; Chiu, L.; Pierce, A. Three-dimensional drug printing: A structured review. J. Am. Pharm. Assoc. 2013, 53, 136–144. [Google Scholar] [CrossRef]
  10. Sandler, N.; Preis, M. Printed Drug-Delivery Systems for Improved Patient Treatment. Trends Pharmacol. Sci. 2016, 37, 1070–1080. [Google Scholar] [CrossRef]
  11. Moulton, S.E.; Wallace, G.G. 3-dimensional (3D) fabricated polymer based drug delivery systems. J. Control. Release 2014, 193, 27–34. [Google Scholar] [CrossRef] [Green Version]
  12. Jamróz, W.; Szafraniec, J.; Kurek, M.; Jachowicz, R. 3D printing in pharmaceutical and medical applications. Pharm. Res. 2018, 35, 176. [Google Scholar] [CrossRef] [Green Version]
  13. Kotta, S.; Nair, A.; Alsabeelah, N. 3D Printing Technology in Drug Delivery: Recent Progress and Application. Curr. Pharm. Des. 2018, 24, 5039–5048. [Google Scholar] [CrossRef]
  14. Mohapatra, S.; Kar, R.K.; Biswal, P.K.; Bindhani, S. Approaches of 3D printing in current drug delivery. Sensors Int. 2021, 3, 100146. [Google Scholar] [CrossRef]
  15. Cunha-Filho, M.; Araújo, M.R.; Gelfuso, G.M.; Gratieri, T. FDM 3D printing of modified drug-delivery systems using hot melt extrusion: A new approach for individualized therapy. Ther. Deliv. 2017, 8, 957–966. [Google Scholar] [CrossRef]
  16. Gueche, Y.A.; Sanchez-Ballester, N.M.; Cailleaux, S.; Bataille, B.; Soulairol, I. Selective laser sintering (Sls), a new chapter in the production of solid oral forms (sofs) by 3d printing. Pharmaceutics 2021, 13, 1212. [Google Scholar] [CrossRef]
  17. Wang, J.; Goyanes, A.; Gaisford, S.; Basit, A.W. Stereolithographic (SLA) 3D printing of oral modified-release dosage forms. Int. J. Pharm. 2016, 503, 207–212. [Google Scholar] [CrossRef]
  18. Daly, R.; Harrington, T.S.; Martin, G.D.; Hutchings, I.M. Inkjet printing for pharmaceutics—A review of research and manufacturing. Int. J. Pharm. 2015, 494, 554–567. [Google Scholar] [CrossRef] [Green Version]
  19. Peng, W.; Datta, P.; Ayan, B.; Ozbolat, V.; Sosnoski, D.; Ozbolat, I.T. 3D bioprinting for drug discovery and development in pharmaceutics. Acta Biomater. 2017, 57, 26–46. [Google Scholar] [CrossRef]
  20. Trenfield, S.J.; Awad, A.; Madla, C.M.; Hatton, G.B.; Firth, J.; Goyanes, A.; Gaisford, S.; Basit, A.W. Shaping the future: Recent advances of 3D printing in drug delivery and healthcare. Expert Opin. Drug Deliv. 2019, 16, 1081–1094. [Google Scholar] [CrossRef]
  21. Rohani Shirvan, A.; Bashari, A.; Hemmatinejad, N. New insight into the fabrication of smart mucoadhesive buccal patches as a novel controlled-drug delivery system. Eur. Polym. J. 2019, 119, 541–550. [Google Scholar] [CrossRef]
  22. Domsta, V.; Seidlitz, A. 3d-printing of drug-eluting implants: An overview of the current developments described in the literature. Molecules 2021, 26, 4066. [Google Scholar] [CrossRef] [PubMed]
  23. Khatri, P.; Shah, M.K.; Vora, N. Formulation strategies for solid oral dosage form using 3D printing technology: A mini-review. J. Drug Deliv. Sci. Technol. 2018, 46, 148–155. [Google Scholar] [CrossRef]
  24. Economidou, S.N.; Lamprou, D.A.; Douroumis, D. 3D printing applications for transdermal drug delivery. Int. J. Pharm. 2018, 544, 415–424. [Google Scholar] [CrossRef]
  25. Wu, B.M.; Borland, S.W.; Giordano, R.A.; Cima, L.G.; Sachs, E.M.; Cima, M.J. Solid free-form fabrication of drug delivery devices. J. Control. Release 1996, 40, 77–87. [Google Scholar] [CrossRef]
  26. Katstra, W.E.; Palazzolo, R.D.; Rowe, C.W.; Giritlioglu, B.; Teung, P.; Cima, M.J. Oral dosage forms fabricated by Three Dimensional Printing. J. Control. Release 2000, 66, 1–9. [Google Scholar] [CrossRef]
  27. Lee, K.J.; Kang, A.; Delfino, J.J.; West, T.G.; Chetty, D.; Monkhouse, D.C.; Yoo, J. Evaluation of Critical Formulation Factors in the Development of a Rapidly Dispersing Captopril Oral Dosage Form. Drug Dev. Ind. Pharm. 2003, 29, 967–979. [Google Scholar] [CrossRef]
  28. Wang, C.C.; Tejwani, M.R.; Roach, W.J.; Kay, J.L.; Yoo, J.; Surprenant, H.L.; Monkhouse, D.C.; Pryor, T.J. Development of near zero-order release dosage forms using three-dimensional printing (3-DPTM) technology. Drug Dev. Ind. Pharm. 2006, 32, 367–376. [Google Scholar] [CrossRef]
  29. Gbureck, U.; Vorndran, E.; Müller, F.A.; Barralet, J.E. Low temperature direct 3D printed bioceramics and biocomposites as drug release matrices. J. Control. Release 2007, 122, 173–180. [Google Scholar] [CrossRef]
  30. Yu, D.G.; Yang, X.L.; Huang, W.D.; Liu, J.; Wang, Y.G.; Xu, H. Tablets With Material Gradients Fabricated by Three-Dimensional Printing. J. Pharm. Biomed. Anal. 2007, 96, 2446–2456. [Google Scholar] [CrossRef]
  31. Yu, D.G.; Branford-White, C.; Ma, Z.H.; Zhu, L.M.; Li, X.Y.; Yang, X.L. Novel drug delivery devices for providing linear release profiles fabricated by 3DP. Int. J. Pharm. 2009, 370, 160–166. [Google Scholar] [CrossRef]
  32. Khaled, S.A.; Burley, J.C.; Alexander, M.R.; Roberts, C.J. Desktop 3D printing of controlled release pharmaceutical bilayer tablets. Int. J. Pharm. 2014, 461, 105–111. [Google Scholar] [CrossRef]
  33. Bhattacharya, S.; Singh, S.K.; Shrestha, S.; Baghel, Y.S.; Maity, D.; Kumar, A.; Gupta, G.D.; Kumar, R. Recent Findings and Development of 3D Printing Technology in Pharmaceutical Formulation Development: An Extensive Review. Int. J. Drug Dev. Res. 2019, 11, 1–14. [Google Scholar]
  34. Goyanes, A.; Buanz, A.B.M.; Hatton, G.B.; Gaisford, S.; Basit, A.W. 3D printing of modified-release aminosalicylate (4-ASA and 5-ASA) tablets. Eur. J. Pharm. Biopharm. 2015, 89, 157–162. [Google Scholar] [CrossRef]
  35. Goyanes, A.; Wang, J.; Buanz, A.; Martínez-Pacheco, R.; Telford, R.; Gaisford, S.; Basit, A.W. 3D Printing of Medicines: Engineering Novel Oral Devices with Unique Design and Drug Release Characteristics. Mol. Pharm. 2015, 12, 4077–4084. [Google Scholar] [CrossRef] [Green Version]
  36. Khaled, S.A.; Burley, J.C.; Alexander, M.R.; Yang, J.; Roberts, C.J. 3D printing of tablets containing multiple drugs with defined release profiles. Int. J. Pharm. 2015, 494, 643–650. [Google Scholar] [CrossRef]
  37. Khaled, S.A.; Burley, J.C.; Alexander, M.R.; Yang, J.; Roberts, C.J. 3D printing of five-in-one dose combination polypill with defined immediate and sustained release profiles. J. Control. Release 2015, 217, 308–314. [Google Scholar] [CrossRef]
  38. Boetker, J.; Water, J.J.; Aho, J.; Arnfast, L.; Bohr, A.; Rantanen, J. Modifying release characteristics from 3D printed drug-eluting products. Eur. J. Pharm. Sci. 2016, 90, 47–52. [Google Scholar] [CrossRef]
  39. Okwuosa, T.C.; Stefaniak, D.; Arafat, B.; Isreb, A.; Wan, K.W.; Alhnan, M.A. A Lower Temperature FDM 3D Printing for the Manufacture of Patient-Specific Immediate Release Tablets. Pharm. Res. 2016, 33, 2704–2712. [Google Scholar] [CrossRef]
  40. Sadia, M.; Sośnicka, A.; Arafat, B.; Isreb, A.; Ahmed, W.; Kelarakis, A.; Alhnan, M.A. Adaptation of pharmaceutical excipients to FDM 3D printing for the fabrication of patient-tailored immediate release tablets. Int. J. Pharm. 2016, 513, 659–668. [Google Scholar] [CrossRef]
  41. Goyanes, A.; Robles Martinez, P.; Buanz, A.; Basit, A.W.; Gaisford, S. Effect of geometry on drug release from 3D printed tablets. Int. J. Pharm. 2015, 494, 657–663. [Google Scholar] [CrossRef] [PubMed]
