Combinatorial microneedle patch with tunable release
kinetics and dual fast-deep/sustained release capabilities
Journal:
Journal of Materials Chemistry B
Manuscript ID
TB-COM-01-2021-000141.R1
Article Type:
Communication
Date Submitted by the
Author:
11-Feb-2021
Complete List of Authors:
Lopez-Ramirez, Miguel; University of California San Diego, Department
of Nanoengineering
Kupor, Daniel; University of California San Diego, Nanoengineering
Marchiori, Leonardo; University of California San Diego, Nanoengineering
Soto, Fernando; University of California San Diego, Department of
NanoEngineering
Rueda, Ricardo; University of California San Diego, Nanoengineering
Reynoso, Maria; University of California San Diego
Narra, Lakshmi; University of California San Diego
Chakravarthy, Krishnan; University of California San Diego Department
of Anesthesiology
Wang, Joseph; University of California San Diego, Department of
Nanoengineering
Journal of Materials Chemistry B
1
Combinatorial microneedle patch with tunable release kinetics and dual
fast-deep/sustained release capabilities
Miguel Angel Lopez-Ramirez,
a
Daniel Kupor,
a
Leonardo Marchiori,
a
Fernando Soto,
a
Ricardo Rueda,
a
Maria Reynoso,
a
Lakshmi Rekha Narra,
b
Krishnan Chakravarthy
*a,b
and Joseph Wang
*a
a
Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United
States.
b
Department of Anesthesiology and Pain Medicine, University of California, San Diego, Health Sciences,
La Jolla, California, 92093, United States.
* Correspondence to: [email protected] and [email protected]
Abstract
Transdermal microneedle (MN) drug delivery patches, comprising water-soluble polymers, have played
an essential role in diverse biomedical applications, but with limited development towards fast deep
release or sustained delivery applications. The effectiveness of such MN delivery patches strongly
depends on the materials from which they are constructed. Herein, we present a dual-action
combinatorial programmable MN patch, comprising of fast and sustained-release MN zones, with
tunable release kinetics towards delivering a wide range of therapeutics over different timeframes in
single application. We demonstrate the fine tuning of MN materials; the patches can be tailored to
deliver a first payload faster and deeper within minutes, while simultaneously delivering a second
payload over long times ranging from weeks to months. The active and rapid burst release relies on
embedding biodegradable Mg microparticle ‘engines’ in dissolvable MNs while the sustained release is
attributed to biocompatible polymers that allow prolonged release in a controllable tunable manner. In
addition, the patches are characterized and optimized for their design, materials and mechanical
properties. These studies indicate that such programmable dual-action versatile MN platform is
expected to improve therapeutic efficacy and patient compliance, achieving powerful benefits by single
patch application at low manufacturing cost.
Page 1 of 24 Journal of Materials Chemistry B
2
Introduction
Advanced microfabrication methods have enabled the development of microneedle (MN) arrays as
novel transdermal drug-delivery devices,
1,2
that are widely used for treating diverse disease conditions
ranging from superficial dermatitis to diabetes and chronic pain.
3-7
MNs are microscale structures that
allow the delivery of drugs within few microns of the skin and are considered minimally invasive with
minimal patient-reported pain compared to traditional hypodermic needles,
8-11
requiring minimal
training from medical practitioners,
12,13
avoiding the cold chain (refrigeration, storage) and are thus
suitable for application in remote locations. Optimal design of MN balances various parameters that
determine the drug delivery efficiency, mechanical strength, or manufacturability. Currently, MN
patches play an essential role in a wide variety of biomedical research applications and are well tolerated
in clinical trials,
14,15
leaving behind only biocompatible, dissolvable and hence safe soluble materials
when compared to the conventional biohazardous waste left from the needle and syringe. Previous
research with MN patches has involved the delivery of small molecules,
16,17
biomacromolecules,
18,19
and
nanoparticles (NPs),
20,21
using either coated or non-coated dissolvable MN’s,
22,23
usually comprised of
highly water-soluble polymer matrixes with narrow development in sustained release applications,
24-30
and limited development in hybrid/programmable MN patches.
31
For example, Nguyen’s team recently
described core-shell MNs that can deliver cargoes in a pulsatile manner, mimicking release via repetitive
bolus injections over long periods of time.
32
Additionally, short-term sustained release microneedles
have been reported by employing chitosan,
33
silk fibroin,
34
and PEGDA/PVP
35
polymeric matrices.
Nevertheless, each disease has diverse needs and requires different delivery strategies, immediate (fast-
acting) or sustained (prolonged) release for over more than a few days. Our ultimate goal is to offer
effective post-operative pain management via a fast-local anesthetic delivery for immediate pain relief
along with co-delivery of a second anesthetic to address prolonged sustained effects. Consequently, the
development of improved, practical and efficient MN patches with tunable dissolution and
combinatorial release kinetics, is necessary and required in a wide variety of biomedical applications, as
they remain broadly unexplored.
30,36-38
Herein, we present a dual-action programmable combinatorial MN patch with tunable fast-acting
and sustained release of different payloads. The novel dual-action patch was engineered and designed
Page 2 of 24Journal of Materials Chemistry B
3
to have two spatially resolved MN compartments with different dissolution rates and a tunable payload
release (Fig. 1). The first compartment (active MNs) made of carboxymethylcellulose (CMC) dissolvable
polymer needles allowed a fast and forceful immediate payload release (within ~5 minutes) by
incorporating biocompatible and degradable active Mg microparticles. Skin insertion of the fast acting
MNs enables instantaneous reaction of the embedded Mg particles with the surrounding interstitial
fluid, resulting in the formation of gas (hydrogen) microbubbles, that induce localized vortex flow fields
and a ‘pumping-like’ action within the application site.
39-42
Simultaneously, the nearby second
compartment (the sustained MN zone) is made of different concentrations of Eudragit®L100, to allow a
controlled and constant sustained payload release over prolonged time periods, ranging from weeks to
months. We have demonstrated recently the utility of active Mg-based ballistic MN for effective in vivo
melanoma tumor eradication.
37,38
Here we combine such ballistic payload delivery with sustained
payload delivery using a single MN patch footprint that offers fast and sustained co-delivery via two
neighboring MN compartment zones.
We envision that this fast-acting/sustained release patch will set the foundation for future MN
research towards next generation patches, able to treat diverse disease conditions per single application
at low manufacturing cost. The ultimate goal of our collaborative research is to apply this unique delivery
capability for the management of pain, considering the current limitations of existing pain-relieving
patch technologies, such as general Mylan generic lidocaine patches,
43
and the lack of effective
transdermal modalities for post-operative settings.
44
Such post-operative pain control will be realized
using a single patch, providing fast and slow tunable delivery of the corresponding anesthetic drugs. Yet,
the new delivery concept goes beyond pain management as it could greatly benefit broad medical
scenarios by delivering different therapeutics with different nature of kinetics and solubility (water
soluble in one compartment, while loading a fat-soluble/organic-soluble molecule in the nearby
compartment). In the following sections we present the detailed characterization of the new
combinatorial MN patch, including its design, fabrication and performance in vitro and ex vivo.
