You are on page 1of 13

Expert Review of Medical Devices

ISSN: 1743-4440 (Print) 1745-2422 (Online) Journal homepage: http://www.tandfonline.com/loi/ierd20

3D printed drug delivery devices: perspectives and


technical challenges

Mirja Palo, Jenny Holländer, Jaakko Suominen, Jouko Yliruusi & Niklas
Sandler

To cite this article: Mirja Palo, Jenny Holländer, Jaakko Suominen, Jouko Yliruusi & Niklas
Sandler (2017) 3D printed drug delivery devices: perspectives and technical challenges, Expert
Review of Medical Devices, 14:9, 685-696, DOI: 10.1080/17434440.2017.1363647

To link to this article: https://doi.org/10.1080/17434440.2017.1363647

Accepted author version posted online: 04


Aug 2017.
Published online: 17 Aug 2017.

Submit your article to this journal

Article views: 319

View related articles

View Crossmark data

Citing articles: 3 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ierd20
EXPERT REVIEW OF MEDICAL DEVICES, 2017
VOL. 14, NO. 9, 685–696
https://doi.org/10.1080/17434440.2017.1363647

REVIEW

3D printed drug delivery devices: perspectives and technical challenges


Mirja Paloa, Jenny Holländera,b, Jaakko Suominena, Jouko Yliruusib and Niklas Sandlera
a
Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; bDivision of Pharmaceutical
Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland

ABSTRACT ARTICLE HISTORY


Introduction: The technological advancements in the pharmaceutical field are constantly improving Received 9 June 2017
and provide various possibilities for meeting the needs of personalized drug therapy. The three- Accepted 1 August 2017
dimensional (3D) printing technology has endless potential in the fabrication of patient-specific drug KEYWORDS
delivery devices (DDD) and dosage forms as the technological development is progressing. Moreover, Fused deposition modelling;
the rapidly evolving research on 3D printed DDD has enabled us to determine several challenges oral drug delivery;
related to the manufacturing and marketing of personalized drug delivery systems. personalized medicine; solid
Areas covered: In this review, an overview is provided on the relevant accomplishments in the freeform fabrication; tailored
development of 3D printed drug delivery systems, as well as the technical challenges surrounding dosage forms;
the 3D printing of personalized drug-loaded medical devices and dosage forms. Furthermore, observa- three-dimensional printing
tions are presented on the future perspectives of pharmaceutical 3D printing.
Expert commentary: The 3D printing has enabled the fabrication of prototypes of DDD with varying
complexity and shows that customization of drug products is possible. There is potential to improve
patient-specific drug therapies of the future using printing technologies. The technological advancements,
new scientific concepts, interdisciplinary work and defined regulatory guidelines will continue to support
and strengthen the prospects of 3D printing as an option in the manufacture of medical products.

1. Introduction
in 1993 [8]. The techniques utilized in the pharmaceutical field are
Since its initial use, three-dimensional (3D) printing technol- SLA, selective laser sintering (SLS), inkjet-based 3DP, extrusion-
ogy has been used as a rapid and cost-effective prototyping based FDM, and pressure-assisted microsyringe (PAM) printing.
technique in a wide range of different applications including The following features are common to all the 3D printing
aerospace, automotive, construction, jewelry, and fashion [1]. technologies [2]:
3D printing techniques have been applied in small production
volumes, such as prototyping, customization, and the manu- ● The object is created from a 3D model, obtained from
facturing of products with complex designs, which are difficult computer-aided design (CAD), computerized tomogra-
to manufacture with traditional methods. phy, or magnetic resonance imaging scan data.
In the medical field, 3D printing has been used in dentistry, ● The 3D model is sent to the printer’s software as a file. In
tissue engineering, and computer modeling of organs [2]. the software, the 3D model is sliced into a layered struc-
However, it is still in its infancy in the pharmaceutical field. ture, which determines how the layers are constructed.
Several research groups have been investigating the potential ● The 3D object is printed by a layer-by-layer approach.
for 3D printed drug delivery devices (DDD) since the early
1990s, but it was not until 2016 when the first 3D printed The presented overview on the 3D printed dosage forms and
pharmaceutical product was approved by the US FDA [3,4]. DDD presented is largely based on the Master’s thesis by
The term 3D printing, also called solid freeform fabrication, Holländer [9]. The review aims to summarize the recent advance-
additive manufacturing, rapid prototyping, and digital fabrication, ments in the pharmaceutical development of 3D drug delivery
has been intensively researched in the medical and the pharma- systems (DDS) and drug-loaded medical devices, as well as the
ceutical field for the past decades. The 3D printing is an umbrella concurrent technological and regulatory challenges. Finally, key
term for about two dozen 3D printing processes, which use remarks are provided on the current state and the future per-
various printer technologies, hundreds of materials, different reso- spectives of 3D printing in pharmaceutical manufacturing.
lutions and speeds [5]. The first 3D printing technique, stereolitho-
graphy (SLA), was invented by Charles Hull in 1986 [6]. Since then,
2. 3D printing technologies in pharmaceutics
several other printing technologies have been developed, for
example fused filament deposition (FDM) by Scott Crump in The SLA technique was one of the first 3D printing techniques
1992 [7] and three-dimensional printing (3DP) by Emanuel Sachs introduced as a commercial system [2,6]. In the SLA process,

CONTACT Niklas Sandler niklas.sandler@abo.fi Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Biocity,
Tykistökatu 6A, FI-20520 Turku, Finland
© 2017 Informa UK Limited, trading as Taylor & Francis Group
686 M. PALO ET AL.

the layered 3D structures are built by solidifying a thin layer of are fed into the printer through a polymer-liquefying heating
the liquid resin on the movable platform and curing the block. The molten polymer is deposited on the build plate
polymer with an ultraviolet (UV) laser beam across the liquid through the printhead [22]. Upon repeating that process,
surface in a defined depth according to the CAD design. layered structures are formed after polymer solidification.
Photoinitiators are added to the system for the conversion of The FDM process uses material for two different purposes: (i)
light energy into chemical energy by free radical formation. to build the object and (ii) to support the overhangs of the
Thus, post-processing with UV-curing is often necessary to model. So far, the APIs have been incorporated in the fila-
improve the mechanical properties of the printed structures, ments mainly by impregnation or hot melt extrusion (HME).
since the conversion of the reactive groups of the resins is The FDM printing of the drug-loaded polymer filaments results
usually incomplete [10]. The original SLA systems use epoxy- usually in a 3D structure, where the drug is (molecularly)
or acrylate-based monomers that are not suitable for the dispersed throughout the polymer matrix [23]. Since FDM is
development of DDS [11]. However, biodegradable macromers a solvent-free method, it usually does not require drying steps
used in tissue engineering, including polycaprolactone (PCL), or posttreatment [24].
poly(propylene fumarate), trimethylene carbonate, and poly(D, The PAM method, also called semi-solid extrusion (EXT), is
L-lactide) (PDLLA), could be suitable also for pharmaceutical another extrusion-based 3D printing technique. It involves a
applications [12]. layer-by-layer deposition of semisolid or paste materials
In SLS, materials in a powder form are sintered by a laser through mechanical, pneumatical or solenoid-based extrusion
beam. The 3D structures are built by the fusion of thin powder [25,26]. A single polymer layer is deposited on the build plate
layers with a high-power pulsed laser beam and by controlling through the nozzle which is moving in the X-Y orientation.
the movement and heating of the SLS fabrication platform [2]. When the layer is completed, the build plate is lowered and
The suitable materials for SLS should be thermally stable below the process is repeated. The PAM systems are often called
their melting temperature to enable the powder fusion by bioprinters with the extrusion-based bioprinting technique,
heating. The SLS printing of 3D structures leaves powder resi- because this method was first utilized to print living cells
dues on the final structures and processing leftovers; thus, and other biologics for the fabrication of living tissue and
requiring posttreatment of the formulations and protocols for organs [26]. To avoid confusion, bioprinting should be used
the waste collection or powder reusage [13]. SLS is traditionally only when printing living cells and biologics, whereas the
used with metal powders; however, polymers like polyamide, technique should be referred to as PAM or EXT 3D printing
polystyrene, polypropylene, and thermoplastic elastomers are when inert materials are printed [22,25,26].
commercially available for SLS [14].
In the 3DP, also called binder jetting, the 3D structure is
3. 3D printing of personalized drug delivery devices
built up with powder as the build material and liquid as the
binding material [2]. Briefly, the 3DP printing process consists Printing technologies are evolving fast and offer various
of two main steps that are repeated to produce a layered potential applications in the pharmaceutical field. One of the
3D structure. First, a layer of powder is spread onto the build benefits of pharmaceutical 3D printing is providing opportu-
plate with a roller. Second, the binding material is jetted onto nities for individualization of drug therapies and customization
a predefined location on the powder layer by movable inkjet of DDD. The overall development concept of 3D printed DDD
printhead(s) and the powder is bound together only in the in personalized medicine is presented on Figure 1.
printed areas. Several commercially available 3DP printers The need for personalized medicine has received growing
have been utilized in the development of printed DDD and attention for many years already. Several patient groups, such
scaffolds [15–19]. as pediatric and geriatric patients could benefit from the
After the first patented FDM technique in 1992, several personalization of the DDD [27,28]. There is a need for custo-
single or multi-extruder systems utilizing the fused filament mized DDD due to the genetic variations in the treatment
fabrication technique have entered the market [20,21]. The responses [29]. The development of diagnostic tests based
FDM experimental desktop printers are relatively cheap and on genetics makes it possible to optimize treatments for
versatile. The FDM process uses solid polymer filaments that every patient.