  42. Goyanes, A.; Chang, H.; Sedough, D.; Hatton, G.B.; Wang, J.; Buanz, A.; Gaisford, S.; Basit, A.W. Fabrication of controlled-release budesonide tablets via desktop (FDM) 3D printing. Int. J. Pharm. 2015, 496, 414–420. [Google Scholar] [CrossRef] [PubMed]
  43. Acosta-Vélez, G.F.; Linsley, C.S.; Craig, M.C.; Wu, B.M. Photocurable bioink for the inkjet 3D pharming of hydrophilic drugs. Bioengineering 2017, 4, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Beck, R.C.R.; Chaves, P.S.; Goyanes, A.; Vukosavljevic, B.; Buanz, A.; Windbergs, M.; Basit, A.W.; Gaisford, S. 3D printed tablets loaded with polymeric nanocapsules: An innovative approach to produce customized drug delivery systems. Int. J. Pharm. 2017, 528, 268–279. [Google Scholar] [CrossRef]
  45. Chai, X.; Chai, H.; Wang, X.; Yang, J.; Li, J.; Zhao, Y.; Cai, W.; Tao, T.; Xiang, X. Fused deposition modeling (FDM) 3D printed tablets for intragastric floating delivery of domperidone. Sci. Rep. 2017, 7, 2829. [Google Scholar] [CrossRef] [Green Version]
  46. Genina, N.; Boetker, J.P.; Colombo, S.; Harmankaya, N.; Rantanen, J.; Bohr, A. Anti-tuberculosis drug combination for controlled oral delivery using 3D printed compartmental dosage forms: From drug product design to in vivo testing. J. Control. Release 2017, 268, 40–48. [Google Scholar] [CrossRef]
  47. Li, Q.; Wen, H.; Jia, D.; Guan, X.; Pan, H.; Yang, Y.; Yu, S.; Zhu, Z.; Xiang, R.; Pan, W. Preparation and investigation of controlled-release glipizide novel oral device with three-dimensional printing. Int. J. Pharm. 2017, 525, 5–11. [Google Scholar] [CrossRef]
  48. Fina, F.; Goyanes, A.; Madla, C.M.; Awad, A.; Trenfield, S.J.; Kuek, J.M.; Patel, P.; Gaisford, S.; Basit, A.W. 3D printing of drug-loaded gyroid lattices using selective laser sintering. Int. J. Pharm. 2018, 547, 44–52. [Google Scholar] [CrossRef]
  49. Holländer, J.; Hakala, R.; Suominen, J.; Moritz, N.; Yliruusi, J.; Sandler, N. 3D printed UV light cured polydimethylsiloxane devices for drug delivery. Int. J. Pharm. 2018, 544, 433–442. [Google Scholar] [CrossRef]
  50. Kollamaram, G.; Croker, D.M.; Walker, G.M.; Goyanes, A.; Basit, A.W.; Gaisford, S. Low temperature fused deposition modeling (FDM) 3D printing of thermolabile drugs. Int. J. Pharm. 2018, 545, 144–152. [Google Scholar] [CrossRef] [Green Version]
  51. Li, Q.; Guan, X.; Cui, M.; Zhu, Z.; Chen, K.; Wen, H.; Jia, D.; Hou, J.; Xu, W.; Yang, X.; et al. Preparation and investigation of novel gastro-floating tablets with 3D extrusion-based printing. Int. J. Pharm. 2018, 535, 325–332. [Google Scholar] [CrossRef]
  52. Sadia, M.; Arafat, B.; Ahmed, W.; Forbes, R.T.; Alhnan, M.A. Channelled tablets: An innovative approach to accelerating drug release from 3D printed tablets. J. Control. Release 2018, 269, 355–363. [Google Scholar] [CrossRef]
  53. Scoutaris, N.; Ross, S.A.; Douroumis, D. 3D Printed “Starmix” Drug Loaded Dosage Forms for Paediatric Applications. Pharm. Res. 2018, 35, 34. [Google Scholar] [CrossRef]
  54. Verstraete, G.; Samaro, A.; Grymonpré, W.; Vanhoorne, V.; Van Snick, B.; Boone, M.N.; Hellemans, T.; Van Hoorebeke, L.; Remon, J.P.; Vervaet, C. 3D printing of high drug loaded dosage forms using thermoplastic polyurethanes. Int. J. Pharm. 2018, 536, 318–325. [Google Scholar] [CrossRef]
  55. Goyanes, A.; Madla, C.M.; Umerji, A.; Duran Piñeiro, G.; Giraldez Montero, J.M.; Lamas Diaz, M.J.; Gonzalez Barcia, M.; Taherali, F.; Sánchez-Pintos, P.; Couce, M.L.; et al. Automated therapy preparation of isoleucine formulations using 3D printing for the treatment of MSUD: First single-centre, prospective, crossover study in patients. Int. J. Pharm. 2019, 567, 118497. [Google Scholar] [CrossRef]
  56. Öblom, H.; Zhang, J.; Pimparade, M.; Speer, I.; Preis, M.; Repka, M.; Sandler, N. 3D-Printed Isoniazid Tablets for the Treatment and Prevention of Tuberculosis—Personalized Dosing and Drug Release. AAPS PharmSciTech 2019, 20, 52. [Google Scholar] [CrossRef] [Green Version]
  57. Robles-Martinez, P.; Xu, X.; Trenfield, S.J.; Awad, A.; Goyanes, A.; Telford, R.; Basit, A.W.; Gaisford, S. 3D printing of a multi-layered polypill containing six drugs using a novel stereolithographic method. Pharmaceutics 2019, 11, 274. [Google Scholar] [CrossRef] [Green Version]
  58. Zidan, A.; Alayoubi, A.; Asfari, S.; Coburn, J.; Ghammraoui, B.; Cruz, C.N.; Ashraf, M. Development of mechanistic models to identify critical formulation and process variables of pastes for 3D printing of modified release tablets. Int. J. Pharm. 2019, 555, 109–123. [Google Scholar] [CrossRef]
  59. Awad, A.; Yao, A.; Trenfield, S.J.; Goyanes, A.; Gaisford, S.; Basit, A.W. 3D printed tablets (Printlets) with braille and moon patterns for visually impaired patients. Pharmaceutics 2020, 12, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Dumpa, N.R.; Bandari, S.; Repka, M.A. Novel gastroretentive floating pulsatile drug delivery system produced via hot-melt extrusion and fused deposition modeling 3D printing. Pharmaceutics 2020, 12, 52. [Google Scholar] [CrossRef] [Green Version]
  61. Gioumouxouzis, C.I.; Tzimtzimis, E.; Katsamenis, O.L.; Dourou, A.; Markopoulou, C.; Bouropoulos, N.; Tzetzis, D.; Fatouros, D.G. Fabrication of an osmotic 3D printed solid dosage form for controlled release of active pharmaceutical ingredients. Eur. J. Pharm. Sci. 2020, 143, 105176. [Google Scholar] [CrossRef] [PubMed]
  62. Karavasili, C.; Gkaragkounis, A.; Moschakis, T.; Ritzoulis, C.; Fatouros, D.G. Pediatric-friendly chocolate-based dosage forms for the oral administration of both hydrophilic and lipophilic drugs fabricated with extrusion-based 3D printing. Eur. J. Pharm. Sci. 2020, 147, 105291. [Google Scholar] [CrossRef] [PubMed]
  63. Real, J.P.; Barberis, M.E.; Camacho, N.M.; Sánchez Bruni, S.; Palma, S.D. Design of novel oral ricobendazole formulation applying melting solidification printing process (MESO-PP): An innovative solvent-free alternative method for 3D printing using a simplified concept and low temperature. Int. J. Pharm. 2020, 587, 119653. [Google Scholar] [CrossRef] [PubMed]
  64. Tsintavi, E.; Rekkas, D.M.; Bettini, R. Partial tablet coating by 3D printing. Int. J. Pharm. 2020, 581, 119298. [Google Scholar] [CrossRef] [PubMed]
  65. Xu, X.; Robles-Martinez, P.; Madla, C.M.; Joubert, F.; Goyanes, A.; Basit, A.W.; Gaisford, S. Stereolithography (SLA) 3D printing of an antihypertensive polyprintlet: Case study of an unexpected photopolymer-drug reaction. Addit. Manuf. 2020, 33, 101071. [Google Scholar] [CrossRef]
  66. Algahtani, M.S.; Mohammed, A.A.; Ahmad, J.; Abdullah, M.M.; Saleh, E. 3d printing of dapagliflozin containing self-nanoemulsifying tablets: Formulation design and in vitro characterization. Pharmaceutics 2021, 13, 993. [Google Scholar] [CrossRef]
  67. Tan, Y.J.N.; Yong, W.P.; Low, H.R.; Kochhar, J.S.; Khanolkar, J.; Lim, T.S.E.; Sun, Y.; Wong, J.Z.E.; Soh, S. Customizable drug tablets with constant release profiles via 3D printing technology. Int. J. Pharm. 2021, 598, 120370. [Google Scholar] [CrossRef]
  68. Yang, Y.; Xu, Y.; Wei, S.; Shan, W. Oral preparations with tunable dissolution behavior based on selective laser sintering technique. Int. J. Pharm. 2021, 593, 120127. [Google Scholar] [CrossRef]