Results and discussion
Design and fabrication of combinatorial MN patches
Page 3 of 24 Journal of Materials Chemistry B
4
The successful realization of the effective combinatorial MN array patch, based on the sequential
fast and slow release of different payloads, requires judicious selection and systematic optimization of
the materials, design and size to impart the desired biocompatibility and mechanical properties, drug
loading capacities and tunable dissolution. The combinatorial MN patch has been engineered to have 2
MN compartment zones, one of which is able to dissolve and rapidly deliver dosages in minutes, while
the second zone is slowly dissolved to release its payload over periods ranging from weeks to months
(Fig. 1a).
Page 4 of 24Journal of Materials Chemistry B
5
Fig. 1 Combinatorial MN patch with programmable (fast/sustained) drug co-delivery. (a) Schematic
illustration of the combinatorial MN patch application process to the skin and dual-stage MN delivery:
Page 5 of 24 Journal of Materials Chemistry B
6
fast-acting and sustained release of two payloads. (b) Illustration of the MN patch composition. (c) Time-
frame schematics displaying the delivery performance of both compartments: a fast-active burst release
of the first therapeutic, along with a prolonged-sustained delivery of a second drug. (d) Digital
fluorescence photograph of a combinatorial MN patch displaying both MN compartments loaded with
FITC and Rh6G. Scale bar: 2.5 mm. (e) Release kinetics representation of a dual-stage MN patch, featuring
the fast delivery of first therapeutic within minutes, while gradually releasing the second drug over
weeks and months.
MN compartments were formulated using different materials to meet adequate dissolution and
delivery times, and their specific composition was optimized for fine tuning the release kinetics (Fig. 1b).
The fast-dissolving MN compartment was formulated using CMC, a water-soluble and safe
polysaccharide,
45
with high biocompatibility and biodegradability in the biomedical field,
46,47
often used
in biosensors.
48
To facilitate MNs to dissolve instantly and ensure a rapid, forceful payload release from
the fast-dissolving compartment we incorporated biocompatible active Mg microparticles within
different CMC polymer matrix concentrations. The embedded particles lead to localized generation of
gas microbubbles and corresponding vortex flow that results in a nearly instantaneous release of the
payload, enabling wider and greater payload distribution and permeation in the application area.
Recently, our previous active MN platform was shown extremely useful for eradicating tumor growth in
a murine melanoma model, greatly improving immunotherapeutic efficacy.
39,40
The sustained release
compartment was designed and formulated of a biocompatible methacrylic acid pH sensitive co-polymer
(Eudragit®L100).
49-53
The delivery and release of the sustained MN compartment can be tailored from
weeks to months by tuning the polymer concentration used in the fabrication process, therefore
obviating the need to load the drug within nanocarriers,
54-56
hence improving the loading. The different
dissolution properties and behavior of the combinatorial MN patch (fast-acting and sustained release)
are clearly illustrated in Fig. 1c. A characteristic combinatorial MN patch displaying both compartments:
fast-acting needles loaded with Fluorescein 5(6)-isothiocyanate (FITC), and prolonged release needles
loaded with Rhodamine 6G (Rh6g) is shown (Fig. 1d). Moreover, the release kinetics concept of the dual-
stage MN patch, featuring the fast delivery of first payload within minutes, while promoting a gradual-
sustained release of the second payload for 30 days is shown in Fig. 1e.
We custom designed and engineered the patch by a mask stereolithography technique (MSLA) with
the use of a high-resolution 3D printer. Master MN (triangular right angle shaped) mold dimensions were
Page 6 of 24Journal of Materials Chemistry B
7
designed to be 550µm in base (triangular base length) x 950µm long (height) arranged in a circular
pattern (Fig. 2a). The outer compartment comprised of two concentric circular arrays of 35 MNs while
the middle compartment of 13 MNs. The base of the array was designed to be 10mm in diameter and
2mm in thickness to ensure facile polydimethylsiloxane (PDMS) micromolding steps. The direct light
process (DLP) fabrication projected a 25W UV light source through a 2K LCD masking (XY resolution: 47
µm), photocuring a liquid acrylate resin material in a 20 µm layer-by-layer additive manufacturing
fashion (detailed fabrication in experimental section).
Fig. 2 3D lithographic MN printing. (a) Drafting of the computer aided design (CAD) model of the
combinatorial MN patch and lithographic 3D printing process. Digital photograph of 3D-printed MN
molds. Scale bar: 2.5 mm. (b) Compositional and morphological analysis of master 3D printed MN molds
assessed by SEM. Scale bar, 1mm and 400µm. (c) Colored SEM displaying both distributed compartments
within the patch: the fast-acting (green) and sustained release (red) arrays. Scale bars: 1.2 mm and 2
mm, respectively. (d) 3D printed MN array post processing steps: UV treatment, de-solvation and
heating, followed by subsequent PDMS micromold fabrication.
An evaluation of different 3D printing parameters to enable controllable dimensions, surface
stepping/smoothness and tip sharpness can be found in Table S1 and Table S2 (ESI†). Master MN arrays
were subjected to a scanning electron micrograph (SEM) to verify needle quality after each printed
Page 7 of 24 Journal of Materials Chemistry B
8
batch; a characteristic SEM of the master combinatorial MN array printed after optimization is shown in
Fig. 2b. Additional SEM images of different printing parameters can be found in Fig. S1 and S2 (ESI†).
Colored SEM image of the master array clearly depicting the spatially resolved MN compartments is
shown in Fig. 2c (fast-acting and sustained MNs in green and red, respectively).
Prior to the fabrication of negative micromolding steps, master MN arrays were subjected to a post
processing procedure to remove all leaching compounds inherent from the acrylate resin used in the
printing process (Fig. 2d). Leaching compounds demonstrated to inhibit the PDMS curing process, not
enabling it to correctly reproduce master mold features, leaving behind not useful silicone molds; molds
required to be subjected under UV light (405 nm) and temperature treated for different times (detailed
fabrication and parameters in Table S3, ESI†). Following post treatment, silicone negative molds were
developed by casting PDMS over post-processed master MN arrays (Fig. 2d).
Tailored fabrication and optimization of dual-stage combinatorial MN patches
We molded combinatorial MNs by casting different materials onto each compartment. Briefly, to
prepare fast-acting MNs we proceeded to pack Mg microparticles (30-100µm in diameter) into mold
cavities by a drop casting process using a Mg-isopropanol suspension. After the solvent was evaporated,
we proceeded to cast an aqueous solution (H
2
O, pH 10.5) to solubilize CMC (2, 3 and 4%, w/v) and FITC,
enabling a slow evaporation under vacuum during the active compartment fabrication, therefore
avoiding Mg particle reactivity (hydrogen bubble generation) throughout the drying process. The cavities
designated for sustained release MNs were subsequently filled under vacuum by casting an organic
solvent (ethanol/isopropanol, 50:50% v/v) to solubilize Rh6G and Eudragit®L100 (4, 8, 16 and 24%, w/v).