Figure 1. The design concept of three-dimensionally (3D) printed drug delivery devices (DDD) for personalized therapy.
EXPERT REVIEW OF MEDICAL DEVICES 687

The 3D printing allows manufacturing DDD with varying oligomers that would be biocompatible and approved for
geometries and/or with modified drug-release profiles that are human use [22]. As an additional drawback, the SLA printers
challenging to achieve by conventional drug manufacturing operate primarily with single materials. Thus, the fabrication of
techniques. Drug-release kinetics can be controlled by the formulations with multiple materials, such as polymer mixtures
DDS design and by the selection of polymers and their com- and drug-loaded structures, is limited [48].
binations. In the printed DDS, the dosing can be adjusted The manufacturing speed and the physical size of the desk-
according to the individual needs of the patient by changing top printers contribute to the suitability of the SLA fabrication
the dimensions of the printed formulation and/or by modify- of personalized DDS at the point of care [48–50]. The high
ing the content of the solid material(s). The facility and versa- accuracy and resolution of the SLA printed structures makes it
tility to independently tailor the drug delivery of multiple an attractive approach for the preparation of DDS with com-
drugs from the 3D printed DDS makes it suitable for patient plex 3D inner structures [10,48]. Furthermore, the UV photo-
groups that need several drugs simultaneously, for example polymerization has shown no significant drug degradation
patients with metabolic diseases [30]. These DDS could be (<5%) after processing [51–53].
obtained by designing formulations with complex inner struc- For pharmaceutical applications, SLA has been studied for the
tures and compartmentalized matrix systems. manufacturing of drug-eluding scaffolds [51], topical patches
Aprecia® Pharmaceuticals, founded in 2003, was the first [53], torus-shaped tablets [52], and microneedles [54–56] with
pharmaceutical company to launch a 3D printed drug product poly(ethylene glycol) diacrylate (PEGDA) as the base material
[3,4]. In 2008, the ZipDose® Technology was invented as a (Figure 2). Goyanes et al. [53] prepared the drug-eluding topical
proprietary 3D printing technique mainly for manufacturing patches with salicylic acid according to a scanned image of an
high-dose and easy-to-swallow products for central nervous individual’s nose (Figure 2(b)). The flexibility of the nose-shaped
system therapeutics. Other pharmaceutical companies, such as patches was adjusted by controlling the degree of crosslinking
GlaxoSmithKline (GSK), have expressed interest in this field as between PEGDA chains with the addition of poly(ethylene
well [31]. Printing techniques have been studied to generate glycol) (PEG) as a filling agent. In transdermal drug delivery, the
high-throughput screening arrays for drug screening and drug micro-SLA method has shown to be suitable for preparing micro-
testing [32]. The collaborations between pharmaceutical and needles with various shapes [56] and antimicrobial coatings
printing companies are aiming to provide cheaper and more [54,55]. The SLA-fabricated DDS are suitable for personalized
personalized healthcare solutions in the future [2]. In addition, medicine with patient-customized size and drug loading at
3D printed human skin and organs offer an alternative for high accuracy and inter-formulation uniformity [52–54].
animal testing in cosmetic and biomedical research [2,33]. Nevertheless, SLA has several technology-driven limitations
Besides 3D printing technologies, the two-dimensional (2D) that restrict its applicability for printing pharmaceuticals (Table 1).
printing technologies, i.e. inkjet and flexographic printing, To be able to manufacture DDD with higher drug loadings, the
have also been applied in the pharmaceutical field. The inkjet API must be adequately soluble in the polymer, which restricts
printers are non-contact systems that are based on either the use of SLA for high-dose DDD. So far, the drugs have been
continuous or drop-on-demand jetting technologies, whereas dissolved in the photopolymer for the preparation of DDD with
flexography is a rotary-type contact printing method [34,35]. low drug loadings of 1–5.9% [51–53]. However, the manufactur-
Reviews on the applications of 2D printing technologies in ing of printable objects with homogenously suspended particles
pharmaceutics have been provided by Kolakovic et al. [36], (with loadings up to 53%) has been reported [10]. This indicates
Alomari et al. [37], and Preis et al. [38]. that the SLA printing of DDS from resins containing undissolved
drug particles at higher drug content could be possible.

3.1. Stereolithography
3.2. Selective laser sintering
The SLA technology has been successfully applied in tissue
engineering [12,39–42] and in prototyping of customized surgi- The SLS technique has been investigated in the pharmaceutical
cal implants [43–47]. However, the use of SLA in pharmaceutical field only by a few research groups [13,57–60]. The designed
applications is limited by the availability of photopolymerizable DDD with customized porosity and microstructures have been

Figure 2. Drug delivery devices by stereolithography (SLA). Images of drug-loaded tablets (a) [52] and topical patches (b) [53], and scanning electron micrographs of
drug-doped microneedles (c) [54]. Reprinted from [52,53] with permission from Elsevier. Reprinted from [54] by permission of John Wiley and Sons, Inc.
688 M. PALO ET AL.

Table 1. Challenges of stereolithography (SLA) printing in the preparation of 3.3. 3D printing


