You are on page 1of 11

Advanced Drug Delivery Reviews 127 (2018) 35–45

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews

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

Device-assisted transdermal drug delivery☆


Hyunjae Lee a,1, Changyeong Song a,b,1, Seungmin Baik a,b,1, Dokyoon Kim a,
Taeghwan Hyeon a,b,⁎, Dae-Hyeong Kim a,b,⁎
a
Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
b
School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea

a r t i c l e i n f o a b s t r a c t

Article history: Transdermal drug delivery is a prospective drug delivery strategy to complement the limitations of conventional
Received 12 May 2017 drug delivery systems including oral and injectable methods. This delivery route allows both convenient and
Received in revised form 19 August 2017 painless drug delivery and a sustained release profile with reduced side effects. However, physiological barriers
Accepted 29 August 2017
in the skin undermine the delivery efficiency of conventional patches, limiting drug candidates to small-mole-
Available online 1 September 2017
cules and lipophilic drugs. Recently, transdermal drug delivery technology has advanced from unsophisticated
Keywords:
methods simply relying on natural diffusion to drug releasing systems that dynamically respond to external stim-
Transdermal drug delivery uli. Furthermore, physical barriers in the skin have been overcome using microneedles, and controlled delivery by
Microneedles wearable biosensors has been enabled ultimately. In this review, we classify the evolution of advanced drug de-
Wearable biosensors livery strategies based on generations and provide a comprehensive overview. Finally, the recent progress in ad-
Controlled drug delivery vanced diagnosis and therapy through customized drug delivery systems based on real-time analysis of
Soft bioelectronics physiological cues is highlighted.
© 2017 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
2. Advancement of transdermal drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
2.1. First generation: diffusion-based transdermal drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.2. Second generation: non-invasive transdermal drug delivery with actuation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.3. Third generation: minimally invasive transdermal drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3. Fourth generation: controlled and feedback-induced transdermal drug delivery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
3.1. Transdermal drug delivery based on physiological/electrophysiological monitoring . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
3.2. Transdermal drug delivery based on biochemical signal monitoring . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
3.3. Smart microneedle systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43

1. Introduction
Abbreviations: RF, radiofrequency; DMF, dimethylformamide; DMSO, dimethyl
sulfoxide; MSN, mesoporous silica nanoparticle; NP, nanoparticle; PCM, phase change
material. The growing demand for patient-friendly therapies has led to the de-
☆ This review is part of the Advanced Drug Delivery Reviews theme issue on "Skin- velopment of transdermal drug delivery that has several advantages
Associated Drug Delivery". over conventional drug delivery methods. For instance, transdermal de-
⁎ Corresponding authors at: Center for Nanoparticle Research, Institute for Basic Science
(IBS), Seoul 08826, Republic of Korea.
livery enables sustained and controlled drug release, and promotes pa-
E-mail addresses: thyeon@snu.ac.kr (T. Hyeon), dkim98@snu.ac.kr (D.-H. Kim). tients' compliance and convenience with its non-invasiveness and
1
These authors contributed equally to this work. painlessness [1]. In particular, transdermal route is a good alternative

http://dx.doi.org/10.1016/j.addr.2017.08.009
0169-409X/© 2017 Elsevier B.V. All rights reserved.
36 H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45

to needle injection or oral intake of medication. Transdermal delivery to adopt an advanced paradigm of device-assisted personalized therapy
often requires a lower dosage of drugs than oral administration. With where the precise control of the amount and duration of a drug dose in
a short diffusive pathway to reach vasculature, transdermal mode response to the patient's physiological condition is a key to maximizing
helps the patients to circumvent side effects due to digestion and me- the therapeutic efficacy (Fig. 1a and b). The controlled and on-demand
tabolism of the drug in the gastrointestinal tract. Transdermal delivery administration of a drug tailored to the needs of each patient requires
is painless and non-invasive, the advantage which is prominent against extensive innovation. Wearable devices may be a viable and useful
intravenous or intramuscular administration. The non-invasiveness al- strategy because they can collect an assortment of physiological [6–
lows repeated administrations of the drug over the same region of the 15], electrophysiological [16–21], and biochemical [22–27] cues, and
body for a long-term treatment, and thus provides patients with com- thereby, facilitate drug delivery via energetic actuations.
pliance and convenience. In this review, we recapitulate the major breakthroughs of four gen-
However, transdermal drug delivery is not a flawless drug transport erations of transdermal drug delivery strategies, providing the reader
method. The stratum corneum, the outermost barrier of the skin against with the detailed advantages and challenges of each generation. In ac-
foreign materials, imposes an obstacle to the transdermal diffusion of cordance with a growing interest in controlled drug release for person-
drugs [2]. Since only a few small-molecule drugs with high lipophilicity alized medical treatment, we also emphasize the recent progress on
can naturally permeate through the hydrophobic stratum corneum, a wearable devices that support device-assisted transdermal drug deliv-
vast majority of hydrophilic drugs have been ruled out against transder- ery. This review concludes with case presentations to demonstrate the
mal delivery. Even after the drug successfully penetrates the stratum wearable biosensors and advanced transdermal delivery schemes that
corneum, it may be degraded prematurely by epidermal enzymes, are seamlessly integrated. Thereby, we hope to present a novel potential
resulting in decreased bioavailability. Latency in the onset of action as protocol and foundation for the next generation of personalized
well as a lack of pharmacokinetic control may also undermine the reli- therapy.
ability of the transdermal delivery.
The endeavor for a better transdermal delivery has evolved towards 2. Advancement of transdermal drug delivery systems
overcoming such limitations, and the innovation has been seen in sever-
al generations, namely, focusing on drug formulation, external actua- Among the anatomical layers and appendageal structures of the skin
tion, and minimally invasive microneedles. The first generation of (Fig. 2a), the epidermis and its outermost stratum corneum are the most
transdermal delivery focused on refining the drug formulation to max- challenging barriers to transdermal delivery [28,29]. The brick-and-
imize its diffusion through the skin as manifested by emulsions, nano- mortar assembly of keratinocytes and intercellular lipids renders the
carriers, and chemical enhancers. The second generation has sought ex- epidermis impregnable to most foreign materials. Therefore, a cursory
ternal forces, such as heat and electricity, to energetically fuel the drug observation of transdermal delivery can regard it as a counterintuitive
to cross the epidermal layers and reach the vasculature. The third gen- strategy for delivering a drug systematically. However, a detailed analy-
eration incorporated microneedles, which puncture the outer skin sis reveals that transdermal delivery provides a number of advantages
layer but preserve the dermis, to provide the drug with a physical short- over oral intake or hypodermic injections. Compared with oral delivery,
cut bypassing the epidermal barriers. The breakthroughs of previous transdermal delivery requires a lower dosage and has fewer side effects
generations have greatly improved the efficiency of transdermal drug because transdermal diffusion circumvents the digestion and first-pass
delivery. metabolism of the active drug in the gastrointestinal tract and liver
Although the betterment of transdermal delivery so far has focused [30]. Moreover, transdermal delivery is less painful, minimally invasive,
on maximizing the delivery efficiency, transdermal drug delivery is and convenient to patients so that the compliance with drug adminis-
yet to see another breakthrough by adopting a paradigm shift towards tration increases to a great extent. Because such advantages offset the
personalized therapy [3–5]. Furthermore, transdermal delivery is set drawbacks of transdermal delivery, studies continue to develop reliable

a b

Body
temperature

Diagnosis

Blood Blood
pressure glucose
Device-assisted
transdermal
drug delivery

Therapy

Microneedle
Iontophoresis
Device-assisted
transdermal drug
delivery Heat

Fig. 1. (a) Schematic illustration of the device-assisted transdermal drug delivery for patient-customized skin-based therapy. (b) The wearable device system conducts real-time
monitoring of vital signs and actuates transdermal drug delivery according to individual health condition.
H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45 37

a Sweat gland pore


b 1st generation c 2nd generation
Drug-loaded
patch External
Hair shaft stimuli
Epidermis

Natural Actuated
Skin diffusion diffusion
Dermis

d 3rd generation e Wearable 4th generation


Actuation device

Subcutaneous
layer
Capillary
Tactile
corpuscle Penetration Signal Drug
Sweat gland duct monitoring delivery

Fig. 2. (a) Schematic illustration of human skin. (b) First generation transdermal drug delivery technology via natural diffusion of drugs. (c) Second generation transdermal drug delivery
technology for actuated drug delivery via external stimulation. (d) Third generation transdermal drug delivery technology for enhanced drug transport via microneedle-mediated
destruction of skin layer and various functionalities accompanying microneedles. (e) Fourth generation transdermal drug delivery technology for patient-customized therapy with the
assistance of wearable devices.