After having both MN compartment cavities filled and dried, molds were allocated inside a conventional
oven to enhance MN strength. Later, an aqueous and ethanol based polyvinylpyrrolidone (PVP, 10%,
w/v) backing solution was applied to the fast-acting and sustained release MN compartments,
respectively. The resulting combinatorial MN patch was then transferred to a slightly larger medical
adhesive. The patch was custom designed to be small, enabling ease of use, facile transportation and
consequently easy storage and handling. Schematics of the steps involved (side and top views) in the
fabrication of combinatorial MN patches are illustrated in Fig. 3a (detailed fabrication in ESI†).
Page 8 of 24Journal of Materials Chemistry B
9
Combinatorial MN patches were formulated from different polymer concentrations. Therefore, we
decided to examine their dissolution properties by imaging them at different times. Time-frame
fluorescence microscopy dissolution images of characteristic fast-acting MN formulations can be found
in Fig. 3b. Fast-acting needle dissolution images (30s intervals) with variable concentration (2, 3 and 4%
CMC) showed that the polymer matrix is dissolved almost instantly when in contact with the solution
(phosphate buffered solution (PBS) pH 7.4). Images clearly show the spontaneous and vigorous H
2
pump
activity from the entrapped Mg microparticles as soon as they are exposed to the solution. From this
perspective, no significant difference was found within groups examined, as all demonstrated complete
MN dissolution within a short period of time displaying rapid particle activation.
Fig. 3 Dual-stage combinatorial MN patch customization for the simultaneous co-delivery of different
drug payloads. (a) Schematics of the steps involved in the fabrication of combinatorial MN patches (side
and top view) for the fast-acting and sustained delivery: active particle loading (I), polymer (CMC,
Eudragit®L100) and payload inclusion (FITC, Rh6G), respectively (II), polymer base and drying (III),
adhesive application (IV) and demolding (V). (b) Fluorescence microscopy time-frame dissolution images
of the “fast-acting” MN compartment, displaying a rapid polymer matrix dissolution driven by the
reactivity of Mg microparticles (30 s intervals) (I) 2% CMC, (II) 3% CMC, (III) 4% CMC, packed with 1mg of
Mg. Scale bar: 400 µm. (c) SEM image of a characteristic fast-acting MN and its corresponding EDX (C,
Page 9 of 24 Journal of Materials Chemistry B
10
Na and Mg). Scale bar: 400 µm. (d) Fluorescence microscopy time-frame dissolution images of the
“sustained release” MN compartment, displaying slow polymer matrix degradation at day 5, 10, 20 and
25. (I) Eudragit®L100 4%, (II) 8%, (III) 16%, and (IV) 24%. Scale bar: 400 µm. (e) SEM image of a
characteristic sustained release MN and its corresponding EDX analysis (C, O and Mg). Scale bar: 400 µm.
A SEM image of a characteristic fast-acting MN is shown in Fig. 3c. Energy Dispersive X-ray (EDX)
elemental analysis of the structure displays the MN composition (Fig. 3c); as expected, elemental
analysis of Mg show that Mg microparticles remain entrapped within the MN structure. In contrast,
sustained release MNs (4, 8, 16 and 24% Eudragit®L100) visually demonstrated to have slow dissolution
properties for days while being exposed to circumvent solution around it (Fig. 3d), where the methacrylic
acid pH sensitive co-polymer starts to dissolve at pH values above 6.0. Correspondingly, sustained
release MNs were characterized by SEM and EDX (Fig. 3e), image shows a smooth MN surface and sharp
tip.
MN patch mechanical performance and delivery modulation
To investigate the optimal formulation of the combinatorial MN patch we performed extensive
characterization of each compartment. To evaluate whether MNs had sufficient mechanical strength to
penetrate the skin, we evaluated the mechanical performance of fast-acting and sustained release MNs
under compression (schematic of the experimental setup shown in Fig. 4a). Results demonstrated that
the MN strength under compression increased in both fast-acting and sustained release compartments
with increasing polymer (CMC/Eudragit®L100) concentration (force displacement curve shown in Fig. 4b
and c, respectively), however, no significant difference was found among the three different polymer
concentrations used as fast-acting MNs, where 3% CMC was the most optimal formulation, considering
polymer solution viscosity and easier fabrication process to fill the silicone negative molds. Moreover,
the area under the curve (AUC) of each MN compartment was estimated up to a displacement of 300µm,
where the higher the AUC value, the stronger the MN (Fig. 4d). Although combinatorial MN patches with
the lowest concentration were weaker, they tolerated compression forces 0.15 N needle
-1
, value
reported and expected to enable efficient and reliable skin penetration.
54
To determine until what degree the drug loading capacity and efficiency of the fast-acting MN
compartment could be compromised upon different amount of Mg microparticles, we examined 3%
Page 10 of 24Journal of Materials Chemistry B
11
CMC fast-acting MNs loaded with 0, 0.5, 1, 2, and 3mg of Mg microparticles. A UV-vis spectrum of the
characteristic model payload (FITC) loaded, and the corresponding calibration curve (inset) is shown in
Fig. 4e. Different formulations of fast-acting MNs were fabricated (0, 0.5, 1, 2, and 3 mg of Mg) and
subjected to complete dissolution. UV-vis spectrum of the release of these arrays can be found in Fig. 4f
and the corresponding number of micrograms loaded in Fig. 4g. As expected, we identified that
regardless of the amount of Mg microparticles loaded within the fast-acting MN array, the relative
available space to load the drug seems to be compromised and significantly reduced (~47% for 0.5mg of
Mg, 49% for 1mg, 64% for 2 mg and 68% for 3mg). However, this experiment set the foundation to
precisely tune the amount of drug that needed to be loaded in each patch; by just adjusting the stock
payload solution we were able to fix the loading micrograms required per patch for subsequent in vitro
release kinetics and ex vivo penetration studies.
Fig. 4 Mechanical properties, drug loading efficiency and MN compartment optimization. (a) Schematic
illustration of the mechanical testing stage employed. (b) Mechanical strength analysis (force
displacement curve) of different active MN formulations: 2%, 3% and 4% carboxymethyl cellulose loaded
Page 11 of 24 Journal of Materials Chemistry B
12
with 1 mg of Mg microparticles. Points represent means ± s.d (n=5 independent experiments). (c)
Mechanical strength analysis (force displacement curve) of diverse sustained release MN formulations:
4%, 8%, 16% and 24% Eudragit®L100. Points represent means ± s.d (n=5 independent experiments). (d)
Area under the curve (AUC) up to a displacement of 300 µm. A higher AUC means stronger MN. Data are
means ± s.d (n=5). (e) Fast-acting MN patch model payload (FITC) absorbance spectrum calibration curve.
