You are on page 1of 10

Journal of Drug Delivery Science and Technology 59 (2020) 101923

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


journal homepage: www.elsevier.com/locate/jddst

Review article

A review of recent advances in microneedle technology for transdermal


drug delivery
Rigved Nagarkar a, *, Mahima Singh a, Hiep X. Nguyen b, Sriramakamal Jonnalagadda a
a
Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, 600 S 43rd Street, Philadelphia, PA, 19104, USA
b
Aurobindo Pharma USA Inc, 2929 Weck Dr, Durham, NC, 27709, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Transdermal devices are used to deliver drugs through the skin. However, limitations such as molecular weight
Microelectromechanical systems and hydrophilicity of the molecules prevent its wider use. Microneedle technology offers an enhanced drug
Biocompatibility delivery option. Depending on the delivery, different types of microneedles such as solid, hollow, dissolving and
Transdermal
coated are discussed. While solid microneedles create micropores in the skin, hollow microneedles provide a
Microneedles
channel into the dermis. Dissolving microneedles have been explored for vaccine delivery while coated micro­
Micromolding
Non-degradable needles use a drug dispersion to effectively load drugs. This review also focuses on the techniques involved in
fabrication of microneedles. Several methods using various solvent techniques, electromechanical systems, laser
ablation and additive manufacturing are discussed. The nature of material used impacts the method used for
fabrication. Various degradable and non-degradable materials used for fabrication are discussed. Microneedles
can be made from metals, silicone, ceramics, synthetic as well as biodegradable polymers like carbohydrates.
Each material has its own advantages and disadvantages. Stainless steel demonstrates good biocompatibility but
are highly corrosive while silicones are easy to fabricate but brittle. Microneedle technology possesses a
tremendous potential considering the variations in types and materials it offers with ease of fabrication and are
the next generation therapeutics.

1. Introduction address the problems associated with transdermal delivery. However,


intradermal injections are associated with limitations such as needle
The transdermal route of drug delivery offers an attractive, non- injuries, phobia, requirements of specially trained staff increasing the
invasive route of drug administration. Drugs administered through the cost of the delivery.
transdermal route show high bioavailability avoiding gastrointestinal Microneedle drug delivery is the answer to the limitations provided
degradation and first-pass metabolism [1]. During transdermal perme­ by the two traditional dosage forms. The technology has been evaluated
ation, drug accumulation in the skin prior to release into blood vessels to deliver not only small molecules [20] but also various macromole­
enables a natural mechanism for sustained drug delivery [2]. Despite cules [21,22], cosmeceuticals [23], and micro/nano-particles [24].
numerous advantages, transdermal drug delivery is limited to a few Recently, the application of coated as well as dissolving microneedles
molecules with certain physicochemical properties. Ideally, a trans­ has been evaluated for noninvasive transdermal vaccination [16], pa­
dermal candidate should have molecular weight less than 500 Da and tient monitoring, and diagnostic applications [25]. Research and
log P within 2–3 [3]. The stratum corneum is the main barrier for development of microneedles have reached the stage of large-scale
transdermal permeation [4]. Several technologies have therefore been manufacturing and commercialization. Microneedles have not only
developed to disrupt the stratum corneum to enhance skin permeability. been researched for transdermal drug delivery but also find their use in
These include iontophoresis [5], sonophoresis [6], magnetophoresis [7], ocular, diagnostic testing and oral delivery. This review focuses on
electroporation [8], and laser-microporation [9]. These methods have microneedling which holds significant promise for bypassing stratum
major applicability and economic limitations. Traditional drug delivery corneum to enhance transdermal drug delivery [26].
systems such as intradermal injections are currently being used to Microneedles (micron-sized needling system) are intended to pierce

* Corresponding author.
E-mail addresses: rnagarkar@mail.usciences.edu (R. Nagarkar), msingh@mail.usciences.edu (M. Singh), Bnguyen@aurobindousa.com (H.X. Nguyen), s.jonnal@
usciences.edu (S. Jonnalagadda).

https://doi.org/10.1016/j.jddst.2020.101923
Received 12 May 2020; Received in revised form 22 June 2020; Accepted 8 July 2020
Available online 16 July 2020
1773-2247/© 2020 Elsevier B.V. All rights reserved.
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

the epidermal layer, creating micro-channels in the skin without causing solutions within a patch then are applied on these microprojections
pain, bleeding, or infection [27]. These channels allow therapeutic [33]. Another variation is the use of a roller containing solid micro­
agents to diffuse into the dermal layer, which is well perfused with blood needles, which pierces the stratum corneum multiple times as the roller
vessels [28]. The length of microneedles can vary to ensure epidermal spins on the skin. Commercially available Derma-rollers are based on
penetration, while avoiding stimulation of the nerve fibers or puncturing this concept and are used for skin pore opening therapies. Ita et al.
blood capillaries [29]. Microneedle delivery does not face limitations (2015) conducted a study to deliver drugs for high blood pressure using
based on the molecular size, since the channels are markedly larger than cylindrical surface Microneedle Systems®. Transdermal flux values were
the typical therapeutic agents. Microneedles (MNs) have been fabricated shown to increase 5 to 8 times after treatment of porcine skin with
by various microfabrication techniques [30], to represent varied mate­ microneedle rollers [34].
rials, geometries, and dimensions (50–900 μm height, 2000 mm 2 sur­
face area). A comparison of microneedles with the conventional 2.1.2. Hollow microneedles
injectable forms of drug delivery is shown in Fig. 1. These microneedle systems are a miniature version of the conven­
Regulatory issues remain a concern, as is the case for any new tional hypodermic needles. A microscopic representation of hollow
technology. This paper reviews a range of critical aspects related to microneedles is shown in Fig. 3 [13]. Drug delivery is achieved through
microneedle technology, including types of microneedle, materials of a pressure-driven flow of liquid formulation [14]. Substantial doses can
construction, geometries, penetration depth, drug release, and irritation be delivered into the dermal layer through this technique [15]. Hollow
possibility. MNs are difficult to fabricate due to their structure and fragility. In one
such study, Davis et al. (2005) fabricated an array of hollow metal
2. Microneedle classification microneedles. These microneedles were studied for the delivery of in­
sulin in diabetic rats [35]. The study concluded that
Microneedles may be broadly classified on the basis of delivery hollow-microneedles display pharmacodynamic profiles that are com­
profile, or the material used in manufacture. parable to a traditional hypodermic injection.