delivery systems and broaden the library of applicable candidate drugs. cycle of diagnosis, actuated drug release, and follow-up diagnosis con-
Novel designs of transdermal patches to increase the skin penetration of stitute a feedback loop that provides patients with a precise and person-
drugs have been widely investigated, and major breakthroughs have ally customized medical treatment.
been observed as the evolving generations of transdermal delivery
systems. 2.1. First generation: diffusion-based transdermal drug delivery
The research on the first generation of transdermal drug delivery
systems focused on tailoring the physicochemical properties of chemi- Transdermal drug delivery is an attractive drug delivery option be-
cal drugs. Drugs for transdermal delivery are either selected or modified cause it offers several advantages. First, dermal layers provide the
to have a high partition coefficient and a low molecular weight for facile drug a shorter route to the bloodstream than gastrointestinal tract
diffusion through the skin barrier (Fig. 2b). The second-generation re- does [1]. Particularly, transdermal shortcut also enhances bioavailability
search focused on improving the skin permeability of drugs using chem- and reduces the dosage because the drug avoids the extensive first-pass
ical enhancers and stimulators through external driving forces (Fig. 2c). metabolism and degradation in liver. Second, patients usually comply
Chemical enhancers [31,32] and emulsion/nano-carriers [33–36] solu- with transdermal drug administration because a drug patch is painless,
bilize the drugs and facilitate their easy penetration into the skin. Exter- non-invasive, and easy to use. A long-lasting patch that provides
nal actuation using heat [37], electricity [38], or noncavitational sustained drug release for up to several days is convenient for patients,
ultrasound [39] also facilitates drug transportation. The third-genera- and its non-invasiveness allows repeated drug application over the
tion research adopted methods that cause microscopic destruction of same part of the body. Freedom from pain, compared particularly to
epidermis to facilitate the delivery of drugs. Radiofrequency (RF) abla- needle-based injections, increases the popularity and acceptance of
tion [40], ultrasound [41], and lasers [42,43] disrupt the stratum the transdermal delivery.
corneum temporarily and enhance drug penetration through the weak- The first generation of simple transdermal patches emerged during
ened barrier. Microneedles are another approach of microinvasive the early 1970s. Following the first approval from the U.S. Food and
transdermal delivery with vastly diverse drug candidates [44–47]. The Drug Administration (FDA) for the use of scopolamine patch for motion
microneedles create micrometer-sized porations in epidermis, which sickness, approximately 19 patches including nicotine, menthol, and es-
serve as an unobstructed pathway for incoming drugs (Fig. 2d). Since tradiol are commercially available to date [52]. However, the number of
the size of the microneedles leaves the dermal and hypodermal layers drugs that are suitable for patch formulation is severely limited because
untouched, the microneedle is considered virtually painless and mini- of the physiological barrier of the epidermis. The vast majority of the
mally invasive. first generation transdermal drugs are highly lipophilic with partition
While breakthroughs in transdermal delivery in previous genera- coefficients greater than 104, small particle sizes, and molecular weights
tions have focused on maximizing the efficiency of drug delivery, the re- no more than 400 Da [53]. The forerunning system of transdermal deliv-
cent growth in personalized medicine requires a new generation of the ery depending on the physicochemical properties of drug molecules has
drug delivery system aimed at controlled and feedback-induced trans- been effective for its therapeutic purposes but with a limited variety in
dermal release of drugs. Wearable devices consisting of sensors and ac- applications. Therefore, extensive efforts to render more drug candi-
tuators are expected to meet such needs [48–50]. Indeed, novel dates transdermally applicable and to further increase their delivery ef-
materials design and device fabrication have spurred the innovation in ficiency had ensued.
wearable devices for biomedical application, providing a new opportu-
nity in personalized healthcare [51]. By precisely monitoring the key in- 2.2. Second generation: non-invasive transdermal drug delivery with
dicators of disease treatment and potential side effects of the delivered actuation
drugs, decisions can be rapidly made on the appropriate amount of drug
to be delivered. The external energy stimulates and controls state-of- The second-generation transdermal delivery strategies seek to max-
the-art transdermal delivery systems. Device-assisted transdermal de- imize the permeability of the drug to the skin using chemical enhancers
livery, an ensemble of soft bioelectronics and a transdermal drug deliv- or external energy sources without damaging the structure of the skin.
ery system, simultaneously controls the release of drugs and monitors Chemical enhancers facilitate the drug to penetrate the skin by
the alteration of the physiological states by the drug (Fig. 2e). The interacting with the proteins comprising the skin and by increasing
38 H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45

a Microemulsion Human skin b c 100


d

Relative release (%)


Solution Nanoparticle Temp. resp. nanogel NPC H+
80 32 oC H+
SC SC 22 oC Farnesol
60 Diclofenac Bacteria
Epidermis Epidermis
40 Biofilm pH 7.2 pH 4.5

Dermis Dermis 20 H+

0 H+
Nucleic acid loaded H+
0.1 µm 100 µm nanoparticle 12 24
Time (hr)

Cumul. release (%)


e 60 f

Rel. amount (µg/mL)


Light on Light off OFF OFF OFF OFF 60 w/o strain
ON ON ON ON
50
40
40 w/ strain
Stretch F F 30
20
20
no light 10
0 w/ light Drug release
0 4 8 12 16 20 24 0
0 100 400 1000
Time (h) Stretching cycles
g IR image Temp. h 70 i Iontophoresis device j Control Iontophoresis
Drug release (%)

50°C 60
50
(+) (-)
40
30 25oC Drug
Current
37oC Drug
20 Skin
Ag NW/SBS heater 0 20 40 60 80 ion
20°C Time (hour)

Fig. 3. (a) TEM image of microemulsion in hydrophilic gel (left). Cross-sectional fluorescent image of dye permeation through human skin (right). (b) Confocal images of solution-based
(left) and nanoparticle (NP)-based (right) nucleic acid delivery via transdermal route. (c) Temperature responsive drug release from nanogel. (d) Schematic illustration of pH-responsive
drug release. (e) Optical images of light responsive release (left). Drug release profile under visible light irradiation (right). (f) Schematic illustration of stretch-triggered drug release (left).
Accumulative drug release during 1000 cycles of finger flexion (right). (g) Infrared camera image of a wearable heater on the wrist. (h) Cumulative release of drugs from PNIPAM
microparticles embedded on heater before (25 °C) and after (37 °C) thermal stimulation. (i) Schematic illustration of iontophoresis treatment. (j) Optical (top) and fluorescent
(bottom) cross-sectional images of the pigskin taken after 1 h or 6 h of iontophoresis treatment.
Adapted and reproduced with permission from (a) Batheja et al. [55], Copyright 2011, Elsevier; (b) Desai et al. [63], Copyright 2013, Elsevier; (c) Carmona-Moran et al. [65], Copyright
2016, Elsevier; (d) Horev et al. [66], Copyright 2015, American Chemical Society; (e) Kim et al. [67], Copyright 2015, Wiley-VCH; (f) Di et al. [68], Copyright 2015, American Chemical
Society; (g) Choi et al. [69], Copyright 2015, American Chemical Society; (h) Bagherifard et al. [70], Copyright 2016, Wiley-VCH; (i) Kim et al. [24], Copyright 2016, American Chemical
Society; (j) Ogawa et al. [71], Copyright 2015, Wiley-VCH.

the drug solubility [1]. Presently, more benign chemical enhancers (e.g., drug from a transdermal patch. For example, drug-loaded hydrogels can
fatty acids, urea, and pyrrolidone) compared to skin irritants such as di- respond to the temperature change induced by light irradiation (Fig. 3e,
methyl sulfoxide (DMSO), dimethylformamide (DMF), and left) [67]. Along with the increase in cumulative delivery, drug transport
oxazolidinone are available to use [54]. can be easily triggered on and off by controlling the external stimuli
Emulsion is one of the major strategies in non-invasive transdermal (Fig. 3e, right). Alternatively, mechanical force can control the release
delivery because they solubilize a wide range of both lipophilic and hy- of the drug from transdermal patch. The patch is fabricated with a
drophilic drugs in a transdermal formulation (Fig. 3a) [55]. The absorp- drug-laden elastomer (Fig. 3f, left), which can be conformally attached
tion profile of an emulsion is determined by the droplet size, to the finger joints [68]. When the joints flex, the patch experiences lat-
composition, and surface charge. Particularly, reducing the size of vehi- eral strain and releases the drug (Fig. 3f, right).
cle particles to micrometer or nanometer level has been a popular strat- External devices, particularly in the wearable forms, can be integrat-
egy because smaller droplets can easily permeate the tight junctions of ed with drug-laden patches. The devices deliver a sufficient level of en-
the skin barrier [56,57]. Recently, nanomaterials have been studied as ergy to promote drug diffusion through the skin. Devices that operate
potential systems for the delivery and controlled release of various with heat and electricity are frequent drug-delivering locomotives. For
drugs (e.g., nucleic acid, antibacterial, and cancer drugs) [58–62]. For ex- example, local heat from a wearable heater (Fig. 3g) can enhance the
ample, drug-loaded nanoparticles (NPs) effectively overcome the epi- skin permeation of the drug [69]. When the heat is applied, solubility
dermal barrier and deliver an encapsulated cocktail of anti- and diffusivity of inorganic and organic drugs increase from the drug-
inflammatory siRNAs and capsaicin. In contrast, the delivery of siRNA- containing layer (Fig. 3h) [70]. The applied heat also increases the circu-
capsaicin cocktail in solution form was hindered by the stratum lation of body fluid, the permeability of blood vessel walls, and the rate-
corneum (Fig. 3b) [63]. The efficiency of drug delivery control can be en- limiting membrane permeability, which in turn increases the perme-
hanced by using nano-sized vehicles responsive to physicochemical ation of the transdermal drugs. For example, the effect of temperature
stimuli [64]. One example is the temperature-responsive nanogel that was evaluated in vitro by estimating the flux of transdermal fentanyl
significantly increases the release of the drug as the temperature ele- (a potent opioid analgesic) at temperatures of 32 and 37 °C. The flux
vates from room temperature to that of the outer skin (Fig. 3c) [65]. of the drug approximately doubled over the 5 °C increment in temper-
The pH-sensitive release of antibacterial drugs into the skin in response ature. Further studies indicate that a temperature change of 5 °C is nec-
to the acidic microenvironment of a virulent biofilm is another approach essary to cause a measurable enhancement in permeability between cell
for the stimuli-responsive drug release (Fig. 3d) [66]. membranes and transport of the drug.
Stimuli-responsive nano-vehicles are incorporated into patches to Iontophoresis uses an electrical current to enhance and control the
yield a synergetic efficacy in transdermal delivery. External stimuli penetration of charged drugs into tissues through the skin barrier (Fig.
(e.g., light, mechanical force, and magnetic field) promote not only a 3i) [24]. The dose is easily controlled by changing the magnitude and
higher rate of drug absorption but also a facile initiation and termination the duration of the stimulating current. Iontophoretic administration
of drug administration [58,67,68]. Visible light can trigger release of the of the drug can be applied to pain relief, diagnosis of inflammation,
H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45 39