(f) Active MN release curves from patches loaded with different amounts of Mg microparticles (0, 0.5, 1,
2 and 3mg). (g) Optimization of the active MN compartment; micrograms of FITC loaded vs Mg
microparticle packing. Bars represent means ± s.d (n=5). (h) Active MN compartment loading efficiency
vs Mg microparticle packing. Bars represent means ± s.d (n=5). (i) Sustained release compartment MN
patch model payload (Rh6G) absorbance spectrum and calibration curve. (j) Sustained release MN
release curves from patches fabricated of different MN formulations: 4%, 8%, 16% and 24%
Eudragit®L100. (k) Sustained release MN compartment optimization: micrograms of Rh6G loaded vs
polymer concentration. Bars represent means ± s.d (n=5). (l) Sustained release MN compartment loading
efficiency vs polymer concentration. Bars represent means ± s.d (n=5).Statistical significances were
calculated by one-way ANOVA with Tukey’s test: *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Additional graph (Fig. 4h) displays the loading efficiency of each fast-acting patch formulation,
where in a very similar manner as they are closely related, the loading efficiency seems to be reduced as
the number of milligrams of Mg increase. Likewise, same characterization and optimization experiments
were developed to different formulations of sustained release MNs. A UV-vis spectrum of the
characteristic model payload (Rh6G) loaded within the sustained compartment, and the corresponding
calibration curve (inset) is shown in Fig. 4e. Fabricated MNs (4, 8, 16 and 24% Eudragit®L100) were
subjected to complete dissolution. UV-vis spectrum of the release from these arrays is shown in Fig. 4j
and the corresponding number of micrograms loaded in Fig. 4k. We identified that the polymer
concentration of Eudragit®L100 and the amount of drug that can be loaded in the MN structure are
interdependent. As the polymer concentration increases from 4-24%, the number of micrograms loaded
per array is significantly reduced (~200µg for 4% L100, ~107µg for 8% L100, ~73µg for 16% L100 and
~50µg for 24% L100). This is expected due to the viscosity increments in the solutions employed (casted
within the cavities of the molds) in the fabrication process; where the lower the concentration, the faster
the organic solvent vaporization, therefore, higher the loading. Even though the fabrication process of
combinatorial MN patches is very well established, the loading will tend to be higher when lower
polymer concentrations are employed. Additional loading efficiency graph of sustained release MN
compartment (Eudragit®L100) formulations is found in Fig. 4l, clearly displaying the same behavior.
Page 12 of 24Journal of Materials Chemistry B
13
Programmable MN dual-stage active/sustained payload delivery and transdermal penetration
Fluorescence imaging of a characteristic combinatorial MN patch is shown in Fig. 5a. The
combinatorial MN patch image is comprised of 3% CMC (1mg/Mg) fast-acting MN and 16%
Eudragit®L100 sustained release MN compartments. Fig. 5a(i) shows a combinatorial MN patch before
being subjected to dissolution (t=0), clearly displaying both compartments. For convenient visualization,
the inner fast-acting needle compartment was loaded with the FITC dye while the outer sustained
release needles were loaded with Rh6g. Fig. 5a(ii) displays an image of the same combinatorial MN patch
at t=1, where fast-acting MNs are dissolved (t=1) within minutes, leaving behind remaining Mg
microparticles in the base of the middle compartment array. Likewise, t=2 (Fig. 5a(iii)) shows the 2
nd
dissolution stage of the patch, where only the adhesive is left, clearly displaying the complete
spatiotemporal dissolution of both compartments.
Fig. 5 Dual-stage tunable release kinetics from combinatorial MN patch. (a) Digital fluorescence
photographs of the different payload delivery stages at t=0 (Before application), t=1 after active
compartment dissolution (burst and fast release of FITC, and t=2 after sustained delivery of Rh6G. Scale
bar: 5 mm. (b) Cumulative release kinetic profiles of different formulations of the active MN
compartment from the combinatorial MN array, using different CMC loadings (2%, 3% and 4%), showing
a fast delivery of FITC over a period of few minutes. Points represent means ± s.d (n=5). (c) Digital
photograph of the MN patch onto cadaver porcine skin and subsequent imaging of the skin after
application, under UV lamp (left) and fluorescence microscopy imaging under GFP channel (right). Scale
bars: 4 and 2 mm, respectively. (d) Colored SEM image depicting an active MN compartment piercing
Page 13 of 24 Journal of Materials Chemistry B
14
porcine skin. Scale bar: 500 µm. (e) Tunable cumulative release profile kinetics from the sustained
compartment of the combinatorial MN array: the prolonged delivery of Rh6G is achieved from weeks to
months (in connection to Eudragit®L100 polymer concentrations of 4%, 8%, 16% and 24%). Points
represent means ± s.d (n=5). (f) Digital photograph of sustained release compartment MN after piercing
porcine cadaver skin, under UV lamp (left) and fluorescence microscopy imaging under RFP channel
(right). Scale bars: 4 and 2.5 mm, respectively. (g) Colored SEM image depicting a sustained MN
compartment piercing porcine cadaver skin. Scale bar: 500 µm. (h) Percentage penetration of active MN
in porcine skin. Bars represent means ± s.d (n=5). (i) Percentage penetration of sustained release MN in
porcine skin. Bars represent means ± s.d (n=5). Statistical significances were calculated by one-way
ANOVA with Tukey’s test: *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Dual release kinetics from combinatorial MN patches was performed in vitro using release media
of saline buffered solution PBS pH 7.4 (chosen to simulate in vivo transdermal pH).
17,57
FITC release from
the fast-acting MN compartment (with 2, 3, and 4% CMC) showed a burst release within the first minutes
(Fig. 5b). All three patches were fairly consistent over time with no significant difference in release within
this short period of time (<8 min). Release percentage of the different patch formulations at 2 min are:
2% CMC (85±7%), 3% CMC (76±10%) and 4% CMC (55±13%) and at 5 min are: 2% CMC (100%), 3% CMC
(96±3%) and 4% CMC (85±4%). This data indicates that the fast-acting MN compartment can deliver rapid
onset of payloads, achieving the target delivery timeframe in few minutes. The hydrogen generation rate
and heat exchange from Mg microparticles strongly depends on the surrounding pH environment (as
demonstrated in Fig. S3, ESI†). Such dependence suggests that the heat release from Mg microparticles
is small and will not compromise the entrapped payload integrity. The 3% CMC fast-acting MN
compartment was manually applied to cadaver porcine skin for a total of 10 min and imaged in Fig. 5c
under UV lamp and fluorescence microscopy imaging under green fluorescence protein (GFP) channel.
A pseudo-colored SEM image displays the penetration of two fast-acting MNs in green to the skin (Fig.