2.1.3. Dissolving microneedles


2.1. Based on variations in drug delivery
Dissolving microneedles operate by a ‘poke and release’ principle.
They are easy to manufacture, and use compared to other microneedles,
Microneedles may be classified as solid, hollow, dissolving, or coated
and have therefore gained tremendous attention in recent years [16].
microneedles (Fig. 2).
Dissolving MNs are made with biodegradable materials such as sugars or
biodegradable polymers and can be loaded with therapeutic agents [17].
2.1.1. Solid microneedles
Once applied, the MNs dissolve into the skin thereby releasing the
Solid microneedles can be used as a skin pretreatment, as they are
conjugated therapeutic agent. These MNs provide an advantage over the
inserted and removed to form micron-scaled pores on the skin surface.
traditional solid and hollow microneedles due to their ease of fabrication
They work on a ‘poke and patch’ principle, as microchannels are
and convenient single-step application principle. Liu et al. fabricated
created. These microchannels enhance the drug permeability by allow­
microarrays consisting of dissolving hyaluronic acid microneedles [36].
ing diffusion from a formulation directly into the dermal layer. Studies
The cumulative permeated amount of dextran increased by a staggering
on rat skin have shown that the micropores generated by microneedles
610 times when applied as a microneedle array, with negligible lag time
remained upon at least 72 h after microneedle treatment, when held
in drug permeation. Dissolving MN’s are being explored for use in de­
under occlusive conditions [31], such as occlusive tape [32]. In the
livery of vaccine in the skin. Carboxymethylcellulose and hyaluronan
absence of occlusion, the micropores closed shortly after microneedle
microneedles were shown to dissolve completely in rat skin within 5 min
application. The microchannels created by solid MN were observed to
of application releasing the antigen-specific IgG payload (Fig. 4) [37].
heal rapidly within 2 h ensuring the absence of secondary infection [31].
A variation of a typical solid microneedle approach is the ‘scrape and
2.1.4. Coated microneedles
patch’ method, whereby microneedles, microprojections, or micro­
Microneedles that work on ‘coat and poke’ principle are coated
blades are scraped over the skin to generate micro-abrasions. Drug

Fig. 1. Comparison of microneedles with traditional injectable form of delivery.

2
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

Fig. 2. Types of microneedles


A. Dissolving microneedles
B. Solid microneedles
C. Coated microneedles
D. Hollow microneedles.

Fig. 3. A microscopic representation of hollow microneedles. This work is licensed under the Creative Commons Attribution 4.0 International License, which permits
unrestricted use, distribution, modification, and reproduction in any medium, provided you. To view a copy of the Creative Commons license, please visit http://cre
ativecommons.org/licenses/by/4.0/.

3
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

Table 1
A detailed list of advantages, disadvantages, and method of delivery of various
MNs.
MN Advantages Disadvantages Method of Reference
classification drug
delivery

Solid Can be made Microneedle Create Li, J et al.


from a range of fracture under micro [10]
materials skin conduits in Ullah, A
Limited surface skin to et al. [11,
area available for which 12]
drug absorption drug is
applied
Hollow High drug load Must be fabricated Pressure Kim, Y
can be injected with strong driven et al. [13]
materials to flow He, X
withstand flow through et al. [14,
pressure needle 15]
Dissolving Easy Only Dissolve Ita, K et al.
manufacturing biodegradable under skin [16]
materials can be to release Kolli, C.S.
used drug et al. [16,
payload 17]
Coated Used for potent Associated with Coating Chen, X
drugs requiring drug loss while drug et al. [18]
low doses manufacturing, release Chen, J
temperature et al. [18,
limitations 19]

in a polyethylene glycol matrix which showed a significantly higher


delivery compared to a commercial product [10]. Coated MNs are less
commonly used compared to other MNs, as they offer a relatively small
surface area for drug absorption.
The method of delivery and materials used will directly impact the
applicability of MNs. Solid MNs are easy to fabricate and use. Depending
on the nature of the material, solid MN potentially can be re-used. For
Fig. 4. Dissolution of carboxymethylcellulose and hyaluronan microneedles in instance, the use of Derma rollers multiple times for pore formation.
rat skin. Coated MNs consist of a coat of drug on solid MN. They are difficult to
This work is licensed under the Creative Commons Attribution 4.0 International fabricate given that the coating should be consistent throughout the MN
License, which permits unrestricted use, distribution, modification, and repro­
surface area. Due to a small surface area, low doses can be delivered with
duction in any medium, provided you. To view a copy of the Creative Commons
this MN type. On the other hand, with hollow MN significant drug
license, please visit http://creativecommons.org/licenses/by/4.0/.
amounts can be injected into the dermis. However, injecting solutions
can be challenging due to the pressure resistance by the opening size of
microneedles. These MNs consists a base of solid microneedles which
the MN and thick tissue present in the dermis. Dissolving MNs are easy to
have a coat with drug solutions/dispersions. Several methods have been
fabricate amongst all other types of MN. They are cheap and can be
studied to coat microneedles [18,19,38]. The most common method is
fabricated by various materials. They are single use MNs which dissolve
dip coating however, it is complicated due to the need for precise control
readily under the skin. Dissolving MNs have been researched signifi­
to assure that the MNs are inserted accurately into the dipping solution
cantly for their use in controlled delivery, vaccine delivery and these
[38]. Fig. 5 shows steps involved in dip-coating technique. Spray coating
MNs could potentially be the future of immunization (Table 1).
is an alternative approach for drug deposition on the microneedles; this
method cannot avoid drug loss through spraying on the substrate of the
2.2. Based on material selection
microneedles array, which is unavailable for drug permeation [18].
Aerosol coating methods have improved efficiency; however the need
MNs may be fabricated from metals, silicone, ceramic, or polymers.
for high temperatures of up to 200 � C may limit the use of thermolabile
In the context of this review, the material of construction has been
drugs [19]. Ma et al. (2018) coated solid MN with suspended lidocaine
classified into two groups, namely non-degradable and degradable. Non-

Fig. 5. Microneedles coated with dip coating technique.