drug delivery devices (DDD).
Topic Challenges The 3DP is a simple, versatile, low cost, and high-speed process
Materials Limited availability of photopolymerizable materials for SLA- that provides various possibilities for the preparation of perso-
printed DDD that would be biocompatible and nontoxic nalized DDS as well as for customizing the composition and
for human use. properties of drug-eluding implants. The physicochemical prop-
API The photopolymerization process and the post-processing
with UV-curing could be unsuitable for the APIs and erties of the dosage forms, and the drug-release profiles could
restrict the SLA applicability in the production of DDD. be adjusted by using different polymers and/or ratios between
Dosage form The fabrication of multi-component formulations is limited, the bulk material, API(s), binding agents, and other excipients.
design especially at higher drug loadings.
Process The printing speed of a conventional SLA process is Furthermore, the appearance and the therapeutic performance
relatively low compared to other 3D printing methods. could be tailored by adjusting the size and shape of the DDD
Cost The SLA printing instruments are comparatively expensive. and by incorporating functional excipients (e.g. drug release-
3D: three-dimensional; API: active pharmaceutical ingredient; DDD: drug deliv- modifying agents, colorants, taste masking agents, etc.), into the
ery device; SLA: stereolithography; UV: ultraviolet.
binder solution or the bulk powder mixture.
Since the printing process is performed at ambient tempera-
prepared with nylon [13], polyamide [57], poly(L-lactic acid) ture, the 3DP is suitable for thermolabile drugs and excipients.
(PLLA) [58], and PCL [58–60] as the polymeric base material. However, printing parameters such as the time interval between
The impact of several process parameters on the porosity of two printing layers, and the distance between the nozzles and
the scaffolds was investigated by Low et al. [13]. In the designed the powder bed must be optimized for successful 3DP printing.
DDD, the inner and outer parts were manufactured with differ- The drawbacks of the 3DP technique are presented in Table 3.
ent porosities. The results showed that the porosity of the DDD The first 3DP pharmaceutical formulations were built up
could be controlled by varying the powder bed temperature, the from PCL, polyethylene oxide, and dye-loaded binder solu-
length between the dense walls and the SLS laser power. Leong tions, demonstrating that 3DP can reproducibly fabricate
et al. [58] demonstrated the versatility of SLS in the manufactur- DDD with controlled release profiles [61]. Since 1996, several
ing of microporous DDD. The SLS printed discs with controlled DDD and implants have been designed and developed to
porosity were obtained by varying the laser power intensity and tailor and control the release of pharmaceuticals for better
the laser scan speed. In another study, the same research group therapeutic effect. For example, Lin et al. [62] manufactured
manufactured four different cylinder-shaped formulations to three different designs for subdermal implantation of ethiny-
investigate how varying the number of dense rings in the struc- lestradiol: (i) the single-channel device, where the drug was
ture impacts the initial burst release of drugs [59]. The initial incorporated in the channel; (ii) the device with the drug
burst release followed by a slow and constant release was homogenously distributed in the polymer matrix; and (iii) the
reported for all designs with the lowest release obtained for device, where the drug was placed under a concentration
the formulation containing the highest number of dense rings. gradient into a polymer matrix. The in vitro and in vivo studies
Salmoria et al. [60] manufactured a reservoir-type DDS with the showed that the drug release from the single-channel device
model compound progesterone by SLS. The drug-loaded DDS (i) stayed at a steady level for the first five weeks before
with the API either within the core or within the core and the
shell of the device showed controlled zero-order drug release.
For the fabrication of tailored DDD, the SLS method pro- Table 3. Challenges of three-dimensional printing (3DP) in the preparation of
vides possibilities for controlling the porosity and the drug- drug delivery devices (DDD).
release kinetics of the 3D printed structures. Nevertheless, the Topic Challenges
contribution of this method to the pharmaceutical manufac- Materials The availability of suitable nontoxic solvents for increasing
turing has remained modest so far. The limited use of SLS in the binder solution capacity for dissolved/solubilized
materials is limited.
pharmaceutical field is driven by the disadvantages related
Dosage form Compared to other methods, the 3DP has lower printing
mainly to the printable materials and the process parameters design resolution and poorer surface quality that can result in
(Table 2). Besides these restrictions, the low interest in SLS has imprecise printing results and affect the DDS/DDD
performance. This could be improved by the additional
also been caused by the lack of affordable printers [2].
post-processing.
Process The preprocessing of the starting powder materials might
be necessary to ensure the suitable particle size
Table 2. Challenges of selective laser sintering (SLS) printing in the preparation distribution and powder flowability for uniform filling of
of drug delivery devices (DDD). the power bed.
Clogging of the printhead might occur during printing of
Topic Challenges nonhomogeneous binder solutions.
Materials Limited availability of pharmaceutical excipients and APIs, which Migration of the binder solution contributes to the
are suitable for SLS without degradation at high energy levels. deflections from the original design and the
Process The production of SLS-fabricated DDD is time consuming. SLS inhomogeneity between the printed DDS/DDD.
printing speed is limited to approximately 1–5 cm/h. The 3DP dosage forms have typically lower inter-product
Posttreatment of the printed DDD is required due to non-sintered homogeneity and higher risk for internal agglomeration.
powder residue on the final structures. The porosity of 3DP products is poorly controllable.
Waste SLS produces significant powder waste after printing process. DDD quality Lower mechanical strength of 3DP products limits their
Cost The SLS instruments are comparatively expensive due to the low packaging, administration, and overall handling
amount of commercially available printers. processes.
API: active pharmaceutical ingredient; DDD: drug delivery device; SLS: selective 3DP: three-dimensional printing or binder jetting; DDD: drug delivery device;
laser sintering. DDS: drug delivery system.
EXPERT REVIEW OF MEDICAL DEVICES 689

declining gradually, whereas other designs (ii, iii) showed a 3DP with pharmaceutical grade excipients as matrix forming
higher drug-release rate from the DDD within the first weeks. agents and binding additives.
The structural integrity of the single-channel DDD (i) was Katstra et al. [15] fabricated cellulose-based delayed-release
maintained during the implantation of 5 months. tablets with chlorpheniramine maleate within the binder solu-
For certain indications, the local delivery of antibiotics is tion. The printed tablets had the API-loaded layers in between
more advantageous compared to the systemic drug delivery, the placebo bottom and top layers. Furthermore, Katstra et al.
providing a relatively higher therapeutic efficacy and a lower [15] used a fluorescein-activated binder solution to evaluate
systemic toxicity. Local delivery of antibiotics by 3DP implants the binder jetting uniformity of the 3DP process. A good
to treat diseases such as chronic osteomyelitis and bone correlation between the fluorescence intensity and the
tuberculosis has been studied by several research groups expected fluorescein content showed that it could be possible
[18,63–69]. For example, drug-eluting 3DP ceramics based on to precisely define the drug amount by controlling the API
tricalcium phosphate (TCP) have been investigated by concentration in the binder solution. Rowe et al. [16] fabri-
Gbureck et al. [63,65] with vancomycin, ofloxacin, tetracycline, cated four different types of complex oral DDS, including
levofloxacin and by Inzana et al. [67] with rifampicin and immediate-extended, breakaway, enteric dual pulsatory, or
vancomycin. The use of TCP-based 3DP systems has several dual pulsatory devices, with chlorpheniramine maleate and
advantages compared to the established polymethylmetha- diclofenac as model APIs. The drug-release rate from the 3DP
crylate drug-eluting scaffolds manufactured by conventional printed DDS could be controlled by the polymer(s) and their
methods: (i) higher release rate due to the porous structure, (ii) concentration(s) either in the powder matrix or in the binder
no need for a second surgery to remove the implant, and (iii) solution [15,71].
the possibility of incorporating several antibiotics [63]. The 3DP technique has shown to be beneficial for produ-
Recently, Wu et al. [18] studied columnar-shaped, doughnut- cing DDS with variable doses, especially at high drug loadings.
shaped, and multilayer doughnut-shaped implants with PLLA In the study by Yu et al. [19], the controlled-release tablets
matrix and isoniazid as the model antibiotic (Figure 3(a)). The were manufactured by adding the API in the powder bed
implants showed an initial burst release, but the release from instead of the binder solution; thus, achieving higher drug
multilayer doughnut-shaped implants stabilized after few days loadings compared to the earlier studies, where the drug
due to the constant effective surface area. In another study, Wu was dissolved in the binder solution. The drug loadings up
et al. [68] manufactured multilayered concentric multidrug to 68% of the weight of the 3DP-printed tablets were
implants containing antimicrobial tobramycin and levofloxacin obtained. The drug-release profiles were influenced by the
in a PDLLA matrix for sustained and programmed drug release. polymers that were incorporated into the DDS core and the
In these multidrug implants, an orderly release of the drugs drug concentration gradient in the tablets. Yu et al. [17]
from the periphery to the center was obtained. The antimicro- further increased the drug loading in the 3DP tablets by
bial drug with effectivity in low concentrations could be utilized incorporating the drug both in the powder bed and in the
in these types of implants [18,68]. However, higher drug binder solution. These acetaminophen tablets were fabricated
loadings could be achieved by dispersing the drug in the with a doughnut-shaped multilayered 3D structure. In addi-
powder bed, compared to the incorporation of the API into tion, they showed that the printing of top and bottom layers
the binder solution. with an inert and impermeable polymer hindered the diffusion
In the personalized DDS design, the 3DP of pharmaceuti- of water and drug, and the incorporation of a release-retar-
cals has been mainly aimed at manufacturing dosage forms dant material into the binder solution in the peripheral region
with layered 3D structures and highly variable drug loadings of the tablets prevented initial burst release and provided a
to control and modify the drug-release profile. Oral dosage linear drug release.
forms, including tablets [15–17,19,71] and fast-disintegrating The high porosity of the 3DP-printed tablets contributes to
tablets (FDTs) [70,72–75], have been successfully prepared by the rapid disintegration of the DDS within the oral cavity. Lee
at al. [72] prepared mannitol-based oral FDTs with captopril,
where the dispersion time for each FDT decreased when the
binder saturation level (binder liquid content per tablet) and
the level of the additive powder decreased. Yu et al. [70,73]
manufactured oral FDTs with a loose powder core to obtain a
fast disintegration and dissolution of acetaminophen that was
dispersed in the powder bed at 40% loading (Figure 3(b)).
Moreover, the first prescription drug product on the US mar-
ket that is manufactured using a 3DP technology has been
formulated to disintegrate within a few seconds in the mouth
when taken with a sip of liquid [3,4,75]. The antiepileptic
Spritam® (levetiracetam) tablets for oral suspension from
Aprecia® Pharmaceuticals are available in four different
Figure 3. Drug delivery devices by three-dimensional printing (3DP). Images of strengths with the API content of 60–90% in the bulk powder
a drug-loaded doughnut-shaped tablet (a) [18] and the cross-section of a fast-
disintegrating drug delivery device with a predefined inner structure (b) [70]. mixture. Levetiracetam, cellulose, mannitol, and colloidal sili-
Reprinted from [70] by permission of John Wiley and Sons, Inc. con dioxide are incorporated in the powder matrix of these
690 M. PALO ET AL.