  69. Jamróz, W.; Kurek, M.; Czech, A.; Szafraniec, J.; Gawlak, K.; Jachowicz, R. 3D printing of tablets containing amorphous aripiprazole by filaments co-extrusion. Eur. J. Pharm. Biopharm. 2018, 131, 44–47. [Google Scholar] [CrossRef]
  70. Raijada, D.; Wac, K.; Greisen, E.; Rantanen, J.; Genina, N. Integration of personalized drug delivery systems into digital health. Adv. Drug Deliv. Rev. 2021, 176, 113857. [Google Scholar] [CrossRef]
  71. Rautamo, M.; Kvarnström, K.; Sivén, M.; Airaksinen, M.; Lahdenne, P.; Sandler, N. Benefits and prerequisites associated with the adoption of oral 3D-printed medicines for pediatric patients: A focus group study among healthcare professionals. Pharmaceutics 2020, 12, 229. [Google Scholar] [CrossRef] [Green Version]
  72. Cui, M.; Pan, H.; Fang, D.; Qiao, S.; Wang, S.; Pan, W. Fabrication of high drug loading levetiracetam tablets using semi-solid extrusion 3D printing. J. Drug Deliv. Sci. Technol. 2020, 57, 101683. [Google Scholar] [CrossRef]
  73. Rowe, C.W.; Katstra, W.E.; Palazzolo, R.D.; Giritlioglu, B.; Teung, P.; Cima, M.J. Multimechanism oral dosage forms fabricated by three dimensional printing. J. Control. Release 2000, 66, 11–17. [Google Scholar] [CrossRef]
  74. Salmoria, G.V.; Klauss, P.; Zepon, K.; Kanis, L.A.; Roesler, C.R.M.; Vieira, L.F. Development of functionally-graded reservoir of PCL/PG by selective laser sintering for drug delivery devices: This paper presents a selective laser sintering-fabricated drug delivery system that contains graded progesterone content. Virtual Phys. Prototyp. 2012, 7, 107–115. [Google Scholar] [CrossRef]
  75. Goyanes, A.; Buanz, A.B.M.; Basit, A.W.; Gaisford, S. Fused-filament 3D printing (3DP) for fabrication of tablets. Int. J. Pharm. 2014, 476, 88–92. [Google Scholar] [CrossRef]
  76. Pietrzak, K.; Isreb, A.; Alhnan, M.A. A flexible-dose dispenser for immediate and extended release 3D printed tablets. Eur. J. Pharm. Biopharm. 2015, 96, 380–387. [Google Scholar] [CrossRef]
  77. Skowyra, J.; Pietrzak, K.; Alhnan, M.A. Fabrication of extended-release patient-tailored prednisolone tablets via fused deposition modelling (FDM) 3D printing. Eur. J. Pharm. Sci. 2015, 68, 11–17. [Google Scholar] [CrossRef]
  78. Alhijjaj, M.; Belton, P.; Qi, S. An investigation into the use of polymer blends to improve the printability of and regulate drug release from pharmaceutical solid dispersions prepared via fused deposition modeling (FDM) 3D printing. Eur. J. Pharm. Biopharm. 2016, 108, 111–125. [Google Scholar] [CrossRef] [Green Version]
  79. Goyanes, A.; Kobayashi, M.; Martínez-Pacheco, R.; Gaisford, S.; Basit, A.W. Fused-filament 3D printing of drug products: Microstructure analysis and drug release characteristics of PVA-based caplets. Int. J. Pharm. 2016, 514, 290–295. [Google Scholar] [CrossRef]
  80. Clark, E.A.; Alexander, M.R.; Irvine, D.J.; Roberts, C.J.; Wallace, M.J.; Sharpe, S.; Yoo, J.; Hague, R.J.M.; Tuck, C.J.; Wildman, R.D. 3D printing of tablets using inkjet with UV photoinitiation. Int. J. Pharm. 2017, 529, 523–530. [Google Scholar] [CrossRef]
  81. Fina, F.; Goyanes, A.; Gaisford, S.; Basit, A.W. Selective laser sintering (SLS) 3D printing of medicines. Int. J. Pharm. 2017, 529, 285–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Goyanes, A.; Fina, F.; Martorana, A.; Sedough, D.; Gaisford, S.; Basit, A.W. Development of modified release 3D printed tablets (printlets) with pharmaceutical excipients using additive manufacturing. Int. J. Pharm. 2017, 527, 21–30. [Google Scholar] [CrossRef] [PubMed]
  83. Goyanes, A.; Scarpa, M.; Kamlow, M.; Gaisford, S.; Basit, A.W.; Orlu, M. Patient acceptability of 3D printed medicines. Int. J. Pharm. 2017, 530, 71–78. [Google Scholar] [CrossRef] [PubMed]
  84. Kyobula, M.; Adedeji, A.; Alexander, M.R.; Saleh, E.; Wildman, R.; Ashcroft, I.; Gellert, P.R.; Roberts, C.J. 3D inkjet printing of tablets exploiting bespoke complex geometries for controlled and tuneable drug release. J. Control. Release 2017, 261, 207–215. [Google Scholar] [CrossRef] [PubMed]
  85. Okwuosa, T.C.; Pereira, B.C.; Arafat, B.; Cieszynska, M.; Isreb, A.; Alhnan, M.A. Fabricating a Shell-Core Delayed Release Tablet Using Dual FDM 3D Printing for Patient-Centred Therapy. Pharm. Res. 2017, 34, 427–437. [Google Scholar] [CrossRef]
  86. Solanki, N.G.; Tahsin, M.; Shah, A.V.; Serajuddin, A.T.M. Formulation of 3D Printed Tablet for Rapid Drug Release by Fused Deposition Modeling: Screening Polymers for Drug Release, Drug-Polymer Miscibility and Printability. J. Pharm. Sci. 2018, 107, 390–401. [Google Scholar] [CrossRef] [Green Version]
  87. Tagami, T.; Fukushige, K.; Ogawa, E.; Hayashi, N.; Ozeki, T. 3D Printing Factors Important for the Fabrication of Polyvinylalcohol Filament-Based Tablets. Biol. Pharm. Bull. 2017, 40, 357–364. [Google Scholar] [CrossRef] [Green Version]
  88. Zhang, J.; Feng, X.; Patil, H.; Tiwari, R.V.; Repka, M.A. Coupling 3D printing with hot-melt extrusion to produce controlled-release tablets. Int. J. Pharm. 2017, 519, 186–197. [Google Scholar] [CrossRef]
  89. Acosta-Vélez, G.F.; Zhu, T.Z.; Linsley, C.S.; Wu, B.M. Photocurable poly(ethylene glycol) as a bioink for the inkjet 3D pharming of hydrophobic drugs. Int. J. Pharm. 2018, 546, 145–153. [Google Scholar] [CrossRef]
  90. Arafat, B.; Wojsz, M.; Isreb, A.; Forbes, R.T.; Isreb, M.; Ahmed, W.; Arafat, T.; Alhnan, M.A. Tablet fragmentation without a disintegrant: A novel design approach for accelerating disintegration and drug release from 3D printed cellulosic tablets. Eur. J. Pharm. Sci. 2018, 118, 191–199. [Google Scholar] [CrossRef]
  91. Arafat, B.; Qinna, N.; Cieszynska, M.; Forbes, R.T.; Alhnan, M.A. Tailored on demand anti-coagulant dosing: An in vitro and in vivo evaluation of 3D printed purpose-designed oral dosage forms. Eur. J. Pharm. Biopharm. 2018, 128, 282–289. [Google Scholar] [CrossRef] [PubMed]
  92. Fina, F.; Madla, C.M.; Goyanes, A.; Zhang, J.; Gaisford, S.; Basit, A.W. Fabricating 3D printed orally disintegrating printlets using selective laser sintering. Int. J. Pharm. 2018, 541, 101–107. [Google Scholar] [CrossRef] [PubMed]
  93. Khaled, S.A.; Alexander, M.R.; Wildman, R.D.; Wallace, M.J.; Sharpe, S.; Yoo, J.; Roberts, C.J. 3D extrusion printing of high drug loading immediate release paracetamol tablets. Int. J. Pharm. 2018, 538, 223–230. [Google Scholar] [CrossRef]
  94. Martinez, P.R.; Goyanes, A.; Basit, A.W.; Gaisford, S. Influence of Geometry on the Drug Release Profiles of Stereolithographic (SLA) 3D-Printed Tablets. AAPS PharmSciTech 2018, 19, 3355–3361. [Google Scholar] [CrossRef]
  95. Trenfield, S.J.; Goyanes, A.; Telford, R.; Wilsdon, D.; Rowland, M.; Gaisford, S.; Basit, A.W. 3D printed drug products: Non-destructive dose verification using a rapid point-and-shoot approach. Int. J. Pharm. 2018, 549, 283–292. [Google Scholar] [CrossRef]
  96. Barakh Ali, S.F.; Mohamed, E.M.; Ozkan, T.; Kuttolamadom, M.A.; Khan, M.A.; Asadi, A.; Rahman, Z. Understanding the effects of formulation and process variables on the printlets quality manufactured by selective laser sintering 3D printing. Int. J. Pharm. 2019, 570, 118651. [Google Scholar] [CrossRef]