4
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

degradable materials include materials such as metals, silicone and ce­ 2.2.1.3. Ceramic. Alumina (Al2O3) is one of the most common type of
ramics, while degradable consists of polysaccharides and biodegradable material used to produce MN [51]. Alumina holds a defined volume of
polymers. active due to the porosity of alumina [52]. Gypsum and Brushite have
also been used to fabricate MNs. A novel material which is made of
2.2.1. Non degradable materials organically modified silicon alkoxides and organic monomers called
Ormocer® has been used to fabricate MNs. It forms a 3D network
2.2.1.1. Metals. Soluvia® is one of the first marketed MN-based prod­ increasing the area for absorption of drug solutions [53]. In a study
uct made from metal. Another product to reach market was a silicone carried out by Ovsianikov et al. (2007) Ormocer® MN were fabricated
based product Micronjet® [39]. Stainless steel and titanium are the most using laser two-photon polymerization at different aspect ratios. The
commonly used metals to fabricate MNs. Metal MNs may be fabricated fabricated MNs penetrated porcine adipose tissue without cracking the
as solid, hollow, or coated microneedles. Fabrication techniques include MNs [54]. Boks et al. fabricated ceramic nanoporous microneedle arrays
three-dimensional laser ablation, laser cutting, wet etching, or metal (npMNA) for delivery of vaccines. npMNA enhanced vaccine efficacy by
electroplating [14]. reaching skin dendritic cells consisting of T and B cell immunity [41].
Li et al. used modified metal injection molding technology to fabri­
cate titanium porous microneedle arrays (TPMA). TPMA exhibited a 2.2.1.4. Synthetic polymers. Several synthetic polymers have been
staggering more than 27 times higher flux of calcein across intact skin researched for use in MNs such as polyvinylpyrrolidone (PVP), and
[11]. Delivery of calcein dye was evaluated by Assad Ullah et al. (2018), polyvinyl alcohol (PVA). Poly (methyl methacrylate) (PMMA) is a
where stainless steel microneedles were modified by surface plasma biocompatible polymer used for MN arrays [55]. In a study performed
treatment. These porous polymer-coated MNs were used to deliver by Nguyen et al. microneedles were fabricated using PVA by micro
lidocaine and rhodamine B dye [12]. An example of a commercially molding technique for delivery of doxorubicin [42].
available hollow stainless steel microneedles are the AdminPen®, which
vary in length from 600 to 1500 μm [40]. Derma rollers made from 2.2.2. Degradable materials
stainless steel or titanium are commercially used for cosmetic purposes.
2.2.2.1. Natural. Carbohydrates are good alternatives as materials for
2.2.1.2. Silicone. Silicone is among the most commonly used material fabrication of microneedles. One of the most important attribute of
for microneedle devices, and has been explored for over two decades carbohydrates is that they are cheap and safe [56]. They have not only
[48]. Silicone MN arrays could be used as primary molds in micro­ an excellent biocompatibility, but also show low toxicity. Some carbo­
molding (Fig. 6). The use of silicone allows to fabricate MNs of different hydrates are known to have significant strength/toughness. All of these
geometry. Narayanan et al. fabricated silicone microneedles using an materials are available at low cost and are easily biodegradable [57].
etching technique [49]. Etching factors such as the use of Tetramethy­ Carbohydrates can be easily molded into MNs using master templates
lammonium Hydroxide (TMAH) in varying concentrations, time and [58]. Carbohydrates can be formulated with active ingredients to form
rate of etching, and temperature were used to produce sharper micro­ an active-carbohydrate mixture. This mixture then can be casted into the
needles with a higher aspect ratio. molds which dissolve upon insertion into the skin to release their drug
Krieger et al. have shown that high aspect ratio microneedle silicone payload [59]. One of the most common sugars used to prepare MN ar­
master molds may also be printed using a 3D printer [50]. rays is Maltose [17]. Maltose is a very common excipient in
FDA-approved parenteral formulations. Gouhua et al. investigated
maltose microneedle-mediated in vitro transdermal delivery of a
monoclonal antibody using human IgG as a model protein. The experi­
ment showed that methylene blue was taken up by microchannels
created by maltose MNs. Cryosections microneedles penetrated the
dermis layer. Human IgG showed an increase in the delivery with in­
crease in different MN geometry like number of arrays and length [60].
In addition to Maltose, various sugars like sucrose, trehalose, mannitol,
and galactose have been used in MN fabrication [61]. The use of ma­
terials such as starch and gelation have also been explored for MN
preparation. Starch-gelatin microneedles completely dissolved after
insertion into the skin within 5 min. The microneedle could easily be
inserted into porcine skin. The depth of penetration was around 200 μm
in porcine skin, which was similar in depth to in-vivo rat skin (200–250
μm). MN mediated hypoglycemic effect in rats was similar when
compared to subcutaneous injection [44].

2.2.2.2. Synthetic. A plethora of synthetic macromolecular materials


have been used for fabrication of MNs. These include polylactic acid
(PLA), polyglycolic acid (PGA), polylactic-co-glycolic acid (PLGA) [45,
47]. For these types of materials, MNs govern the drug release which in
turn depends upon the composition of the MN materials [46,62].

2.2.3. Hydrogel forming microneedles


Hydrogel forming microneedles can be called as in-situ microneedles
where a polymer is crosslinked in dry hydrogel state. When injected into
the skin, they absorb water from interstitial fluid causing MNs to swell.
The swollen hydrogel network produces conduits which act as drug
reservoirs and control the drug delivery. It also maintains the integrity of
the MNs in the skin. These MNs are mostly fabricated by molding process
Fig. 6. Silicone microneedle array template.

5
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

which also includes a crosslinking step [63]. Many different materials aqueous solutions could successfully cover the entire mold surface
have been studied for hydrogel forming MNs. The most common being without generation of voids. The authors fabricated dissolving MNs (5%
polysaccharides [64]. Other polymers like Poly(ethylene glycol) dia­ solids content) with atomized spraying into silicon micromolds [43].
crylate (PEGDA) and PVA [63], polyacrylic acid-co-maleic acid (PAMA)
[65], methylvinylether-co-maleic acid [66] have been studied for 3.1.3. Droplet-born air blowing (DAB)
hydrogel forming MNs. The main advantage of hydrogel forming In this method, polymer droplet is given the shape of a microneedle
microneedle is delivery of high doses of drugs due to swell ability of the with the use of air blowing, allowing for gentle conditions without the
MN base (Table 2). use of external harsh conditions such as heat and UV irradiation [70].
Several types of materials have been used for fabrication of MNs. The process starts with dispensing of solution on two different plates,
Depending on the material, MNs can be fabricated with non-degradable upper and lower. The two plates are kept in contact and gradually pulled
or degradable materials. While the choice of materials from degradable apart thus elongating the viscous solution. The elongated viscous solu­
or non-degradable material is vast, one needs to consider supporting tion is then exposed to blowing air under controlled conditions. The
aspects that can impact the material of construction. Factors like method required shape is given during this drying step. The fabrication method
of fabrication available, drug and excipients of choice, drug compati­ was proposed by Kim et al. A single polymer drop per MN is used to
bility with material and fabrication process, final use, type of delivery control the dimension and concentration of the droplet as well as the
have an influence of the selection of material. controlled drug loading without the drug loss. This process has been
used in the fabrication of insulin loaded dissolving microneedles by the
3. Fabrication techniques authors where a successful use of insulin loaded dissolving microneedles
reduced blood glucose levels in diabetic mice [71].
Skin is an elastic organ which possesses significant tensile strength.
In order for MNs to effectively penetrate, they must have optimum 3.1.4. Pulling pipettes
strength to penetrate without cracking or breaking during insertion. Pulling pipettes technique is only applicable for fabrication of hollow
Hence, fabricating microneedles is a challenging task. The fabrication glass MNs. Glass is exposed to high temperatures, after which it MNs
methodology varies based on material of choice, MN geometry, and skin arrays are created by pulling the hot glass with micropipette puller. This
depth requirement [67]. For instance, solvent-casting may be appro­ technique can be made more robust and consistent by the use of pro­
priate for polymeric microneedles, whereas microelectromechanical grammable puller [72].
systems are better utilized to design metal microneedles. The fabrication
techniques have been classified based on the technology used for
manufacture. 3.2. Electromagnetic/microsystem/laser technology

