Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

International Journal of Biological Macromolecules 170 (2021) 602–621

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules

journal homepage: http://www.elsevier.com/locate/ijbiomac

Review

Biodegradable microneedles fabricated with carbohydrates and proteins:


Revolutionary approach for transdermal drug delivery
Neha Dabholkar a,1, Srividya Gorantla a,1, Tejashree Waghule a, Vamshi Krishna Rapalli a, Avinash Kothuru b,
Sanket Goel b, Gautam Singhvi a,⁎
a
Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, Pilani Campus, Rajasthan, India
b
Department of Electrical and Electronics Engineering, Principal Investigator: MEMS, Microfluidics and Nanoelectronics Lab, Birla Institute of Technology and Science (BITS), Pilani, Hyderabad
Campus, Hyderabad, India

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

Article history: There has been a surge in the use of transdermal drug delivery systems (TDDS) for the past few years. The market
Received 1 October 2020 of TDDS is expected to reach USD 7.1 billion by 2023, from USD 5.7 billion in 2018, at a CAGR of 4.5%.
Received in revised form 22 December 2020 Microneedles (MNs) are a novel class of TDDS with advantages of reduced pain, low infection risk, ease of appli-
Accepted 23 December 2020
cation, controlled release of therapeutic agents, and enhanced bioavailability. Biodegradable MNs fabricated from
Available online 31 December 2020
natural polymers have become the center of attention among formulation scientists because of their recognized
Keywords:
biodegradability, biocompatibility, ease of fabrication, and sustainable character. In this review, we summarize
Biodegradable microneedles the various polysaccharides and polypeptide based biomaterials that are used to fabricate biodegradable MNs.
Pullulan Particular emphasis is given to cellulose and its derivatives, starch, and complex carbohydrate polymers such
Hyaluronic acid as alginates, chitosan, chondroitin sulfate, xanthan gum, pullulan, and hyaluronic acid. Additionally, novel
Silk fibroin biopolymers protein-based polymers such as zein, collagen, gelatin, fish scale and silk fibroin (polyamino acid) biopolymers
Transdermal drug delivery application in transdermal drug delivery have also been discussed. The current review will provide a unique per-
spective to the readers on the developments of biodegradable MNs composed of carbohydrates and protein poly-
mers with their clinical applications and patent status.
© 2020 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 603
2. Mechanism of drug delivery from MNs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
3. Types of microneedles and drug delivery approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
3.1. Solid microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
3.2. Coated microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 605
3.3. Dissolving microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 606
3.4. Hollow microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 606
3.5. Hydrogel forming microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 607
3.6. Biodegradable microneedles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 607
4. Biodegradable microneedles: mechanism, natural polymers used for their fabrication, and applications. . . . . . . . . . . . . . . . . . . . . . 608
4.1. Polysaccharides or carbohydrate polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 608
4.1.1. Alginate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 608
4.1.2. Cellulose and its derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 609
4.1.3. Chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 609
4.1.4. Chondroitin sulfate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 609
4.1.5. Hyaluronic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 609
4.1.6. Sugars . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 609

⁎ Corresponding author.
E-mail address: gautam.singhvi@pilani.bits-pilani.ac.in (G. Singhvi).
1
Authors contributed equally.

https://doi.org/10.1016/j.ijbiomac.2020.12.177
0141-8130/© 2020 Elsevier B.V. All rights reserved.
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

4.1.7. Xanthan gum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610


4.1.8. Pullulan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
4.1.9. Bletilla striata . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
4.2. Polypeptides or protein polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
4.2.1. Zein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
4.2.2. Collagen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
4.2.3. Gelatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
4.2.4. Fish scale polymer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 613
4.2.5. Silk fibroin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 613
4.3. Polysaccharide and polypeptide composites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 613
5. Biocompatibility and biodegradation of the polysaccharide and polypeptide MNs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 614
6. Clinical applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 615
7. Patent status . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 615
8. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 616
9. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 617
Declaration of competing interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 617
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 617

1. Introduction compatibility, improved patient compliance, and termination of dose


simply by removing the patch [5,6].
Human skin is a protective barrier that only allows one-way chan- The most commonly used TDDS includes hypodermic needles and
nelization. It prevents the entry of external matter into the body. Thus, transdermal patches. However, the needles are less accepted by patients
the skin resists absorption of chemicals that come in contact with it; because of the pain associated with their use, and transdermal patches
making drug delivery through the skin cumbersome [1,2]. Since the ap- may be limited to delivery of small molecules as they have to diffuse
proval of the first transdermal drug delivery system (TDDS), Transderm through several layers before reaching the systemic circulation [7].
SCOP (for travel-associated nausea and vomiting) in 1979 by the FDA, MNs combine the advantages and overcome the shortcomings of hypo-
there has been a surge in the use of TDDS [3]. The market of TDDS is ex- dermic needles and transdermal patches. Fig. 1 depicts drug delivery by
pected to reach USD 7.1 billion by 2023, from USD 5.7 billion in 2018, at various transdermal delivery systems. A comparative description be-
a CAGR (Compound Annual Growth Rate) of 4.5% [4]. tween topical cream, transdermal patch, hypodermic needle, and
The term ‘transdermal delivery’ is used for the diffusion of a solute microneedle drug delivery systems is presented in Table 1 [7].
through the different layers of the skin to reach the systemic circulation The use of microneedles (MNs) is a novel, third-generation approach
and elicit a therapeutic effect. For a drug to be delivered through the to optimize TDDS [8]. MN delivery system was first proposed as a US
skin by passive transport, it must be lipophilic and have a molecular patent by Gerstel and Place in 1971 [9,10]. The MNs should be fabricated
weight <500 Da. This limits the transdermal delivery of drugs. Active with suitable length and depth devoid of stimulating the underlying
and passive approaches have been used to optimize TDDS. In the active dermis and blood vessels. Typically, MNs are 250–1000 μm in length,
approach, drug delivery is enhanced using physical methods. In the pas- 50–250 μm in width, and 1–25 μm in diameter. When the MNs are ar-
sive approach, the drug or the formulation is optimized to improve skin ranged in arrays on a patch backing, it is known as a MN patch [11].
permeation. However, the passive approach cannot be used for drugs The common terminology and parameters used in describing the archi-
having molecular weight >500 Da [3,5]. Transdermal delivery of drugs tecture of a typical MN patch are presented in Fig. 2(a) [12]. A MN setup
offers certain advantages including avoidance of drug degradation in has four components: drug reservoir, micropump, microflow sensor,
the gastrointestinal tract (GIT), overcoming intra- and inter-subject var- and MNs. The drug reservoir stores and retains drug formulation.
iability, retention of the drug at the therapeutic concentration for a Micropumps inject the drug formulation from the reservoir to the nee-
prolonged period, minimization of dosing frequency, enhanced dle. The flow sensor monitors and regulates the drug flow through MNs

Fig. 1. Comparison of topical cream, hypodermic needle, microneedle patch and transdermal patch (Reprinted with permission from ref. [7], copyright © Elsevier).

603
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Table 1
Comparison between topical cream, transdermal patch, hypodermic needle, and microneedle drug delivery systems.

Topical cream Transdermal patch Hypodermic needle Microneedle

Description Emulsion/emulgel/cream/ointments Adhesive patch to be placed on the Fine, hollow tube having a Micron size needles are aligned on the surface
skin. sharp tip with small of a small patch.
opening at the end.
Onset of action Slow Slow Faster Faster
Pain Painless Painless Painful Painless
Bioavailability Poor Insufficient Sufficient Sufficient
Patient compliance less Better less Better
Self-administration Possible Possible Not possible possible
Mechanism of drug Permeation through skin pores. Drug has to cross stratum corneum Drug placed directly in the Bypass stratum corneum, and drug placed
deliver barrier, thus poor diffusion of large dermis. directly into epidermis or dermis, resulting in
molecules. enhanced permeability.