  97. Goyanes, A.; Allahham, N.; Trenfield, S.J.; Stoyanov, E.; Gaisford, S.; Basit, A.W. Direct powder extrusion 3D printing: Fabrication of drug products using a novel single-step process. Int. J. Pharm. 2019, 567, 118471. [Google Scholar] [CrossRef]
  98. Ilyés, K.; Balogh, A.; Casian, T.; Igricz, T.; Borbás, E.; Démuth, B.; Vass, P.; Menyhárt, L.; Kovács, N.K.; Marosi, G.; et al. 3D floating tablets: Appropriate 3D design from the perspective of different in vitro dissolution testing methodologies. Int. J. Pharm. 2019, 567, 118433. [Google Scholar] [CrossRef]
  99. Isreb, A.; Baj, K.; Wojsz, M.; Isreb, M.; Peak, M.; Alhnan, M.A. 3D printed oral theophylline doses with innovative ‘radiator-like’ design: Impact of polyethylene oxide (PEO) molecular weight. Int. J. Pharm. 2019, 564, 98–105. [Google Scholar] [CrossRef]
  100. Krause, J.; Bogdahn, M.; Schneider, F.; Koziolek, M.; Weitschies, W. Design and characterization of a novel 3D printed pressure-controlled drug delivery system. Eur. J. Pharm. Sci. 2019, 140, 105060. [Google Scholar] [CrossRef]
  101. Lamichhane, S.; Park, J.; Sohn, D.H.; Lee, S. Customized Novel Design of 3D Printed Pregabalin Tablets for Intra-Gastric Floating and Controlled Release Using Fused Deposition Modeling. Pharmaceutics 2019, 11, 564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Shin, S.; Kim, T.H.; Jeong, S.W.; Chung, S.E.; Lee, D.Y.; Kim, D.H.; Shin, B.S. Development of a gastroretentive delivery system for acyclovir by 3D printing technology and its in vivo pharmacokinetic evaluation in Beagle dogs. PLoS ONE 2019, 14, e0216875. [Google Scholar] [CrossRef] [PubMed]
  103. Wen, H.; He, B.; Wang, H.; Chen, F.; Li, P.; Cui, M.; Li, Q.; Pan, W.; Yang, X. Structure-Based Gastro-Retentive and Controlled-Release Drug Delivery with Novel 3D Printing. AAPS PharmSciTech 2019, 20, 68. [Google Scholar] [CrossRef] [PubMed]
  104. Xu, X.; Zhao, J.; Wang, M.; Wang, L.; Yang, J. 3D Printed Polyvinyl Alcohol Tablets with Multiple Release Profiles. Sci. Rep. 2019, 9, 12487. [Google Scholar] [CrossRef]
  105. Allahham, N.; Fina, F.; Marcuta, C.; Kraschew, L.; Mohr, W.; Gaisford, S.; Basit, A.W.; Goyanes, A. Selective laser sintering 3D printing of orally disintegrating printlets containing ondansetron. Pharmaceutics 2020, 12, 110. [Google Scholar] [CrossRef] [Green Version]
  106. Cheng, Y.; Qin, H.; Acevedo, N.C.; Jiang, X.; Shi, X. 3D printing of extended-release tablets of theophylline using hydroxypropyl methylcellulose (HPMC) hydrogels. Int. J. Pharm. 2020, 591, 119983. [Google Scholar] [CrossRef]
  107. Fanous, M.; Gold, S.; Muller, S.; Hirsch, S.; Ogorka, J.; Imanidis, G. Simplification of fused deposition modeling 3D-printing paradigm: Feasibility of 1-step direct powder printing for immediate release dosage form production. Int. J. Pharm. 2020, 578, 119124. [Google Scholar] [CrossRef]
  108. Hamed, R.; Mohamed, E.M.; Rahman, Z.; Khan, M.A. 3D-printing of lopinavir printlets by selective laser sintering and quantification of crystalline fraction by XRPD-chemometric models. Int. J. Pharm. 2020, 592, 120059. [Google Scholar] [CrossRef]
  109. Jamróz, W.; Kurek, M.; Szafraniec-Szczęsny, J.; Czech, A.; Gawlak, K.; Knapik-Kowalczuk, J.; Leszczyński, B.; Wróbel, A.; Paluch, M.; Jachowicz, R. Speed it up, slow it down…An issue of bicalutamide release from 3D printed tablets. Eur. J. Pharm. Sci. 2020, 143, 105169. [Google Scholar] [CrossRef]
  110. Januskaite, P.; Xu, X.; Ranmal, S.R.; Gaisford, S.; Basit, A.W.; Tuleu, C.; Goyanes, A. I spy with my little eye: A paediatric visual preferences survey of 3d printed tablets. Pharmaceutics 2020, 12, 1100. [Google Scholar] [CrossRef]
  111. Mohamed, E.M.; Barakh Ali, S.F.; Rahman, Z.; Dharani, S.; Ozkan, T.; Kuttolamadom, M.A.; Khan, M.A. Formulation Optimization of Selective Laser Sintering 3D-Printed Tablets of Clindamycin Palmitate Hydrochloride by Response Surface Methodology. AAPS PharmSciTech 2020, 21, 13–15. [Google Scholar] [CrossRef]
  112. Ong, J.J.; Awad, A.; Martorana, A.; Gaisford, S.; Stoyanov, E.; Basit, A.W.; Goyanes, A. 3D printed opioid medicines with alcohol-resistant and abuse-deterrent properties. Int. J. Pharm. 2020, 579, 119169. [Google Scholar] [CrossRef]
  113. Vo, A.Q.; Zhang, J.; Nyavanandi, D.; Bandari, S.; Repka, M.A. Hot melt extrusion paired fused deposition modeling 3D printing to develop hydroxypropyl cellulose based floating tablets of cinnarizine. Carbohydr. Polym. 2020, 246, 116519. [Google Scholar] [CrossRef]
  114. Boniatti, J.; Januskaite, P.; da Fonseca, L.B.; Viçosa, A.L.; Amendoeira, F.C.; Tuleu, C.; Basit, A.W.; Goyanes, A.; Ré, M.I. Direct powder extrusion 3d printing of praziquantel to overcome neglected disease formulation challenges in paediatric populations. Pharmaceutics 2021, 13, 1114. [Google Scholar] [CrossRef]
  115. Davis, D.A.; Thakkar, R.; Su, Y.; Williams, R.O.; Maniruzzaman, M. Selective Laser Sintering 3-Dimensional Printing as a Single Step Process to Prepare Amorphous Solid Dispersion Dosage Forms for Improved Solubility and Dissolution Rate. J. Pharm. Sci. 2021, 110, 1432–1443. [Google Scholar] [CrossRef]
  116. Johannesson, J.; Khan, J.; Hubert, M.; Teleki, A.; Bergström, C.A.S. 3D-printing of solid lipid tablets from emulsion gels. Int. J. Pharm. 2021, 597, 120304. [Google Scholar] [CrossRef]
  117. Viidik, L.; Vesala, J.; Laitinen, R.; Korhonen, O.; Ketolainen, J.; Aruväli, J.; Kirsimäe, K.; Kogermann, K.; Heinämäki, J.; Laidmäe, I.; et al. Preparation and characterization of hot-melt extruded polycaprolactone-based filaments intended for 3D-printing of tablets. Eur. J. Pharm. Sci. 2021, 158, 105619. [Google Scholar] [CrossRef] [PubMed]
  118. Xu, X.; Seijo-Rabina, A.; Awad, A.; Rial, C.; Gaisford, S.; Basit, A.W.; Goyanes, A. Smartphone-enabled 3D printing of medicines. Int. J. Pharm. 2021, 609, 121199. [Google Scholar] [CrossRef]
  119. Melocchi, A.; Parietti, F.; Loreti, G.; Maroni, A.; Gazzaniga, A.; Zema, L. 3D printing by fused deposition modeling (FDM) of a swellable/erodible capsular device for oral pulsatile release of drugs. J. Drug Deliv. Sci. Technol. 2015, 30, 360–367. [Google Scholar] [CrossRef]
  120. Maroni, A.; Melocchi, A.; Parietti, F.; Foppoli, A.; Zema, L.; Gazzaniga, A. 3D printed multi-compartment capsular devices for two-pulse oral drug delivery. J. Control. Release 2017, 268, 10–18. [Google Scholar] [CrossRef]
  121. Auvinen, V.V.; Virtanen, J.; Merivaara, A.; Virtanen, V.; Laurén, P.; Tuukkanen, S.; Laaksonen, T. Modulating sustained drug release from nanocellulose hydrogel by adjusting the inner geometry of implantable capsules. J. Drug Deliv. Sci. Technol. 2020, 57, 101625. [Google Scholar] [CrossRef]
  122. Melocchi, A.; Parietti, F.; Maroni, A.; Foppoli, A.; Gazzaniga, A.; Zema, L. Hot-melt extruded filaments based on pharmaceutical grade polymers for 3D printing by fused deposition modeling. Int. J. Pharm. 2016, 509, 255–263. [Google Scholar] [CrossRef] [PubMed]
  123. Goyanes, A.; Fernández-Ferreiro, A.; Majeed, A.; Gomez-Lado, N.; Awad, A.; Luaces-Rodríguez, A.; Gaisford, S.; Aguiar, P.; Basit, A.W. PET/CT imaging of 3D printed devices in the gastrointestinal tract of rodents. Int. J. Pharm. 2018, 536, 158–164. [Google Scholar] [CrossRef]