3.2.1. Microelectromechanical systems (MEMS)


3.1. Fabrication of polymeric MN using solvent MEMS has been traditionally used for creation of integrated circuit
technology. MEMS is divided into following main processes of (1)
3.1.1. Micromolding deposition, (2) patterning, (3) etching [73].
Micromolding is utilized to fabricate various polymers, ranging from
pectin (natural polymers) to PVA and PVP(synthetic or semisynthetic (1) Deposition includes formation of films on a base material called
polymers) [29]. The technique utilizes multiple approaches to uniformly substrate. The films are formed by a physical or chemical process.
pour a liquid polymeric solution onto mold, followed by elimination of In the physical vapor deposition (PVD) technique, the materials
air voids. The removal of air voids can be facilitated by use of a vacuum are removed from the source and deposited on the substrate
(or alternatively, a centrifuge), followed by drying in an oven, and final whereas in the chemical vapor deposition (CVD) technique hot
removal from the mold [68]. substrate and the carrier gases are reacted chemically the in the
chamber.
3.1.2. Atomized spraying to fill molds (2) The patterning process includes transferring microneedle geom­
Continuous manufacturing microneedles with micromolding tech­ etry pattern, e.g. height, base width, tip angle, bore size details on
niques can be challenging due to centrifugation or vacuuming step the film. Lithography is a common process used for patterning.
involved [69]. Atomized spraying the polymer solution into the molds Briefly, a source of radiation such as light is impinged upon a
can overcome this challenge. McGrath et al. showed that atomization of photosensitive substrate thereby transferring a microneedle
pattern. The source of radiation can be different such as electron
Table 2 beam lithography, ion beam lithography or x-ray lithography
A summary of products made with different types of microneedles. [74]. The commonly used is photolithography. Generally
Product Type of MN Reference following sequential steps are involved in the mask transfer onto
the photosensitive-coated substrate: The first step involves for­
Vaccine Nano-porous ceramics, Boks et al.
carboxymethylcellulose, hyaluronan [41] mation of oxide layer by heating in the presence of steam or
Doxorubicin PVA micro mold Nguyen et al. humidified oxygen. This is followed by spin coating of a thin layer
(chemotherapeutic [42] of an organic polymer, known as a photosensitive/photoresist or
agent) resist material (sensitive to UV radiation) [75]. Then heat is
Dissolving microarrays Hyaluronic acid Liu et al. [36]
Insulin delivery Hollow metal Davis et al.
applied to the resist layer to remove solvent. This is followed by
[35]. exposure to UV lamp, through a mask, allowing a nearly perfect
Derma Rollers Solid MN Ita et al. [34] transfer (in other words “printing”) of the mask image onto the
Lidocaine Solid MN with PEG matrix Ma et al. [10] resist-coated wafers.
Soluvia Metal Donnelly
(3) Etching uses a harsh caustic agent to etch out the unprotected
et al. [39]
Micronjet Silicon Donnelly parts of the substrate to form a microneedle design of interest.
et al. [39] Etching can be done by a wet and dry technique [76].
Calcein TPMA Li et al. [11] 3.1 The dry process may involve etching methods like reactive
AdminPen Hollow stainless steel Yuzhakov ion or ion-beam milling. Dry etching process produces solid
et al. [40]
and hollow microneedles [29].

6
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

3.2 Wet itching: In this process, the substrate is submerged in the Table 3
chemical liquid which helps in elimination of the extra ma­ A summary of products made from various materials is shown in Table 3.
terial [77]. Wet itching can be done at same rate (isotropic) Material Common/commercial Fabrication method Reference
or different rates (anisotropic). It is used to produce metallic product
and silicon arrays. Metals Stainless steel Laser cutting Indermun S et al.
[30]
3.2.2. Laser cutting Titanium Injection molding Li, J. et al. [11]
In this method, an infra-red laser is used to cut metallic sheets in the Silicone TMAH Etching Tarbox, T.N et al.
[43]
shape of microneedles. AutoCAD software is used to design the geometry Natural Maltose, Starch Micromolding Kolli, C.S. et al.
of the MN. The laser beam then removes the metal sheet per the design [17],
which produces in plane MNs. The fabricated MNs are cleaned and Ling, M.H et al.
bended at 90� angle of the sheet. The tips are then tuned by electro­ [44]
Synthetic PLA 3D additive Camovi�c, M. et al.
polishing. This method is used for manufacturing metallic MNs. The
manufacturing [45]
most frequently employed material is stainless steel [30]. PGA Lithography, Park, J.H et al.
micromolding [46]
3.2.3. Laser ablation PLGA Micromolding Nguyen, H.X et al.
As the name suggests, in this method laser is impinged on a metal [47]