tablets, and the binder solution contains mainly water, PVP, Table 4. Challenges of fused deposition modeling (FDM) in the preparation of
drug delivery devices (DDD).
Tween 20, and glycerol [4].
Topic Challenges
Materials Due to the heating step in the FDM printing, the materials
3.4. Fused deposition modeling are limited to thermoplastic polymers.
API Traditional drug-loaded filaments from thermoplastic
FDM is perhaps the most extensively investigated 3D printing polymers that require high processing temperatures are
generally not suitable for the preparation of DDS with
technique in the pharmaceutical field. The flexibility of FDM thermolabile APIs.
allows manufacturing of drug-eluting devices and DDS with Dosage form The design of complex structures might require printing of
various geometries (Figure 4(a)) and/or modified drug-release design support structures that need to be removed during post-
processing.
profiles for patient-specific treatments at high reproducibility Process FDM printing process requires pre-preparation of printable
[24,76]. The drug dosing could easily be adjusted through the filaments.
adjustment of the physical parameters of the designed DDS Compared to HME, the drug incorporation into the polymer
filaments by impregnation is time consuming and results
(Figure 4(b)) [76] or by changing the infill percentage of the in a lower drug loading yield.
formulated dosage forms [80,81]. Despite its various applica- API: active pharmaceutical ingredient; DDD: drug delivery device; DDS: drug
tions, several challenges in the printing of DDD have also been delivery system; FDM: fused deposition modeling.
identified over the years (Table 4).
In the FDM-printed products, the drug is incorporated
into the printable filaments prior to printing. The impreg- of the drugs has been reported to be dependent on the shape
nation of APIs into polyvinyl alcohol (PVA) filaments has of the tablets, particularly on the surface area/volume
been investigated for printing tablets with different shapes ratio [77].
at various drug loadings [24,80,81]. The impregnation pro- In addition to the original feedstock material PVA, other
cess is based on passive diffusion and requires saturated or pharmaceutical grade polymers have also been investigated for
highly concentrated drug solutions; therefore, it is an FDM, including PVP [86], methacrylic polymers [76,84,85,87],
expensive and time-consuming process [22]. Another draw- cellulose derivatives [85], polyvinyl caprolactam-polyvinyl acet-
back is that the drugs are mainly incorporated on the sur- ate-polyethylene glycol graft copolymer (Soluplus®) [84,85], and
face of the filaments, resulting in a low loading percentage PVA-polyethylene glycol graft copolymer (Kollicoat®) [85].
[24,80,81]. The higher solubility the drug has in the impreg- In addition to single drug systems, FDM method is feasible
nation liquid, the smaller percentage of the drug is loaded for producing DDS with multiple APIs. For example, PVA-based
into the polymer filament [81]. caplets with two APIs (acetaminophen and caffeine) printed as
Compared to the impregnation method, HME can be used layered caplets or caplet-in-caplet systems have been success-
to prepare filaments with higher drug loading in the range of fully manufactured with a multi-extruder FDM printers [82].
5–40% [76,77,82–87]. In the HME process, the melted polymer Furthermore, fabricating DDD from drug-free filaments and
is mixed with the drug and other excipients. Depending on filling them with APIs after printing, allows changing the API
the properties of the drug, i.e. its melting point and solubility or the formulation without affecting the printing process [88–
in the polymer, the drug either (i) dissolves, (ii) melts, or (iii) 90]. These alternative FDM-printed DDS could be suitable also
disperses in the molten polymer [23]. Tablets with different for thermolabile APIs [88–90]. For example, Markl et al. [88]
shapes (e.g. caplets, cubes, pyramids, torus, etc.) and drug- fabricated one compartment cylindrical devices from PVA or
release profiles have been manufactured from extruded PVA poly(lactic acid) (PLA) with carbamazepine powder filling, and
filaments with drug loadings of 5–10% [77,82,83]. The higher two-compartment cylindrical devices from PVA with saquina-
drug loading reduced the quality of the filaments and/or the vir and halofantrine within the inner and outer compartments,
DDS [77,82,83]. However, the addition of plasticizers could respectively [88]. Besides the design aspects of the formula-
help to increase the drug loading of the HME filaments com- tions, Markl et al. [88] showed that X-ray computed microto-
pared to the original PVA filaments [84,85]. The release profile mography and terahertz pulsed imaging were suitable for

Figure 4. Drug delivery devices by fused deposition modelling (FDM). Images of printed drug delivery systems (DDS) with adjustable dosing by scaling (a) [76] and
different geometries (b) [77], and printed prototypes of intrauterine systems and filaments with 15% of drug loading (c) [78,79]. Reprinted from [76–79] with
permission from Elsevier.
EXPERT REVIEW OF MEDICAL DEVICES 691