[8]. At a commercial level, MNs have been used for the delivery of small transepidermal route, the molecules pass through the stratum
and large drug molecules. MNs pierce the stratum corneum of the skin corneum; intracellularly through the corneocytes, and intercellularly
and produce micron-sized pore channels, which act as a direct route through the intercellular spaces. The intracellular route allows penetra-
for drug delivery [11]. tion of polar or hydrophilic solutes, whereas the intercellular route al-
Chen et al. reported that MNs maintained the concentration of insu- lows penetration of non-polar or hydrophobic solutes. In the
lin for a longer period compared to s.c. injection. The maximum change transappendegeal route, the molecules pass across the hair follicles,
in glucose level was obtained after 1 h of administration of the MNs and and via sweat glands [33–36]. Understanding of kinetics is necessary
s.c. injection. The blood sugar continued to decrease in the subcutane- to design a TDDS. Percutaneous absorption is an important step in trans-
ous injection group, reaching its lowest level after 2 h. In contrast, in dermal delivery. The penetration of molecules into various layers of skin
the MN group, the lowest level was reached after 3 h [13]. MNs are and their permeation into the systemic circulation is termed as percuta-
also superior to transdermal patches. In a study conducted by neous absorption. It is a step-wise process that involves (i) penetration,
Wermeling et al., the systemic administration of a hydrophilic drug, nal- (ii) partitioning from the stratum corneum into the epidermis, (iii) dif-
trexone, was demonstrated using MNs. Upon transdermal delivery of fusion into the upper dermis, (iv) permeation, and (v) absorption into
naltrexone using the transdermal patch, no plasma levels were detected the systemic circulation [33].
even after 72 h of application. However, upon pretreatment with MNs, The mechanism of drug delivery from MNs is based on mechanical
steady-state plasma concentration was obtained within 2 h of applica- disruption of the skin and delivery of drugs within the epidermis.
tion and the concentration was retained for at least 48 h. Thus, MNs Thus, the drug reaches the target site readily bypassing the stratum
can provide a rapid onset of action [14]. The advantages of MNs include corneum barrier. The active substance is dissolved/dispersed/entrapped
ease of administration of large molecules [15,16], minimally painful and in the MNs. After application, the MNs either create micron-sized chan-
invasive, decreased microbial penetration compared to a hypodermic nels in the skin or completely degrade in the skin releasing the encapsu-
needles [17], targeting of specific skin area [18], dose reduction, lated drug without any hazardous residue left behind [37]. The MNs
accurate dosing [19], rapid drug delivery by coupling with electrically pierce the upper skin layers and enable the delivery of the drug directly
controlled micropump [20,21], rapid onset of action and self- into the epidermis/upper dermis region which is otherwise not possible
administration. MNs have been used for the transdermal delivery of a by passive diffusion alone. As micron-sized channels are created, even
wide array of therapeutic agents including, vaccines [22–24], insulin macromolecules like hormones, peptides, and immunobiologicals can
[25,26], hormones [27], enzymes [28], contraceptives [29], and anti- be delivered efficiently.
cancer drugs [30,31].
Despite having these many advantages, MNs possess some limita-
3. Types of microneedles and drug delivery approaches
tions. Solid MNs fabricated by silicon, glass, metal, ceramics are used
as first-generation MNs. These are used for transdermal drug delivery
Microneedle is a micron-sized needle with appropriate size ranges
due to its tremendous mechanical strength and biocompatibility. The
i.e. 150–1500 μ (height), 50–250 μ (base width), and 1–25 μ (tip diam-
solid MNs fabricated by silicon, metal, ceramics can overcome the skin
eter) [38,39]. It directly pierces the stratum corneum barrier without
barriers but exhibits limitations such as limited drug loading, skin
stimulating nerves and damaging blood vessels. The above-mentioned
break up, and costly fabrication procedure. The material used to fabri-
dimensions are mostly used for transdermal drug delivery because of
cate MNs, affects its permeability, flexibility, and toughness. In recent
advantages such as improved drug bioavailability, safety, minimally in-
days, to overcome these limitations, polymers are the most widely
vasive, and proficient drug delivery. Fig. 2(b) shows the transdermal
used materials for MNs fabrication [32]. The biomaterials obtained
and lymphatic drug delivery using microneedle [12]. Morphologically,
from various plant and animal kingdoms such as polysaccharides and
MNs can be classified into five types: solid, coated, dissolving, hollow,
protein-based biopolymers have attracted significant attention in the
and hydrogel-forming MNs [40]. Each type of MN has its own way of de-
realm of MNs research. This review provides a view on the types of
livering drugs into the epidermis. Different types of microneedles with
MNs and drug delivery approaches properties of polysaccharides and
their drug delivery methods are presented in Fig. 2(c) [12].
protein-based biopolymers which make them suitable for MNs fabrica-
tion. Additionally, it gives the recent update on the applications of MNs
fabricated from cellulose and its derivatives, sugars, complex carbohy- 3.1. Solid microneedles
drates and protein biopolymers in drug delivery.
Solid MNs use the “poke-and- patch” approach. Solid MNs are made
from different types of metals like silicon, stainless steel and titanium.
2. Mechanism of drug delivery from MNs They are mostly used for pretreatment of the skin to create micropores.
Upon subsequent application of a drug patch, the drug enters the skin
There are two routes of penetration of drug across the intact with decreased resistance [9,20,32,41]. In a study, super-short solid sil-
skin; transepidermal route and transappendegeal route. In the icon MNs (70–80 μm) were found to be better than sharp-tipped MNs in

604
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Fig. 2. (a) Schematic highlighting common terminology and parameters used in describing the architecture of a typical microneedle patch. A microneedle patch consists of micron
projections which are commonly known as microneedles supported on backing layer (or baseplate). The tip of a microneedle (as shown in the inset) may be characterized by the apex
angle or radius of curvature which dictate the skin insertion capability. (b) Lymphotropic drug delivery using microneedles. Microneedles with typical lengths of 250–1000 μm would
penetrate the epidermis layer, reaching the dermis layer of the skin. The dermis is rich in blood and lymph vessels and the drug released in this layer will readily have exposure to
various types of immune cells. (c) Types of microneedles: Solid, hollow, coated, dissolving and hydrogel-forming microneedle (Reprinted with permission from ref. [12], copyright ©
Elsevier).

enhancing skin permeation. Also, they are retained in the skin for a lon- a coated solid microstructured transdermal system (sMTS, developed
ger period compared to sharp MNs [42]. Narayanan and co-workers fab- by 3 M) for a potent peptide A and investigated the preclinical evalua-
ricated solid silicon MNs and coated them with gold to improve their tion using two different coating solutions. Approximately 250 μg of
mechanical strength (Vickers hardness value: 3800 Hv) and biocompat- the peptide could be coated on 1.27 cm2 patch containing 316 MNs
ibility [43]. Solid MNs made of metals and silicon have been especially with precision. The absolute bioavailability for MNs coated with solu-
used in gene therapy [44]. Dermaroller® is a commercially available tion 1 and solution 2 was found to be 88% and 74%, respectively,
solid MN device that has been widely used in dermatology [45]. Thus, which was similar to s.c. bioavailability of 75%. The stability of the pep-
the solid MNs could be a promising candidate for increasing drug avail- tide at room temperature was improved. Additionally, skin irritation
ability in transdermal drug delivery systems. studies on porcine skin demonstrated minimal skin irritation [46]. Li
and co-workers coated MNs individually, instead of coating all MNs
3.2. Coated microneedles with the same formulation. Thus, individually coated MNs delivered
therapeutic agents with different physicochemical characteristics. The
Coated MNs use the “coat-and-poke” approach. The biomolecules or number of coating layers was modified to adjust the MN loading. It
medications are coated on the MNs [9,20,32,41]. Kapoor et al. developed was found that polyvinyl pyrrolidone (PVP) coating dissolved within