plate to form a bulge. Light pulses are used to give a defined shape to the
bulge to form a MN. Similar to laser cutting, laser ablation is also used polyacrylic acid to manufacture microneedle arrays using CLIP. Pre­
for fabricating solid metallic arrays. Metals like tantalum are also pared microneedles were shown to penetrate murine skin and release
fabricated [78]. rhodamine dye. The CLIP process can be basically used to design any
microneedle patch in two to 10 min [75]. The CLIP approach provides
3.2.4. Deep x-ray lithography huge flexibility for microneedle design and rapid prototyping that en­
In this technique a polymer solution is coated and cured on silicon ables a varied number of different parameters such as size, shape, and
wafer. An array of triangular columns and channels are generated by composition to be investigated in a throughput process (Table 3).
exposing the silicone wafer to vertical deep x-ray. The x-ray mask and
the substrate are continuously adjusted and aligned to provide the 4. Current biocompatibility data
desired MN geometry [79]. This technique is known for high precision
hollow MNs. Metallic, polymeric and ceramic and glass MN arrays can Biocompatibility and safety are of prime importance when consid­
be fabricated. ering the material of construction for microneedles. Stainless steel is an
iron-based alloy that contains about 11–30 wt percent of chromium and
3.3. Additive manufacturing varying amounts of nickel, which affects its properties and biocompat­
ibility. Stainless steel grade 316L demonstrates good biocompatibility
3.3.1. Two photon polymerization and therefore, is commonly used for surgical processes [80]. However, it
The two-photon polymerization is a layer-by-layer fabrication pro­ suffers a high corrosion rate when compared to other metals, such as
cess of three-dimensional structures that offers high throughput at a low titanium alloys [81]. Titanium alloys have superior corrosion resistance
cost. Femtosecond or picosecond laser pulses for the polymerization of as shown by insignificant mutagenicity, indicating that titanium alloys
resins into microneedle structure [74]. The polymerization is initiated are safe for humans and animals [82]. Silicones are widely used as im­
by two-photon absorption which generates an energy that is centered at plants but there have been concerns with their biocompatibility. Sili­
the laser focal point. The desired structure is initially sliced with the aid cones are brittle in nature and possess a risk of being fractured in the
of a computer-aided design (CAD) model. Next, each layer is written in skin [83]. The presence of silicone in skin can be hazardous due to sil­
the photosensitive resin by restoring the path of the laser for the contour icone- and glass-related granulomas [84].
to be filled in for each layer. After fabrication of the MN, resin is washed Ceramics have been used as replacement of musculoskeletal systems.
with a solvent and cured using ultraviolet light [68]. The main reason behind this is due to biocompatibility and high strength
of ceramics. The use of alumina for bone and dental implants is very
3.3.2. Microstereolithography (μSL) common from the past two decades [85]. Therefore, its biocompatibility
Micro-stereolithography (μSL) is a high throughput fabrication has been extensively studied. Some studies showed the long term effects
technique. It generates 3D microstructures in a bottom-up addition of bone implants with risk of aluminum release [86].
fashion. This technology has been widely used in biomedical and tissue Natural sugars have been extensively used in many drug delivery
engineering. The fabrication spatially controlled solidification of a systems, as they are safe. Carbohydrates have been known to be
liquid by photopolymerization [30]. Laser beam creates MN pattern on extremely biocompatible, and abundantly used for different types of
the surface of a resin which causes the resin to have definite depth. This application. Due to their excellent biocompatibility, they have been used
helps to adhere to a support platform. Photo-polymerization of the first for different applications like tissue engineering [87], nanocomposites
layer is followed by separation and realignment after which recoating of [88] and wound care [89].
the first built layer takes place. The pattern is cured in this second layer. Biocompatible materials are beneficial than other material as they
These steps are repeated to construct a solid, three-dimensional object. offer an additional advantage of controllable biodegradation rate.
Saturated aliphatic polymers are frequently used for decades in the field
3.3.3. Continuous liquid interface (CLIP) of bone tissue engineering. It is common to use PLA, PGA, and PLGA in
CLIP is used to computationally design microneedles. It is a novel scaffold application [90].
approach which utilizes continuous additive method versus a traditional
layer by layer approach mentioned above. The main advantage of CLIP 5. Geometry of microneedles and penetration depth in skin
over the traditional approach is that it fabricates microneedles with a
significantly higher resolution. It eliminates the separation and Since their inception, there have been many different designs of MNs
realignment which helps to build efficiency producing MNs at a faster with different surface areas since their first manufacture. As expected,
rate. Johnson et al. used trimethylolpropane triacrylate, polyethylene the cumulative delivery of the drug is directly proportional to the
glycol dimethacrylate, polycaprolactone trimethacrylate and

7
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

surface area of microneedle. This suggests the necessity to consider and 6. Applications of microneedle drug delivery
optimize the surface area which in turn is largely affected by its length
while designing the array system of microneedles. Most commonly the The effectiveness of a transdermal delivery is determined by the
length of MNs is fabricated in range from 150 to 1500 μm with a 50–250 ability of a molecule to penetrate into the skin. Numerous molecules
μm base width, and 1–25 μm tip diameter [91]. have faced challenges in manufacturing and application, mainly pep­
The penetration of skin by MNs is dependent on several factors. Due tides and vaccines, due to their molecular weight. MNs have not only
to the presence of protein elastin, skin possesses elasticity which can shown a successful intradermal delivery of such molecules but in some
prevent MN from penetration. Skin can fold around MN during its cases were also deemed more effective at delivering lower doses than the
application if the needles are blunt and short. Skin also contains protein current traditional delivery systems for the same desired clinical effect.
keratin which gives the strength to the skin. Due to this robustness, MN For instance, a similar change in hemagglutinin inhibition antibody titer
insertion forces may exceed the tensile forces of the MN causing them to was found with less than half the drug concentration when delivered
fracture and/or break. This can happen in the MNs are too long and/or with MN compared to traditional intra-muscular injection [95]. The
made of relatively weak materials [92]. Therefore, microneedle geom­ success of the MN systems was evident from the approval from many
etry is very crucial for efficient MN penetration and delivery into the regulatory agencies for seasonal influenza vaccine delivered by MN
skin. MNs are evaluated for different shapes like cylindrical, rectangular, [96–98]. MN have been studied to address several skin conditions such
pyramidal, conical, octagonal and quadrangular with varying needle as viral warts [99], dermatitis [100], and psoriasis [101]. MN have also
lengths and widths as shown in Fig. 7 [75]. The low density of micro­ shown their efficacy in treatment of skin cancer [102]. MN have not only
needle arrays is better than the dense MN array which can cause ‘bed of been studied in intradermal delivery, but they also find application in
nails’ effect. ocular drug delivery. MN have shown to effectively treat ocular diseases
Al-Qallaf et al. investigated that the drug concentration in the blood like glaucoma [103], macular degeneration [104], Uveitis [105] and
increases with increase in MN length and surface area. Overall, the retinal vascular occlusion [106]. Currently, most microneedles that are
optimum MN design should be long enough without breaking or causing available on the market are indicated for cosmetic purposes. Derma­
any pain [92]. roller helps to improve health of skin, while dissolving microneeldes are
Studies have concluded that an increase in the length of microneedle used to treat acne.
increases the surface area and activates cells in the dermis layer thus
increasing the drug absorption [93]. A study carried out by Romgens 7. Current research, challenges and future trends
et al. showed that the greater number of cells are activated in the dermis
with increase in MN length. Therefore by exactly knowing the depth and Microneedles have been constantly evolving since their invention in
knowledge of skin cells and layers, MN length can be adapted to activate 1970s. Since then, leaps and bounds of progress has been made in the
Langerhans cells or dermal dendritic cells residing in the dermis [93]. field of MNs. MN mode of drug delivery has seen the use of established
Increasing the microneedle length may have some limitations. Gill materials such as glass and stainless steel to many different poly­
et al. showed that the longer the length of the microneedle, the more the saccharides. A thorough understanding and development of microelec­
pain is generated. For instance, when 400 MNs (150 μm in length) were tronics industry has made the fabrication technique easy and
inserted in human volunteers, it was deemed painless against a 2-mm commercially productive. Various polymers have been utilized for pro­
deep insertion of a 26-gauge hypodermic needle. Scraping the skin at duction of MNs. A precise control on the drug delivery has been enabled
a depth of 50–200 μm in length was found to be painless. The authors through these systems. MNs opens up opportunities and potentials to
compared stimulated pain to a 26-gauge hypodermic needle in healthy give a new face to the realm of drug delivery for various sizes of mole­
human subjects and concluded that the microneedle geometry has in­ cules. However, these delivery systems present several challenges. For
fluences on pain. As the length increased the pain was reported to in­ instance, as MNs are injected into the skin with drugs delivered directly
crease [94]. Thus, a detailed evaluation on MN geometry is required to into systemic circulation, do these systems need to be sterile? With
ensure optimum penetration without fracture with minimal pain. sterility in consideration, this limits the number of materials that can be
used. Also, the type of sterilization technique used will impact the

Fig. 7. Different shapes of microneedles developed This work is licensed under the Creative Commons Attribution 4.0 International License, which permits unre­
stricted use, distribution, modification, and reproduction in any medium, provided you. To view a copy of the Creative Commons license, please visit http://creative
commons.org/licenses/by/4.0/.