chronic edema, cystic fibrosis, and cosmetic applications. A weak elec- level of energy at a designated location to penetrate the skin (Fig. 4c)
trical current below 500 μA cm−2 is applied to the skin area of a few cen- [80,81]. Gold nanorods absorb near-infrared light and generate an in-
timeter squares to prevent pain, irritation, or skin burns. The electrical tense heat, which can damage the outer layer of the skin. The pulsed
field increases the rate of drug transport compared to that of passive dif- laser only increases the temperature of the skin surface and causes
fusion-based methods (Fig. 3j) [71]. Recently, a cryoelectrophoresis less damage to the inner skin than continuous-wave laser does (Fig.
technique was introduced to increase the electrophoretic efficacy by ap- 4d) [82–84].
plying a strong current to locally frozen skin to eliminate the risk of skin Although RF ablation, sonophoresis, and laser ablation methods
burns [72]. However, the non-invasive transdermal delivery does not show reasonably high potentials, they need bulky and expensive equip-
drastically increase the efficiency of drug delivery, particularly for high ment and thus can only be performed at clinics. As an alternative,
molecular weight (e.g., proteins) and hydrophilic drugs. microneedles have attracted considerable attention because they can
transport drugs in a minimally invasive manner via the transdermal
2.3. Third generation: minimally invasive transdermal drug delivery route, and are relatively simple and cheap [74,85]. Microneedles are
easier to use and less painful to the skin than high-power energy modal-
As the limitations of non-invasive transdermal delivery have been ities. In addition, microneedles enable the controlled and sustained
recognized, an alternative approach of minimally invasive transdermal release of drugs depending on the patients' needs. Microneedles are di-
delivery is rapidly adopted. Minimal invasion of the skin involves vided into several types according to their shape and use including solid,
destroying the stratum corneum so that epidermal disruption is limited surface-coated, dissolvable, hollow-shaped, and smart microneedle
to clinically safe level while a large number of hydrophilic drugs and [86]. Solid microneedles are fabricated from hard materials such as sili-
macromolecules are more effectively delivered into the inner skin con and metals (Fig. 4e) [87–90]. The hardness of the materials enables
[73]. High power energy modalities (e.g., radiofrequency (RF), ultra- the solid microneedles to easily create micropores, which are the micro-
sound, and laser) and various types of microneedles are currently inlet through which drugs are transferred. The micropores are recov-
used to disrupt or puncture the skin and enhance drug delivery. Howev- ered a short period after the creation and, therefore, additional patches
er, destruction of the skin should be minimized and carefully controlled or drugs (e.g., diclofenac) can be used to extend the duration of the
to allow its rapid recovery [74]. A comparative study using ibuprofen as formed micropores.
a model drug indicates that the biodistribution and the bioavailability of Microneedles may also incorporate a drug coated onto the surface of
differently formulated microneedles can be significantly different, and the solid needle substrate (Fig. 4f) [91–93]. Since the coating layer de-
minimally invasive microneedles have advantages over non-invasive creases the physical strength and sharpness of needles, the drug load
iontophoresis in terms of bioavailability [75]. on the needle surface is limited to a small amount. Accordingly, coated
Disruption of the epidermis by high power energy broadens the microneedles are used for treatments where large amounts of drugs
range of drugs that can be delivered through the skin [73]. Because a sig- are not required to be delivered (e.g., vaccine delivery and cosmetic pro-
nificantly high level of energy is required to damage the outer layers of cedures) [94,95].
skin, it is necessary to carefully control the amplitude and exposure time Dissolvable microneedles are formulated from water-soluble and
of the applied energy to prevent unwanted damage to the inner skin biocompatible polymers. In dissolvable microneedles, drugs are embed-
layers. RF ablation uses a high-frequency electrical current (100– ded in the polymeric matrix and released as the matrix degrades or dis-
500 kHz) for localized heating of the outer skin layer (Fig. 4a) [76]. solves slowly in body fluids (Fig. 4g) [96–103]. The temporal drug
Sonophoresis uses ultrasound which generates heat and promotes release profile can be controlled from several seconds to several months
drug delivery into the skin [41,77,78]. Recently, double frequency ultra- by selecting appropriate polymers. However, there are also challenges
sound was used to enhance the skin permeability and shorten the treat- associated with this technique. For example, the use of ultraviolet
ment time compared to single frequency ultrasound (Fig. 4b) [79]. Laser (UV) irradiation or elevated temperature to cure the polymer for
ablation is also commonly used in cosmetics for procedures such as hair microneedles during the fabrication process can denature the drugs
removal, skin resurfacing, and acne treatment. The laser targets a high loaded in the polymeric precursor. The drug quantity that can be loaded

a Feedback Control
b Double frequency ultrasound c Ablation Laser poration d
RF energy US1
Ablated area CW-laser Pulsed laser
US2
Au NR

Skin 300 µm 300 µm

e Solid microneedle f Coated microneedle g Dissolvable microneedle h Hollow microneedle

2 mm

100 µm 100 µm 100 µm 100 µm 100 µm

Fig. 4. (a) Schematic illustration of radiofrequency (RF) ablation on skin. (b) Schematic illustration of double frequency ultrasonic treatment for transdermal drug delivery. (c) Histological
image of ablated skin after 30 min of delivery of sulforhodamine (left). Optical image of skin after laser poration (right). (d) Schematic illustration of gold nanorod (AuNR) assisted
photothermal ablation with continuous-wave (CW) (left) and pulsed laser (right). (e) SEM image of silicon-based solid microneedles. Optical image of solid microneedle array (inset).
(f) Optical microscopic image of sucrose-coated polylactic acid microneedles. Schematic illustration of coated microneedles (inset). (g) Optical image of dissolvable microneedles (left).
Fluorescence microscopic image of porcine skin after poration of microneedles (right). (h) SEM image of hollow microneedles.
Adapted and reproduced with permission from (a) Sintov et al. [76], Copyright 2003, Elsevier; (b) Schoellhammer et al. [79], Copyright 2012, Elsevier; (c) Lee et al. [80], Copyright 2011,
Elsevier and Weiss et al. [81], Copyright 2012, Elsevier; (d) Tang et al. [82], Copyright 2013, Elsevier; (e) Li et al. [90], Copyright 2010, Elsevier; (f) DeMuth et al. [92], Copyright 2013, Nature
Publishing Group; (g) Sullivan et al. [97], Copyright 2010, Nature Publishing Group; (h) Vazquez et al. [104], Copyright 2014, Elsevier.
40 H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45

into the polymeric matrix is also limited because a high percentage of the sensors subsequently gather information on the therapeutic efficacy
drug in the precursor mixture would undermine the mechanical of the drug. The synergetic performance of wearable devices and drug-
strength of the needles. delivering patches according to a complete feedback loop provides a
Hollow microneedles have a hollow core inside the needle through novel platform for personalized therapy.
which the drug solution flows (Fig. 4h) [104–107]. The drug solution To maximize the performance of device-assisted transdermal drug
can be delivered passively by diffusion or actively by pressure-driven delivery, it is crucial to safeguard a conformal contact between the de-
flow through the needle hole. A drug reservoir attached to the back of vice and the skin which stems from the stretchability and flexibility of
the microneedles releases the drug by the action of a pump. Hollow the device. The softness and deformability of the system minimize the
microneedles have a merit in controlling the amount of injected drug. local detachment and microscopic spaces between the device and the
However, one disadvantage of this approach is that additional equip- skin due to the curvilinear morphology of the skin and bodily move-
ment (e.g., drug reservoir and pump) assisting the microneedles is re- ments [16]. Conformal integration of a device onto the skin also pro-
quired for drug delivery [73]. motes precise measurements of the biological signals with minimal
With increasing variety of applicable drug candidates, microneedles motion-induced noise, enhancing the accuracy of wearable sensors
represent a major breakthrough in transdermal delivery in terms of ef- monitoring physiological (e.g., blood pressure, strain, and temperature)
ficiency and versatility. However, the fabrication process for mass pro- [6–15], electrophysiological (e.g., electrocardiogram and electroenceph-
duction is yet to be established. Furthermore, even the microneedles alogram) [16–21], and biochemical (e.g., pH, blood glucose, and oxygen
share the same limitation of transdermal patches in which the only levels) [22–27] signals from the skin. Based on the collected informa-
way to control the offset of drug release is to detach the patches. A tion, a co-integrated actuator controls the drug diffusion rate. This pro-
more dynamic control on the release profile (e.g., pulsatile, variable, cess is also enhanced by the conformal contact because a stable
and feedback-controlled) of the drug could be envisioned with incorpo- interface between the patch and the skin ensures homogeneous and ef-
ration of other technological elements. ficient energy transfer, and provides a uniform drug release profile
throughout the entire patch area [111,112].
3. Fourth generation: controlled and feedback-induced transdermal
drug delivery 3.1. Transdermal drug delivery based on physiological/electrophysiological
monitoring
Personalized therapy is distinguished from conventional medical
treatments by its ability to optimize the treatment based on each To diagnose the health condition of an individual, an assortment of
individual's pathophysiological conditions [49]. Establishment of per- physical and electrical cues must be collected first. The collection of pre-
sonalized therapy requires systematic control of the administered cise health information with high spatiotemporal resolution is impor-
dose based on an accurate real-time observation of the patient's physi- tant in controlling drug delivery. Since the surface of the skin is soft,
ological parameters to determine the progression of disease and efficacy curvilinear and movable, skin-mounted sensors necessitate physical
of the drug. In response to an increasing need for personalized treat- characteristics similar to those of the skin to obtain high-quality infor-
ment, the advanced transdermal delivery system empowered by soft mation without motion artifacts or background noises from the skin
bioelectronics has been spotlighted as a strategy for the next generation surface. Flexible and stretchable devices in ultrathin and serpentine
drug delivery method. structures facilitate a conformal contact with the skin, which is crucial
Rapid advances in soft and ultrathin devices in wearable forms have for high fidelity measurement of bodily signals [48–50]. Therefore, soft
facilitated the seamless integration of bioelectronic devices in a skin- wearable biosensors that are attached conformally to skin contours en-
mounted patch domain with unprecedented functionalities [108–110]. able the precise real-time monitoring of an individual's physiological
Specifically, sensors accurately measure the physiological [6–15], elec- biomarker without hindering bodily motions. Accurate diagnosis
trophysiological [16–21], and biochemical [22–27] signals, actuators based on the conformal contact also increases the efficacy of controlled
transfer energy to the drug-laden patch in a controlled manner, and drug release in response to physiological feedback.

a b c Patch Surface (°C) d 5 min/40 °C 60 min/40 °C


Strain gauge Heater 45
42
39
m-silica Skin
Heat Patch 36 Thermal
diffusion
TiO2 NM/Au NPs 33
Drug
RRAM 5 mm 40 nm Interface 100 µm 100 µm
30
Penetration depth (µm)

e f Iontophoresis g 3 cycles 12 cycles h 400


Iontophoresis
Skin Amplifiers
Memories + electrode

_
-0 300 Thermal diffusion
Control
200
- 100
100

- 200 (µm) 0
ECG Drugs 0 3 6 9 12
electrodes 5 mm 1 mm 30 nm # of stimulus

Fig. 5. (a) Optical image of a wearable electronic patch which can detect movement disorder and store detected signals. (b) Transdermal drug delivery using a wearable heater. Drug
loaded in mesoporous silica NPs (MSNs) is released by thermal stimulation. (c) Finite element modelling of the three-dimensional thermal profile of a heater on the patch and at the
interface between the patch and the human skin. (d) Cross-sectional fluorescence images show enhanced drug diffusion into pig skin via thermal actuation. (e) Skin-mounted device
used for heart-rate measurement, signal amplification and data storage. (f) Transdermal drug delivery using the iontophoresis. (g) Cross-sectional fluorescence images for the drug
(doxorubicin) diffusion into the mouse skin with different stimuli of iontophoresis; 3 cycles (left) and 12 cycles (right). (h) Stimulus number versus drug diffusion length into the
mouse skin with different actuators (black: none, blue: thermal actuator, and red: iontophoresis electrode).
Adapted and reproduced with permission from (a)–(d) Son et al. [3], Copyright 2014, Nature Publishing Group; (e) Kim et al. [19], Copyright 2016, AAAS; (f)–(h) Choi et al. [5], Copyright
2016, Wiley-VCH.
H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45 41