605
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

30 s in phosphate buffer solution. They reported that PVP K15 dissolved dissolving MNs [51]. In short, compared to solid MNs, the dissolving
even faster compared to PVP K30 [47]. The only limitation of the coated MNs are easy to prepare, biocompatible, provide high drug loading,
MNs is that these give rise to safety concerns due to the biohazardous and are appropriate and safe for transdermal drug delivery. Ling and
waste left behind after use. his coworkers developed a rapidly dissolving MN patch composed of
starch and gelatin for the systematic transdermal delivery of insulin.
3.3. Dissolving microneedles The MNs consisted of 225 (15 * 15) pyramidal needles with a height
of 600 μm, base width of 300 μm. They reported the starch/gelatin
Dissolving MNs use the “poke-and-release” approach. They are fab- MNs rapidly dissolved and released the encapsulated payload within
ricated by encapsulating the drug into water-soluble materials (polyvi- 5 min after skin insertion. The results indicated that the insulin-loaded
nyl pyrrolidone, chondroitin sulfate), sugars (maltose, dextran), or MNs efficiently reduced blood glucose levels in diabetic rats with a rel-
biodegradable polymers (polylactic acid, chitosan). Upon insertion of ative pharmacological availability and relative bioavailability of 92.2 ±
MNs in the skin, either dissolution or polymer degradation takes 3.5% and 92.3 ± 7.2%, respectively. Thus, dissolving starch/gelatin MNs
place, subsequently releasing the drug. Hence, the MN is not to be re- could be a promising approach for the transdermal delivery of various
moved after insertion. This provides a one-step application process con- biomolecules. The dissolution mechanism of rapidly dissolvable
venient for patients [9,20,32,41]. The choice of MN material affects the starch/gelatin microneedles are presented in Fig. 3 [52].
drug release from the MNs. MNs can be used to attain controlled release.
In a study, it was found that biodegradable MNs can provide controlled 3.4. Hollow microneedles
release of the drug for hours to months [48]. The drug release rate and
duration can also be regulated by controlling the rate of degradation Hollow MNs use the “poke-and-flow” approach. They consist of a
of polymers in the case of biodegradable MNs [28,49]. space that is filled with the drug solution or dispersion. Holes are pres-
Lahiji and co-workers developed tissue interlocking dissolving MNs ent at the MN tips. Upon insertion, the drug is directly deposited in the
using rhodamine B as the model drug. Sharp tip, narrow neck, and wide epidermal layer [9,20,32,41]. Norman et al. achieved targeted transder-
body of the MNs resulted in the tissue-interlocking property. The mini- mal injection using poly(lactic acid) hollow MNs. Upon injection of the
mum force required to penetrate the skin was less for the tissue- MNs, 90% of the material was targeted within the skin with negligible
interlocking MNs (0.1±0.07 N) compared to conventional MNs (0.19 leakage onto the surrounding surface [53]. Mishra and co-workers fab-
±0.06 N) [50]. The elastic nature of the skin prevents dissolving MNs ricated hollow MNs using SU-8 on a silicon substrate. The maximum
from complete insertion and accurate drug delivery into the skin. To en- compressive force was found to be 0.27 N per needle, which allowed
counter this issue, a patchless, micro-pillar integrated dissolving MN successful penetration through the rat and mice skin without any break-
was fabricated. Polymethyl methacrylate was used to make the micro- age. Microfluidic characterizations demonstrated hollow MN lumen de-
pillars and hyaluronic acid was used to fabricate the MNs. Interestingly, velopment with a flow rate of 0.93 μL per sec at 2 KPa pressure
the patchless MNs increased the skin penetration of the drug up to difference at the inlet [54]. The unique characteristics of hollow MNs
97.78% ± 2.22%, compared with 44.44% ± 7.85% in the conventional such as delivering quantitative drugs at different pressure-driven flow

Fig. 3. Schematic illustration of transdermal delivery of insulin using starch/gelatin microneedles (MNs), which can rapidly dissolve in the skin to release encapsulated insulin (Reprinted
with permission from ref. [52], copyright © Elsevier).

606
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

rates, delivering the drug directly into the epidermis, appropriate me- biodegradable polymer forms a matrix that degrades in the skin. Thus,
chanical strength makes them potential candidates for transdermal no residues from the MNs remain in the body [55,56]. Li et al. developed
drug delivery. solid polylactic acid MNs for pretreatment of the skin to enhance drug
permeation and absorption. Insulin was used as the model drug to
3.5. Hydrogel forming microneedles study in vivo activity in diabetic mice. Due to the increased permeability
of insulin upon MNs pretreatment, the blood glucose levels of the mice
Hydrogel-forming MNs are a recent addition to the MN delivery sys- were reduced to 29% of the administered level at 5 h [57]. Park et al. de-
tems. They are also known as swellable MNs. Super-swelling polymers veloped multilayered polymeric MNs composed of poly(lactic-co-
are used to fabricate the MNs. When swellable MNs are inserted into glycolic acid) (PLGA) and polyvinyl pyrrolidone exhibited controlled
the skin, the biological fluids are absorbed by the MNs because of the drug release. Bovine serum albumin (BSA) was used as the model
polymeric cross-linking and hydrophilic groups. Thus, the high concen- drug, and spray deposition was used to form the multiple layers. In
tration of the drug from the MNs is released into the circulation. The vitro release studies demonstrated that BSA in the polyvinyl pyrrolidone
major disadvantage of these MNs is poor mechanical strength. Attain- layer was released within 30 min, whereas 60% of BSA in PLGA was re-
ment of balance between the mechanical strength and swellability by leased within 1 h, followed by slow continuous release over 7 days. This
choosing appropriate polymeric material and process for fabrication of approach could be used to modulate the drug release profile using bio-
MNs is required for optimal use of swellable MNs [7,32]. The advantages degradable polymers [58].
and disadvantages of the different types of MNs have been listed in Biodegradable MNs can be easily fabricated using a variety of
Table 2 [7,9,20,32,41]. methods such as microinjection, templating, or lithography at room
Considering the advantages of dissolving/ biodegradable MNs over temperature. They are particularly suitable for delivery of drugs
other types, the different materials used for their preparation and the [59–61], vaccines [23,62,63], DNA [44,64], and proteins. MNs can be
recent studies will be elaborated further. The current scenario of the used to substitute conventional sustained-delivery systems. However,
therapeutic applications of biodegradable MNs will be understood. they must remain in the skin to utilize the biodegradability of the poly-
mer effectively [48,55]. Recently, PLGA MNs were used to formulate in-
3.6. Biodegradable microneedles tradermal implants for etonogestrel (third-generation progesterone).
The pharmacokinetic studies in rats demonstrated that the plasma
Biodegradable MNs use different kinds of biodegradable polymers etonogestrel level was detectable until 36 h. The AUC0→48h only
including polylactic acid, polyglycolic acid, poly(lactide-co-glycolide) accounted for 37.8% of AUC0→∞, suggesting a sustained effect [65]. Bio-
(PLGA), or chitosan. The drug is encapsulated within the biodegradable degradable MNs can be used for the controlled intradermal release of la-
MNs, followed by insertion in the skin. After application, the bile molecules such as enzymes [66]. Tsioris et al. developed silk MNs for

Table 2
Classification of microneedles [7,9,20,32,39].

Type of MNs Approach Characteristics Advantages Disadvantages

Enhances • Poor dose accuracy


• Mechanical • Requires healing after applica-
Creates microchannels in the skin. Upon subsequent application of strength tion
Solid MN Poke-and-patch
a drug patch, the drug enters skin with decreased resistance. • Physical sta- • Extends time of occlusion
bility • Reuse may cause infection
• Drug loading • Poor biocompatibility
Enhances
• Poor biocompatibility
• Stability of • Limited dose
Coated MN Coat-and-poke Enhance dissolution of biomolecules coated on the MN. drug • Peeling during insertion
• Mechanical • Formulation migration
strength
• Enhances sta-
bility of drug
• Low-cost
manufactur- • Limited dose
ing • Poor mechanical strength,
Dissolving MN Poke-and –release Fabricated with biodegradable polymers or sugars.
• Ease of biocompatibility, and physical
manufactur- stability
ing
• Controlled
drug release
• Sample
extraction • Clogging
Consists of a vacant space inside. Drug solution or dispersion can be
Hollow MN Poke-and-flow • Dose accuracy • Ingressing body fluids
filled in the hollow MNs.
• Constant flow • Requires prefilled syringe
rate
• Biodegradable
• Ease of
manufactur-
ing
• Poor mechanical strength and
Hydrogel-forming/swellable • Improves
– Absorb the biological fluids and swell. Excellent swellability. physical stability
MN drug loading
• Ingressing body fluids
• Controlled
drug release
• Easy
sterilization

607
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

the delivery of the horseradish peroxidase enzyme. The results revealed 4.1. Polysaccharides or carbohydrate polymers
that by controlling the secondary structure of silk protein, the drug re-
lease rate decreased by 5.6-fold [28]. The polysaccharides that are used to fabricate MNs include cellulose
The biodegradable MNs gained significance over the other MNs due and its derivatives, starch, and complex carbohydrate polymers such as
to advantages such as precise loading, intact removal of skin, and the at- alginates, chitosan, chondroitin sulfate, xanthan gum, pullulan, and
tachment of the drug reservoir. The natural biopolymers as a material hyaluronic acid.
have been used in medical applications for centuries, and have gained
immense importance in drug delivery [67]. The following section dis-
cusses the various polysaccharides and polypeptides-based biomate- 4.1.1. Alginate
rials used to fabricate biodegradable MNs. Additionally, Alginate is a polyanionic polysaccharide comprising of β-D-
biocompatibility and biodegradation of these biomaterials and their ap- mannuronic acid and α-L-guluronic acid. Alginates are obtained from
plications in transdermal drug delivery will be discussed thoroughly. algae and bacteria. The commercial source of alginates is brown algae
(comprising 40% of the dry matter). Durvillea, Sargassum, Macrocytis,
Eclonia, Ascophyllum, Laminaria, and Lessonia species are used for com-
4. Biodegradable microneedles: mechanism, natural polymers used mercial production. Bacteria belonging to Azotobacter, and Pseudomonas
for their fabrication, and applications species are also used for the production of alginates. These bacteria pro-
duce alginate-like polysaccharides as an extracellular material. How-
Majority of the biodegradable MNs are manufactured using natural ever, this method is not economically feasible and is used only for
biopolymers that degrade after skin insertion [67]. The degradation pro- small-scale research studies [69–71]. Alginate is biodegradable and bio-
cess takes place over a prolonged time in a biological environment compatible. However, its use for the fabrication of MNs has been with-
under the influence of enzymes. The major advantages of this degrada- held due to its poor mechanical strength. Despite this, initial efforts
tion include formation of nontoxic metabolites, and their elimination suggested that alginate MNs can create microholes in the skin without
from the body by natural metabolic pathways [68]. The degradation any deformation [72,96,97]. The “poke-and-release” strategy of alginate
process takes a longer time compared to dissolvable MNs. Hence, biode- MNs was found to be very helpful because it prevents MNs reuse, the
gradable MNs are gaining significance for the sustained delivery of ther- risk of biohazards, and cross-contaminations. Demir et al. developed so-
apeutics. The release rate of incorporated therapeutic from these MNs is dium alginate MNs for the transdermal delivery of bovine serum albu-
managed by modulating the degradation rate of the chosen biodegrad- min. They observed that the MNs patch increased the permeation of
able polymer [56]. bovine serum albumin across human abdominal skin by 15.4 fold com-
The sources and properties of natural polymers such as polysaccha- pared to the needle-less sodium alginate patch [97]. Cross-linked algi-
rides (e.g. cellulose, starch), and complex carbohydrate polymers that nate blended with maltose was used to prepare MNs for transdermal
make them suitable for the fabrication of biodegradable MNs are com- delivery of insulin. The MNs exhibited enhanced mechanical properties
piled in Table 3 [69–95]. due to the addition of maltose [98].