qualitative and quantitative characterization of the microstruc- The mechanical properties of the printed dosage forms com-
tures of the 3D printed oral devices. plied with the USP specifications for weight variation, thick-
Despite the successful manufacturing process, the thermal ness, hardness, and friability. Expectedly, the hardness and the
analysis of the FDM formulations has indicated that high friability of the printed tablets differed significantly from the
processing temperatures can lead to significant degradation commercial compressed tablets due to the lack of compres-
of thermolabile APIs [81,87]. However, a suitable polymer sion force in the 3D printing process.
system could help to stabilize the crystalline API within its In other studies, Khaled et al. [30,97] demonstrated the
matrix at temperatures above the melting point of the API feasibility of PAM printing in the manufacturing of compart-
[77]. Nevertheless, for thermolabile drugs, polymer filaments mentalize tablets with multiple drugs. The three-compartment
with melting point below the degradation temperature of the tablets [97] and five-in-one polypills (Figure 5) [30] were
API are needed despite the short heating time in the designed to physically separate the drugs to avoid incompat-
printhead. ibility issues and to achieve a desired independent control of
A lot of research has also been published on FDM printed the drug release. The mechanical properties of the designed
scaffolds and implants, whereas the amount of scientific polypills were acceptable for handling without any loss of
publications on drug-loaded implants or medical devices structural integrity [30,97].
prepared by FDM printing is limited [78,79,91–94]. Printed PAM has also been applied to develop carrier systems for
devices from drug-loaded PLA filaments with antimicrobial targeted cell therapy. For example, Song et al. [98] manufactured
nitrofurantoin have been studied to investigate the potential 3D printed drug carriers for the transplantation of xenogeneic
of producing custom-made devices (catheters) and implants cells. The PAM printing was performed with cyclosporin A loaded
(discs, beads) for controlled drug release and personalized PLGA microspheres, which were mixed with an alginate hydrogel.
drug therapy [91–93]. Intrauterine systems with anti-inflam- The 3D structure was strengthened by blending the drug-loaded
matory indomethacin as the model drug have been prepared hydrogel with a PCL/PLGA structure during the printing.
by FDM from filaments based on slowly biodegradable PCL Drug-eluting scaffolds manufactured by PAM have been
[78] and a nondegradable polymer ethylene-vinyl acetate investigated by Zhu et al. [99] and Martínez-Vázquez et al.
(EVA) [79] (Figure 4(c)). Furthermore, Goyanes et al. [53] [100]. Zhu et al. [99] manufactured dual drug-eluting implan-
manufactured personalized-shaped patches containing sal- table scaffolds for the treatment of osteoarticular tuberculosis
icylic acid with aid of 3D scanning technology and FDM with isoniazid and rifampicin. The prepared scaffolds exhibited
printing. The nose-shaped structures were printable from higher mechanical strength, drug loading capacity, and pro-
PLA filaments, whereas PCL filaments were suitable for pro- longed drug-release profile compared to the commercial cal-
ducing circular patches [53]. cium phosphate scaffolds, where the drugs were absorbed on
In general, FDM method allows preparing DDD with good the surface of the scaffolds by a dropping method.
mechanical properties at relatively high resolution. Therefore, Most chemotherapeutic drugs have poor aqueous solubility
sufficient control of the mass of the printed units and the high and can cause severe adverse effects if administered systemati-
dosing accuracy of the APIs can be achieved [24]. Nevertheless, cally; therefore, they are usually delivered locally as injections or
the FDM printing process is a complex interplay between many implants. Yi et al. [101] printed a local delivery patch for the
variables that need optimizing for each new feedstock material delivery of the chemotherapeutic 5-fluorouracil. A high drug-
and the printed DDD design. loaded paste, containing a polymer blend of PLGA and PCL, was
extruded as a biodegradable patch and implanted in pancreatic
cancer model mice. The drug release from the patches was
3.5. Pressure-assisted microsyringe
adjusted by altering the surface area, porosity and thickness of
The PAM extrusion method has found use in the pharmaceu- the patch.
tical field quite recently. It is advantageous in producing per- PAM is a suitable printing technique for many biocompatible
sonalized DDS with multiple drugs and in fabricating DDD and materials [102]. Nevertheless, the PAM printing process has also
drug-eluding scaffolds from pastes, hydrogels and other vis- some significant manufacturing limitations [102,103] (Table 5).
cous polymer systems. Materials with viscosity ranging from 30 to > 6 × 107 mPa·s have
Rattanakit et al. [95] were the first to utilize a PAM-based been exploited in the PAM printers. Despite of that, the drug
laboratory-constructed printer for the preparation of DDD. The loading capacity of semi-solids is limited by the rheological prop-
biodegradable DDD were printed with a PVA-based paste erties of the materials and might require significant adjustment of
containing dexamethasone that was deposited either on the printing parameters. The utilized printing temperatures are
poly(lactide-co-glycolide) (PLGA) films or sandwich printed moderate and the printing is often performed at room tempera-
with PLGA to fabricate DDD with a scroll configuration. The ture; therefore, PAM is highly suitable printing method for thermo-
drug-release kinetics depended on the geometry of the DDD, labile APIs. Furthermore, changing between the different materials
e.g. the thickness of the printed film, and showed a controlled could be easily achieved with multi-head extruder systems.
long-term release up to 4 months.
The PAM printer with a multi-head extruder system can be
4. Regulatory perspective on 3D printing of drug
highly beneficial for fabricating layered DDS with multiple
delivery devices
drug-release profiles. For example, Khaled et al. [96] prepared
bilayer tablets with guaifenesin to mimic the drug-release Although it was not until recently FDA approved the first 3D
profiles of commercially available guaifenesin bilayer tablets. printed oral dosage form submitted under the 505b(2) pathway,
692 M. PALO ET AL.

Figure 5. Drug delivery devices by pressure-assisted microsyringe (PAM) printing. Image of drug delivery systems (DDS) with multiple drugs (left) and the schematic
structural diagram of the DDS design (right) [30]. Reprinted from [30] with permission from Elsevier.

Table 5. Challenges of pressure-assisted microsyringe (PAM) printing in the derived from the aspects of personalized medicine [106,107].
preparation of drug delivery devices (DDD).
Due to the complexity of 3D printed DDD and dosage forms,
Topic Challenges many issues on the liability, intellectual rights and data pro-
Materials The polymers with crosslinking mechanism or shear- tection need to be addressed to protect manufacturers and
thinning properties are highly preferred.
The printability of the materials is affected by their the end-users. The exploitation instructions, appropriate sto-
rheological properties. The applied extrusion forces are rage conditions and safety of the computationally designed
dependent on the viscosity of the semisolids. dosage forms and medical devices, as well as the patient-
API The APIs are required to be dissolved or uniformly dispersed
in the semisolid printing material. related data, must be ensured to avoid possible liability
Dosage form The printing resolution of PAM printing is limited by the problems related to the misuse of the information and to
design nozzle size of the microsyringes. minimize the risk for manufacturing errors. As with any new
Process Drying of the printed formulations might be required before
final solid formulations are obtained. This could be production line/system, the validity procedures should be
avoided by crosslinking during printing. strictly determined and the responsible bodies should be
The PAM printing process is relatively slow extrusion appointed to avoid any fraud or negligence. It must be men-
technique.
DDD quality The mechanical strength and durability of the printed tioned that with the advent of improved online analytical
formulations is low. Posttreatment, e.g. by crosslinking, systems, such as different kind of spectrometers, there are
might be applicable to improve these properties. good prospects for integrating quality control tools to printers
API: active pharmaceutical ingredient; DDD: drug delivery device; PAM:pressure- in an inexpensive way to monitor the quality of every
assisted microsyringe.
printed DDD.
In the European Union legal framework, the marketed
medicinal products are subjected to the Directive 2001/83/EC
the FDA has already for 10 years reviewed and cleared many 3D related to the medicinal products for human use, when the 3D
printed non-implantable and implantable medical devices printed devices are either ‘prepared industrially or manufac-
through the 510(k) process. The medical devices that have been tured by a method involving an industrial process’ [108].
approved by FDA have all been reviewed and accepted according Medicinal products that do not fall under that category should
to the existing regulations by recognizing similarities and differ- be evaluated on case-by-case basis. The 3D printed DDS may
ences with existing technologies [104]. Naturally, the printed DDD also be defined as magistral formula, officinal formula, or
must be made in accordance with the current chemistry, manu- investigational medicinal products, if the appropriate regula-
facturing and control (CMC) standards as set forth in FDAs 21 CFR tory requirements are fulfilled. In addition, it should be noted
200 & 300 series and other relevant guidance, and thus follow the that the recently published European Union regulations on
same pathway and manufacturing requirements as conventional medical devices that will come into force in spring 2020 will
medical devices/products. define the rules and acceptance criteria for the medical
On 10 May 2016, FDA issued a draft guidance document for devices and in vitro diagnostic medical devices. To date,
the additive manufactured devices, where the design, manu- there are no valid legislations, neither national nor interna-
facturing, and testing issues are taken into account [105]. The tional, that would cover the 3D printed DDS and DDD as a
scope of the guidance is to describe the flow of the manufac- separate category of medicinal products.
turing process and to outline the critical design, manufactur-
ing and post-processing parameters. Additionally, the draft
guidance outlines the process validation and device testing
5. Expert commentary
considerations. The current pharmaceutical regulations are
being reassessed to resolve issues related to the process 3D printing is in an intriguing state from the pharmaceutical
units, pharmaceutical ink formulations, printers, printing development viewpoint. The rapidly increasing research in the
designs, and the final printed DDS. field has led to the fabrication of complex prototypes of DDS
In personalized drug therapy, the marketable 3D printed and DDD with the aim to provide customized drug products
medicinal products will also face several regulatory hurdles and to improve the patient-specific drug therapy. However,
EXPERT REVIEW OF MEDICAL DEVICES 693