Physiological signals such as body temperature [6], and dynamic in- motifs based on bio-inspired interfaces provide sufficient adhesion to
formation including tremors [3,9,10] and cardiac movements [5,7,8,19] the skin so that the devices do not require adhesive glues, which are po-
are the key information required for diagnosing the health condition of tential skin irritants in the long-term. Based on the measured electrical
an individual. Collected signals can be used for patient-customized signals, the integrated drug actuators can efficiently deliver the loaded
transdermal drug delivery. For example, a multifunctional and wearable drug through the skin (Fig. 5f) [5]. For electrostatically charged drugs,
patch continuously monitors movement disorders using silicon mem- iontophoresis can serve as an efficient delivery method. In a study,
brane-based strain gauges, and stores the recorded information on the MSNs were used as drug delivery vehicles to prevent oxidation, dena-
activities and tremors in the memory module (Fig. 5a) [3]. Continuously turation, and unstimulated diffusion of drugs. Iontophoretic delivery ac-
operating sensors collect vital signals and the cumulative information is celerates the deep permeation of the drug into the skin, mediated by the
processed to operate a co-integrated drug delivery unit (Fig. 5b). Ac- charge repulsion between the electrodes and drugs (Fig. 5g). Iontopho-
cording to the patient's health condition, wearable drug actuators trig- retic actuation of drug-loaded MSNs enhanced the dermal penetration
ger drug delivery through the skin. To maximize the amount of loaded significantly compared to those of the control and thermal diffusion
drugs, mesoporous silica NPs (MSNs) serve as the drug delivery vehicles (Fig. 5h). Iontophoresis also has advantages over thermal diffusion by
in the patch because their large surface area allows greater drug adsorp- preventing low-temperature burns on the skin and thermal denatur-
tion. The programmable thermal actuators control the drug delivery ation of the loaded drugs.
through the skin by enhancing drug diffusion (Fig. 5c). The generated
heat degrades the binding of the MSNs and the loaded drugs, which sub- 3.2. Transdermal drug delivery based on biochemical signal monitoring
sequently diffuse transdermally (Fig. 5d). The integrated temperature
sensor conducts real-time monitoring to prevent any low-temperature Biochemical analysis of biofluids can provide ample information on
burns. an individual's health condition and disease progression [22–27]. For
Electrical signal is also an integral part of health monitoring that en- wearable devices, sweat is a frequently used specimen to monitor the
ables relevant information to be collected continuously. The time-re- physiological and biochemical conditions such as glucose level because
solved amplitude and wave pattern of electrophysiological signals it contains voluminous metabolic data (Fig. 6a) [113–116]. In accor-
such as those of an electromyogram, electroencephalogram, and elec- dance with the versatility of sweat analysis in disease diagnosis, wear-
trocardiogram reflect the real-time condition of the muscles, brain, able sweat sensors are developed to conveniently measure the
and heart, respectively. These signals can be detected from the outer analytes of interest.
skin noninvasively using wearable electrodes and a co-integrated am- Wearable and disposable chemical sensors are recent examples of
plifier (Fig. 5e) [19]. To investigate the reliable long-term monitoring colorimetric and electrochemical analyses. Colorimetric analysis is
of signals, studies have focused on the interface between wearable de- widely used for simple and disposable sensing instruments because
vice and biological tissues to establish a robust mechanical coupling the presence of biomarkers is easily confirmed by color changes (Fig.
for a highly sensitive and accurate biometric detection [5,8]. The design 6b) [117,118]. Although this method can be used to monitor various

a Sensor Glucose, b Colorimetric sweat sensor c Tattoo-type glucose sensor d


Lactate, ions

Smart
5 mm 5 mm wristband
Sweat duct Sweat

e Sweat glucose f Integrated Sweat glucose


Temp. Heater
monitoring patch patch system sensor
e-

Hand
pH Sens. Glu. Sens.
Sweat 5 mm [Wearable diabetes patch] [Sweat analysis] [Transdermal drug delivery]

8 35
g R.T. h Initial i j
Rel. release (a.u.)

T>Tc: transition Release No patch w/o drug


Blood Glu. (mM)

30 Dose1 Dose2
PCM 6 w/ heater
25
Left on 4 20
PVP/
drug drug 15 * *
2 **
elution
10 **
Left/right on 0 N=4
i ii iii iv v vi vii viii 5
0 2 4 6
2 mm Heater actuation Time (hr)

Fig. 6. (a) Schematic illustration of sweat monitoring system on the skin. Biomarkers such as glucose, lactate, and ions secrete from epithelial cells along the sweat ducts inside the skin,
reflecting a subject's physiological state. (b) Optical image of colorimetric sweat sensor mounted on the forearm. (c) Optical image of a sweat glucose monitoring tattoo device applied to a
human subject. (d) Optical image of smart wristband on subject's wrist, integrating the multiplexed sweat sensor array. (e) Optical image of multifunctional electrochemical devices on the
human skin with perspiration. (f) Schematic illustrations of operation process of skin-based diabetes monitoring and therapy system. (g) Schematic illustrations of drug-loaded
bioresorbable microneedles. The phase change material (PCM) coating prevents unwanted drug release before programmed thermal actuation. (h) Optical images of the stepwise
dissolution of the microneedles for drug dose control. (i) Multistage drug release profile by using three-channel thermal actuator for stepwise transdermal drug delivery. (j) Blood
glucose levels of the db/db mice for the treated groups (microneedles with the drugs) and control groups (without the patch, microneedle without the drugs).
Adapted and reproduced with permission from (a) Heikenfeld [113], Copyright 2016, Nature Publishing Group; (b) Koh et al. [117], Copyright 2016, AAAS; (c) Bandodkar et al. [114],
Copyright 2015, American Chemical Society; (d) Gao et al. [26], Copyright 2016, Nature Publishing Group; (e)–(h) Lee et al. [115], Copyright 2016, Nature Publishing Group; (i) and (j) Lee
et al. [116], Copyright 2017, AAAS.
42 H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45

biomarkers such as perspiration rate, total sweat loss, pH, chlorides, and microneedles (Fig. 6h). The amount of delivered drug can be controlled
lactates, it is difficult to accurately quantify the color changes to deter- by the area of the applied heating, which is determined by the integrat-
mine the concentration of the biomarkers. Wearable electrochemical ed wearable biosensors. Temperature sensors near the heater simulta-
sensors have been devised to quantifiably detect biomarkers in the neously monitor the temperature variation of the heaters to
sweat. The concentration of target biomarkers is directly measured sequentially control the drug release and prevent low-temperature
from the electrical signals of the redox reaction of the markers in a burns. By using additional PCMs combined with drugs and multichannel
short time. The wearable electrochemical sensors fabricated by screen heaters, the drug delivery system features a precisely controlled trans-
printing have simple structures and enable the facile measurement of dermal release of drugs at up to six different release amounts (Fig. 6i).
a single biomarker (Fig. 6c) [114]. An array of electrochemical sensors The effectiveness of the wearable microneedle system was demonstrat-
is also available for the simultaneous detection of multiple biomarkers ed in diabetic (db/db) mice by triggering controlled transcutaneous
(Fig. 6d) [26]. The information acquired from the sweat is wirelessly col- drug release using thermal actuators. Animal experiments with diabetic
lected and continuously stored in mobile devices for long-term health mice confirmed the efficacy of the microneedles in reducing blood glu-
monitoring. cose levels (Fig. 6j).
Accurate monitoring of the sweat glucose level is of primary interest
because of its direct correlation to the blood glucose level [119]. Further- 3.3. Smart microneedle systems
more, this correlation inspired the development of a skin-based diabe-
tes control system that combines sweat glucose detection with Smart microneedles represent the culminating point of advanced
microneedle-based drug delivery for minimally invasive blood glucose transdermal delivery since they incorporate the aforementioned ele-
regulation (Fig. 6e) [115,116]. Conventional blood glucose monitoring ments of sensors, actuators, and enhanced drug formulation into a com-
for diabetes inflicts pain and stress during the blood withdrawal and plete feedback loop of personalized therapy. Sensors monitor the
drug injection steps. Therefore, device-assisted glucose monitoring physiological conditions and trigger the medication, and actuators ma-
and feedback therapy could improve the quality of life of patients with nipulate the release profile and dose of the administered drug. Without
diabetes by managing blood glucose level more conveniently. The min- further medical intervention, automated microneedle patches continue
imally invasive glucose monitoring system is initiated by perspiration to respond to the progression of the disease until symptoms are allevi-
after the patch is applied (Fig. 6f, left). To measure the sweat glucose ated. Smart microneedles can provide a patient-friendly and feedback-
level more accurately, data collected from the glucose sensor are controlled drug delivery, which has not been accomplished with previ-
corrected by integrated pH, temperature, and humidity sensors. It en- ous microneedle systems. Microneedles that are responsive to external
sures that corrections are made for the activity variation of glucose ox- stimuli (e.g., heat, laser, and mechanical strain) provide an on/off trigger
idase, which is the primary detection moiety (Fig. 6f, middle). When for drug administration, which can be easily controlled by patients [44,
hyperglycemia occurs, the microneedles are triggered by the sensor sig- 68]. An example is the mechanically responsive microneedles whose
nal to transdermally administer the antidiabetic drug (Fig. 6f, right). drug release is modulated by lateral strain (Fig. 7a, left) [68]. The
In the case of conventional microneedles, it was difficult to control microneedles that release the drug payload following lateral pulling
the duration and amount of the drug dose released because the drug dif- were successfully tested in vivo (Fig. 7a, right). Smart NPs that are re-
fusion is irreversible once the microneedles are inserted into the skin. In sponsive to biochemical stimuli (e.g., pH change, glucose concentration,
addition, data to determine the drug delivery rate are unavailable. Re- and hypoxia) can also be loaded in microneedles (Fig. 7b, left) [46,120,
cent studies have demonstrated the prospect of controlled transdermal 121]. Microneedles facilitate the entry of NPs into the bloodstream, and
drug release based on real-time health monitoring using a wearable de- the delivered NPs respond to the patient's physiological state and re-
vice-assisted thermo-responsive microneedles system [115,116]. Phase lease the drugs accordingly. The concept has been demonstrated by
change materials (PCMs) protect the drug and dissolvable microneedle NPs containing glucose oxidase, which release insulin in response to a
matrix from biofluids below the critical melting temperature (Fig. 6g). high blood glucose concentration to modulate its level (Fig. 7b, right)
When multi-channel heaters initiate patterned thermal actuation, the [120]. Thrombin-responsive microneedle array can prevent the un-
PCM coating on the microneedles melts, and thereby, the transdermal wanted blood coagulation as thrombin-cleavable peptides interlinked
drug delivery proceeds by partial or total dissolution of the within hyaluronic acid hydrogel scaffolds trigger the drug release

a Microneedle b
Blood glu. (mg/dl)

Without strain With strain Glu. 600


F 500
400
300
Force 200 GRV(E,I)
Force GRV(0.5E,I)
100
GRV(I)
F 0 Insulin Blank
Skin 0 2 4 6 8 10 12
Drug release 200 µm Time (hr)

c 400 0 U/ml Thrombin d 100mg/dL 400mg/dL


Heparin rel. (a.u.)