Table 3
Carbohydrate polymers, their sources, and their properties that make them suitable for fabrication of microneedles.

Carbohydrate Sources Properties suitable for fabrication of MNs References


polymers

Alginate -Brown algae: Durvillea, Sargassum, Macrocytis, Eclonia, -Microhole creation in the skin without producing any deformation [69–72]
Ascophyllum, Laminaria, and Lessonia species Thickening agent
-Bacteria: Azotobacter, and Pseudomonas species
Cellulose and -Wood -Durability [73–75]
its derivatives -Algae: Chaetamorpha melagonicum, Valonia ventricosa -Transparency
-Bacteria: Acetobacter, Agrobacterium, Rhizobium, Sarcina -Elasticity
species -Viscosity
-Bacterial cellulose has higher water absorption and retention capacity, low
solubility, high mechanical strength, resistance to degradation, and nano-scaled
and uniform fiber network.
Chitosan Crustaceans such as crabs, shrimps, lobsters, and krill. -Dissolvable [76–79]
-Swellable
-Absorbs and retains large amounts of water
- Natural antibacterial property
-Wound healing
Chondroitin Porcine skin and rib cartilage, bovine trachea, and shark -Water soluble [80–82]
sulfate (CS) cartilage. -Possesses bioactivity
Hyaluronic acid -Animal sources: Rooster combs, bovine synovial fluid, human -Versatility [83–87]
(HA) umbilical cord, and vitreous humor of cattle. -Enhanced permeability
-Bacterial production: Streptococci sp., Agrobacterium sp. -Sustained release
Bacillus subtilis, and Escherichia coli. -Skin hydration
-In vitro: Enzymes from Streptococcus pyogenes and Pasteurella -Viscoelasticity
multocida. -Tissue re-epithelization
Sugars Sugar cane and beet are major sources. Other sources include -Non-toxicity [88–92]
honey, corn, fruits and vegetables. -Minimal cytotoxicity
-Affordability
-Water soluble
-Antimicrobial properties
Xanthan gum Bacteria: Xanthomonas campestris, Xanthomonas arboricola -Excellent solubility in water [93–95]
(XG) and Xanthomonas axonopodis. -Retards drug release
-Time independent release kinetics
-Viscosity enhancement

608
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

4.1.2. Cellulose and its derivatives prolonged release of rhodamine B for >72 h. Therefore, these MNs can
Cellulose is the most abundant structural component of herbal cells be suitably used for the delivery of peptides [108]. Thiolated chitosan
and tissues. The primary source of cellulose is wood. Commercially, cel- can also be used for preparing MN patch [104]. Chi et al. developed a
lulose is obtained from wood or cotton. The other sources of cellulose temperature-responsive chitosan MNs patch for wound healing. This
include algae and bacteria [73,74]. The cellulose obtained by biosynthe- smart delivery system is a novel approach that can be utilized in
sis in living organisms consists of glucose monomers only that are wound healing [109].
linked by β-(1,4) linkages [99].
The two important cellulose derivatives are cellulose ethers and cel- 4.1.4. Chondroitin sulfate
lulose esters. Cellulose ethers possess commercially important proper- Chondroitin sulfate (CS) is a sulfated glycosaminoglycan consisting
ties of solubility, viscosity, surface activity, and stability [74]. of disaccharide units of d-glucuronic acid and N-acetyl-galactosamine
Carboxymethyl cellulose (CMC) is one of the most important cellulose linked by β-(1,3) glycosidic bonds. There are two major CS, A and C, in
ethers. It is highly soluble in water and produces a clear solution, mak- which the sulfate group is present at different positions. The sulfate
ing it a pharmaceutically acceptable polymer [100]. Cellulose esters are group is present in the 4th position in CS A and the 6th position in CS
water-insoluble film-forming polymers. Examples of cellulose esters in- C. The sulfate group provides hydrophilicity to CS [80,81]. CS is a
clude cellulose acetate, cellulose acetate phthalate and cellulose nitrate major component of cartilage and imparts elasticity to the cartilage.
[74]. Park et al. mixed CMC with amylopectin to enhance the mechani- CS can be predominantly obtained from porcine skin and rib cartilage,
cal and dissolution properties of MNs for the controlled release of drugs bovine trachea, and shark cartilage [82]. The concentration and compo-
[101]. Garcia et al. fabricated self-sterilizing CMC MNs loaded with silver sition of CS depend on the source, terrestrial or marine, from which it is
nanoparticles. These MNs can be used as vehicles for vaccine and drug obtained. CS exhibits antioxidant, anticoagulant, anti-inflammatory,
delivery [102]. The havoc caused by SARS-CoV-2 is being witnessed on and anti-thrombus activities making it beneficial in joint-related pathol-
the global level. Kim et al. developed a MNs-assisted recombinant coro- ogies [110]. CS is a water-soluble and biodegradable polysaccharide
navirus vaccine using CMC (molecular weight: 90 kDa) by a three-stage [111]. Poirier et al. developed a dissolvable MN array composed of CS
MNs manufacturing process. The results demonstrated that significantly and hydroxyethyl starch. It was loaded with hepatitis B surface antigen
high SARS-CoV-2 IgG responses were detected as early as week 2 for all with QS-21 saponin as an adjuvant. The antigenicity was retained at
subunit vaccines compared to control groups [24]. 37 °C and 45 °C. Only a 10% loss was observed at 50 °C after 6 months
[112]. Siqi Liu et al. fabricated recombinant Staphylococcal enterotoxin
4.1.3. Chitosan B-loaded dissolvable MNs composed of CS (2%) and trehalose (0.8%).
Chitosan is a marine polysaccharide obtained by chemical purifica- The MNs easily penetrated the skin within 5 min with a skin penetration
tion of chitin. Chitin is extracted from crustaceans such as crabs, depth of 260 μm. The MNs extended the antigen retention time in vivo
shrimps, lobsters, and krill. Chitosan is obtained commercially by significantly [113].
deacetylation of chitin. Chitosan is also found in the cell walls of certain
fungi [76,78]. Chitosan is a linear biopolymer formed by D-glucosamine 4.1.5. Hyaluronic acid
and N-acetyl-D-glucosamine linked by β-(1,4) bond [76,77]. The molec- Hyaluronic acid (HA) or hyaluronan is a linear, non-sulfated glycos-
ular weight of chitosan is between 300 and 1000 kDa (source depen- aminoglycan. It consists of N-acetyl-D-glucosamine and D-glucuronic
dent). Properties such as molecular weight, degree of deacetylation, acid linked by β-(1,4) glycosidic bonds [84]. The strength of MNs is an
and moisture content determine the stability and the mechanism of essential characteristic for dermal penetration. The increase in the HA
degradation of a material. It was reported that the lower molecular molecular weight and concentration leads to reinforcement of the
weight-based chitosan exhibits low mechanical strength. To overcome three-dimensional polymer network [114].
the limitations of low mechanical strength of chitosan MNs, they were HA is present in the extracellular matrix of connective tissue of most
mounted on to PLGA as a supporting array [103]. In another study, vertebrates. It is synthesized by a class of proteins known as HA
thiolated chitosan was selected for MN fabrication, as the presence of synthases [115]. High concentrations of HA are found in the skin, umbil-
the thiol group exhibits high mechanical strength and high-water up- ical cord, synovial fluid of joints, and vitreous humor of the eye. 50% of
take ability. This enhanced the overall swelling properties of the poly- HA in the human body is present in the skin [83]. It has been found
mer. The MNs prepared with 2% thiolated chitosan exhibited ease of that rooster combs contain 7.5 mg/g of HA, which is the highest re-
fabrication, and enhanced sharpness and tensile strength of the needle ported in an animal tissue [116]. HA is also present in the pathogenic
compared to 1% and thiolated chitosan. High concentration (3% microbe Streptococci and non-pathogenic microbes including Enterococ-
thiolated chitosan) was difficult to pour and fill the micropores of the cus faecalis, and Bacillus subtilis [83,117]. HA enhances the permeability
template [104]. and helps in achieving sustained release. Zhu et al. have described the
Chitosan is bioabsorbable. However, it is poorly soluble in aqueous possible mechanisms producing the aforementioned effects [84]. HA
solutions at neutral pH. It is degraded slowly by lysozymes and possesses many unique advantages, allowing its use in TDDS. HA is a
chitosanases. Chitosan behaves like a hydrogel because it absorbs and suitable material for making MNs. This is because of its biocompatibility
retains large amounts of water, allowing it to swell without dissolving and biodegradability. Also, because of its versatile nature, many differ-
completely [76]. Additionally, it possesses unique properties such as ent formulations such as solutions, nanorods and nanostructured lipids
natural antibacterial and wound healing properties [105]. Chen et al. de- can be easily loaded [84,118–120].
veloped chitosan MN patches using bovine serum albumin (BSA) as the HA based MNs were used to enhance lymphatic delivery via the
model drug. In vitro studies performed on porcine skin demonstrated transdermal route. Lymphatic delivery can be used essentially for vacci-
that BSA was released in a sustained manner for at least 8 days. Alexa nation and tumor chemotherapy [121]. Liu et al. used HA based MNs for
Fluor 488-labeled BSA MNs were applied on the rat skin in vivo. BSA dif- the transdermal delivery of insulin. It was found to be effective and safe
fused gradually into the dermal layer and the fluorescence was ob- for use in the clinic [26]. A combination of various approaches can help
served at a depth of 300 μm [106]. Chen et al. developed embeddable to optimize transdermal drug delivery. Hao et al. combined the ap-
MNs system using poly(L-lactide-co-D,L-lactide) as a supporting array proaches of near-infrared responsive PEGylated nanorods and HA MNs
for sustained intradermal release of encapsulated antigens [103]. Sadeqi for transdermal delivery of doxorubicin [120].
et al. presented fabrication of solid and hollow chitosan MNs using the
cost-effective method of cross-over lines laser engraving [107]. 4.1.6. Sugars
Chandrasekharan et al. prepared trifluoroacetic acid-treated water- Sugar, which commonly refers to sucrose, is a natural water-soluble
soluble chitosan for MN-assisted delivery. The MNs patch showed carbohydrate. Sugar can be obtained from various sources including