the introduction of 3D printing into the pharmaceutical indus- Key issues


try has barely started with the first commercial product reach-
● Concepts of 3D printing of pharmaceuticals have evolved
ing the US market in 2016. A considerable increase in the
rapidly over the last few decades. The fabrication of 3D
activity and the interest in the field by the pharmaceutical
printed drug delivery devices (DDD) has enabled the devel-
industry can be seen after the approval of Spritam® by FDA.
opment and production of formulations with complex
With the technological advancements as well as the new
structures that are difficult or impossible to achieve with
concepts generated in the scientific community, solid dosage
traditional manufacturing techniques.
form design and the manufacturing concepts have the possi-
● The 3D printing provides various opportunities for the indi-
bility to develop and evolve. The researchers have been coura-
vidualization of drug therapy and for the patient-specific
geous in synthesizing and testing the feasibility of novel
tailoring of DDD and drug delivery systems (DDS).
materials, and developing dosage forms and complex designs
● The 3D printing is an umbrella term for several 3D printing
for advanced and more accurate drug therapy. Since the
methods, which are based on different technological pro-
applications of 3D printing expand over a vast range of dis-
cesses and use various types of materials. The main techni-
ciplines from electronics to medicine, the scientific collabora-
ques applied in pharmaceutics are fused deposition
tions and interdisciplinary work could help to establish smart
modelling (FDM), jetting-based three-dimensional printing
pathways for achieving novel therapeutic solutions.
(3DP), pressure-assisted microsyringe (PAM) extrusion print-
After approximately two decades, the possibilities and lim-
ing, stereolithography (SLA) and selective laser sinter-
itations in the 3D printing of pharmaceuticals have started to
ing (SLS).
form, which has also been the driving force for reviewing
● In general, the 3D printed products are produced according
regulatory documents to provide up-to-date guidelines for
to a computer-aided design (CAD) that is sliced into prin-
the pharmaceutical manufacturers. Nevertheless, it is expected
table layers. A layer-by-layer fabrication approach is used to
to take several years, before the legal aspects of pharmaceu-
obtain the final solid 3D structures.
tical printing are clearly understood, and 3D printing can be
● The 3D printing is suitable for the fabrication of DDS with
considered a natural part of the pharmaceutical manufactur-
tailored doses, multiple drugs and/or controlled drug
ing system. It remains to be seen whether the compounding
release kinetics. The personalization of the 3D printed
route will play role in bringing the technology and drug
DDS can be achieved through the customized design of
manufacture closer to the patient.
the DDS, including the use of dosage forms with various
geometries, the scaling of the dosage form dimensions and
the formulation of compartmentalized dosage forms.
6. Five-year view ● The technological challenges of pharmaceutical 3D printing
The development of 3D printed medical devices is expected to are mainly related to the limited availability of suitable
progress by the knowledge-driven design within the next materials, the incompatibility of the drugs with the printing
5 years and beyond. The experimental manufacturing of DDS conditions and the need for post-processing.
and other medical products by advanced and emerging tech- ● The fabrication of 3D printed DDD and DDS must be in
nologies, e.g. 3D printing, will continue to increase at a steady accordance with the current pharmaceutical manufacturing
pace. As the utilization of 3D printing evolves, viable business standards for conventional medical devices/products.
models that allow improved therapies by printing will be Currently, the 3D printed DDD and DDS are not a separate
founded. This approach will be especially suitable for certain category of medicinal products.
therapies, where there is a need for tailoring of drug delivery, ● In the future, the scientific collaborations within the 3D
dosing and/or the geometry of the devices. printing community in academia and industry will be cru-
From the research and technical development point of cial for establishing smart solutions and pathways for novel
view, the production units with integrated quality control therapeutic concepts to improve treatments in specific
systems will start to emerge on the market to permit printers areas. The technological solutions with the hardware, soft-
to be used closer to the patient at hospitals and pharmacies. ware and integrated quality control units aimed specifically
This allows quality-by-design pharmaceutical manufacturing for pharmaceutical applications would immensely support
and real-time monitoring at the production unit and the the progress of 3D printing within the field.
development of high quality dosage forms and devices.
Furthermore, different 3D printing methods are expected to
support the development of complex delivery systems with
Acknowledgments
easily controllable parameters for personalized drug therapy.
We will most likely see printers and printheads developed for The authors would like to thank Risto Hakala, Joonas Mikkonen and
specific products rather than having one-printer-fits-all solu- Christine Talling from Bayer Oy for review of the text.
tions. From a technological aspect, tailored printable materials
might be needed; 3D printers and software have to be
designed in a manner that they meet the specific needs of Funding
pharmaceutical manufacturing to ease the progress of imple-
menting 3D printing for better treatments. This paper was funded by the Abo Akademi University.
694 M. PALO ET AL.

Declaration of interest 24. Skowyra J, Pietrzak K, Alhnan MA. Fabrication of extended-release