0.5 U/ml 12
Insulin rel. (µg/ml)

300 1 U/ml
10
200 8
Skin
100 6
Disass
Thrombin -embly 4
0
2
0 1 2 3 4 12 Dual-sensitive Insignificant
Time (hr) 0
Heparin release d-GRP inflammation Time

Fig. 7. (a) Schematic illustration of strain-responsive microneedles (left). Optical image of stretch-triggered drug release on shaved dorsal skin of mouse (right). (b) SEM image of smart
NP-loaded microneedles (left). Schematic illustration of glucose-responsive drug release (inset). Changes in the level of blood glucose after treatment with microneedle patch (right). (c)
Schematic illustration of thrombin responsive microneedle array (left). Release profile of heparin at different thrombin concentrations (right). (d) Schematic illustration of hypoxia and
H2O2 dual-sensitive microneedles for delivery of insulin (left). Pulsatile release profile of insulin as a function of glucose concentrations (right).
Adapted and reproduced with permission from (a) Di et al. [68], Copyright 2015, Elsevier; American Chemical Society; (b) Yu et al. [120], Copyright 2012, National Academy of Sciences; (c)
Zhang et al. [122], Copyright 2017, Wiley-VCH; (d) Yu et al. [123] Copyright 2017, American Chemical Society.
H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45 43

against the increased level of thrombin in a controlled and on-demand [3] D. Son, J. Lee, S. Qiao, R. Ghaffari, J. Kim, J.E. Lee, C. Song, S.J. Kim, D.J. Lee, S.W. Jun, S.
Yang, M. Park, J. Shin, K. Do, M. Lee, K. Kang, C.S. Hwang, N. Lu, T. Hyeon, D.H. Kim,
manner (Fig. 7c, left) [122]. Release of heparin in the thrombin respon- Multifunctional wearable devices for diagnosis and therapy of movement disor-
sive hydrogels is promoted as the concentration of thrombin is in- ders, Nat. Nanotechnol. 9 (2014) 397–404.
creased (Fig. 7c, right). For an enhanced glucose-responsive delivery [4] M.K. Choi, I. Park, D.C. Kim, E. Joh, O.K. Park, J. Kim, M. Kim, C. Choi, J. Yang, K.W.
Cho, J.-H. Hwang, J.-M. Nam, T. Hyeon, J.H. Kim, D.-H. Kim, Thermally controlled,
of insulin, dual-responsive (hypoxia and H2O2) vesicles are embedded patterned graphene transfer printing for transparent and wearable electronic/op-
in the microneedle array (Fig. 7d, left) [123]. This microneedle array is toelectronic system, Adv. Funct. Mater. 25 (2015) 7109–7118.
highly sensitive to glucose concentrations and shows a pulsatile release [5] M.K. Choi, O.K. Park, C. Choi, S. Qiao, R. Ghaffari, J. Kim, D.J. Lee, M. Kim, W. Hyun,
S.J. Kim, H.J. Hwang, S.H. Kwon, T. Hyeon, N. Lu, D.H. Kim, Cephalopod-inspired
of insulin according to the glucose concentrations (Fig. 7d, right). The miniaturized suction cups for smart medical skin, Adv. Healthc. Mater. 5 (2016)
next-generation transdermal drug delivery systems are a considerable 80–87.
improvement over wearable devices that have a limited ability to detect [6] R.C. Webb, A.P. Bonifas, A. Behnaz, Y. Zhang, K.J. Yu, H. Cheng, M. Shi, Z. Bian, Z. Liu,
Y.S. Kim, W.H. Yeo, J.S. Park, J. Song, Y. Li, Y. Huang, A.M. Gorbach, J.A. Rogers, Ultra-
or treat disease conditions. Furthermore, the next-generation transder-
thin conformal devices for precise and continuous thermal characterization of
mal drug delivery represents the integration of multiple functionalities human skin, Nat. Mater. 12 (2013) 938–944.
in a single device. In addition, the interactive and complementary co-ap- [7] G. Schwartz, B.C. Tee, J. Mei, A.L. Appleton, D.H. Kim, H. Wang, Z. Bao, Flexible poly-
plication of sensing and therapy to enable patient-tailored transdermal mer transistors with high pressure sensitivity for application in electronic skin and
health monitoring, Nat. Commun. 4 (2013) 1859.
drug delivery will likely be available in the foreseeable future through [8] C. Pang, J.H. Koo, A. Nguyen, J.M. Caves, M.G. Kim, A. Chortos, K. Kim, P.J. Wang, J.B.
the next-generation systems. Tok, Z. Bao, Highly skin-conformal microhairy sensor for pulse signal amplification,
Adv. Mater. 27 (2015) 634–640.
[9] J. Kim, M. Lee, H.J. Shim, R. Ghaffari, H.R. Cho, D. Son, Y.H. Jung, M. Soh, C. Choi, S.
Jung, K. Chu, D. Jeon, S.T. Lee, J.H. Kim, S.H. Choi, T. Hyeon, D.H. Kim, Stretchable sil-
4. Conclusion
icon nanoribbon electronics for skin prosthesis, Nat. Commun. 5 (2014) 5747.
[10] S. Lim, D. Son, J. Kim, Y.B. Lee, J.-K. Song, S. Choi, D.J. Lee, J.H. Kim, M. Lee, T. Hyeon,
The skin, which has long been perceived as unsuitable for drug deliv- D.-H. Kim, Transparent and stretchable interactive human machine interface based
ery, currently widens the vista on the possibilities for drug delivery on patterned graphene heterostructures, Adv. Funct. Mater. 25 (2015) 375–383.
[11] S. Jung, J.H. Kim, J. Kim, S. Choi, J. Lee, I. Park, T. Hyeon, D.H. Kim, Reverse-micelle-
strategies with its physiological significance and convenience. Studies induced porous pressure-sensitive rubber for wearable human–machine inter-
to overcome the challenges of the skin barrier against drug permeation faces, Adv. Mater. 26 (2014) 4825–4830.
and improve the efficiency of transdermal drug delivery have shown [12] S. Jung, J. Lee, T. Hyeon, M. Lee, D.H. Kim, Fabric-based integrated energy devices
for wearable activity monitors, Adv. Mater. 26 (2014) 6329–6334.
considerable success as demonstrated by the first, second, and third [13] H. Yang, D. Qi, Z. Liu, B.K. Chandran, T. Wang, J. Yu, X. Chen, Soft thermal sensor
generations of transdermal delivery strategies. From simple patches to with mechanical adaptability, Adv. Mater. 28 (2016) 9175–9181.
intricate microneedles, diverse approaches to improve the efficiency [14] T. Wang, Y. Guo, P. Wan, X. Sun, H. Zhang, Z. Yu, X. Chen, A flexible transparent col-
orimetric wrist strap sensor, Nano 9 (2017) 869–874.
of drug delivery are still in progress. Amidst the rising popularity of [15] M. Kaltenbrunner, T. Sekitani, J. Reeder, T. Yokota, K. Kuribara, T. Tokuhara, M.
the transdermal drug delivery, the device-assisted transdermal delivery Drack, R. Schwodiauer, I. Graz, S. Bauer-Gogonea, S. Bauer, T. Someya, An ultra-
provides a new paradigm for drug delivery in response to the real-time lightweight design for imperceptible plastic electronics, Nature 499 (2013)
458–463.
progression of disease.
[16] D.-H. Kim, N. Lu, R. Ma, Y.-S. Kim, R.-H. Kim, S. Wang, J. Wu, S.M. Won, H. Tao, A.
The diagnosis of the health status of patients using wearable sensors Islam, Epidermal electronics, Science 333 (2011) 838–843.
and the drug administration that is finely and timely controlled by actu- [17] S. Xu, Y. Zhang, L. Jia, K.E. Mathewson, K.-I. Jang, J. Kim, H. Fu, X. Huang, P. Chava, R.
Wang, Soft microfluidic assemblies of sensors, circuits, and radios for the skin, Sci-
ators are combined with the detection of physiological changes by sen-
ence 344 (2014) 70–74.
sors to provide a complete feedback therapy. This strategy is expected to [18] K.-I. Jang, H.U. Chung, S. Xu, C.H. Lee, H. Luan, J. Jeong, H. Cheng, G.-T. Kim, S.Y. Han,
enhance the quality and effectiveness of medical therapy. Since the pre- J.W. Lee, Soft network composite materials with deterministic and bio-inspired de-
vious generations of the transdermal drug delivery technology have fo- signs, Nat. Commun. 6 (2015).
[19] J. Kim, D. Son, M. Lee, C. Song, J.-K. Song, J.H. Koo, D.J. Lee, H.J. Shim, J.H. Kim, M. Lee,
cused on maximizing the efficiency of delivery, this novel technological A wearable multiplexed silicon nonvolatile memory array using nanocrystal
platform complements the drawbacks of previous transdermal delivery charge confinement, Sci. Adv. 2 (2016), e1501101.
systems and even yields synergetic results when the most advanced [20] T. Sekitani, T. Yokota, K. Kuribara, M. Kaltenbrunner, T. Fukushima, Y. Inoue, M.
Sekino, T. Isoyama, Y. Abe, H. Onodera, Ultraflexible organic amplifier with bio-
transdermal patches and devices are combined altogether. compatible gel electrodes, Nat. Commun. 7 (2016).
Since the incorporation of wearable sensors and advanced transder- [21] J. Park, S. Choi, A.H. Janardhan, S.-Y. Lee, S. Raut, J. Soares, K. Shin, S. Yang, C. Lee, K.-
mal patch formulations is still in the early stage, there is a spacious room W. Kang, Electromechanical cardioplasty using a wrapped elasto-conductive epi-
cardial mesh, Sci. Transl. Med. 8 (2016) 344ra386.
for future research aimed at improving the wearable devices and rigor- [22] A.J. Bandodkar, V.W. Hung, W. Jia, G. Valdes-Ramirez, J.R. Windmiller, A.G.
ously optimizing the reliability of the system, as well as their application Martinez, J. Ramirez, G. Chan, K. Kerman, J. Wang, Tattoo-based potentiometric
to assorted disease models. Continued efforts to enhance the delivery of ion-selective sensors for epidermal pH monitoring, Analyst 138 (2013)
123–128.
drugs that are considered unsuitable for administration through the
[23] W. Jia, A.J. Bandodkar, G. Valdes-Ramirez, J.R. Windmiller, Z. Yang, J. Ramirez, G.
skin (e.g., peptides, nucleic acids, and macromolecules) must also be Chan, J. Wang, Electrochemical tattoo biosensors for real-time noninvasive lactate
carried out to broaden the library of transdermally applicable drugs. Al- monitoring in human perspiration, Anal. Chem. 85 (2013) 6553–6560.
[24] J. Kim, I. Jeerapan, S. Imani, T.N. Cho, A. Bandodkar, S. Cinti, P.P. Mercier, J. Wang,
though the patient-customized transdermal drug delivery still requires
Noninvasive alcohol monitoring using a wearable tattoo-based iontophoretic-bio-
much development, this approach holds great promise for the conve- sensing system, ACS Sens. 1 (2016) 1011–1019.
nient management of diseases in daily life and a novel paradigm for [25] D.P. Rose, M.E. Ratterman, D.K. Griffin, L. Hou, N. Kelley-Loughnane, R.R. Naik, J.A.
healthcare in the future. Hagen, I. Papautsky, J.C. Heikenfeld, Adhesive RFID sensor patch for monitoring
of sweat electrolytes, IEEE Trans. Biomed. Eng. 62 (2015) 1457–1465.
[26] W. Gao, S. Emaminejad, H.Y. Nyein, S. Challa, K. Chen, A. Peck, H.M. Fahad, H. Ota,
H. Shiraki, D. Kiriya, D.H. Lien, G.A. Brooks, R.W. Davis, A. Javey, Fully integrated
Acknowledgement wearable sensor arrays for multiplexed in situ perspiration analysis, Nature 529
(2016) 509–514.
[27] W. Gao, H.Y.Y. Nyein, Z. Shahpar, H.M. Fahad, K. Chen, S. Emaminejad, Y. Gao, L.-C.
This work was supported by IBS-R006-D1, and IBS-R006-A1.
Tai, H. Ota, E. Wu, J. Bullock, Y. Zeng, D.-H. Lien, A. Javey, Wearable microsensor
array for multiplexed heavy metal monitoring of body fluids, ACS Sens. 1 (2016)
References 866–874.
[28] H. Rein, Experimental electroendosmotic studies on living human skin, Z. Biol. 81
[1] M.R. Prausnitz, R. Langer, Transdermal drug delivery, Nat. Biotechnol. 26 (2008) (1924) 125–140.
1261–1268. [29] R.J. Scheuplein, I.H. Blank, Permeability of the skin, Physiol. Rev. 51 (1971)
[2] A.W. Oosten, J.A. Abrantes, S. Jonsson, P. de Bruijn, E.J. Kuip, A. Falcao, C.C. van der 702–747.
Rijt, R.H. Mathijssen, Treatment with subcutaneous and transdermal fentanyl: re- [30] M.P. Cramer, S.R. Saks, Translating safety, efficacy and compliance into economic
sults from a population pharmacokinetic study in cancer patients, Eur. J. Clin. value for controlled release dosage forms, PharmacoEconomics 5 (1994) 482–504.
Pharmacol. 72 (2016) 459–467. [31] M.E. Lane, Skin penetration enhancers, Int. J. Pharm. 447 (2013) 12–21.
44 H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45