609
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

honey, corn, fruits, and vegetables; sugar cane and beet being the com- 4.1.9. Bletilla striata
mercial sources [88]. Sugars are safe and affordable. Sugars also exhibit Bletilla striata is a water-soluble polysaccharide extracted from tu-
antimicrobial properties against infection and wound causing patho- bers of Bletilla striata. It is a glucomannan comprising of α-mannose,
gens [7,122]. The commonest sugar used to fabricate MNs is maltose. β-mannose, and β-glucose. This polysaccharide is mostly used in the
Other alternatives include trehalose, galactose, sucrose, mannitol, and fabrication of gels, adhesives, and scaffolds because of its non-toxicity,
xylitol. However, sugars present problem in the processing (require- less cost, good stability, biocompatibility and biodegradability proper-
ment of thermal treatment), storage, and application on the skin ties [131]. Hu et al. developed rhodamine loaded dissolving MNs using
[89,90]. Yalcintas et al. conducted in vitro study to investigate the cyto- Bletilla striata polysaccharide. The texture analysis and histological ex-
toxicity of various carbohydrate-based materials such as glucose, treha- amination demonstrated that the MNs had sufficient mechanical
lose, CMC, maltodextrin, and HA. The results obtained from different strength to be inserted into the skin. After the removal of MNs, slight ir-
qualitative and quantitative analyses suggested that trehalose, CMC, ritation appeared on the skin but recovered within 24 h. In vitro studies
maltodextrin, and HA do not pose cytotoxicity challenges and thus can indicated that the rhodamine B-loaded MNs could provide sustained re-
be used to make MNs. However, glucose should be used at low concen- lease of the drug [132].
trations; as it was found to be toxic to the cells at concentrations above The summary of applications of MNs fabricated from polysaccha-
50 mg/mL [91]. Milewski et al. developed MNs for the delivery of an in- rides or complex carbohydrate polymers have been listed in Table 4.
vestigational peptide using MicroCor®. The coating solution contained
the peptide and sugars (dextran and sorbitol) [123]. Sugars such as tre- 4.2. Polypeptides or protein polymers
halose [63] and sucrose [124] have also been used to stabilize the for-
mulations for effective loading on the MNs. The polypeptide or protein biopolymers that are used to fabricate
MNs include, zein [30,158], collagen [159], gelatin [59,160,161], fish
scale biopolymer [60,162–164], and silk [28,165–168].
4.1.7. Xanthan gum
Xanthan gum (XG) is a high molecular weight heteropolysaccharide
4.2.1. Zein
secreted by the bacteria Xanthomonas campestris. XG can also be ob-
Zein is a water-insoluble prolamine protein found in maize. Zein is
tained from Xanthomonas arboricola and Xanthomonas axonopodis. The
generally recognized as a safe (GRAS) approved biodegradable polymer
bacteria produce XG at the cell wall surface by an enzymatic process
and sustainable agropolymer. Recently, it has been used in the fabrica-
during its normal life cycle [93,94]. XG consists of β-(1,4)-D-
tion of fibers, textile, plastic, chewing gums, and an adhesive. Bhatnagar
glucopyranose glucan backbone similar to cellulose with side chains of
et al. investigated the effect of physicochemical properties of chemo-
(3,1)-α-linked D-mannopyranose-(2,1)-β-D-glucuronic acid-(4,1)-β-D-
therapeutic agents (tamoxifen and gemcitabine) in loading behavior,
mannopyranose on alternating residues [125]. XG possesses good bio-
drug disposition within the skin, and permeation across the skin by
compatibility. It is soluble in both hot and cold water [93]. XG causes re-
zein microneedles. The loading capacity for gemcitabine and tamoxifen
tardation of drug release and can offer time independent release
was found to be 1459 ± 74 μg and 607 ± 21 μg respectively. The skin
kinetics [94,125]. For coating MNs successfully, different coating
permeation study on porcine skin revealed that after 48 h application
methods are used which require different viscosities and surface ten-
of zein MNs, tamoxifen was retained on viable epidermis whereas the
sion. Different excipients have been added to the coating solutions to
gemcitabine microneedles showed the greatest percentage permeation
obtain these desirable properties. XG has been used to increase the vis-
across excised skin via poke and patch approach [30].
cosity of the coating solution of MNs [95]. Increased viscosity leads to
improvement in coating thickness [126]. Kim et al. developed MNs
4.2.2. Collagen
coated with influenza virus-like particle vaccine. XG (1% w/v) was
Collagen is a protein with amino acids such as arginine, glycine, pro-
used as a viscosity enhancer in the coating solution. XG produced better
line, and hydroxyproline. It possesses high mechanical strength. Due to
hemagglutinin activity than carboxymethyl cellulose. However, the
its non-immunogenic, non-toxic, biodegradable, and biocompatible na-
coating dose of XG was significantly lower compared to carboxymethyl
ture, it is mostly applied for tissue engineering [159]. Aditya et al. pre-
cellulose. Hence, carboxymethyl cellulose was used as the viscosity en-
pared dissolvable collagen microneedles of tryptophan blue (model
hancer [127]. Choi et al. used XG (0.075% w/v) as the viscosity enhancer
drug) using a micro-molding technique. They investigated the effect of
with trehalose solution to prepare MNs coated with the whole
process parameters like biopolymers w/v ratio, MN height, pressure
inactivated influenza vaccine. Due to the addition of XG, the hemagglu-
drop, and duration of solidification on the production of MN. They re-
tinin activity increased significantly to 81% compared to trehalose alone
vealed that the duration of the microneedle inside the vacuum oven in-
which was 50–60% [128].
fluences the microneedle height. Similarly, increasing PVP
concentration increases solution viscosity and enhances drug loading,
4.1.8. Pullulan but it makes it difficult to penetrate the mold cavities [159].
Pullulan is a hydrophilic linear biopolymer obtained from yeast-like
fungus Aureobasidium pullulans and composed α-(1→6) maltotriose 4.2.3. Gelatin
residues. Due to the presence of more hydroxyl groups, the chemical Gelatin is a mixture of proteins derived by acid hydrolysis (type A
modification of pullulan made it easy. Thus, pullulan and pullulan deriv- gelatin) and alkaline hydrolysis (type B gelatin) of collagen. Gelatin
atives possess special characteristics like good film-forming ability, ad- has a molecular weight ranging from 15 to 400 kDa. The low molecular
hesiveness, and excellent mechanical performance and plays a key weight gelatin forms soft gels; high molecular weight gelatin provides
role in biomedical applications [129]. Vora et al. developed a dissolvable suitable strength to the MNs [169]. Gelatin is readily available, inexpen-
MN of pullulan for transdermal drug delivery of small molecule (meth- sive, and can be easily fabricated into different shapes. Recently gelatin
ylene blue and fluorescein sodium) and with biomolecules (fluorescein was studied in fields of bio-industry such as tissue engineering
isothiocyanate labeled bovine serum albumin and insulin). The ex vivo (cell-interactive coatings), drug delivery, biological glues, and tissue
porcine skin permeation studies demonstrated that 95–100% of MNs scaffolds. Gelatin is not utilized in fiber production because of its strong
penetrated, which is similar to results attained with solid MNs. The pre- hydrogen bonding and its potential to harden at low temperatures
pared dissolvable pullulan MNs successfully delivered the small mole- [170]. Yu et al. prepared gelatin and hydroxyapatite MNs for the deliv-
cule and biomolecules across neonatal porcine skin. Therefore, the ery of insulin. They reported that s.c. injection of insulin resulted in a
pullulan MNs patch could be efficient to penetrate the stratum corneum sharp increase of the plasma insulin concentration with the highest
[130]. value at 103.5 ± 9.5 μIU/mL in ~1 h, and it decreased rapidly to a