patient-tailored prednisolone tablets via Fused Deposition Modelling
The authors have no relevant affiliations or financial involvement with any (FDM) 3D printing. Eur J Pharm Sci. 2015;68:11–17.
organization or entity with a financial interest in or financial conflict with 25. Alhnan MA, Okwuosa TC, Sadia M, et al. Emergence of 3D printed
the subject matter or materials discussed in the manuscript. This includes dosage forms: opportunities and challenges. Pharm Res.
employment, consultancies, honoraria, stock ownership or options, expert 2016;33:1817–1832.
testimony, grants or patents received or pending, or royalties. •• Review on 3D printing of oral solid dosage forms.
26. Ozbolat IT, Hospodiuk M. Current advances and future perspectives
in extrusion-based bioprinting. Biomaterials. 2016;76:321–343.
References 27. Sandler N, Preis M. Printed drug-delivery systems for improved
patient treatment. Trends Pharmacol Sci. 2016;37:1070–1080.
Papers of special note have been highlighted as either of interest (•) or of •• Review on printing of personalized drug products.
considerable interest (••) to readers. 28. Preis M, Öblom H. 3D-printed drugs for children — are we ready
1. Berman B. 3-D printing: the new industrial revolution. Bus Horiz. yet? AAPS PharmSciTech. 2017;18:303–308.
2012;55:155–162. 29. Hamburg MA, Collins FS. The path to personalized medicine. N
2. 3D Printing Industry [Internet]. 2017 [cited 2017 Apr 10]. Available Engl J Med. 2010;363:301–304.
from: https://3dprintingindustry.com/ 30. Khaled SA, Burley JC, Alexander MR, et al. 3D printing of five-in-one
3. Aprecia Pharmaceuticals [Internet]. 2017 [cited 2017 Apr 10]. dose combination polypill with defined immediate and sustained
Available from: www.aprecia.com. release profiles. J Control Release. 2015;217:308–314.
4. Spritam [Internet]. 2017 [cited 2017 Apr 12]. Available from: www. 31. Sanderson K. 3D printing: the future of manufacturing. Pharm J.
spritam.com. 2015;294:7865.
•• The first commercially available 3D printed drug product. 32. Gudapati H, Dey M, Ozbolat I. A comprehensive review on droplet-
5. Ventola CL. Medical applications for 3D printing: current and pro- based bioprinting: past, present and future. Biomaterials.
jected uses. P T. 2014;39:704–711. 2016;102:20–42.
6. Hull CW Apparatus for Production of Three-Dimensional Objects by 33. Organovo [Internet]. 2017 [cited 2017 Apr 10]. Available from:
Stereolithography.United States patent US 4,575,330. 1986. http://organovo.com/.
7. Crump SS Apparatus and Method for Creating Three-Dimensional 34. Daly R, Harrington TS, Martin GD, et al. Inkjet printing for pharma-
Objects.United States patent US 5,121,329. 1992. ceutics – a review of research and manufacturing. Int J Pharm.
8. Sachs EM, Haggerty JS, Cima MJ, et al. Three-Dimensional Printing 2015;494:554–567.
Techniques.United States patent US 5,204,055. 1993. p. 1–14. 35. Genina N, Fors D, Vakili H, et al. Tailoring controlled-release oral
9. Holländer J. 3D printed UV light cured polydimethylsiloxane scaf- dosage forms by combining inkjet and flexographic printing tech-
folds for drug delivery. Helsinki, Finland: University of Helsinki; niques. Eur J Pharm Sci. 2012;47:615–623.
2017. 36. Kolakovic R, Viitala T, Ihalainen P, et al. Printing technologies in
•• The Master’s thesis that the review is based on. fabrication of drug delivery systems. Expert Opin Drug Deliv.
10. Melchels FPW, Feijen J, Grijpma DW. A review on stereolithography 2013;10:1711–1723.
and its applications in biomedical engineering. Biomaterials. 37. Alomari M, Mohamed FH, Basit AW, et al. Personalised dosing: printing a
2010;31:6121–6130. dose of one’s own medicine. Int J Pharm. 2015;494:568–577.
11. Chia HN, Wu BM. Recent advances in 3D printing of biomaterials. J • Review on 2D printing of personalized drug products.
Biol Eng. 2015;9:4. 38. Preis M, Breitkreutz J, Sandler N. Perspective: concepts of printing
12. Skoog SA, Goering PL, Narayan RJ. Stereolithography in tissue technologies for oral film formulations. Int J Pharm. 2015;494:578–584.
engineering. J Mater Sci Mater Med. 2014;25:845–856. 39. Elomaa L, Teixeira S, Hakala R, et al. Preparation of poly(ε-
13. Low KH, Leong KF, Chua CK, et al. Characterization of SLS parts for Caprolactone)-based tissue engineering scaffolds by stereolithogra-
drug delivery devices. Rapid Prototyp J. 2001;7:262–268. phy. Acta Biomater. 2011;7:3850–3856.
14. Mazzoli A. Selective laser sintering in biomedical engineering. Med 40. Gauvin R, Chen Y-C, Lee JW, et al. Microfabrication of complex
Biol Eng Comput. 2013;51:245–256. porous tissue engineering scaffolds using 3D projection stereo-
15. Katstra WE, Palazzolo RD, Rowe CW, et al. Oral dosage forms fabricated lithography. Biomaterials. 2012;33:3824–3834.
by three dimensional printingTM. J Control Release. 2000;66:1–9. 41. Lu Y, Chen S. Projection printing of 3-dimensional tissue scaffolds.
16. Rowe CW, Katstra WE, Palazzolo RD, et al. Multimechanism oral dosage In: Liebschner MAK, editor. Computer Aided Tissue Engineering.
forms fabricated by three dimensional printingTM. J Control Release. New York, NY: Springer Science+Business Media; 2012. p. 289–302.
2000;66:11–17. 42. Pereira RF, Bártolo PJ. 3D photo-fabrication for tissue engineering
17. Yu DG, Branford-White C, Ma ZH, et al. Novel drug delivery devices and drug delivery. Engineering. 2015;1:90–112.
for providing linear release profiles fabricated by 3DP. Int J Pharm. 43. Popov VK, Evseev AV, Ivanov AL, et al. Laser stereolithography
2009;370:160–166. and supercritical fluid processing for custom-designed implant
18. Wu G, Wu W, Zheng Q, et al. Experimental study of PLLA/INH slow fabrication. J Mater Sci Mater Med. 2004;15:123–128.
release implant fabricated by three dimensional printing technique 44. Lal K, White GS, Morea DN, et al. Use of stereolithographic
and drug release characteristics in vitro. Biomed Eng Online. templates for surgical and prosthodontic implant planning
2014;13:97. and placement. Part I. The Concept. J Prosthodont.
19. Yu DG, Yang XL, Huang WD, et al. Tablets with material gradients 2006;15:51–58.
fabricated by three-dimensional printing. J Pharm Sci. 2007;96:2446– 45. Lal K, White GS, Morea DN, et al. Use of stereolithographic tem-
2456. plates for surgical and prosthodontic implant planning and place-
20. Stratasys [Internet]. 2017 [cited 2017 Apr 19]. Available from: http:// ment. Part II. A clinical report. J Prosthodont. 2006;15:117–122.
www.stratasys.com. 46. Nayar S, Bhuminathan S, Bhat WM. Rapid prototyping and stereo-
21. Turner BN, Strong R, Gold SA. A review of melt extrusion additive lithography in dentistry. J Pharm Bioallied Sci. 2015;7:218–219.
manufacturing processes: I. Process design and modeling. Rapid 47. Parthasarathy J. 3D modeling, custom implants and its future per-
Prototyp J. 2014;20:192–204. spectives in craniofacial surgery. Ann Maxillofac Surg. 2014;4:9–18.
22. Goole J, Amighi K. 3D printing in pharmaceutics: a new tool for design- 48. Formlabs [Internet]. 2017 [cited 2017 Apr 10]. Available from:
ing customized drug delivery systems. Int J Pharm. 2016;499:376–394. https://formlabs.com/.
•• Review on technological aspects of pharmaceutical 3D printing. 49. Tumbleston JR, Shirvanyants D, Ermoshkin N, et al. Continuous
23. Moulton SE, Wallace GG. 3-dimensional (3D) fabricated polymer liquid interface production of 3D objects. Science (80-.).
based drug delivery systems. J Control Release. 2014;193:27–34. 2015;347:1349–1352.
EXPERT REVIEW OF MEDICAL DEVICES 695