8
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

microneedle type. For instance, heat sterilization can be utilized for solid [3] S. Wiedersberg, R.H. Guy, Transdermal drug delivery: 30þ years of war and still
fighting, J. Contr. Release 190 (2014) 150–156.
microneedles made from metals while a gamma ray sterilization would
[4] D.J.P. Sharma, Microneedles: an approach in transdermal drug delivery: a review
be the choice for dissolving microneedles. Challenges with respect to 6 (1) (2018) 7–15.
dose delivery are also evident as relatively low doses can be used with [5] B.W. Barry, Novel mechanisms and devices to enable successful transdermal drug
the MN technology. Additionally, repeated use of MNs at the same site delivery, Eur. J. Pharmaceut. Sci. 14 (2) (2001) 101–114.
[6] J. Kushner IV, D. Blankschtein, R. Langer, Heterogeneity in skin treated with low-
may lead to erythema, swelling, and skin irritation. MNs can potentially frequency ultrasound, Eur. J. Pharmaceut. Sci. 97 (10) (2008) 4119–4128.
cause and infection if the skin pores remain open for a longer time while [7] S.N. Murthy, S.M. Sammeta, C. Bowers, Magnetophoresis for enhancing
some MN delivery systems pose a challenge of leaking. At this time, a transdermal drug delivery: mechanistic studies and patch design, J. Contr.
Release 148 (2) (2010) 197–203.
plethora of research is carried out on materials and techniques to [8] M.R. Prausnitz, et al., Electroporation of mammalian skin: a mechanism to
improve the current MN delivery. In the coming years, more MN prod­ enhance transdermal drug delivery 90 (22) (1993) 10504–10508.
ucts will be commercialized based on the volume of on-going clinical [9] Y. Song, et al., Transdermal delivery of human growth hormone via laser-
generated micropores 8 (2) (2018) 450–460.
studies. A detailed research needs to be performed for regulatory [10] X. Ma, et al., Delicate assembly of ultrathin hydroxyapatite nanobelts with
approval of these systems as they act as the bridge between transdermal nanoneedles directed by dissolved cellulose, Inorg. Chem. 57 (8) (2018)
and injectable dosage form. 4516–4523.
[11] J. Li, et al., Fabrication of a Ti porous microneedle array by metal injection
Several clinical trials have been performed on MN technology. The molding for transdermal drug delivery 12 (2) (2017).
first study was done in 2001 by Kaushik el at using silicon microneedles [12] A. Ullah, C.M. Kim, G.M. Kim, Porous polymer coatings on metal microneedles for
to compare pain index in human volunteer subjects [107]. Since then, enhanced drug delivery, R Soc Open Sci 5 (4) (2018) 171609.
[13] Y. Li, et al., Fabrication of sharp silicon hollow microneedles by deep-reactive ion
different researchers and companies have done comprehensive clinical
etching towards minimally invasive diagnostics 5 (1) (2019) 1–11.
trials using different active ingredients with MN systems. A similar study [14] Y.-C. Kim, J.-H. Park, M.R. Prausnitz, Microneedles for drug and vaccine delivery,
was performed on the oral cavity of 30 human volunteers to determine Adv. Drug Deliv. Rev. 64 (14) (2012) 1547–1568.
the pain index. In another study carried out by Ornelas et al. anesthetic [15] X. He, et al., Microneedle system for transdermal drug and vaccine delivery:
devices, safety, and prospects 17 (4) (2019), 1559325819878585.
effect from lidocaine was found to be produced in half the time in human [16] K.J.B. Ita, Pharmacotherapy, Dissolving microneedles for transdermal drug
volunteers using MN compared to topical cream [108]. MN applications delivery, Advances and challenges 93 (2017) 1116–1127.
have also extended to pediatric populations. In a recent study, the effects [17] C.S. Kolli, A.K.J.P.r. Banga, Characterization of solid maltose microneedles and
their use for transdermal delivery 25 (1) (2008) 104–113.
of MN in 34 children was assessed by applying placebo MN patches. [18] X. Chen, et al., Dry-coated microprojection array patches for targeted delivery of
NanoPass technology is actively involved in clinical trials of their immunotherapeutics to the skin 139 (3) (2009) 212–220.
micronjet injection system. There are two Phase I studies on-going in the [19] J. Chen, et al., Controllable coating of microneedles for transdermal drug delivery
41 (3) (2015) 415–422.
field of allergy immunotherapy for evaluation of safety and immune [20] B. Chiang, et al., Distribution of particles, small molecules and polymeric
response. Another on-going phase II study involves delivery of dendritic formulation excipients in the suprachoroidal space after microneedle injection
cells targeting viral vector and recombinant protein in combination with 153 (2016) 101–109.
[21] A.P. Raphael, et al., Formulations for microprojection/microneedle vaccine
atezolizumab for treatment of metastatic sarcoma [109]. delivery: structure, strength and release profiles 225 (2016) 40–52.
[22] M.R. Prausnitz, Y. Gomaa, W.J.N.m. Li, Microneedle patch drug delivery in the
8. Conclusions gut 25 (10) (2019) 1471–1472.
[23] S. Jung, H. Lee, S. Li, A study on the effect of spirulina-containing cosmetics using
micro-needle, J. Korea Acad. 18 (6) (2017) 269–276.
A framework has been presented in this paper to review and discuss [24] G. Du, et al., Intradermal vaccination with hollow microneedles: a comparative
the categories, materials, and geometries of microneedles, an innovative study of various protein antigen and adjuvant encapsulated nanoparticles 266
(2017) 109–118.
and promising technology to enhance the skin permeability. Biocom­ [25] L.R. Pires, et al., A perspective on microneedle-based drug delivery and
patibility issues and selection criteria of materials of construction of diagnostics in paediatrics 9 (4) (2019) 49.
microneedles were also elaborated. The design and dimensions of [26] Y. Hao, et al., Microneedles-based transdermal drug delivery systems: a review 13
(12) (2017) 1581–1597.
microneedles have been indicated as the primary parameters to be [27] S. Brown, et al., Minimally invasive technique for measuring transdermal glucose
customized for the optimal performance of the needle system. Micro­ with a fluorescent biosensor 410 (27) (2018) 7249–7260.
needle length could be adjusted to achieve a desired depth of penetra­ [28] H.L. Quinn, et al., The role of microneedles for drug and vaccine delivery 11 (11)
(2014) 1769–1780.
tion to facilitate the drug release and maximize the drug delivery
[29] M.R. Prausnitz, Microneedles for transdermal drug delivery, Adv. Drug Deliver
enhancement. The discussion in this paper suggests that biodegradable Rev. 56 (5) (2004) 581–587.
microneedles appear superior to non-biodegradable needles in almost all [30] S. Indermun, et al., Current advances in the fabrication of microneedles for
transdermal delivery 185 (2014) 130–138.
aspects except the risk of possible skin-related toxicity. Microneedling
[31] J. Gupta, et al., Kinetics of skin resealing after insertion of microneedles in human
technology has progressed to a possibility of large-scale production and subjects 154 (2) (2011) 148–155.
microneedle-based drug delivery systems are expected to appear on the [32] H. Kalluri, C.S. Kolli, A.K. Banga, Characterization of microchannels created by
pharmaceutical and cosmeceutical market in the near future. metal microneedles: formation and closure, AAPS J. 13 (3) (2011) 473–481.
[33] K. van der Maaden, W. Jiskoot, J. Bouwstra, Microneedle technologies for (trans)
dermal drug and vaccine delivery, J. Contr. Release 161 (2) (2012) 645–655.
[34] K.J.P. Ita, Transdermal delivery of drugs with microneedles—potential and
Declaration of competing interest challenges 7 (3) (2015) 90–105.
[35] S.P. Davis, et al., Hollow metal microneedles for insulin delivery to diabetic rats
52 (5) (2005) 909–915.
The authors declare no conflict of interest. [36] S. Liu, et al., Transdermal delivery of relatively high molecular weight drugs
using novel self-dissolving microneedle arrays fabricated from hyaluronic acid
and their characteristics and safety after application to the skin 86 (2) (2014)
Acknowledgments
267–276.
[37] A. Ono, et al., Development of novel faster-dissolving microneedle patches for
Financial supports from Philadelphia College of Pharmacy, Univer­ transcutaneous vaccine delivery 9 (3) (2017) 27.
sity of the Sciences are gratefully acknowledged. [38] H.S. Gill, M.R. Prausnitz, Coated microneedles for transdermal delivery, J. Contr.
Release 117 (2) (2007) 227–237.
[39] R.F. Donnelly, et al., Hydrogel-forming microneedle arrays exhibit antimicrobial
References properties: potential for enhanced patient safety 451 (1–2) (2013) 76–91.
[40] V.V. Yuzhakov, The AdminPenTM microneedle device for painless & convenient
drug delivery, J. Drug Deliv. Technol. 10 (4) (2010) 32–36.
[1] E. Larraneta, et al., Microneedle arrays as transdermal and intradermal drug
[41] M.A. Boks, et al., Controlled release of a model vaccine by nanoporous ceramic
delivery systems: materials science, manufacture and commercial development
microneedle arrays 491 (1–2) (2015) 375–383.
104 (2016) 1–32.
[2] H. Marwah, et al., Permeation enhancer strategies in transdermal drug delivery
23 (2) (2016) 564–578.