5d); these images show that MNs penetrated skin efficiently. Furthermore, the release kinetics from the
sustained (slow) release MN compartment (4, 8, 16 and 24% Eudragit®L100) showed no initial burst
release on day 1 (Fig. 5e), with a constant Rh6G release over time (ranging from ~0.4 to ~3.8% Rh6G
release per day, depending on the polymer concentration). By using 4% Eudragit®L100, the payload was
released within 28 days, while using higher Eudragit®L100 concentrations resulted in longer time-frame
delivery period exceeding 90 days (combinatorial MN release absorbance curves are shown in Fig. S4,
supporting information). These release profiles clearly illustrate the ability to tune the sustained release
for period ranging from weeks to months through control of the Eudragit®L100 loading. Additional data
Page 14 of 24Journal of Materials Chemistry B
15
on an alternative delivery system by the microencapsulation of Rh6G was developed by synthetizing
PLGA microparticles; in contrast to our combinatorial patch, PLGA microparticles deliver full dosages
within ~40 days, corresponding data of the release and procedures can be found in Fig. S5 (ESI†). The
16% Eudragit®L100 sustained MN compartment was manually applied to cadaver porcine skin and
imaged in Fig. 5f under UV lamp and fluorescence microscopy imaging under RFP channel. Images display
that MNs penetrated skin efficiently; additionally, a pseudo-colored SEM image displays the penetration
of three fast-acting MNs in red to the skin (Fig. 5g). The combinatorial microneedle composition and
design allow the transfer base (PVP) to detach from the needles post application (>10 min) and thus to
reduce the risk of infection while increasing patient compliance and leaving behind no sharp waste. (as
illustrated in Fig. S6, ESI†). Combinatorial MN patches were applied to porcine skin and the penetration
efficiency of both fast-acting and sustained compartments is shown in graph (Fig. 5 h and i, respectively).
The 2% CMC/4% L100 composition resulted in penetration efficiency lower than 50% (directly related to
the percentage of drug delivered within the skin). Nevertheless, by increasing the polymer
concentration, the skin penetration improved, regardless of the polymer used in the fabrication process.
The majority of needles were able to breach dermal barriers in an efficient matter, delivering the payload
and remained fully embedded under the skin surface. Although our combinatorial MN patches are
currently based on manual administration, we plan to develop and use applicators future studies.
Conclusions
We developed a combinatorial MN patch with dual and tunable release kinetics, aimed at providing
greater access in delivering wide range of therapeutics with variable target delivery timeframes in a
single application. The patch was fabricated to be comprised of two MN compartment zones: a fast-
acting and a sustained release compartment. The dual release properties of the patch with different
dissolution time frame is attributed to the ability of the patch to load several drugs (even with different
solubility and incompatible) within the same array, but spatially resolved, due to the additive
manufacturing process employed. MN compartments in such combinatorial array can thus be
engineered and tailored to present different release kinetics based on the specific materials and
compositions used in fabrication. More encouragingly, we demonstrate that the combinatorial patch can
be programmed to deliver drug payloads faster in minutes, greatly enhancing the release when
Page 15 of 24 Journal of Materials Chemistry B
16
compared to passive MNs, but additionally, can deliver dosages that last over the course of weeks-to-
months, for up to >90 days, with constant release over time. We envision that using different active
microengine materials will allow tailoring the burst release profiles of the new ballistic MN. This novel
combinatorial MN patch provides flexibility in the loading capacity, as needed to reach clinically relevant
doses, and is expected to improve therapeutic efficacy and improve patient compliance towards
substantially enhanced pain management. In the absence of effective treatment modalities for
neuropathic pain, we envision that the new patch has the potential to improve control of postoperative
pain of current therapy platforms, achieving powerful benefits by single patch application, decreasing
daily injections, localizing the treatment, thus increasing efficacy. The new concept is expected to benefit
a wide range of medical situations, beyond pain management.
Materials and methods
MN CAD/CAM design
Drafting of the computer aided design (CAD) model of the printed part was custom designed in a
modeling program (Solidworks version 2019-2020) and ran in an operated Microsoft system-based
computer. The microneedle array was designed without considering having PDMS walls between the
different MN compartments (microchannels in the master mold), to facilitate the micromolding process
of the combinatorial microneedles (easier removal of polymer solution from the base). Models were
transferred to a free open-source SLA/DLP/LCD 3D printer software to adjust, prepare and edit the 3D
model (Chitubox-Version 1.6.3) prior slicing and printing. Specifically, the developing of supports and
lattices for each model with the corresponding dimensions and connections to prevent printing failure.
Computer aided manufacturing (CAM) was developed by AnyCubic Photon Slicer Software with the
parameters listed in Table 1 and 2.
3D lithographic MN printing
The direct light processing (DLP) fabrication of MNs was developed with the use of an AnyCubic Photon
UV LCD 3D Printer. The DLP 3D printer projected a 25W UV light source through a 2K LCD masking
photocuring a liquid resin material (black colored AnyCubic acrylate resin). The 3D printed models were
mass produced and cured in a layer-by-layer fashion (20-50µm layers). Printed models were removed
Page 16 of 24Journal of Materials Chemistry B
17
from the 115 x 65mm metal build plate and supports trimmed. The MN models were tippled rinsed with
2-propanol (Sigma Aldrich) and further placed within an ultrasonic bath to remove uncured resin
material in excess.
MN post processing
Post processing of the printed MN mold was performed by an additional post curing step under a 60W
Mercury UV Curing Lamp (Moai Peopoly) for 30 min. Subsequently, MNs in a metal plate (10cm x 10 cm;
2 mm thickness) were heat treated at a fixed temperature (120
o
C) for different times (3-30 min) in a
conventional oven. Detailed parameters of post processing are listed in Table S3, supporting information.
PDMS MN micromold fabrication
The polydimethylsiloxane (PDMS) SYLGARD® 184 (Ellsworth Adhesives) micromolding process of 3D
printed MNs allowed the fabrication of reusable negative templates for the mass production of
combinatorial polymeric MN arrays. The fabrication of silicone MN molds was done by the following
procedure: PDMS (12g, 86/14; base/curing agent) solution was casted over a combinatorial MN array
attached to a crystal-clear borosilicate petri dish glass. Afterwards, PDMS was degassed in a sealed
desiccator connected to a vacuum pump for 15 min at 23 in Hg. PDMS was set to cure (30 min at 85°C)
in an oven and further demolded from the master combinatorial MN array, thus obtaining the final
combinatorial negative mold. Negative molds were resized with the use of a stainless-steel blade cut and
triple cleaned/washed with hand soap; subsequently allocated in an ultrasonic bath, sterilized at 80
o
C
and stored in a sealed container prior use.
PLGA microparticle synthesis
The fabrication method of the Rhodamine 6G (Rh6G, Sigma Aldrich; λex/λem, 526/555 nm)-loaded
poly(lactic-co-glycolic acid) (PLGA, MW=50/50, Sigma Aldrich) microparticles was based on an emulsion
solvent evaporation technique. Briefly, PLGA (30mg) was dissolved in Ethyl Acetate (1mL, Sigma Aldrich).