  124. Melocchi, A.; Parietti, F.; Maccagnan, S.; Ortenzi, M.A.; Antenucci, S.; Briatico-Vangosa, F.; Maroni, A.; Gazzaniga, A.; Zema, L. Industrial Development of a 3D-Printed Nutraceutical Delivery Platform in the Form of a Multicompartment HPC Capsule. AAPS PharmSciTech 2018, 19, 3343–3354. [Google Scholar] [CrossRef]
  125. Buanz, A.B.M.; Saunders, M.H.; Basit, A.W.; Gaisford, S. Preparation of personalized-dose salbutamol sulphate oral films with thermal ink-jet printing. Pharm. Res. 2011, 28, 2386–2392. [Google Scholar] [CrossRef]
  126. Genina, N.; Janßen, E.M.; Breitenbach, A.; Breitkreutz, J.; Sandler, N. Evaluation of different substrates for inkjet printing of rasagiline mesylate. Eur. J. Pharm. Biopharm. 2013, 85, 1075–1083. [Google Scholar] [CrossRef]
  127. Planchette, C.; Pichler, H.; Wimmer-Teubenbacher, M.; Gruber, M.; Gruber-Woelfler, H.; Mohr, S.; Tetyczka, C.; Hsiao, W.K.; Paudel, A.; Roblegg, E.; et al. Printing medicines as orodispersible dosage forms: Effect of substrate on the printed micro-structure. Int. J. Pharm. 2016, 509, 518–527. [Google Scholar] [CrossRef]
  128. Vakili, H.; Nyman, J.O.; Genina, N.; Preis, M.; Sandler, N. Application of a colorimetric technique in quality control for printed pediatric orodispersible drug delivery systems containing propranolol hydrochloride. Int. J. Pharm. 2016, 511, 606–618. [Google Scholar] [CrossRef]
  129. Jamróz, W.; Kurek, M.; Łyszczarz, E.; Szafraniec, J.; Knapik-Kowalczuk, J.; Syrek, K.; Paluch, M.; Jachowicz, R. 3D printed orodispersible films with Aripiprazole. Int. J. Pharm. 2017, 533, 413–420. [Google Scholar] [CrossRef]
  130. Elbl, J.; Gajdziok, J.; Kolarczyk, J. 3D printing of multilayered orodispersible films with in-process drying. Int. J. Pharm. 2020, 575, 118883. [Google Scholar] [CrossRef]
  131. Sjöholm, E.; Sandler, N. Additive manufacturing of personalized orodispersible warfarin films. Int. J. Pharm. 2019, 564, 117–123. [Google Scholar] [CrossRef]
  132. Tian, Y.; Orlu, M.; Woerdenbag, H.J.; Scarpa, M.; Kiefer, O.; Kottke, D.; Sjöholm, E.; Öblom, H.; Sandler, N.; Hinrichs, W.L.J.; et al. Oromucosal films: From patient centricity to production by printing techniques. Expert Opin. Drug Deliv. 2019, 16, 981–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Yan, T.T.; Lv, Z.F.; Tian, P.; Lin, M.M.; Lin, W.; Huang, S.Y.; Chen, Y.Z. Semi-solid extrusion 3D printing ODFs: An individual drug delivery system for small scale pharmacy. Drug Dev. Ind. Pharm. 2020, 46, 531–538. [Google Scholar] [CrossRef]
  134. Genina, N.; Fors, D.; Vakili, H.; Ihalainen, P.; Pohjala, L.; Ehlers, H.; Kassamakov, I.; Haeggström, E.; Vuorela, P.; Peltonen, J.; et al. Tailoring controlled-release oral dosage forms by combining inkjet and flexographic printing techniques. Eur. J. Pharm. Sci. 2012, 47, 615–623. [Google Scholar] [CrossRef]
  135. Ehtezazi, T.; Algellay, M.; Islam, Y.; Roberts, M.; Dempster, N.M.; Sarker, S.D. The Application of 3D Printing in the Formulation of Multilayered Fast Dissolving Oral Films. J. Pharm. Sci. 2018, 107, 1076–1085. [Google Scholar] [CrossRef]
  136. Conceição, J.; Farto-Vaamonde, X.; Goyanes, A.; Adeoye, O.; Concheiro, A.; Cabral-Marques, H.; Sousa Lobo, J.M.; Alvarez-Lorenzo, C. Hydroxypropyl-β-cyclodextrin-based fast dissolving carbamazepine printlets prepared by semisolid extrusion 3D printing. Carbohydr. Polym. 2019, 221, 55–62. [Google Scholar] [CrossRef]
  137. Eleftheriadis, G.K.; Ritzoulis, C.; Bouropoulos, N.; Tzetzis, D.; Andreadis, D.A.; Boetker, J.; Rantanen, J.; Fatouros, D.G. Unidirectional drug release from 3D printed mucoadhesive buccal films using FDM technology: In vitro and ex vivo evaluation. Eur. J. Pharm. Biopharm. 2019, 144, 180–192. [Google Scholar] [CrossRef]
  138. Öblom, H.; Sjöholm, E.; Rautamo, M.; Sandler, N. Towards printed pediatric medicines in hospital pharmacies: Comparison of 2d and 3d-printed orodispersiblewarfarin films with conventional oral powders in unit dose sachets. Pharmaceutics 2019, 11, 334. [Google Scholar] [CrossRef] [Green Version]
  139. Sjöholm, E.; Mathiyalagan, R.; Prakash, D.R.; Lindfors, L.; Wang, Q.; Wang, X.; Ojala, S.; Sandler, N. 3D-Printed Veterinary Dosage Forms—A Comparative Study of Three Semi-Solid Extrusion 3D Printers. Pharmaceutics 2020, 12, 1239. [Google Scholar] [CrossRef]
  140. Germini, G.; Peltonen, L. 3D Printing of Drug Nanocrystals for Film Formulations. Molecules 2021, 26, 3941. [Google Scholar] [CrossRef]
  141. Huang, W.; Zheng, Q.; Sun, W.; Xu, H.; Yang, X. Levofloxacin implants with predefined microstructure fabricated by three-dimensional printing technique. Int. J. Pharm. 2007, 339, 33–38. [Google Scholar] [CrossRef] [PubMed]
  142. Wu, W.; Zheng, Q.; Guo, X.; Sun, J.; Liu, Y. A programmed release multi-drug implant fabricated by three-dimensional printing technology for bone tuberculosis therapy. Biomed. Mater. 2009, 4, 065005. [Google Scholar] [CrossRef]
  143. Rattanakit, P.; Moulton, S.E.; Santiago, K.S.; Liawruangrath, S.; Wallace, G.G. Extrusion printed polymer structures: A facile and versatile approach to tailored drug delivery platforms. Int. J. Pharm. 2012, 422, 254–263. [Google Scholar] [CrossRef] [PubMed]
  144. Genina, N.; Holländer, J.; Jukarainen, H.; Mäkilä, E.; Salonen, J.; Sandler, N. Ethylene vinyl acetate (EVA) as a new drug carrier for 3D printed medical drug delivery devices. Eur. J. Pharm. Sci. 2016, 90, 53–63. [Google Scholar] [CrossRef] [PubMed]
  145. 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] [PubMed]
  146. Kempin, W.; Franz, C.; Koster, L.C.; Schneider, F.; Bogdahn, M.; Weitschies, W.; Seidlitz, A. Assessment of different polymers and drug loads for fused deposition modeling of drug loaded implants. Eur. J. Pharm. Biopharm. 2017, 115, 84–93. [Google Scholar] [CrossRef]
  147. Qamar, N.; Abbas, N.; Irfan, M.; Hussain, A.; Arshad, M.S.; Latif, S.; Mehmood, F.; Ghori, M.U. Personalized 3D printed ciprofloxacin impregnated meshes for the management of hernia. J. Drug Deliv. Sci. Technol. 2019, 53, 101164. [Google Scholar] [CrossRef]
  148. 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] [Green Version]
  149. Wang, Y.; Sun, L.; Mei, Z.; Zhang, F.; He, M.; Fletcher, C.; Wang, F.; Yang, J.; Bi, D.; Jiang, Y.; et al. 3D printed biodegradable implants as an individualized drug delivery system for local chemotherapy of osteosarcoma. Mater. Des. 2020, 186, 108336. [Google Scholar] [CrossRef]
  150. Cui, M.; Pan, H.; Li, L.; Fang, D.; Sun, H.; Qiao, S.; Li, X.; Pan, W. Exploration and Preparation of Patient-specific Ciprofloxacin Implants Drug Delivery System Via 3D Printing Technologies. J. Pharm. Sci. 2021, 110, 3678–3689. [Google Scholar] [CrossRef]
  151. Lim, S.H.; Kathuria, H.; Tan, J.J.Y.; Kang, L. 3D printed drug delivery and testing systems—A passing fad or the future? Adv. Drug Deliv. Rev. 2018, 132, 139–168. [Google Scholar] [CrossRef]
  152. Kumar, A.; Pillai, J. Implantable Drug Delivery Systems: An Overview; Elsevier: Amsterdam, The Netherlands, 2018; ISBN 9780128136669. [Google Scholar]