610
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Table 4
Summary of applications of microneedles fabricated from polysaccharides and complex carbohydrate polymers.

Carbohydrate Model drug Application Results References


polymer

Alginate Bovine serum albumin (BSA) Peptide and protein delivery MN patch increased the permeation of BSA across human abdominal [97]
skin by 15.4 fold compared to needle-less sodium alginate patch.
Alginate blended with HA was used to fabricate MNs. The relative
Insulin Diabetes mellitus [133]
bioavailability of insulin from MN patches was found to be 92.9 ± 7%.
Alginate blended with maltose was used to fabricate MNs. The MNs
Insulin Diabetes mellitus exhibited enhanced mechanical properties due to addition of maltose [98]
(highest failure force: 0.41 N/ needle approximately.).
Cellulose and Ascorbic acid (Vitamin C) Cosmetic products Amylopectin was mixed with carboxymethyl cellulose (CMC) to [101]
its enhance the mechanical and dissolution properties of MNs. Also, an
derivative enhanced anti-oxidant activity of ascorbic acid was obtained.
1% sodium CMC was used to prepare MNs. MNs formulated with
Influenza vaccine Influenza vaccination calcium heptagluconate and arginine had no significant loss in activity [22]
for influenza vaccine during storage at RT for 6 months.
The CMC MNs were self-sterilizing. Zones of inhibition developed
Vehicles for vaccine and drug
Silver nanoparticles within 24 h around nanosilver-loaded MNs but not around the plain [102]
delivery
CMC MNs.
CMC was used to prepare the base of MNs. Sustained release of the
Finasteride (FNS) Androgenetic alopecia implanted FNS powder for 3 days was obtained with only one [134]
application of FNS powder-carrying MNs.
12% CMC was used to fabricate MN backbone. The MNs delivered VA
Valproic acid (VA) Hair regrowth with higher accuracy than topical application and stimulated wound [135]
re-epithelialization signals involved in hair follicle regrowth.
Pups born to mice immunized with Ad5.ZIKV-Efl were aprotected
Zika virus vaccines: Ad5.ZIKV-Efl and against the infection without weight loss or neurological signs. Pups
Zika virus vaccination [23]
ZIKV-rEfl born to dams immunized with MNA-ZIKV-rEfl were partially protected
(50%).
CyA MNs were fabricated using hydroxypropyl cellulose. The MNs
penetrated skin successfully with up to 50% CyA content. Upon [136]
Local delivery of insoluble administration of 10% CyA to rats, CyA MNs showed Tmax of 8 h, Cmax
Cyclosporin A (CyA)
drugs, Psoriasis treatment of 15.9 ng/mL, and AUC of 686, compared to Tmax of 2 h, Cmax of
18.205 ng/mL, and AUC of 254 for oral administration of solubilized
CyA.
Carboxy methyl cellulose was used to fabricate the base of the MNs
and hydroxypropylmethyl cellulose was used to make the tips of MNs.
Donepezil hydrochloride Alzheimer's disease [137]
95% of the drug was delivered into the porcine skin after 5 min of
insertion.
Chitosan Rhodamine B Transdermal delivery of The MN patch made of water soluble chitosan showed prolonged [108]
hydrophilic drugs and release of rhodamine B for >72 h. The MNs exhibited sufficient
proteins mechanical strength.
MNs had sufficient mechanical strength for insertion at 250 μm and
200 μm in the porcine skin (in vitro) and rat respectively. In vitro
Transdermal delivery of
studies revealed that BSA was released in a sustained manner for at
Bovine serum albumin macromolecules such as [106]
least 8 days. In vivo studies demonstrated BSA diffusion gradually into
proteins
the dermal layer and the fluorescence was observed at a depth of
300 μm.
Upon embedding the ovalbumin-loaded MNs in rat skin in vivo, the
MNs degraded gradually and prolonged ovalbumin exposure for upto
Ovalbumin Intradermal vaccination [103]
14 days. Compared to i.m. vaccination, the MNs demonstrated higher
ovalbumin-specific antibody response that lasted for at least 6 weeks.
In vitro studies demonstrated better skin penetration following
immunization by MNs. In vivo studies demonstrated comparable
Tetanus toxoid Immunization induction of IgG and IgG1 titer and higher IgG2a titer by MNs [138]
compared to commercial tetanus toxoid. Also, MNs generated higher
Th1 cytokines.
The MNs can be loaded with about 73.27±2.75 μg of the drug. The
penetrated wound in Switzerland albino mice healed completely
within 7 days. After applying the MNs, serum luteinizing hormone
Androgen deprivation
Leuprolide acetate level increased and declined within the first 7 days. However, serum [139]
therapy for prostate cancer
testosterone levels increased, reached a peak on the 14th day and
declined on the 21st day. The castration level can be maintained for
2 weeks.
The layer-by-layer coated pH-sensitive MNs were more efficient than
Diphtheria toxoid Dermal immunization s.c. injection. The EC50 dose of the toxoid was 3 fold lower upon the [140]
use of MNs compared to s.c. injection.
Rats immunized with low-dose ovalbumin (200 μg)-loaded MNs had
Ovalbumin Low-dose immunization high levels of antibody levels for 18 weeks persistently, demonstrating [141]
2.5 fold sparing in the i.m. injection dose (500 μg) of ovalbumin.
Chitosan lactate MNs can load large quantities of drug and
– Solid or liquid drugs demonstrate good puncture performance. There is no need of chemical [142]
modification of the drugs before loading.
The MN patch demonstrated 84% of skin penetration without any
Sustained delivery of
Tacrolimus breakage. In vitro and ex vivo permeation studies demonstrated [104]
tacrolimus
sustained release (85%) of tacrolimus from MNs compared to controls.

(continued on next page)

611
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Table 4 (continued)