Subsequently, Rh6G (5mg) was dispersed in the polymer solution, and further poured into a 2% PVA
aqueous solution (10mL). Solution was emulsified with the use of a high-speed ultrasonic homogenizer
Page 17 of 24 Journal of Materials Chemistry B
18
(Ultrasonics) by three 3s (100W) pulses. The organic phase was allowed to evaporate by continuous
mixing (900 rpm) for 18h at ambient temperature. PLGA microparticles were collected by centrifugal
force (10,000 rpm) for 7 min and tripled washed with deionized water. Microparticles were suspended
in deionized water and then used for MN patch fabrication and particle characterization.
Combinatorial polymeric MN micromolding
The fabrication of the combinatorial (active/sustained) polymeric MN patch was realized by a
micromolding approach following the next procedure. The fabrication process of this bi-compartmental
specific microneedle array design restricted us in fabricating each polymeric microneedle compartment
in a sequential specific form (inner to outer order). Briefly, the compartment designated for the active
MNs was primarily loaded and packed with Mg microparticles (0, 0.5, 1, 2, or 3 mg) by employing a 2-
propanol Mg microparticle suspension (50 mg mL
-1
, catalog #FMW40, TangShanWeiHao Magnesium
Powder Co., Ltd China), followed by casting a 3% (w/v) sodium carboxymethyl cellulose (CMC, 50 µL,
average MW=250K) aqueous polymer solution (pH 10.5 to prevent Mg microparticle reaction and
corresponding hydrogen bubbles generation) supplemented with Fluorescein 5(6)-isothiocyanate (FITC,
0.2mg mL
-1
; λex/λem, 492/518 nm). Molds were allocated under a sealed desiccator connected to a
vacuum pump (23 in Hg) for 5 min. Bubbles were removed from the mold and the process was repeated
by triplicate. Process was repeated 5 times. Excess of solution was removed from the mold by swiping it
with the use of a blade cut and saved for the next repetition. The active compartment was allowed to
dry for 15 min between each addition and kept at room temperature for 30 min prior the following
compartment fabrication. The procedure of the sustained release compartment fabrication was
performed by loading it with a polymeric blend of a 4%, 8%, 16% and 24% (100µL, Ethanol/2-propanol;
50/50% v/v) solution of Eudragit®L100 supplemented with Rh6G (0.5mg). Similarly, this allowed us to
take advantage of the design and wipe easily the excess of remaining polymer solution from the mold in
a circular motion without compromising the previously made compartment and saved for next mold
fabrication (process repeated by triplicate). The sustained release compartment was further allowed to
dry for 15 min at room temperature. The finished combinatorial MN mold was placed inside a
conventional oven for 2h at 70°C. For the cumulative release kinetics experiments a layer of
polyvinylpyrrolidone (PVP, MW=360K) was added as a support base on top of the active or sustain
Page 18 of 24Journal of Materials Chemistry B
19
release compartment by casting an ethanol (50 µL, Sigma Aldrich) based or water based (100µL) 10%
(w/v) PVP solution respectively and let to dry overnight. Finally, the active/sustained combinatorial MN
patch was obtained by a demolding procedure; in brief, after drying, a 1 mm circular adhesive (3M) was
adapted to the backing of the MN patches and demolded. The reason behind the PVP layers was to
efficiently detach the needles for release measurements and to prove mechanical stability. Prior use, the
combinatorial MN patches were stored in a sealed container at room temperature. CMC, PVP,
Eudragit®L100 polymer solutions were prepared using a dual asymmetric centrifugal mixer (Flacktek
Speedmixer, DAC 150.1 KV-K, FlackTek, SC, U. S. A.), speed of 2500 rpm for 5 min (by triplicate).
Combinatorial MN patches loaded with PLGA microparticles were fabricated following the same
procedure. Nevertheless, instead of Rhd6G alone, PLGA@Rh6G microparticle suspension (30µL) was
infiltrated within the cavities of the silicon molds, and polyvinylpyrrolidone (PVP) was casted on top as
the matrix MN material. Even though the polymer matrix is made of a water-soluble material,
PLGA@Rh6G microparticles provided sustained release over prolonged periods of time.
MN patch imaging characterization
Fluorescent microscopy images of the combinatorial MN patches were developed with the use of a
fluorescent microscope (EVOS FL coupled with 2x and 4x objectives and GFP and RFP filters) for the
imaging of Rh6G and FITC. SEM images were performed by a FEI Quanta 250 ESEM instrument (Hillsboro,
Oregon, USA). Prior imaging, the 3D printed, and polymeric combinatorial MN patches were sputtered
with Iridium in an Emitech K575X Sputter Coater, providing a fine grain metal deposition; samples were
imaged at acceleration voltages between 3-5 keV. Bright field and colored images were obtained by a
3.5X-180X Simul-Focal Stereo Zoom Microscope coupled with an 18MP digital camera. Digital fluorescent
images of the combinatorial MN patch before and after dissolution were carried out with a digital camera
Nikon D7000 coupled with a 40mm 2.8G macro lens; MN patches were placed under a portable UV lamp
projecting a 365nm wavelength light.
MN patch dissolution experiments
To image the dissolution in solution of our combinatorial MN, 3 continuous MN (active or sustained,
loaded with FITC or Rh6G, respectively) were attached horizontally to a clear glass slide with 4 acrylate
walls to contain solution. To capture the dissolution in real time, Phosphate buffered solution (pH 7.4)
Page 19 of 24 Journal of Materials Chemistry B
20
was administrated to the MN setup, followed by time-set point frame images with use of a fluorescent
microscope (EVOS FL coupled with a 4x objective and GFP-RFP filters).
MN patch mechanical strength study
Fast-acting and sustained release MN arrays from the patch were set under a mechanical compression
test. The set up used was comprised of a Force Gauge Model M4-20 system Mark0-10 Series 4, a metal
plate and a stepping-motor controlled biaxial stage. In brief, each MN array was set under a constant
load, and the displacement of the base plate in reference to each needle height was monitored and
plotted. The fracture (failure) force was determined by a notorious drop in force.
Release Kinetics Experiments
Fast-acting and sustained release MN patches were subjected to a release kinetics study. MN patches
were dissolved in 1000 µL of PBS pH 7.4 at 37.5 °C for different set-time points: the fast-acting MN (from
1-10 min) and sustained release compartment (from 1-90 days). Prior each measurement samples were
centrifuged at 4000 rpm for 5 min, and the solution release amount was measured by UV-vis
spectrophotometry (UV-2450 Shimadzu spectrophotometer) in an average of 3-4 days. After each
measurement, 1mL of PBS pH 7.4 solution was replaced within the sealed container. Each measurement
was conducted within a 400-700 nm spectrum window. The patches release was plotted vs time. Data
was analyzed and charts were generated using Prism 7 (GraphPad software).