  153. Wu, G.; Wu, W.; Zheng, Q.; Li, J.; Zhou, J.; Hu, Z. Experimental study of PLLA/INH slow release implant fabricated by three dimensional printing technique and drug release characteristics in vitro. Biomed. Eng. Online 2014, 13, 97. [Google Scholar] [CrossRef] [Green Version]
  154. Min, Z.; Kun, L.; Yufang, Z.; Jianhua, Z.; Xiaojian, Y. 3D-printed hierarchical scaffold for localized isoniazid/rifampin drug delivery and osteoarticular tuberculosis therapy. Acta Biomater. 2015, 16, 145–155. [Google Scholar]
  155. Water, J.J.; Bohr, A.; Boetker, J.; Aho, J.; Sandler, N.; Nielsen, H.M.; Rantanen, J. Three-dimensional printing of drug-eluting implants: Preparation of an antimicrobial polylactide feedstock material. J. Pharm. Sci. 2015, 104, 1099–1107. [Google Scholar] [CrossRef]
  156. Benmassaoud, M.M.; Kohama, C.; Kim, T.W.B.; Kadlowec, J.A.; Foltiny, B.; Mercurio, T.; Ranganathan, S.I. Efficacy of eluted antibiotics through 3D printed femoral implants. Biomed. Microdevices 2019, 21, 51. [Google Scholar] [CrossRef]
  157. Arany, P.; Papp, I.; Zichar, M.; Csontos, M.; Elek, J.; Regdon, G.; Budai, I.; Béres, M.; Gesztelyi, R.; Fehér, P.; et al. In Vitro Tests of FDM 3D-Printed Diclofenac Sodium-Containing Implants. Molecules 2020, 25, 5889. [Google Scholar] [CrossRef]
  158. Xu, X.; Goyanes, A.; Trenfield, S.J.; Diaz-Gomez, L.; Alvarez-Lorenzo, C.; Gaisford, S.; Basit, A.W. Stereolithography (SLA) 3D printing of a bladder device for intravesical drug delivery. Mater. Sci. Eng. C 2021, 120, 111773. [Google Scholar] [CrossRef]
  159. Xu, X.; Awwad, S.; Diaz-Gomez, L.; Alvarez-Lorenzo, C.; Brocchini, S.; Gaisford, S.; Goyanes, A.; Basit, A.W. 3D Printed Punctal Plugs for Controlled Ocular Drug Delivery. Pharmaceutics 2021, 13, 1421. [Google Scholar] [CrossRef]
  160. Goyanes, A.; Det-Amornrat, U.; Wang, J.; Basit, A.W.; Gaisford, S. 3D scanning and 3D printing as innovative technologies for fabricating personalized topical drug delivery systems. J. Control. Release 2016, 234, 41–48. [Google Scholar] [CrossRef] [PubMed]
  161. Yi, H.G.; Choi, Y.J.; Kang, K.S.; Hong, J.M.; Pati, R.G.; Park, M.N.; Shim, I.K.; Lee, C.M.; Kim, S.C.; Cho, D.W. A 3D-printed local drug delivery patch for pancreatic cancer growth suppression. J. Control. Release 2016, 238, 231–241. [Google Scholar] [CrossRef]
  162. Wang, J.C.; Zheng, H.; Chang, M.W.; Ahmad, Z.; Li, J.S. Preparation of active 3D film patches via aligned fiber electrohydrodynamic (EHD) printing. Sci. Rep. 2017, 7, srep43924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Chaudhari, V.S.; Malakar, T.K.; Murty, U.S.; Banerjee, S. Extruded filaments derived 3D printed medicated skin patch to mitigate destructive pulmonary tuberculosis: Design to delivery. Expert Opin. Drug Deliv. 2021, 18, 301–313. [Google Scholar] [CrossRef] [PubMed]
  164. Ovsianikov, A.; Chichkov, B.; Mente, P.; Monteiro-Riviere, N.A.; Doraiswamy, A.; Narayan, R.J. Two photon polymerization of polymer-ceramic hybrid materials for transdermal drug delivery. Int. J. Appl. Ceram. Technol. 2007, 4, 22–29. [Google Scholar] [CrossRef]
  165. Boehm, R.D.; Miller, P.R.; Schell, W.A.; Perfect, J.R.; Narayan, R.J. Inkjet printing of amphotericin B onto biodegradable microneedles using piezoelectric inkjet printing. Jom 2013, 65, 525–533. [Google Scholar] [CrossRef]
  166. Boehm, R.D.; Miller, P.R.; Daniels, J.; Stafslien, S.; Narayan, R.J. Inkjet printing for pharmaceutical applications. Mater. Today 2014, 17, 247–252. [Google Scholar] [CrossRef]
  167. Lu, Y.; Mantha, S.N.; Crowder, D.C.; Chinchilla, S.; Shah, K.N.; Yun, Y.H.; Wicker, R.B.; Choi, J.-W. Microstereolithography and characterization of microneedle arrays Microstereolithography and characterization of poly (propylene fumarate) -based drug-loaded microneedle arrays. Biofabrication 2015, 7, 045001. [Google Scholar] [CrossRef]
  168. Ross, S.; Scoutaris, N.; Lamprou, D.; Mallinson, D.; Douroumis, D. Inkjet printing of insulin microneedles for transdermal delivery. Drug Deliv. Transl. Res. 2015, 5, 451–461. [Google Scholar] [CrossRef] [Green Version]
  169. Lim, S.H.; Ng, J.Y.; Kang, L. Three-dimensional printing of a microneedle array on personalized curved surfaces for dual-pronged treatment of trigger finger. Biofabrication 2017, 9, 015010. [Google Scholar] [CrossRef]
  170. Pere, C.P.P.; Economidou, S.N.; Lall, G.; Ziraud, C.; Boateng, J.S.; Alexander, B.D.; Lamprou, D.A.; Douroumis, D. 3D printed microneedles for insulin skin delivery. Int. J. Pharm. 2018, 544, 425–432. [Google Scholar] [CrossRef] [Green Version]
  171. Economidou, S.N.; Pere, C.P.P.; Reid, A.; Uddin, M.J.; Windmill, J.F.C.; Lamprou, D.A.; Douroumis, D. 3D printed microneedle patches using stereolithography (SLA)for intradermal insulin delivery. Mater. Sci. Eng. C 2019, 102, 743–755. [Google Scholar] [CrossRef]
  172. Chiappini, C.; De Rosa, E.; Martinez, J.O.; Liu, X.; Steele, J.; Stevens, M.M.; Tasciotti, E. Biodegradable silicon nanoneedles delivering nucleic acids intracellularly induce localized in vivo neovascularization. Nat. Mater. 2015, 14, 532–539. [Google Scholar] [CrossRef]
  173. Uddin, M.J.; Scoutaris, N.; Klepetsanis, P.; Chowdhry, B.; Prausnitz, M.R.; Douroumis, D. Inkjet printing of transdermal microneedles for the delivery of anticancer agents. Int. J. Pharm. 2015, 494, 593–602. [Google Scholar] [CrossRef] [Green Version]
  174. Farias, C.; Lyman, R.; Hemingway, C.; Chau, H.; Mahacek, A.; Bouzos, E.; Mobed-Miremadi, M. Three-dimensional (3D) printed microneedles for microencapsulated cell extrusion. Bioengineering 2018, 5, 59. [Google Scholar] [CrossRef] [Green Version]
  175. Luzuriaga, M.A.; Berry, D.R.; Reagan, J.C.; Smaldone, R.A.; Gassensmith, J.J. Biodegradable 3D printed polymer microneedles for transdermal drug delivery. Lab Chip 2018, 18, 1223–1230. [Google Scholar] [CrossRef]
  176. Tappa, K.; Jammalamadaka, U.; Ballard, D.H.; Bruno, T.; Israel, M.R.; Vemula, H.; Meacham, J.M.; Mills, D.K.; Woodard, P.K.; Weisman, J.A. Medication eluting devices for the field of OBGYN (MEDOBGYN): 3D printed biodegradable hormone eluting constructs, a proof of concept study. PLoS ONE 2017, 12, e0182929. [Google Scholar] [CrossRef]
  177. Varan, C.; Wickström, H.; Sandler, N.; Aktaş, Y.; Bilensoy, E. Inkjet printing of antiviral PCL nanoparticles and anticancer cyclodextrin inclusion complexes on bioadhesive film for cervical administration. Int. J. Pharm. 2017, 531, 701–713. [Google Scholar] [CrossRef]
  178. Arany, P.; Papp, I.; Zichar, M.; Regdon, G.; Béres, M.; Szalóki, M.; Kovács, R.; Fehér, P.; Ujhelyi, Z.; Vecsernyés, M.; et al. Manufacturing and Examination of Vaginal Drug Delivery System by FDM 3D Printing. Pharmaceutics 2021, 13, 1714. [Google Scholar] [CrossRef]
  179. Weisman, J.A.; Ballard, D.H.; Jammalamadaka, U.; Tappa, K.; Sumerel, J.; D’Agostino, H.B.; Mills, D.K.; Woodard, P.K. 3D printed antibiotic and chemotherapeutic eluting catheters for potential use in interventional radiology: In vitro proof of concept study. Acad. Radiol. 2019, 26, 270–274. [Google Scholar] [CrossRef]
  180. Espey, E.; Ogburn, T. Long-acting reversible contraceptives: Intrauterine devices and the contraceptive implant. Obstet. Gynecol. 2011, 117, 705–719. [Google Scholar] [CrossRef]