[32] A. Alexander, S. Dwivedi, T.K. Giri Ajazuddin, S. Saraf, S. Saraf, D.K. Tripathi, Ap- [60] S. Marepally, C.H. Boakye, A.R. Patel, C. Godugu, R. Doddapaneni, P.R. Desai, M.
proaches for breaking the barriers of drug permeation through transdermal drug Singh, Topical administration of dual siRNAs using fusogenic lipid nanoparticles
delivery, J. Control. Release 164 (2012) 26–40. for treating psoriatic-like plaques, Nanomedicine 9 (2014) 2157–2174.
[33] T.W. Prow, J.E. Grice, L.L. Lin, R. Faye, M. Butler, W. Becker, E.M. Wurm, C. Yoong, [61] D. Zheng, D.A. Giljohann, D.L. Chen, M.D. Massich, X.-Q. Wang, H. Iordanov, C.A.
T.A. Robertson, H.P. Soyer, M.S. Roberts, Nanoparticles and microparticles for skin Mirkin, A.S. Paller, Topical delivery of siRNA-based spherical nucleic acid nanopar-
drug delivery, Adv. Drug Deliv. Rev. 63 (2011) 470–491. ticle conjugates for gene regulation, Proc. Natl. Acad. Sci. U. S. A. 109 (2012)
[34] S. Ge, Y. Lin, H. Lu, Q. Li, J. He, B. Chen, C. Wu, Y. Xu, Percutaneous delivery of 11975–11980.
econazole using microemulsion as vehicle: formulation, evaluation and vesicle- [62] D. Son, J. Lee, D.J. Lee, R. Ghaffari, S. Yun, S.J. Kim, J.E. Lee, H.R. Cho, S. Yoon, S. Yang,
skin interaction, Int. J. Pharm. 465 (2014) 120–131. Bioresorbable electronic stent integrated with therapeutic nanoparticles for
[35] F. Lai, R. Pireddu, F. Corrias, A.M. Fadda, D. Valenti, E. Pini, C. Sinico, endovascular diseases, ACS Nano 9 (2015) 5937–5946.
Nanosuspension improves tretinoin photostability and delivery to the skin, Int. J. [63] P.R. Desai, S. Marepally, A.R. Patel, C. Voshavar, A. Chaudhuri, M. Singh, Topical de-
Pharm. 458 (2013) 104–109. livery of anti-TNFalpha siRNA and capsaicin via novel lipid-polymer hybrid nano-
[36] Y.S. Elnaggar, W.M. El-Refaie, M.A. El-Massik, O.Y. Abdallah, Lecithin-based nano- particles efficiently inhibits skin inflammation in vivo, J. Control. Release 170
structured gels for skin delivery: an update on state of art and recent applications, (2013) 51–63.
J. Control. Release 180 (2014) 10–24. [64] D. Ling, H. Xia, W. Park, M.J. Hackett, C. Song, K. Na, K.M. Hui, T. Hyeon, pH-sensitive
[37] F. Teodorescu, G. Queniat, C. Foulon, M. Lecoeur, A. Barras, S. Boulahneche, M.S. nanoformulated triptolide as a targeted therapeutic strategy for hepatocellular car-
Medjram, T. Hubert, A. Abderrahmani, R. Boukherroub, S. Szunerits, Transdermal cinoma, ACS Nano 8 (2014) 8027–8039.
skin patch based on reduced graphene oxide: a new approach for photothermal [65] C.A. Carmona-Moran, O. Zavgorodnya, A.D. Penman, E. Kharlampieva, S.L. Bridges
triggered permeation of ondansetron across porcine skin, J. Control. Release 245 Jr., R.W. Hergenrother, J.A. Singh, T.M. Wick, Development of gellan gum contain-
(2017) 137–146. ing formulations for transdermal drug delivery: component evaluation and con-
[38] J. Cazares-Delgadillo, A. Ganem-Rondero, V. Merino, Y.N. Kalia, Controlled trans- trolled drug release using temperature responsive nanogels, Int. J. Pharm. 509
dermal iontophoresis for poly-pharmacotherapy: simultaneous delivery of (2016) 465–476.
granisetron, metoclopramide and dexamethasone sodium phosphate in vitro and [66] B. Horev, M.I. Klein, G. Hwang, Y. Li, D. Kim, H. Koo, D.S. Benoit, pH-activated nano-
in vivo, Eur. J. Pharm. Sci. 85 (2016) 31–38. particles for controlled topical delivery of farnesol to disrupt oral biofilm virulence,
[39] T.A. Pereira, D.N. Ramos, R.F. Lopez, Hydrogel increases localized transport regions ACS Nano 9 (2015) 2390.
and skin permeability during low frequency ultrasound treatment, Sci Rep 7 [67] H. Kim, H. Lee, K.Y. Seong, E. Lee, S.Y. Yang, J. Yoon, Visible light-triggered on-de-
(2017) 44236. mand drug release from hybrid hydrogels and its application in transdermal
[40] J. Kim, J.H. Jang, J.H. Lee, J.K. Choi, W.R. Park, I.H. Bae, J. Bae, J.W. Park, Enhanced patches, Adv. Healthc. Mater. 4 (2015) 2071–2077.
topical delivery of small hydrophilic or lipophilic active agents and epidermal [68] J. Di, S. Yao, Y. Ye, Z. Cui, J. Yu, T.K. Ghosh, Y. Zhu, Z. Gu, Stretch-triggered drug de-
growth factor by fractional radiofrequency microporation, Pharm. Res. 29 (2012) livery from wearable elastomer films containing therapeutic depots, ACS Nano 9
2017–2029. (2015) 9407–9415.
[41] C.M. Schoellhammer, S. Srinivasan, R. Barman, S.H. Mo, B.E. Polat, R. Langer, [69] S. Choi, J. Park, W. Hyun, J. Kim, J. Kim, Y.B. Lee, C. Song, H.J. Hwang, J.H. Kim, T.
D. Blankschtein, Applicability and safety of dual-frequency ultrasonic treat- Hyeon, Stretchable heater using ligand-exchanged silver nanowire nanocomposite
ment for the transdermal delivery of drugs, J. Control. Release 202 (2015) for wearable articular thermotherapy, ACS Nano 9 (2015) 6626–6633.
93–100. [70] S. Bagherifard, A. Tamayol, P. Mostafalu, M. Akbari, M. Comotto, N. Annabi, M.
[42] M. Haedersdal, A.M. Erlendsson, U. Paasch, R.R. Anderson, Translational medicine Ghaderi, S. Sonkusale, M.R. Dokmeci, A. Khademhosseini, Dermal patch with inte-
in the field of ablative fractional laser (AFXL)-assisted drug delivery: a critical re- grated flexible heater for on demand drug delivery, Adv. Healthc. Mater. 5 (2016)
view from basics to current clinical status, J. Am. Acad. Dermatol. 74 (2016) 175–184.
981–1004. [71] Y. Ogawa, K. Kato, T. Miyake, K. Nagamine, T. Ofuji, S. Yoshino, M. Nishizawa, Or-
[43] W.R. Lee, S.C. Shen, I.A. Aljuffali, Y.C. Li, J.Y. Fang, Impact of different vehicles for ganic transdermal iontophoresis patch with built-in biofuel cell, Adv. Healthc.
laser-assisted drug permeation via skin: full-surface versus fractional ablation, Mater. 4 (2015) 506–510.
Pharm. Res. 31 (2014) 382–393. [72] A. Aloisi, M. Matera, R. Potenza, G. Santoro, C. Tuve, A. Messina, Cryoelectrophoresis:
[44] M.C. Chen, Z.W. Lin, M.H. Ling, Near-infrared light-activatable microneedle system painless administration of drugs through a suitable association of thermal and electri-
for treating superficial tumors by combination of chemotherapy and photothermal cal techniques, AIP Conference Proceedings, AIP 2000, pp. 19–22.