9
R. Nagarkar et al. Journal of Drug Delivery Science and Technology 59 (2020) 101923

[42] H.X. Nguyen, et al., Poly (vinyl alcohol) microneedles: fabrication, [76] S. More, T. Ghadge, S. Dhole, Microneedle: an advanced technique in transdermal
characterization, and application for transdermal drug delivery of doxorubicin, drug delivery system, Asia J. Res. Pharmaceut. Sci. 3 (3) (2013) 141–148.
Eur. J. Pharm. Biopharm. 129 (2018) 88–103. [77] Y. Yoon, et al., Fabrication of a microneedle/CNT hierarchical micro/nano
[43] T.N. Tarbox, et al., An update on coating/manufacturing techniques of surface electrochemical sensor and its in-vitro glucose sensing characterization 13
microneedles 8 (6) (2018) 1828–1843. (12) (2013) 16672–16681.
[44] M.-H. Ling, M.-C.J.A.b. Chen, Dissolving polymer microneedle patches for rapid [78] R.F. Donnelly, et al., Microneedle arrays allow lower microbial penetration than
and efficient transdermal delivery of insulin to diabetic rats 9 (11) (2013) hypodermic needles in vitro 26 (11) (2009) 2513–2522.
8952–8961. [79] S.J. Moon, et al., Fabrication of microneedle array using LIGA and hot embossing
[45] M. Camovi�c, et al., Coated 3d printed PLA microneedles as transdermal drug process 11 (4–5) (2005) 311–318.
delivery systems, in: International Conference on Medical and Biological [80] J.R. Davis, Stainless Steels, ASM international, 1994.
Engineering, Springer, 2019. [81] L.C. Zhang, L.Y. Chen, A review on biomedical titanium alloys: recent progress
[46] J.-H. Park, M.G. Allen, M.R. Prausnitz, Biodegradable polymer microneedles: and prospect, Adv. Eng. Mater. 21 (4) (2019) 1801215.
fabrication, mechanics and transdermal drug delivery, J. Contr. Release 104 (1) [82] Q. Chen, G.A.J.M.S. Thouas, E.R. Reports, Metallic implant biomaterials 87
(2005) 51–66. (2015) 1–57.
[47] H.X. Nguyen, A.K. Banga, Delivery of methotrexate and characterization of skin [83] C.J.B.m. O’Mahony, Structural characterization and in-vivo reliability evaluation
treated by fabricated PLGA microneedles and fractional ablative laser, Pharm. of silicon microneedles 16 (3) (2014) 333–343.
Res. (N. Y.) 35 (3) (2018) 68. [84] P.J.C.o. Christel, r. research, Biocompatibility of surgical-grade dense
[48] S. Henry, et al., Microfabricated microneedles: a novel approach to transdermal polycrystalline alumina, 1992, pp. 10–18, 282.
drug delivery 87 (8) (1998) 922–925. [85] M. Navarro, et al., Biomaterials in orthopaedics 5 (27) (2008) 1137–1158.
[49] S.P. Narayanan, S. Raghavan, Solid silicon microneedles for drug delivery [86] M. Lewandowska-Szumiel, J.J.C.m. Kolnender, Aluminium release as a new factor
applications, Int. J. Adv. Manuf. Technol. 93 (1–4) (2017) 407–422. in the estimation of alumina bioceramic implants 5 (2–4) (1990) 167–175.
[50] K.J. Krieger, et al., Simple and customizable method for fabrication of high-aspect [87] A. Carvalho, et al., Control of osmotic pressure to improve cell viability in cell-
ratio microneedle molds using low-cost 3D printing 5 (1) (2019) 1–14. laden tissue engineering constructs 12 (2) (2018) e1063–e1067.
[51] S. Bystrova, R.J.M.e. Luttge, Micromolding for ceramic microneedle arrays 88 (8) [88] T.D. Andreeva, et al., Hybrid graphene oxide/polysaccharide nanocomposites
(2011) 1681–1684. with controllable surface properties and biocompatibility 181 (2018) 78–85.
[52] F. Verbaan, et al., Improved piercing of microneedle arrays in dermatomed [89] A. Basu, et al., Ion-crosslinked wood-derived nanocellulose hydrogels with
human skin by an impact insertion method 128 (1) (2008) 80–88. tunable antibacterial properties: candidate materials for advanced wound care
[53] S. Gittard, et al., Pulsed laser deposition of antimicrobial silver coating on applications 181 (2018) 345–350.
Ormocer® microneedles 1 (4) (2009), 041001. [90] M.I. Sabir, X. Xu, L. Li, A review on biodegradable polymeric materials for bone
[54] A. Ovsianikov, et al., Two photon polymerization of polymer–ceramic hybrid tissue engineering applications, Journal of materials science 44 (21) (2009)
materials for transdermal drug delivery 4 (1) (2007) 22–29. 5713–5724.
[55] S.-O. Choi, et al., An electrically active microneedle array for electroporation 12 [91] M. Pearton, et al., Microneedle delivery of plasmid DNA to living human skin:
(2) (2010) 263–273. formulation coating, skin insertion and gene expression 160 (3) (2012) 561–569.
[56] T. Miyano, et al., Sugar micro needles as transdermic drug delivery system 7 (3) [92] B. Al-Qallaf, D.B Das, Optimization of square microneedle arrays for increasing
(2005) 185–188. drug permeability in skin, Chem. Eng. Sci. 63 (9) (2008) 2523–2535.
[57] X. Hong, et al., Dissolving and biodegradable microneedle technologies for [93] A.M. R€ omgens, et al., Predicting the optimal geometry of microneedles and their
transdermal sustained delivery of drug and vaccine 7 (2013) 945. array for dermal vaccination using a computational model 19 (15) (2016)