Carbohydrate Model drug Application Results References


polymer

Immunostaining demonstrated excellent anti-inflammatory effect and


Vascular endothelial growth factor Wound healing [109]
remarkable collagen deposition compared to control.
The layer-by-layer MN patches of poly (vinyl alcohol) and chitosan
demonstrated a good photothermal capacity causing a 12 °C increase
Cancer chemo-photothermal
Doxorubicin and AuMSS nanorods in temperature under near-IR (808 nm, 1.7 W/cm2 for 5 min). In [143]
therapy
combination with the pH sensitivity of chitosan, it can be used to
control release of doxorubicin.
Chondroitin Staphylococcal enterotoxin B (SEB) Staphylococcal Enterotoxin B The dissolvable MNs were composed of CS (2%) and trehalose (0.8%). [113]
sulfate (CS) toxic shock syndrome and The MNs easily penetrated the skin within 5 min with a skin
food poisoning penetration depth of 260 μm. The MNs extended the antigen retention
time in vivo significantly.
The antigenicity was retained at 37 °C and 45 °C. Only a 10% loss was
Hepatitis B surface antigen Hepatitis B vaccination [112]
observed at 50 °C after 6 months.
Sodium CS was used to prepare MNs. In in vitro conditions, LA was
stable in the MN patch and 102.2 ± 1.9–95.3 ± 1.9% recoveries were
Leuprolide acetate (LA) Prostate cancer [31]
obtained for 3 months. In rats, the relative bioavailability of LA from
the MNs against s.c. solution was 105.6 ± 13.5%.
CS was used in the coating solution of MN. The MN had the potential to
H1N1 and H5N1 influenza
Split virion and whole virus particle enclose the dose of 15 μg of hemagglutinin, which is the conventional [144]
virus vaccine
dose of vaccine (data not shown).
The ex vivo evaluation demonstrated that the penetration depth of
MNs in rats was higher than 70 μm. The cumulative penetration of
Neurotoxin Rheumatoid arthritis neurotoxin in MNs could reach 95.8% in 4 h. In contrast, the [145]
neurotoxin solution could hardly penetrate skin. The MNs were stable
for 3 months.
Insertion of the MNs delivered ovalbumin into the skin within 3 min
Ovalbumin Percutaneous vaccination [146]
effectively.
The bioavailabilities of insulin from MNs were 72.1 ± 11.6% (for 2
Insulin Diabetes mellitus patches) and 72.4 ± 8.3% (for 4 patches). Insulin was stable in the MNs [25]
for 1 month at 4 °C. The % recovery was 99.2 ± 13.9%.
The bioavailability of rhGH and desmopressin from MNs was found to
Recombinant human growth hormone
Peptide/ protein delivery be 72.8 ± 4.2–89.9 ± 10.0% and 90.0 ± 15.4–93.1 ± 10.3%, respectively. [147]
(rhGH) and desmopressin
The MNs were stable for one month at −80 °C and 4 °C.
The AUC, Tmax, and Cmax of SH-loaded MNs were found to be 33.07 ±
Transdermal delivery of
Sinomenine hydrochloride (SH) 1.62 μg·h·mL−1, 9.74 ± 0.95 h, and 2.19 ± 0.72 μg·mL−1, [148]
sinomenine hydrochloride
respectively.
Blood glucose monitoring The adhesion time increased. The coefficient of determination R2
– [149]
device increased to 0.750.
Hyaluronic Magnesium ascorbyl phosphate (MAP) Transdermal delivery of MAP Enhanced permeation of MAP using MN (96.8 ± 3.9 μg/cm2) [86]
acid (HA) compared to passive delivery of MAP (44.9 ± 16.3 μg/cm2).
The conjugates loaded into the MNs were localized near the
Hyaluronic acid-antigenic peptide Prophylactic cancer administration site and were retained for >24 h. A single vaccination
[150]
conjugate immunotherapy with MN patch resulted in a statistically significant tumor growth
inhibition in B16 melanoma mice model.
PK parameters; Cmax, Tmax and AUC of MNs was similar to that of
Insulin Diabetes mellitus subcutaneous injection. After storage for one month at −40, 4, 20, and [26]
40 °C, >90% insulin remained in the MNs.
Systemic delivery of Maximum amount of artemether delivered into skin was 72.67±2.69%
Artemether [151]
poorly-water soluble drugs of the initial dose that was loaded on MNs.
Remarkable anti-tumor efficacy; no recurrence was observed. Good
Human epidermoid cancer
Doxorubicin heating transfer efficacy was observed. The temperature of tumors [120]
therapy
treated with MNs rise to 60 °C within 1 min.
40% HA was used to fabricate MNs. The complexes increased the
Polydatin/hydroxypropyl-β-cyclodextrin aqueous solubility of polydatin to 124.47 mg/mL. The combination of
Acute gout arthritis [152]
inclusion complexes inclusion complexes and MN approach enhanced transdermal delivery
of polydatin.
Long term stability of MNs. In vivo studies demonstrated that in 3 out
Photodynamic therapy of
5-Aminolevulinic Acid of 5 mice, tumors continued to shrink and completely disappeared at [153]
superficial tumors
day 14.
Alleviation of psoriasis-like skin inflammation in mice. Methotrexate
Methotrexate Psoriasis [154]
MNs were more efficacious than same dose administered orally.
MN enhanced bioavailability of doxorubicin. The Cmax value of
doxorubicin-loaded transferosome in MN was 153.3 ng/mL which was
-Chemotherapy of tumor
Doxorubicin 3 folds higher than only transferosome. The absolute bioavailability of [121]
-Immune-therapy for tumor
DOXQ loaded transferosome in MN was 79.9%; 3 times higher than
epidermal diffusion of only transferosome.
The pruritus visual analogue scale was significantly lower for MN
compared to topical application. Topical steroid application followed
Topical steroids Prurigo nodularis [85]
by application of MN patch could be a useful strategy for treating
refractory skin diseases.
Sugars Human IgG Monoclonal antibody Maltose MNs: Methylene blue was taken up by microchannels [155]
delivery indicating disruption of the stratum corneum and reaching the dermis.
Immunohistochemical studies demonstrated that IgG follows
microchannels for transport across the skin.
Sinomenine hydrochloride Transdermal delivery of Maltose MNs: In vivo transdermal penetration studies of hydrogel and [61]

612
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Table 4 (continued)

Carbohydrate Model drug Application Results References


polymer

sinomenine hydrochloride MN arrays demonstrated the following pharmacokinetic parameters;


for rheumatoid arthritis AUC0−t and AUC0−inf were 5.38 ± 0.53 μg (mL h)−1 and 35.35 ±
1.27 μg (mL h)−1, 5.75 ± 0.49 μg (mL h)−1 and 45.36 ± 2.36 μg
(mL h)−1, respectively. The MN array had slight irritation to the rat
skin.
25% w/v trehalose: The studies conducted in rhesus macaques
indicated that MNs are similarly immunogenic as i.m. vaccination
Adjuvant-free hepatitis B vaccine Hepatitis B vaccination [62]
without adjuvant. However, MNs are less immunogenic than i.m.
vaccination with alum adjuvant.
Trehalose MNs: % drug recovery of pentagastrin and sincalide
Pentagastrin and sincalide analogues Peptide delivery analogues from the MNs was 89.4 ± 10.61% and 85.7 ± 7.6% compared [156]
to 56.4 ± 9.37% and 90.2 ± 7.7%, respectively in the control.
Trehalose MNs: MNs retained most of the activity after storage for up
Human growth hormone Biopharmaceuticals delivery to 15 months at RT. After insertion into rat skin, pharmacokinetics of [27]
human growth hormone were similar to conventional s.c. injection.
Trehalose MNs: The MN patch-induced neutralizing antibody
Trivalent Sabin inactivated poliovirus
Polio vaccination responses were comparable to higher vaccine doses delivered i.m. for [157]
vaccine
type 1 and type 3 poliovirus serotype.
Xanthan gum Influenza virus-like particle vaccine Influenza vaccine XG (1% w/v) was used as a viscosity enhancer in the coating solution. [127]
(XG) XG produced better hemagglutinin activity than CMC. However,
coating dose of XG was significantly lower compared to CMC.
XG (0.075% w/v) was used as the viscosity enhancer with trehalose
solution to prepare MNs coated with whole inactivated influenza
Whole inactivated influenza vaccine Influenza vaccine vaccine. Due to the addition of XG, the hemagglutinin activity [128]
increased significantly to (81%) compared to trehalose alone
(50–60%).
Pullulan Insulin Diabetes mellitus The secondary structure of insulin was maintained upon storage at [129]
4 °C, 20 °C, and 40 °C. The MNs delivered 87% of insulin, 120 min after
insertion into human abdominal skin in in vitro.