PLGA-Rh6G microparticle release study
PLGA@Rh6G microparticles were dissolved in 1000 µL of PBS pH 7.4 at 37.5 °C for different set-time
points: (from 1-45 days). Prior to each measurement, samples were centrifuged at 4000 rpm for 5 min
and the release amount was measured by UV-vis spectrophotometry (UV-2450 Shimadzu
spectrophotometer). Each measurement was conducted within a 400-700 nm spectrum window. Particle
release was plotted vs time. Data was analyzed and charts were generated using Prism 7 (GraphPad
software).
Skin Penetration Studies
Page 20 of 24Journal of Materials Chemistry B
21
Both fast-acting and sustained release MN patches were set to pierce porcine cadaver skin. The skin
thickness was sliced to be ~3.0mm in thickness and was stored in a sealed container with PBS (pH 7.4)
at room temperature prior use. The combinatorial MN patches were applied to the skin manually.
Conflicts of interest
The authors declare no conflict of interest
Acknowledgements
This work was supported by the NSF grant 1937653 to J.W. and by the UCSD Center of Wearable Sensors.
M.A.-L.R., F.S. and M.R acknowledge the UC MEXUS-CONACYT Doctoral Fellowships. L. M acknowledges
FAPESP fellowship 2019/16608-4TOC. Some schematics were created with biorender.com.
References
1 E. Larrañeta, M.-T. McCrudden, A.-J. Courtenay and R.-F. Donnelly, Pharm. Res., 2016, 33, 1055–
1073.
2 D.-D. Zhu, X.-P. Zhang, B.-L. Zhang, Y.-Y. Hao and X.-D. Guo, Adv. Ther., 2020, 3, 2000033.
3 H. Lee, T.-K. Choi, Y.-B. Lee, H.-R. Cho, R. Ghaffari, L. Wang, H.-J. Choi, T.-D. Chung, N. Lu, T. Hyeon
and S.-H. Choi, Nat. Nanotechnol., 2016, 11, 566–572.
4 W. Chen, R. Tian, C. Xu, B.-C. Yung, G. Wang, Y. Liu, Q. Ni, F. Zhang, Z. Zhou, J. Wang and G. Niu,
Nat. Commun., 2017, 8, 1777.
5 R. Jamaledin, C.-K. Yiu, E.-N. Zare, L.-N. Niu, R. Vecchione, G. Chen, Z. Gu, F.-R. Tay and P.
Makvandi, Adv. Mater., 2020. 32, 2002129.
6 M. Yin, L. Xiao, Q. Liu, S.-Y. Kwon, Y. Zhang, P.-R. Sharma, L. Jin, X. Li and B. Xu, Adv. Healthc.
Mater., 2019, 8, 1901170.
7 X. Xie, C. Pascual, C. Lieu, S. Oh, J. Wang, B. Zou, J. Xie, Z. Li, J. Xie, D.-C. Yeomans and M.-X. Wu,
ACS nano, 2017, 11, 395–406
8 J. Arya and M.-R. Prausnitz, J. Control. Release, 2016, 240, 135–141.
9 H. Lee, C. Song, S. Baik, D. Kim, T. Hyeon and D.-H. Kim, Adv. Drug Deliv. Rev., 2018, 127, 35–45.
Page 21 of 24 Journal of Materials Chemistry B
22
10 K. Van der Maaden, W. Jiskoot and J. Bouwstra, J. Control. Release, 2012, 161, 645–655.
11 S. Marshall, L. J. Sahm and A. C. Moore, Hum. Vaccin. Immunother., 2016, 12, 2975–2983.
12 J.-J. Norman, J.-M. Arya, M.-A. McClain, P.-M. Frew, M.-I. Meltzer and M.-R. Prausnitz, Vaccine,
2014, 32, 1856–1862.
13 N.-G. Rouphael, M. Paine, R. Mosley, S. Henry, D.-V. McAllister, H. Kalluri, W. Pewin, P.-M. Frew,
T. Yu, N.-J. Thornburg, S. Kabbani, L. Lai, E.-V. Vassilieva, I. Skountzou, R.-W. Compans, M.-J.
Mulligan and M.-R. Prausnitz, Lancet, 2017, 390, 649–658.
14 S. Bhatnagar, K. Dave and V.-V.-K. Venuganti, J. Control. Release, 2017, 260, 164–182.
15 T.-A. Petukhova, L.-A. Hassoun, N. Foolad, M. Barath and R.-K. Sivamani, JAMA Dermatol., 2017,
153, 637–643.
16 R.-F. Donnelly, D.-I. Morrow, M.-T. McCrudden, A.-Z. Alkilani, E.-M. Vicente-Pérez, C. O'Mahony,
P. González-Vázquez, P.-A. McCarron and A.-D. Woolfson, Photochem. Photobiol., 2014, 90, 641–
647.
17 R.-F. Donnelly, T.-R.-R. Singh, M.-J. Garland, K. Migalska, R. Majithiya, C.-M. McCrudden, P.-L.
Kole, T.-M.-T. Mahmood, H.-O. McCarthy and A.-D. Woolfson, Adv. Funct. Mater., 2012, 22,
4879–4890.
18 Y.-C. Ryu, D.-I. Kim, S.-H. Kim, H.-M.-D. Wang and B.-H. Hwang, Biotechnol. Bioprocess Eng., 2018,
23, 286–292.
19 M.-T. McCrudden, A.-Z. Alkilani, A.-J. Courtenay, C.-M. McCrudden, B. McCloskey, C. Walker, N.
Alshraiedeh, R.-E. Lutton, B.-F. Gilmore, A.-D. Woolfson and R.-F. Donnelly, Drug Deliv. Transl.
Res., 2015, 5, 3–14.
20 C. Wang, Y. Ye, G.-M. Hochu, H. Sadeghifar and Z. Gu. Nano Lett., 2016, 16, 2334–2340.
21 P.-C. DeMuth, X. Su, R.-E. Samuel, P.-T. Hammond and D.-J. Irvine, Adv. Mater., 2010, 22, 4851–
4856.
22 U. Angkawinitwong, A.-J. Courtenay, A.-M. Rodgers, E. Larrañeta, H.-O. McCarthy, S. Brocchini,
R.-F. Donnelly and G.-R. Williams, ACS Appl. Mater. Interfaces, 2020, 12, 12478–12488.
23 X. Lan, J. She, D.-A. Lin, Y. Xu, X. Li, W.-F. Yang, V.-W.-Y. Lui, L. Jin, X. Xie, Y.-X. Su, ACS Appl. Mater.
Interfaces, 2018, 10, 33060–33069.
24 J.-H. Park, M.-G. Allen and M.-R. Prausnitz, Pharmaceutical research, 2006, 23, 1008–1019.
Page 22 of 24Journal of Materials Chemistry B
23
25 Y. Zhang, Q. Liu, J. Yu, S. Yu, J. Wang, L. Qiang and Z. Gu, ACS nano, 2017, 11, 9223–92030.
26 W. Li, R.-N. Terry, J. Tang, M.-R. Feng, S.-P. Schwendeman and M.-R. Prausnitz, Nat. Biomed. Eng.,
2019, 3, 220–229.