therapy, ACS Nano 10 (2016) 93–101. [73] T. Gratieri, I. Alberti, M. Lapteva, Y.N. Kalia, Next generation intra- and transdermal
[45] S. Hirobe, H. Azukizawa, T. Hanafusa, K. Matsuo, Y.S. Quan, F. Kamiyama, I. therapeutic systems: using non- and minimally-invasive technologies to increase
Katayama, N. Okada, S. Nakagawa, Clinical study and stability assessment of a drug delivery into and across the skin, Eur. J. Pharm. Sci. 50 (2013) 609–622.
novel transcutaneous influenza vaccination using a dissolving microneedle [74] K. van der Maaden, W. Jiskoot, J. Bouwstra, Microneedle technologies for (trans)-
patch, Biomaterials 57 (2015) 50–58. dermal drug and vaccine delivery, J. Control. Release 161 (2012) 645–655.
[46] C. Wang, Y. Ye, G.M. Hochu, H. Sadeghifar, Z. Gu, Enhanced cancer immunotherapy [75] J. Kennedy, E. Larrañeta, M.T. McCrudden, C.M. McCrudden, A.J. Brady, S.J. Fallows, H.O.
by microneedle patch-assisted delivery of anti-PD1 antibody, Nano Lett. 16 (2016) McCarthy, A. Kissenpfennig, R.F. Donnelly, In vivo studies investigating biodistribution
2334–2340. of nanoparticle-encapsulated rhodamine B delivered via dissolving microneedles,
[47] C. Edens, N.C. Dybdahl-Sissoko, W.C. Weldon, M.S. Oberste, M.R. Prausnitz, J. Control. Release (2017) http://dx.doi.org/10.1016/j.jconrel.2017.04.022.
Inactivated polio vaccination using a microneedle patch is immunogenic in the [76] A.C. Sintov, I. Krymberk, D. Daniel, T. Hannan, Z.E. Sohn, G. Levin, Radiofrequency-
rhesus macaque, Vaccine 33 (2015) 4683–4690. driven skin microchanneling as a new way for electrically assisted transdermal de-
[48] T. Someya, Z. Bao, G.G. Malliaras, The rise of plastic bioelectronics, Nature 540 livery of hydrophilic drugs, J. Control. Release 89 (2003) 311–320.
(2016) 379–385. [77] B.E. Polat, D. Hart, R. Langer, D. Blankschtein, Ultrasound-mediated transdermal
[49] S. Choi, H. Lee, R. Ghaffari, T. Hyeon, D.H. Kim, Recent advances in flexible and drug delivery: mechanisms, scope, and emerging trends, J. Control. Release 152
stretchable bio-electronic devices integrated with nanomaterials, Adv. Mater. 28 (2011) 330–348.
(2016) 4203–4218. [78] S. Mitragotri, D. Blankschtein, R. Langer, Ultrasound-mediated transdermal protein
[50] J. Kim, R. Ghaffari, D.-H. Kim, The quest for miniaturized soft bioelectronic devices, delivery, Science 269 (1995) 850.
Nat. Biomed. Eng. 1 (2017) 0049. [79] C.M. Schoellhammer, B.E. Polat, J. Mendenhall, R. Maa, B. Jones, D.P. Hart, R.
[51] J. Yang, M.K. Choi, D.H. Kim, T. Hyeon, Designed assembly and integration of colloi- Langer, D. Blankschtein, Rapid skin permeabilization by the simultaneous ap-
dal nanocrystals for device applications, Adv. Mater. 28 (2016) 1176–1207. plication of dual-frequency, high-intensity ultrasound, J. Control. Release 163
[52] D.W. Sifton, Physicians' Desk Reference, Medical Economics/Thomson Healthcare, (2012) 154–160.
2002. [80] J.W. Lee, P. Gadiraju, J.H. Park, M.G. Allen, M.R. Prausnitz, Microsecond thermal ab-
[53] M.R. Prausnitz, S. Mitragotri, R. Langer, Current status and future potential of trans- lation of skin for transdermal drug delivery, J. Control. Release 154 (2011) 58–68.
dermal drug delivery, Nat. Rev. Drug Discov. 3 (2004) 115–124. [81] R. Weiss, M. Hessenberger, S. Kitzmuller, D. Bach, E.E. Weinberger, W.D.
[54] V. Mathur, Y. Satrawala, M. Rajput, Physical and chemical penetration enhancers in Krautgartner, C. Hauser-Kronberger, B. Malissen, C. Boehler, Y.N. Kalia, J.
transdermal drug delivery system, Asian J. Pharm. 4 (2010) 173. Thalhamer, S. Scheiblhofer, Transcutaneous vaccination via laser microporation, J.
[55] P. Batheja, L. Sheihet, J. Kohn, A.J. Singer, B. Michniak-Kohn, Topical drug delivery Control. Release 162 (2012) 391–399.
by a polymeric nanosphere gel: formulation optimization and in vitro and in [82] H. Tang, H. Kobayashi, Y. Niidome, T. Mori, Y. Katayama, T. Niidome, CW/pulsed
vivo skin distribution studies, J. Control. Release 149 (2011) 159–167. NIR irradiation of gold nanorods: effect on transdermal protein delivery mediated
[56] H. Chen, X. Chang, T. Weng, X. Zhao, Z. Gao, Y. Yang, H. Xu, X. Yang, A study of by photothermal ablation, J. Control. Release 171 (2013) 178–183.
microemulsion systems for transdermal delivery of triptolide, J. Control. Release [83] S. Ramadan, L. Guo, Y. Li, B. Yan, W. Lu, Hollow copper sulfide nanoparticle-medi-
98 (2004) 427–436. ated transdermal drug delivery, Small 8 (2012) 3143–3150.
[57] P.L. Honeywell-Nguyen, J.A. Bouwstra, Vesicles as a tool for transdermal and der- [84] H.S. Jung, W.H. Kong, D.K. Sung, M.-Y. Lee, S.E. Beack, D.H. Keum, K.S. Kim, S.H. Yun,
mal delivery, Drug Discov. Today Technol. 2 (2005) 67–74. S.K. Hahn, Nanographene oxide-hyaluronic acid conjugate for photothermal abla-
[58] H. Lee, Y. Lee, C. Song, H.R. Cho, R. Ghaffari, T.K. Choi, K.H. Kim, Y.B. Lee, D. Ling, H. tion therapy of skin cancer, ACS Nano 8 (2014) 260–268.
Lee, S.J. Yu, S.H. Choi, T. Hyeon, D.H. Kim, An endoscope with integrated transpar- [85] Y.C. Kim, J.H. Park, M.R. Prausnitz, Microneedles for drug and vaccine delivery, Adv.
ent bioelectronics and theranostic nanoparticles for colon cancer treatment, Nat. Drug Deliv. Rev. 64 (2012) 1547–1568.
Commun. 6 (2015) 10059. [86] T.-M. Tuan-Mahmood, M.T. McCrudden, B.M. Torrisi, E. McAlister, M.J. Garland,
[59] J. Shin, K. Shin, H. Lee, J.B. Nam, J.E. Jung, J.H. Ryu, J.H. Han, K.D. Suh, Y.J. Kim, J. Shim, T.R.R. Singh, R.F. Donnelly, Microneedles for intradermal and transdermal drug de-
J. Kim, S.H. Han, K. Char, Y.K. Kim, J.H. Chung, M.J. Lee, B.C. Kang, J.W. Kim, Non-in- livery, Eur. J. Pharm. Sci. 50 (2013) 623–637.
vasive transdermal delivery route using electrostatically interactive biocompatible [87] K. Lee, H.C. Lee, D.S. Lee, H. Jung, Drawing lithography: three-dimensional fabrica-
nanocapsules, Adv. Mater. 22 (2010) 739–743. tion of an ultrahigh-aspect-ratio microneedle, Adv. Mater. 22 (2010) 483–486.
H. Lee et al. / Advanced Drug Delivery Reviews 127 (2018) 35–45 45