[58] J.W. Lee, J.-H. Park, M.R.J.B. Prausnitz, Dissolving microneedles for transdermal 1599–1609.
drug delivery 29 (13) (2008) 2113–2124. [94] H.S. Gill, et al., Effect of microneedle design on pain in human subjects 24 (7)
[59] R.F. Donnelly, et al., Processing difficulties and instability of carbohydrate (2008) 585.
microneedle arrays 35 (10) (2009) 1242–1254. [95] P. Van Damme, et al., Safety and efficacy of a novel microneedle device for dose
[60] G. Li, et al., In vitro transdermal delivery of therapeutic antibodies using maltose sparing intradermal influenza vaccination in healthy adults 27 (3) (2009)
microneedles 368 (1–2) (2009) 109–115. 454–459.
[61] C. Martin, et al., Low temperature fabrication of biodegradable sugar glass [96] R.L. Atmar, S.M. Patel, W.A. Keitel, Intanza®: a new intradermal vaccine for
microneedles for transdermal drug delivery applications 158 (1) (2012) 93–101. seasonal influenza, Expet Rev. Vaccine 9 (12) (2010) 1399–1409.
[62] R.F. Donnelly, et al., Microneedle-mediated Transdermal and Intradermal Drug [97] G. Icardi, et al., Current evidence on intradermal influenza vaccines administered
Delivery, John Wiley & Sons, 2012. by Soluvia™ licensed micro injection system 8 (1) (2012) 67–75.
[63] D. Kuang, et al., Silk fibroin/polyvinyl pyrrolidone interpenetrating polymer [98] L. Sticchi, et al., The intradermal vaccination: past experiences and current
network hydrogels 10 (2) (2018) 153. perspectives 51 (1) (2010) 7–14.
[64] R.F. Donnelly, et al., Hydrogel-forming microneedle arrays for enhanced [99] K. Konicke, E.J.D.S. Olasz, Successful treatment of recalcitrant plantar warts with
transdermal drug delivery 22 (23) (2012) 4879–4890. bleomycin and microneedling 42 (8) (2016) 1007–1008.
[65] N.N. Aung, et al., Fabrication, characterization and comparison of α-arbutin [100] R. Paleco, et al., Enhancement of the in vitro penetration of quercetin through pig
loaded dissolving and hydrogel forming microneedles, 2020, p. 119508. skin by combined microneedles and lipid microparticles 472 (1–2) (2014)
[66] E.M. Migdadi, et al., Hydrogel-forming microneedles enhance transdermal 206–213.
delivery of metformin hydrochloride 285 (2018) 142–151. [101] A.B. Gottlieb, et al., Pharmacodynamic and pharmacokinetic response to anti-
[67] R.F. Donnelly, T.R.R. Singh, A.D.J.D.d. Woolfson, Microneedle-based drug tumor necrosis factor-α monoclonal antibody (infliximab) treatment of moderate
delivery systems: microfabrication, drug delivery, and safety 17 (4) (2010) to severe psoriasis vulgaris 48 (1) (2003) 68–75.
187–207. [102] R.F. Donnelly, et al., Microneedle-mediated intradermal delivery of 5-aminole­
[68] S.D. Gittard, et al., Fabrication of polymer microneedles using a two-photon vulinic acid: potential for enhanced topical photodynamic therapy 129 (3) (2008)
polymerization and micromolding process 3 (2) (2009) 304–311. 154–162.
[69] M.G. McGrath, et al., Production of dissolvable microneedles using an atomised [103] J. Jiang, et al., Coated microneedles for drug delivery to the eye 48 (9) (2007)
spray process: effect of microneedle composition on skin penetration 86 (2) 4038–4043.
(2014) 200–211. [104] K. Kadonosono, et al., Displacement of submacular hemorrhages in age-related
[70] M. Ogundele, H. Okafor, Transdermal drug delivery: microneedles, their macular degeneration with subretinal tissue plasminogen activator and air 122
fabrication and current trends in delivery methods, Pharmaceut. Res. Int. (2017) (1) (2015) 123–128.
1–14. [105] B.C. Gilger, et al., Treatment of acute posterior uveitis in a porcine model by
[71] J.D. Kim, et al., Droplet-born air blowing: novel dissolving microneedle injection of triamcinolone acetonide into the suprachoroidal space using
fabrication 170 (3) (2013) 430–436. microneedles 54 (4) (2013) 2483–2492.
[72] J. Gupta, et al., Rapid pharmacokinetics of intradermal insulin administered [106] K. Kadonosono, et al., Intra-retinal arterial cannulation using a microneedle for
using microneedles in type 1 diabetes subjects 13 (4) (2011) 451–456. central retinal artery occlusion, Sci. Rep. 8 (1) (2018) 1360.
[73] A.R. Razali, Y.J.P.E. Qin, A review on micro-manufacturing, microforming and [107] S. Kaushik, et al., Lack of pain associated with microfabricated microneedles 92
their key issues 53 (2013) 665–672. (2) (2001) 502–504.
[74] S.D. Gittard, et al., Two-photon polymerization of microneedles for transdermal [108] J. Ornelas, et al., Effect of microneedle pretreatment on topical anesthesia: a
drug delivery 7 (4) (2010) 513–533. randomized clinical trial 152 (4) (2016) 476–477.
[75] A.R. Johnson, et al., Single-step fabrication of computationally designed [109] NanoPass. https://www.nanopass.com/clinical/.
microneedles by continuous liquid interface production 11 (9) (2016).

10

You might also like