27 W. Li, J. Tang, R.-N. Terry, S. Li, A. Brunie, R.-L. Callahan, R.-K. Noel, C.-A. Rodríguez, S.-P.
Schwendeman and M.-R. Prausnitz, Sci. Adv., 2019, 5, eaaw8145.
28 A.-V. Boopathy, A. Mandal, D.-W. Kulp, S. Menis, N.-R. Bennett, H.-C. Watkins, W. Wang, J.-T.
Martin, N.-T. Thai, Y. He, W.-R. Schief, P.-T. Hammond and D.-J. Irvine, PNAS, 2019, 116, 16473–
16478.
29 S. Wang, M. Zhu, L. Zhao, D. Kuang, S.-C. Kundu and S. Lu, ACS Biomater. Sci. Eng., 2019, 5, 1887–
1894.
30 H. Kathuria, D. Lim, J. Cai, B.-G. Chung and L. Kang, ACS Biomater. Sci. Eng., 2020, 6, 5061–5068.
31 K.-T.-M. Tran, T.-D. Gavitt, N.-J. Farrell, E.-J. Curry, A.-B. Mara, A. Patel, L. Brown, S. Kilpatrick, R.
Piotrowska, N. Mishra, S.-M. Szczepanek and T.-D. Nguyen, Nat. Biomed. Eng., 2020, DOI
10.1038/s41551-020-00650-4
32 Z. Wang, J. Wang, H. Li, J. Yu, G. Chen, A.-R. Kahkoska, V. Wu, Y. Zeng, D. Wen, J.-R. Miedema,
J.-B. Buse and Z. Gu, PNAS, 2020, 117, 29512–29517.
33 M. C. Chen, S. F. Huang, K. Y. Lai, and M. H. Ling, Biomaterials, 2013, 34, 30773086.
34 P. C. DeMuth, Y. Min, D. J. Irvine, and P. T. Hammond, Adv. Healthc. Mater., 2014, 3, pp.4758.
35 Y. Gao, M. Hou, R. Yang, L. Zhang, Z. Xu, Y. Kang and P. Xue, Macromol. Mater. Eng., 2018, 303,
1800233.
36 E. Drucker, P.-G. Alcabes and P.-A. Marx, Lancet, 2000, 358, 1989–1992.
37 D. Ding, J. Pan, S. H. Lim, S. Amini, L. Kang, and A. Miserez, J. Mater. Chem. B, 2017, 5, 8467–8478.
38 C. Di Natale, D. De Rosa, M. Profeta, R. Jamaledin, A. Attanasio, E. Lagreca, P. L. Scognamiglio, P.
A. Netti and R. Vecchione, J. Mater. Chem. B, 2021, 9, 392–403.
39 M.-A. Lopez-Ramirez, F. Soto, C. Wang, R. Rueda, S. Shukla, C. Silva-Lopez, D. Kupor, D.-A.
McBride, J.-K. Pokorski, A. Nourhani, N.-F. Steinmetz, N.-J. Shah and J. Wang, Adv. Mater., 2020,
32, 1905740.
40 C.-E. Boone, C. Wang, M.-A. Lopez-Ramirez, V. Beiss, S. Shukla, P.-L. Chariou, D. Kupor, R. Rueda,
J. Wang and N.-F. Steinmetz, ACS Appl. Nano Mater., 2020, 3, 8037–8051.
Page 23 of 24 Journal of Materials Chemistry B
24
41 B. Esteban-Fernández de Ávila, P. Angsantikul, J. Li, M. A. Lopez-Ramirez, D. E. Ramírez-Herrera,
S. Thamphiwatana, C. Chen, J. Delezuk, R. Samakapiruk, V. Ramez, M. Obonyo, Z. Liangfang and
J. Wang, Nat. Comm. 2017, 8, 1–9.
42 A. Nourhani, E. Karshalev, F. Soto, and J. Wang, Research. 2020, 2020,1–9.
43 A. Devers and B.-S. Galer, Clin. J. Pain, 2000, 16, 205–208.
44 J. Kennedy, J.-M. Roll, T. Schraudner, S. Murphy and S. McPherson, J. Pain, 2014, 15, 979–984.
45 B. Mandal, D. Das, A.-P. Rameshbabu, S. Dhara and S. Pal, RSC Adv., 2016, 6, 19605–19611.
46 K. Bekkour, D.-S. Waterhouse and S.-S. Wadhwa, Food Res. Int., 2014, 66, 247–256.
47 R. Biswal and R.-P. Singh, Carbohydr. Polym., 2004, 57, 379–387.
48 Y. Chenga, B. Fengb, X. Yanga, P. Yanga, Y. Dinga and Y. Chena, Sens. Actuators, B., 2013, 182,
288–293.
49 B. Mukherjee, S. Mahapatra, R. Gupta, B. Patra, A. Tiwari and P. Arora, Eur. J. Pharm., 2005, 59,
475–483.
50 P. Minghetti, A. Casiraghi, F. Cilurzo and L. Montanari, Eur. J. Pharm., 2000, 10, 111–117.
51 S. Tokunaga, K. Ono, S. Ito, T. Sharmin, T. Kato, K. Irie, K. Mishima, T. Satho, T. Harada, T.-M. Aida
and K. Mishima, J. Supercrit. Fluids, 2021, 167, 105044.
52 M. Cetin, A. Atila and Y. Kadioglu, AAPS PharmSciTech., 2010, 11, 1250–1256.
53 B. Esteban-Fernández de Ávila, M.-A. Lopez-Ramirez, R. Mundaca-Uribe, X. Wei, D.-E.
Ramírez-Herrera, E. Karshalev, B. Nguyen, R.-H. Fang, L. Zhang and J. Wang, Adv. Mater., 2020,
32, 2000091.
54 X. Lan, W. Zhu, X. Huang, Y. Yu, H. Xiao, L. Jin, J.-J. Pu, X. Xie, J. She, V.-W.-Y. Lui and H.-J. Chen,
Nanoscale, 2020, 12, 18885–18898.
55 M. Wang, Y. Han, X. Yu, L. Liang, H. Chang, D.-C. Yeo, C. Wiraja, M.-L. Wee, L. Liu, X. Liu and C. Xu,
Adv. Healthc. Mater., 2020, 9, 1900635.
56 M.-R. Prausnitz, Adv. Drug Deliv. Rev., 2004, 56, 581–587.
57 L. Wang, Z. Fan, J. Zhang, Y. Changyi, C. Huang, Y. Gu, Z. Xu, Z. Tang, W. Lu, X. Wei and C. Li, Int. J.
Cancer, 2015, 136, E107
Page 24 of 24Journal of Materials Chemistry B