[88] J.B. Carey, F.E. Pearson, A. Vrdoljak, M.G. McGrath, A.M. Crean, P.T. Walsh, T. Doody, delivery in human skin using a hollow microneedle device, J. Control. Release
C. O'Mahony, A.V. Hill, A.C. Moore, Microneedle array design determines the induc- (2017) http://dx.doi.org/10.1016/j.jconrel.2017.02.028.
tion of protective memory CD8+ T cell responses induced by a recombinant live [106] P. Schipper, K. van der Maaden, S. Romeijn, C. Oomens, G. Kersten, W. Jiskoot, J.
malaria vaccine in mice, PLoS One 6 (2011), e22442. Bouwstra, Repeated fractional intradermal dosing of an inactivated polio vaccine
[89] Z. Ding, F.J. Verbaan, M. Bivas-Benita, L. Bungener, A. Huckriede, D.J. van den Berg, by a single hollow microneedle leads to superior immune responses, J. Control. Re-
G. Kersten, J.A. Bouwstra, Microneedle arrays for the transcutaneous immunization lease 242 (2016) 141–147.
of diphtheria and influenza in BALB/c mice, J. Control. Release 136 (2009) 71–78. [107] H. Jun, M.R. Han, N.G. Kang, J.H. Park, J.H. Park, Use of hollow microneedles for
[90] W.Z. Li, M.R. Huo, J.P. Zhou, Y.Q. Zhou, B.H. Hao, T. Liu, Y. Zhang, Super-short solid targeted delivery of phenylephrine to treat fecal incontinence, J. Control. Release
silicon microneedles for transdermal drug delivery applications, Int. J. Pharm. 389 207 (2015) 1–6.
(2010) 122–129. [108] M.K. Choi, J. Yang, K. Kang, D.C. Kim, C. Choi, C. Park, S.J. Kim, S.I. Chae, T.H. Kim, J.H.
[91] H.S. Gill, M.R. Prausnitz, Coated microneedles for transdermal delivery, J. Control. Kim, T. Hyeon, D.H. Kim, Wearable red-green-blue quantum dot light-emitting
Release 117 (2007) 227–237. diode array using high-resolution intaglio transfer printing, Nat. Commun. 6
[92] P.C. DeMuth, A.V. Li, P. Abbink, J. Liu, H. Li, K.A. Stanley, K.M. Smith, C.L. Lavine, M.S. (2015) 7149.
Seaman, J.A. Kramer, A.D. Miller, W. Abraham, H. Suh, J. Elkhader, P.T. Hammond, [109] D. Son, J.H. Koo, J.-K. Song, J. Kim, M. Lee, H.J. Shim, M. Park, M. Lee, J.H. Kim, D.-H.
D.H. Barouch, D.J. Irvine, Vaccine delivery with microneedle skin patches in nonhu- Kim, Stretchable carbon nanotube charge-trap floating-gate memory and logic de-
man primates, Nat. Biotechnol. 31 (2013) 1082–1085. vices for wearable electronics, ACS Nano 9 (2015) 5585–5593.
[93] G.J. Fernando, J. Zhang, H.-I. Ng, O.L. Haigh, S.R. Yukiko, M.A. Kendall, Influenza nu- [110] T. Yokota, P. Zalar, M. Kaltenbrunner, H. Jinno, N. Matsuhisa, H. Kitanosako, Y.
cleoprotein DNA vaccination by a skin targeted, dry coated, densely packed micro- Tachibana, W. Yukita, M. Koizumi, T. Someya, Ultraflexible organic photonic skin,
projection array (Nanopatch) induces potent antibody and CD8+ T cell responses, Sci. Adv. 2 (2016), e1501856.
J. Control. Release 237 (2016) 35–41. [111] H. Wang, G. Pastorin, C. Lee, Toward self-powered wearable adhesive skin patch
[94] A. Vrdoljak, M.G. McGrath, J.B. Carey, S.J. Draper, A.V. Hill, C. O'Mahony, A.M. Crean, with bendable microneedle array for transdermal drug delivery, Adv. Sci. 3
A.C. Moore, Coated microneedle arrays for transcutaneous delivery of live virus (2016) 1500441.
vaccines, J. Control. Release 159 (2012) 34–42. [112] H. Tao, S.W. Hwang, B. Marelli, B. An, J.E. Moreau, M. Yang, M.A. Brenckle, S. Kim,
[95] Y. Chen, B.Z. Chen, Q.L. Wang, X. Jin, X.D. Guo, Fabrication of coated polymer D.L. Kaplan, J.A. Rogers, F.G. Omenetto, Silk-based resorbable electronic devices
microneedles for transdermal drug delivery, J. Control. Release (2017) http://dx. for remotely controlled therapy and in vivo infection abatement, Proc. Natl. Acad.
doi.org/10.1016/j.jconrel.2017.03.383. Sci. U. S. A. 111 (2014) 17385–17389.
[96] J.W. Lee, J.H. Park, M.R. Prausnitz, Dissolving microneedles for transdermal drug [113] J. Heikenfeld, Technological leap for sweat sensing, Nature 529 (2016) 475–476.
delivery, Biomaterials 29 (2008) 2113–2124. [114] A.J. Bandodkar, W. Jia, C. Yardimci, X. Wang, J. Ramirez, J. Wang, Tattoo-based non-
[97] S.P. Sullivan, D.G. Koutsonanos, M. Del Pilar Martin, J.W. Lee, V. Zarnitsyn, S.O. Choi, invasive glucose monitoring: a proof-of-concept study, Anal. Chem. 87 (2015)
N. Murthy, R.W. Compans, I. Skountzou, M.R. Prausnitz, Dissolving polymer 394–398.
microneedle patches for influenza vaccination, Nat. Med. 16 (2010) 915–920. [115] H. Lee, T.K. Choi, Y.B. Lee, H.R. Cho, R. Ghaffari, L. Wang, H.J. Choi, T.D. Chung, N. Lu,
[98] A. Vrdoljak, E.A. Allen, F. Ferrara, N.J. Temperton, A.M. Crean, A.C. Moore, Induction T. Hyeon, S.H. Choi, D.H. Kim, A graphene-based electrochemical device with
of broad immunity by thermostabilised vaccines incorporated in dissolvable thermoresponsive microneedles for diabetes monitoring and therapy, Nat.
microneedles using novel fabrication methods, J. Control. Release 225 (2016) Nanotechnol. 11 (2016) 566–572.
192–204. [116] H. Lee, C. Song, Y.S. Hong, M.S. Kim, H.R. Cho, T. Kang, K. Shin, S.H. Choi, T. Hyeon,
[99] V. Bachy, C. Hervouet, P.D. Becker, L. Chorro, L.M. Carlin, S. Herath, T. Papagatsias, D.-H. Kim, Wearable/disposable sweat-based glucose monitoring device with mul-
J.B. Barbaroux, S.J. Oh, A. Benlahrech, T. Athanasopoulos, G. Dickson, S. Patterson, tistage transdermal drug delivery module, Sci. Adv. 3 (2017), e1601314.
S.Y. Kwon, F. Geissmann, L.S. Klavinskis, Langerin negative dendritic cells promote [117] A. Koh, D. Kang, Y. Xue, S. Lee, R.M. Pielak, J. Kim, T. Hwang, S. Min, A. Banks, P.
potent CD8+ T-cell priming by skin delivery of live adenovirus vaccine Bastien, A soft, wearable microfluidic device for the capture, storage, and colori-
microneedle arrays, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 3041–3046. metric sensing of sweat, Sci. Transl. Med. 8 (2016) 366ra165.
[100] E. Korkmaz, E.E. Friedrich, M.H. Ramadan, G. Erdos, A.R. Mathers, O.B. Ozdoganlar, [118] J. Choi, D. Kang, S. Han, S.B. Kim, J.A. Rogers, Thin, soft, skin-mounted microfluidic
N.R. Washburn, L.D. Falo Jr., Tip-loaded dissolvable microneedle arrays effectively networks with capillary bursting valves for chrono-sampling of sweat, Adv.
deliver polymer-conjugated antibody inhibitors of tumor-necrosis-factor-alpha Healthc. Mater. 6 (2017).
into human skin, J. Pharm. Sci. 105 (2016) 3453–3457. [119] J. Moyer, D. Wilson, I. Finkelshtein, B. Wong, R. Potts, Correlation between sweat
[101] Z. Zhu, X. Ye, Z. Ku, Q. Liu, C. Shen, H. Luo, H. Luan, C. Zhang, S. Tian, C. Lim, Z. glucose and blood glucose in subjects with diabetes, Diabetes Technol. Ther. 14
Huang, H. Wang, Transcutaneous immunization via rapidly dissolvable (2012) 398–402.
microneedles protects against hand-foot-and-mouth disease caused by enterovi- [120] J. Yu, Y. Zhang, Y. Ye, R. DiSanto, W. Sun, D. Ranson, F.S. Ligler, J.B. Buse, Z. Gu,
rus 71, J. Control. Release 243 (2016) 291–302. Microneedle-array patches loaded with hypoxia-sensitive vesicles provide fast
[102] M.T. McCrudden, A.Z. Alkilani, C.M. McCrudden, E. McAlister, H.O. McCarthy, A.D. glucose-responsive insulin delivery, Proc. Natl. Acad. Sci. U. S. A. 112 (2015)
Woolfson, R.F. Donnelly, Design and physicochemical characterisation of novel dis- 8260–8265.
solving polymeric microneedle arrays for transdermal delivery of high dose, low [121] Y. Ye, J. Yu, C. Wang, N.Y. Nguyen, G.M. Walker, J.B. Buse, Z. Gu, Microneedles inte-
molecular weight drugs, J. Control. Release 180 (2014) 71–80. grated with pancreatic cells and synthetic glucose-signal amplifiers for smart insu-
[103] A.P. Raphael, T.W. Prow, M.L. Crichton, X. Chen, G.J. Fernando, M.A. Kendall, lin delivery, Adv. Mater. 28 (2016) 3115–3121.
Targeted, needle-free vaccinations in skin using multilayered, densely-packed dis- [122] Y. Zhang, J. Yu, J. Wang, N.J. Hanne, Z. Cui, C. Qian, C. Wang, H. Xin, J.H. Cole, C.M.
solving microprojection arrays, Small 6 (2010) 1785–1793. Gallippi, Thrombin-responsive transcutaneous patch for auto-anticoagulant regu-
[104] P. Vazquez, G. Herzog, C. O'Mahony, J. O'Brien, J. Scully, A. Blake, C. O'Mathuna, P. lation, Adv. Mater. 29 (2017).
Galvin, Microscopic gel–liquid interfaces supported by hollow microneedle array [123] J. Yu, C. Qian, Y. Zhang, Z. Cui, Y. Zhu, Q. Shen, F.S. Ligler, J.B. Buse, Z. Gu, Hypoxia
for voltammetric drug detection, Sensors Actuators B Chem. 201 (2014) 572–578. and H2O2 dual-sensitive vesicles for enhanced glucose-responsive insulin delivery,
[105] M. Dul, M. Stefanidou, P. Porta, J. Serve, C. O'Mahony, B. Malissen, S. Henri, Y. Levin, Nano Lett. 17 (2017) 733–739.
E. Kochba, F.S. Wong, C. Dayan, S.A. Coulman, J.C. Birchall, Hydrodynamic gene

You might also like