  181. Holländer, J.; Genina, N.; Jukarainen, H.; Khajeheian, M.; Rosling, A.; Mäkilä, E.; Sandler, N. Three-Dimensional Printed PCL-Based Implantable Prototypes of Medical Devices for Controlled Drug Delivery. J. Pharm. Sci. 2016, 105, 2665–2676. [Google Scholar] [CrossRef] [Green Version]
  182. Varan, C.; Şen, M.; Sandler, N.; Aktaş, Y.; Bilensoy, E. Mechanical characterization and ex vivo evaluation of anticancer and antiviral drug printed bioadhesive film for the treatment of cervical cancer. Eur. J. Pharm. Sci. 2019, 130, 114–123. [Google Scholar] [CrossRef] [PubMed]
  183. Pardeike, J.; Strohmeier, D.M.; Schrödl, N.; Voura, C.; Gruber, M.; Khinast, J.G.; Zimmer, A. Nanosuspensions as advanced printing ink for accurate dosing of poorly soluble drugs in personalized medicines. Int. J. Pharm. 2011, 420, 93–100. [Google Scholar] [CrossRef] [PubMed]
  184. Scoutaris, N.; Alexander, M.R.; Gellert, P.R.; Roberts, C.J. Inkjet printing as a novel medicine formulation technique. J. Control. Release 2011, 156, 179–185. [Google Scholar] [CrossRef] [PubMed]
  185. Gu, Y.; Chen, X.; Lee, J.H.; Monteiro, D.A.; Wang, H.; Lee, W.Y. Inkjet printed antibiotic- and calcium-eluting bioresorbable nanocomposite micropatterns for orthopedic implants. Acta Biomater. 2012, 8, 424–431. [Google Scholar] [CrossRef]
  186. Lee, B.K.; Yun, Y.H.; Choi, J.S.; Choi, Y.C.; Kim, J.D.; Cho, Y.W. Fabrication of drug-loaded polymer microparticles with arbitrary geometries using a piezoelectric inkjet printing system. Int. J. Pharm. 2012, 427, 305–310. [Google Scholar] [CrossRef]
  187. Kitson, P.J.; Glatzel, S.; Cronin, L. The digital code driven autonomous synthesis of ibuprofen automated in a 3D-printer-based robot. Beilstein J. Org. Chem. 2016, 12, 2776–2783. [Google Scholar] [CrossRef] [Green Version]
  188. Long, J.; Etxeberria, A.E.; Nand, A.V.; Bunt, C.R.; Ray, S.; Seyfoddin, A. A 3D printed chitosan-pectin hydrogel wound dressing for lidocaine hydrochloride delivery. Mater. Sci. Eng. C 2019, 104, 109873. [Google Scholar] [CrossRef]
  189. Seoane-Viaño, I.; Gómez-Lado, N.; Lázare-Iglesias, H.; García-Otero, X.; Antúnez-López, J.R.; Ruibal, Á.; Varela-Correa, J.J.; Aguiar, P.; Basit, A.W.; Otero-Espinar, F.J.; et al. 3D printed tacrolimus rectal formulations ameliorate colitis in an experimental animal model of inflammatory bowel disease. Biomedicines 2020, 8, 563. [Google Scholar] [CrossRef]
  190. Seoane-Viaño, I.; Ong, J.J.; Luzardo-Álvarez, A.; González-Barcia, M.; Basit, A.W.; Otero-Espinar, F.J.; Goyanes, A. 3D printed tacrolimus suppositories for the treatment of ulcerative colitis. Asian J. Pharm. Sci. 2021, 16, 110–119. [Google Scholar] [CrossRef]
  191. Quanjin, M.; Rejab, M.R.M.; Idris, M.S.; Kumar, N.M.; Abdullah, M.H.; Reddy, G.R. Recent 3D and 4D intelligent printing technologies: A comparative review and future perspective. Procedia Comput. Sci. 2020, 167, 1210–1219. [Google Scholar] [CrossRef]
Figure 1. Cross-section of the acetaminophen-containing matrix tablets based on the authors’ figure. The different colors label dissimilar compartments [30].
Figure 1. Cross-section of the acetaminophen-containing matrix tablets based on the authors’ figure. The different colors label dissimilar compartments [30].
Pharmaceutics 14 01312 g001
Figure 2. Cross-section of the constructed drug dosage forms in the article of Goyanes et al. (a) Sectioned multilayer tablet, (b) sectioned DuoCaplet (caplet in caplet) [35].
Figure 2. Cross-section of the constructed drug dosage forms in the article of Goyanes et al. (a) Sectioned multilayer tablet, (b) sectioned DuoCaplet (caplet in caplet) [35].
Pharmaceutics 14 01312 g002
Figure 3. Cross-section of the printed polypills where three diverse compartments were created (labeled with three nonidentical colors) [36].
Figure 3. Cross-section of the printed polypills where three diverse compartments were created (labeled with three nonidentical colors) [36].
Pharmaceutics 14 01312 g003
Figure 4. Cross-section of the polypills where ASA and HCT formulations were located in the upper immediate release compartments (labeled with blue and orange rectangles) and atenolol, pravastatin, and ramipril formulations were in three distinct extended release compartments (labeled with yellow, green, and peach blossom). The three compartments were the same size but could be visualized like this because of the original design of the “cake slice” [37].
Figure 4. Cross-section of the polypills where ASA and HCT formulations were located in the upper immediate release compartments (labeled with blue and orange rectangles) and atenolol, pravastatin, and ramipril formulations were in three distinct extended release compartments (labeled with yellow, green, and peach blossom). The three compartments were the same size but could be visualized like this because of the original design of the “cake slice” [37].
Pharmaceutics 14 01312 g004
Figure 5. Flow chart on the described tablet manufacturing methods and main breakthroughs between 1996 and 2016 [34,35,36,37,39,40,41,42].
Figure 5. Flow chart on the described tablet manufacturing methods and main breakthroughs between 1996 and 2016 [34,35,36,37,39,40,41,42].
Pharmaceutics 14 01312 g005
Figure 6. Cross-section of the dual-compartmental dosage form designed by Genina et al. In the research, isoniazid (white colored) and rifampicin (red colored) were hot-melt extruded and then 3D printed into the polymeric cap (brown colored) and closed with a cap (blue colored) [46].
Figure 6. Cross-section of the dual-compartmental dosage form designed by Genina et al. In the research, isoniazid (white colored) and rifampicin (red colored) were hot-melt extruded and then 3D printed into the polymeric cap (brown colored) and closed with a cap (blue colored) [46].
Pharmaceutics 14 01312 g006
Figure 7. Cross-section of the 3D-printed gastro-floating tablets with 30% infill percentage rate [51].
Figure 7. Cross-section of the 3D-printed gastro-floating tablets with 30% infill percentage rate [51].
Pharmaceutics 14 01312 g007
Figure 8. Cross-section of the 3D-printed channeled tablets. Each white square represents a channel. (a) Channels parallel to the longer side; (b) channels parallel to the shorter side [52].
Figure 8. Cross-section of the 3D-printed channeled tablets. Each white square represents a channel. (a) Channels parallel to the longer side; (b) channels parallel to the shorter side [52].
Pharmaceutics 14 01312 g008
Figure 9. Flow chart of the described tablet manufacturing methods and main breakthroughs in 2017 and 2018.
Figure 9. Flow chart of the described tablet manufacturing methods and main breakthroughs in 2017 and 2018.
Pharmaceutics 14 01312 g009
Figure 10. Cross-section of the cylinder-shaped polypill. Each color represents diverse API-containing layers: naproxen—yellow; aspirin—purple; paracetamol—orange; caffeine—red; chloramphenicol—green; and prednisolone—blue [57].
Figure 10. Cross-section of the cylinder-shaped polypill. Each color represents diverse API-containing layers: naproxen—yellow; aspirin—purple; paracetamol—orange; caffeine—red; chloramphenicol—green; and prednisolone—blue [57].
Pharmaceutics 14 01312 g010
Figure 11. Flow chart of the described tablet manufacturing methods and main breakthroughs since 2019.