50. Carbon [Internet]. 2017 [cited 2017 Apr 11]. Available from: www. 73. Yu D-G, Branford-White C, Yang Y-C, et al. A novel fast disintegrat-
carbon3D.com. ing tablet fabricated by three-dimensional printing. Drug Dev Ind
51. Vehse M, Petersen S, Sternberg K, et al. Drug delivery from poly Pharm. 2009;35:1530–1536.
(ethylene glycol) diacrylate scaffolds produced by DLC based 74. Pryce Lewis WE, Rowe CW, Cima MJ, et al. System for Manufacturing
micro-stereolithography. Macromol Symp. 2014;346:43–47. Controlled Release Dosage Forms, Such as Zero-Order Release Profile
52. Wang J, Goyanes A, Gaisford S, et al. Stereolithographic (SLA) Dosage Form Manufactured by Three-Dimensional Printing.United
3D printing of oral modified-release dosage forms. Int J Pharm. States patent US 7,820,201 [Internet]. 2010. p. 1–54. Available from:
2016;503:207–212. https://www.google.ch/patents/US7820201.
53. Goyanes A, Det-Amornrat U, Wang J, et al. 3D scanning and 3D 75. Jacob J, Coyle N, West TG, et al. Rapid Disperse Dosage Form
printing as innovative technologies for fabricating personalized Containing Levetiracetam.United States patent US 9,339,489.
topical drug delivery systems. J Control Release. 2016;234:41–48. Google Patents; 2016.
54. Gittard SD, Ovsianikov A, Akar H, et al. Two photon polymerization- 76. Pietrzak K, Isreb A, Alhnan MA. A flexible-dose dispenser for
micromolding of polyethylene glycol-gentamicin sulfate microneedles. immediate and extended release 3D printed tablets. Eur J Pharm
Adv Eng Mater. 2010;12:77–82. Biopharm. 2015;96:380–387.
55. Gittard SD, Miller PR, Jin C, et al. Deposition of antimicrobial coatings on 77. Goyanes A, Robles Martinez P, Buanz A, et al. Effect of geome-
microstereolithography-fabricated microneedles. Jom. 2011;63:59–68. try on drug release from 3D printed tablets. Int J Pharm.
56. Ali Z, Türeyen EB, Karpat Y, et al. Fabrication of polymer micro 2015;494:657–663.
needles for transdermal drug delivery system using DLP based 78. Holländer J, Genina N, Jukarainen H, et al. Three-dimensional
projection stereo-lithography. Procedia CIRP. 2016;42:87–90. printed PCL-based implantable prototypes of medical devices for
57. Cheah CM, Leong KF, Chua CK, et al. Characterization of microfea- controlled drug delivery. J Pharm Sci. 2016;105:2665–2676.
tures in selective laser sintered drug delivery devices. Proc Inst 79. Genina N, Holländer J, Jukarainen H, et al. Ethylene Vinyl Acetate
Mech Eng H. 2002;216:369–383. (EVA) as a new drug carrier for 3d printed medical drug delivery
58. Leong KF, Chua CK, Gui WS, et al. Building porous biopolymeric devices. Eur J Pharm Sci. 2016;90:53–63.
microstructures for controlled drug delivery devices using selective 80. Goyanes A, Buanz ABM, Basit AW, et al. Fused-FILAMENT 3D print-
laser sintering. Int J Adv Manuf Technol. 2006;31:483–489. ing (3DP) for fabrication of tablets. Int J Pharm. 2014;476:88–92.
59. Leong KF, Wiria FE, Chua CK, et al. Characterization of a poly-ε- 81. Goyanes A, Buanz ABM, Hatton GB, et al. 3D printing of modified-
caprolactone polymeric drug delivery device built by selective release aminosalicylate (4-ASA and 5-ASA) tablets. Eur J Pharm
laser sintering. Biomed Mater Eng. 2007;17:147–157. Biopharm. 2015;89:157–162.
60. Salmoria GV, Klauss P, Zepon K, et al. Development of functionally- 82. Goyanes A, Wang J, Buanz A, et al. 3D printing of medicines:
graded reservoir of PCL/PG by selective laser sintering for drug engineering novel oral devices with unique design and drug
delivery devices. Virtual Phys Prototyp. 2012;7:107–115. release characteristics. Mol Pharm. 2015;12:4077–4084.
61. Wu BM, Borland SW, Giordano RA, et al. Solid free-form fabrication 83. Goyanes A, Chang H, Sedough D, et al. Fabrication of controlled-
of drug delivery devices. J Control Release. 1996;40:77–87. release budesonide tablets via desktop (FDM) 3D printing. Int J
62. Lin S, Chao PY, Chien YW, et al. In vitro and in vivo evaluations of Pharm. 2015;496:414–420.
biodegradable implants for hormone replacement therapy: effect of 84. Alhijjaj M, Belton P, Qi S. An investigation into the use of polymer blends
system design and PK-PD relationship. AAPS PharmSciTech. 2001;2:16. to improve the printability of and regulate drug release from pharma-
63. Gbureck U, Vorndran E, Müller FA, et al. Low temperature direct 3D ceutical solid dispersions prepared via Fused Deposition Modeling
printed bioceramics and biocomposites as drug release matrices. J (FDM) 3D printing. Eur J Pharm Biopharm. 2016;108:111–125.
Control Release. 2007;122:173–180. 85. Melocchi A, Parietti F, Maroni A, et al. Hot-melt extruded filaments
64. Huang W, Zheng Q, Sun W, et al. Levofloxacin implants with based on pharmaceutical grade polymers for 3D printing by fused
predefined microstructure fabricated by three-dimensional printing deposition modeling. Int J Pharm. 2016;509:255–263.
technique. Int J Pharm. 2007;339:33–38. 86. Okwuosa TC, Stefaniak D, Arafat B, et al. A lower temperature FDM
65. Gbureck U, Vorndran E, Barralet JE. Modeling vancomycin 3D printing for the manufacture of patient-specific immediate
release kinetics from microporous calcium phosphate ceramics release tablets. Pharm Res. 2016;33:2704–2712.
comparing static and dynamic immersion conditions. Acta 87. Sadia M, Sośnicka A, Arafat B, et al. Adaptation of pharmaceutical
Biomater. 2008;4:1480–1486. excipients to FDM 3D printing for the fabrication of patient-tailored
66. Wu W, Zheng Q, Guo X, et al. The controlled-releasing drug implant immediate release tablets. Int J Pharm. 2016;513:659–668.
based on the three dimensional printing technology: fabrication 88. Markl D, Zeitler JA, Rasch C, et al. Analysis of 3D prints by X-ray
and properties of drug releasing in vivo. J Wuhan Univ Technol computed microtomography and terahertz pulsed imaging. Pharm
Mater Sci Ed. 2009;24:977–981. Res. 2017;34:1037–1052.
67. Inzana JA, Trombetta RP, Schwarz EM, et al. 3D printed bioceramics for 89. Melocchi A, Parietti F, Loreti G, et al. 3D printing by Fused Deposition
dual antibiotic delivery to treat implant-associated bone infection. Eur Modeling (FDM) of a swellable/erodible capsular device for oral pul-
Cells Mater. 2015;30:232–247. satile release of drugs. J Drug Deliv Sci Technol. 2015;30:360–367.
68. Wu W, Ye C, Zheng Q, et al. A therapeutic delivery system for chronic 90. Lim SH, Chia SMY, Kang L, et al. Three-dimensional printing of
osteomyelitis via a multi-drug implant based on three-dimensional carbamazepine sustained-release scaffold. J Pharm Sci.
printing technology. J Biomater Appl. 2016;31:250–260. 2016;105:2155–2163.
69. Wu W, Zheng Q, Guo X, et al. A programmed release multi-drug 91. Sandler N, Salmela I, Fallarero A, et al. Towards fabrication of 3D
implant fabricated by three-dimensional printing technology for printed medical devices to prevent biofilm formation. Int J Pharm.
bone tuberculosis therapy. Biomed Mater. 2009;4:65005. 2014;459:62–64.
70. Yu D-G, Shen -X-X, Branford-White C, et al. Novel oral fast- 92. Water JJ, Bohr A, Boetker J, et al. Three-dimensional pprinting of drug-
disintegrating drug delivery devices with predefined inner eluting implants: preparation of an antimicrobial polylactide feedstock
structure fabricated by three-dimensional printing. J Pharm material. J Pharm Sci. 2015;104:1099–1107.
Pharmacol. 2009;61:323–329. 93. Boetker J, Water JJ, Aho J, et al. Modifying release characteristics
71. Wang -C-C, Tejwani MR, Roach WJ, et al. Development of near zero- from 3D printed drug-eluting products. Eur J Pharm Sci.
order release dosage forms using three-dimensional printing 2016;90:47–52.
(3-DPTM) technology. Drug Dev Ind Pharm. 2006;32:367–376. 94. Weisman JA, Nicholson JC, Tappa K, et al. Antibiotic and che-
72. Lee K-J, Kang A, Delfino JJ, et al. Evaluation of critical formulation motherapeutic enhanced three-dimensional printer filaments and
factors in the development of a rapidly dispersing captopril oral constructs for biomedical applications. Int J Nanomedicine.
dosage form. Drug Dev Ind Pharm. 2003;29:967–979. 2015;10:357–370.
696 M. PALO ET AL.

95. Rattanakit P, Moulton SE, Santiago KS, et al. Extrusion printed polymer 102. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat
structures: a facile and versatile approach to tailored drug delivery Biotechnol. 2014;32:773–785.
platforms. Int J Pharm. 2012;422:254–263. 103. Bandyopadhyay A, Bose S, Das S. 3D Printing of Biomaterials.
96. Khaled SA, Burley JC, Alexander MR, et al. Desktop 3D printing of MRS Bull. 2015;40:108–114.
controlled release pharmaceutical bilayer tablets. Int J Pharm. 104. Di Prima M, Coburn J, Hwang D, et al. Additively manufactured
2014;461:105–111. medical products – the FDA perspective. 3D Print Med.
97. Khaled SA, Burley JC, Alexander MR, et al. 3D printing of tablets contain- 2015;2:1–6.
ing multiple drugs with defined release profiles. Int J Pharm. 105. Technical Considerations for Additive Manufactured Devices.
2015;494:643–650. Draft guidance for industry and food and drug administration
98. Song T-H, Jang J, Choi Y-J, et al. 3D-printed drug/cell carrier staff [Internet]. [cited 2017 June 6]. 2016. Available from: http://
enabling effective release of cyclosporin a for xenogeneic cell- www.fda.gov/downloads/MedicalDevices/
based therapy. Cell Transplant. 2015;24:2513–2525. DeviceRegulationandGuidance/GuidanceDocuments/
99. Zhu M, Li K, Zhu Y, et al. 3D-printed hierarchical scaffold for localized UCM499809.pdf.
Isoniazid/Rifampin drug delivery and osteoarticular tuberculosis ther- •• Draft guidance for 3D printing of medical devices.
apy. Acta Biomater. 2015;16:145–155. 106. Beaver N, Koch G, Konski A, et al. Personalized medicine: a new para-
100. Martínez-Vázquez FJ, Cabañas MV, Paris JL, et al. Fabrication of digm. Assoc Corp Couns. 2016. doi: 10.2174/
Novel Si-Doped Hydroxyapatite/Gelatine Scaffolds by rapid proto- 1389201018666170224105600
typing for drug delivery and bone regeneration. Acta Biomater. 107. Anaya J-M, Duarte-Rey C, Sarmiento-Monroy JC, et al.
2015;15:200–209. Personalized medicine. Closing the gap between knowledge
101. Yi H, Choi Y, Shin K, et al. A 3D-printed local drug delivery patch for and clinical practice. Autoimmun Rev. 2016;15:833–842.
pancreatic cancer growth suppression. J Control Release. 108. Directive 2001/83/EC. Brussels: The European Parliament and of the
2016;238:231–241. Council of European Union; 2001.

You might also like