Figure 11. Flow chart of the described tablet manufacturing methods and main breakthroughs since 2019.
Pharmaceutics 14 01312 g011
Figure 12. Cross-section of the designed capsules. (a) With the same wall thickness, (b) with different wall thicknesses [120].
Figure 12. Cross-section of the designed capsules. (a) With the same wall thickness, (b) with different wall thicknesses [120].
Pharmaceutics 14 01312 g012
Figure 13. Cross-sectional design of the microneedle array [167].
Figure 13. Cross-sectional design of the microneedle array [167].
Pharmaceutics 14 01312 g013
Figure 14. The designed vaginal drug delivery system by our research group [178].
Figure 14. The designed vaginal drug delivery system by our research group [178].
Pharmaceutics 14 01312 g014
Table 3. The grouping of the manufactured capsules based on the publication year and then in alphabetical order.
Table 3. The grouping of the manufactured capsules based on the publication year and then in alphabetical order.
YearType of 3D PrintingType of PolymerType of APIArticle
2015FDMHPCno (yellow and blue dye)Melocchi et al. [119]
2016FDMPLA, EC, HPC, HPMC, HPMCAS, various Eugradit, PEO, PVA, Soluplus, PEG 400 and 8000acetaminophen, furosemideMelocchi et al. [122]
2017FDM, InkjetPLA, PVA, polymer formulationsno (yellow and blue dye)Maroni et al. [120]
2018FDMPVA-PEG, HPC, ECFluorodeoxyglucose (18F-FDG) TRACERlab MX synthesizer (GE Healthcare®)Basit et al. [123]
FDMHPC, PLAcaffeine, blue and yellow dyeMelocchi et al. [124]
2020FDMPLAmetoprolol, nadalololAuviven et al. [121]
Table 4. The grouping of the manufactured orodispersible films based on the publication year and then in alphabetical order.
Table 4. The grouping of the manufactured orodispersible films based on the publication year and then in alphabetical order.
YearType of 3D PrintingType of PolymerType of APIArticle
2011thermal inkjet printingno needsalbutamol sulphateBuanz et al. [125]
2012inkjet and flexographic printingECriboflavin, propranololGenina et al. [134]
2013thermal inkjet printingcrospovidone (Kollidon CL-M)rasagiline mesylateGenina et al. [126]
2016inkjet printingPEGylated PLGAsodium picosulphatePlanchette et al. [127]
inkjet printingHPCpropranolol hydrochlorideVakili et al. [128]
2017FDMPVAaripiprazoleJamróz et al. [129]
2018FDMPVA, PEO, PEGibuprofen, paracetamolEhtezazi et al. [135]
2019semi-solid extrusionhydroxypropyl-β-cyclodextrin, cellulosecarbamazepineConceição et al. [136]
FDMPVAdiclofenac sodiumEleftheriadis et al. [137]
EXT, IJPHPCwarfarinÖblom et al. [138]
BiobotPVAwarfarinSjöholm et al. [131]
2020modified FDMmaltodextrin, HECbenzydamine hydrochlorideElbl et al. [130]
inkjet printingPEO, HPCprednisoloneSjöholm et al. [139]
semi-solid extrusionHPMClevocetirizineYan et al. [133]
2021multitool 3D printerHPMCindomethacinGermini et al. [140]
Table 5. The grouping of the manufactured implants based on the publication year and then in alphabetical order.
Table 5. The grouping of the manufactured implants based on the publication year and then in alphabetical order.
YearType of 3D PrintingType of PolymerType of APIArticle
2007inkjet printingL-PLAlevofloxacinHuang et al. [141]
2009inkjet printingPDLLArifampicin, isoniazidWu et al. [142]
2012extrusion based 3D printingPLGA, PVAdexamethasoneRattanakit et al. [143]
2014powder bindingPLLAisoniazidWu et al. [153]
20153D-Bioplotter system(3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx)isoniazid, rifampinMin et al. [154]
FDM (MakerBot®)PLAnitrofurantoinWater et al. [155]
2016FDMPCL containing EVAindomethacinGenina et al. [144]
inkjet powder printingPDLLAlevofloxacin, tobramycinWu et al. [145]
2017FDMEudragit RS, PCL, PLLA, ECquinineKempin et al. [146]
2019FDM, DMLSPLA, PCL, titanium dioxidedoxycyclineBenmassaoud et al. [156]
FDMPP, PVAciprofloxacinQamar et al. [147]
FDMPLAgentamicin, methotrexateTappa et al. [148]
2020FDMPLA, antibacterial PLA, PETG, PMMAdiclofenac sodiumArany et al. [157]
powder bed printingPLLAcisplatin, ifosfamid, methotrexate, doxorubicinWang et al. [149]
2021SSE, FDMPLAciprofloxacinCui et al. [150]
SLAElastic ResinlidocaineXu et al. [158]
DLPPEGDA, PEG 400dexamethasone, phenyl bis phosphine oxide, β-caroteneXu et al. [159]
Table 6. The grouping of some manufactured TTSs based on the publication year and then in alphabetical order.
Table 6. The grouping of some manufactured TTSs based on the publication year and then in alphabetical order.
YearType of 3D PrintingType of PolymerType of APIArticle
2016FDM, SLAFlex EcoPLA, PCLsalicylic acidGoyanes et al. [160]
inhouse extrusion-based 3D printer—multi-head deposition systemPLA, PCL5-fluorouracilYi et al. [161]
2017EHDPCL, PCL/PVPtetracycline hydrochlorideWang et al. [162]
2021FDMPVPquercetinChaudhari et al. [163]
Table 7. The grouping of some manufactured microneedles based on the publication year and then in alphabetical order.
Table 7. The grouping of some manufactured microneedles based on the publication year and then in alphabetical order.
YearType of 3D PrintingType of PolymerType of APIArticle
2007femtosecond laser two photon polymerizationOrmocer®noneOvsianikov et al. [164]
2013piezoelectric inkjet printingPDMS, PMMAamphotericin BBoehm et al. [165]
2014piezoelectric inkjet printingGantrez® AN 169 BF (poly(methyl vinyl ether-co-maleic anhydride))miconazoleBoehm et al. [166]
2015multi-material microstereolithography (μSL)poly(propylene fumarate)dacarbazineLu et al. [167]
inkjet printingpolyvinyl caprolactame-polyvinyl cetatepolyethylene glycol (SOL), poly(2-ethyl-2-oxazoline) (POX)insulinRoss et al. [168]
inkjet printingSoluplus®5-fluorouracilUddin et al. [173]
2017DLP3DMCastable resindiclofenac sodiumLim et al. [169]
2018SLAmedium viscosity alginateblue dye, HepG2 cell encapsulationFarias et al. [174]
FDMPLAfluoresceinLuzuriaga et al. [175]
inkjet printerDental SGinsulinPere et al. [170]
2019SLADental SGinsulinEconomidou et al. [171]
Table 8. The grouping of the manufactured vaginal drug delivery systems based on the publication year and then in alphabetical order.
Table 8. The grouping of the manufactured vaginal drug delivery systems based on the publication year and then in alphabetical order.
Drug Delivery TypeYearType of 3D PrintingType of PolymerType of APIArticle
IUD, subcutaneous rod2016FDMEVA, PCLindomethacinGenina et al. [144]
IUSFDMPCLindomethacinHolländer et al. [181]
Mesh, IUD, subdermal implant2017FDMPCLestrogen, progesteroneTappa et al. [176]
Bioadhesive filminkjet printingPCL, PEG-PCLpaclitaxel, cidofovirVaran et al. [177]
Bioadhesive film2019inkjet printingPEG-PCLpaclitaxel, cidofovirVaran et al. [182]
Intravaginal ring2021FDMTPUchloramphenicol, metronidazoleArany et al. [178]
Table 9. The grouping of the manufactured micro- and nanoscale drug delivery systems based on the publication year and then in alphabetical order.
Table 9. The grouping of the manufactured micro- and nanoscale drug delivery systems based on the publication year and then in alphabetical order.
Drug Delivery TypeYearType of 3D PrintingType of PolymerType of APIArticle
Nanosuspension2011inkjet-based micro dosing dispenser headnonefolic acidPardeike et al. [183]
Micron-sized dried depositsinkjet printingPVPfelodipineScoutaris et al. [184]
Micropatterns2012inkjet printingPLGArifampicinGu et al. [185]
Microparticlespiezoelectric inkjet printingPLGApaclitaxelLee et al. [186]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bácskay, I.; Ujhelyi, Z.; Fehér, P.; Arany, P. The Evolution of the 3D-Printed Drug Delivery Systems: A Review. Pharmaceutics 2022, 14, 1312. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics14071312

AMA Style

Bácskay I, Ujhelyi Z, Fehér P, Arany P. The Evolution of the 3D-Printed Drug Delivery Systems: A Review. Pharmaceutics. 2022; 14(7):1312. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics14071312

Chicago/Turabian Style

Bácskay, Ildikó, Zoltán Ujhelyi, Pálma Fehér, and Petra Arany. 2022. "The Evolution of the 3D-Printed Drug Delivery Systems: A Review" Pharmaceutics 14, no. 7: 1312. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics14071312

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