lower level within 4 h. In contrast, prepared MN patches application ex- 4.3. Polysaccharide and polypeptide composites
hibited the highest plasma insulin level (190.7 ± 9.6 μIU/mL) for 2 h for
high loading insulin at 20 IU. Thus, the plasma insulin level could be The polysaccharides and polypeptides with lower molecular weight
maintained for a longer time using MNs compared to s.c. injection [160]. may not exhibit the proper mechanical strength for dermal penetration.
It has been reported that higher molecular weight provides better
strength, and can help attain the required MN geometry [173]. In a
4.2.4. Fish scale polymer
study, CMC arrays on thermal treatment led to decreased water content,
The fish scales naturally consist of protein polymers such as collagen
forming hydrogel in the mold. Moreover, continuous heating of the hy-
fibrils, which are arranged in orthogonal plywood structure in a hy-
drogel led to shrinkage. Upon increase in the concentration of CMC, hy-
droxyapatite matrix. The fish scale biopolymer is extracted as collagen
drogel formation was faster with higher shrinkage. The alginate arrays
peptides from the skin surface of fishes by enzymatic, acid, and thermal
disclosed further shrinkage compared to HA and CMC. In the reported
hydrolysis. The biopolymers are water-soluble and possess the excellent
study, MNs prepared using CMC alone were not adequate for use. The
film-forming ability. Hence these can be used in the micromold fabrica-
needles formed were fragile with significant deformation on application
tion [162]. Olatunji et al. developed a methylene blue loaded fish scale
of minimum weight (5 N). The addition of amylopectin increased me-
polymer extracted from the tilapia (Oreochromiss sp.). They reported
chanical strength of the MNs. The higher concentration of amylopectin
that fish scale MNs successfully penetrated the porcine skin and dis-
resulted in enhanced mechanical strength. MNs mixed with amylopec-
solved gradually at 0, 60, 120, and 180 s after insertion. The mechanical
tin hold >90% of the original height [101,174]. Thus, polysaccharide and
analysis study demonstrated that the MNs with tip radius between 10
polypeptide composites were considered as an attractive solution to in-
and 100 μm could withstand 0.12 N of force per MN without any defor-
crease the molecular weight of the polymers, and obtain the desired
mation [162].
mechanical strength and MN geometry.
In another study, Zhang and co-workers fabricated MNs from algi-
4.2.5. Silk fibroin nate and maltose for the transdermal delivery of insulin. The MNs
Silk fibroin is well known purified biocompatible polymer obtained were fabricated by the template method. To enhance the mechanical
from the proteins produced by Bombyx mori [171]. Silk fibers are usually strength of the MNs, 15% w/w maltose monohydrate was added. The
polyamino acid-based fibrous proteins. Silk proteins have acquired im- MNs exhibited strong mechanical properties with the highest failure
portance in the medical field because of their innate properties like good force of 0.41 N per MN. The MNs were then applied for the transdermal
mechanical strength, resilience in the biological environment, biocom- delivery of insulin on diabetic Sprague-Dawley rats. Compared to the
patibility, and biodegradability. Silk has been applied in various biomed- subcutaneous injection, the relative bioavailability of the composite
ical fields like fibers, scaffolds, and hydrogels [172]. You et al. prepared MNs was found to be 93.7 ± 4.7% [175].
methylene blue loaded rapidly dissolving fibroin MNs). The results re- Cheng et al. fabricated HA composite MNs loaded with curcumin mi-
vealed that silk fibroin MNs dissolved within 1 min under the skin to re- celles. Different excipients were used to increase the plasticity of HA and
lease the drug. The dissolved fibroins were sharp and strong enough to facilitate the formation of MNs, such as polyvinyl pyrrolidone, gelatin,
penetrate the porcine skin and generated non-inflammatory amino acid and sodium carboxymethyl starch. It was found that the HA composite
degradation products that are used in cell metabolic functions [165]. MNs containing gelatin had low hardness and mechanical strength.
The summary of applications of MNs fabricated from poly- The HA composite MNs containing polyvinyl pyrrolidone were brittle
peptides or protein polymers have been listed in Table 5 and tended to fracture. Ultimately, sodium carboxymethyl starch was
[28,59–60,158,161,163,166–168]. selected as the composite for HA MNs. The concentration of HA solution

613
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Table 5
Summary of applications of microneedles fabricated from polypeptide or protein polymers.

Polypeptide Model drug Application Results References


polymer

Immunization studies demonstrated significantly (p < 0.001) greater antibody titers (IgG1, IgG2a, and total
Zein Ovalbumin Vaccination [158]
IgG) after the application of MNs compared with the control.
MNs were composed of gelatin and calcium sulfate. Excellent mechanical strength that can reach upto 0.4 N
Diabetes
Insulin for each MN. Compared to s.c. injection, hypoglycemic effect was observed for longer time in vivo because [161]
mellitus
of the MNs.
Gelatin
Gelatin methacryloyl was used to fabricate MNs. MNs without crosslinking released over 80% of
Doxorubicin Cancer doxorubicin in 30 min. Only a quarter of doxorubicin was released in MNs that were crosslinked for 60 s. [59]
Thus, the rate of drug release can be regulated by adjusting the degree of crosslinking.
Low temperature press method was used to fabricate MNs. The MNs could withstand up to 0.136 N of force
Methylene blue -
per MN. The cross-linked MNs swelled to 340% of their original mass in phosphate buffer solution. Within [163]
Ferrous gluconate -
Fish scale 24 h, 34.5% of the loaded drug was released, while the non cross-linked MNs dissolved within minutes.
Franz diffusion cell study in porcine skin demonstrated that the drug permeation rate increased from 2.5 to
Lidocaine – [60]
7.5% w/w after 36 h. Pseudo steady-state profile is observed from 5.0 to 10.0% w/w lidocaine-loaded MN.
Horseradish
The drug release rate decreased by 5.6-fold by controlling the silk protein secondary structure. Antibiotic
peroxidase Enzyme delivery [28]
loaded silk demonstrated a 10-fold reduction of bacterial density after MN application.
enzyme
Sustained
Insulin delivery of In vitro release studies demonstrated that the release time of drug from MNs was maintained up to 60 h. [166]
Silk fibroin
insulin
In vitro studies indicated that 2-ethoxyethanol modified silk MNs easily pierced porcine skin with a depth of
– – [167]
~200 μm and transformed into semisolid hydrogels with 50–700 nm porous network.
Methanol treatment enhanced the mechanical strength of the MNs up to about 200% depending on the
– – [168]
treatment duration. Methanol exposure time can effectively control drug release rates from the MNs.

was selected as 200 mg/mL and the proportion of HA to sodium intuition of good biodegradability and biocompatibility [179].
carboxymethyl starch was selected as 2:1. The MN matrix thus prepared Hyaluronic acid and chondroitin sulphate are GAGs, which are the
had satisfactory viscosity and fluidity and excellent mechanical proper- major components of the extracellular matrix in the skin and the carti-
ties. The MNs had sharp tips with a height of 600 μm, with regular ap- lage of joints. Theydegrade in the body by lysosomal enzymes and free
pearance, without bubbles or broken MNs [176]. radicals in the tissue matrix. Further, hyaluronic acid-based MN array
Chiu et al. developed composite MNs of sodium HA tip and chitosan MicroHyala® are commercially available for cosmetic use
base to release biphasic antigen. Upon insertion of the composite MNs [173,180,181].
in the skin, the sodium HA tip dissolved within the skin and rapidly re- The cellulose ethers and cellulose esters are non-toxic, biocompati-
leased the antigen acting as a primer. The chitosan base remained in the ble, biodegradable, and are extensively used in tissue engineering, diag-
dermis, prolonging the antigen's release for 4 weeks, acting as a booster nostics, drug delivery, and wound healing, enabling application of these
dose. It was observed that single immunization with the composite MNs biopolymers as MNs. Similarly, alginate is a novel hydrogel biopolymer
containing ovalbumin, both the T helper type 1 and 2 were stimulated in that has gained application in tissue engineering and drug delivery
rats. When compared to the traditional subcutaneous vaccination, the [182].Chitosan is biocompatible, but its clearance is based on molecular
MNs produced higher and more durable antibody response. Thus, the weight. Chitosan of low and medium molecular weight can be degraded
unique rapid and sustained release properties of the composite MNs into monomers by N-acetyl-beta-D-glucosaminidase and cleared by the
allow its use as a primer and booster vaccine formulation [177]. Ling kidney. In contrast, chitosan of high molecular weight can be degraded
et al. fabricated dissolving composite MNs of starch and gelatin for the into suitable fragments by proteases for renal clearance [183]. Dextran
transdermal delivery of insulin. Upon insertion into the skin, the MNs is a high molecular weight polymer degraded by amylases and elimi-
dissolved within 5 min and quickly released insulin. Histological exam- nated via kidney [184]. Starch-based amylopectin is biocompatible,
ination demonstrated that the MNs could be inserted into porcine skin but does not degrade quickly in the body. As the amylose content of
to a depth of 200 μm approximately in vitro, and 200–250 μm depth starch increases, the elongation strength also increases. However, starch
into the rat skin in vivo. Compared to subcutaneous injection, the com- has not been explored much for the fabrication of MNs [185]. Xanthan
posite MNs demonstrated relative pharmacological availability and bio- gum is biocompatible as it is used in the food, cosmetic, and pharmaceu-
availability of 92%. Even after storing the MNs at 25 °C or 37 °C for tical fields. Marketed by Cargill under the name Santiaxane™, this poly-
1 month, >90% of the loaded insulin remained in the MNs. Thus, the mer has been used as a controlled release agent in solid dosage forms,
starch and gelatin composite MNs allowed rapid release of insulin [178]. and as a thickening agent in semi-solid, liquid and topical formulations
[186,187]. Pullulan is degraded by α-amylase enzyme, which is mainly
5. Biocompatibility and biodegradation of the polysaccharide and secreted by salivary glands and pancreas. Pullulan was declared as GRAS
polypeptide MNs and gained significance in the cosmetic market (skincare) due to its
non-toxic, biocompatible, and eco-friendly nature. As it resembles the
The biocompatibility of MNs means that the MNs do not elicit any natural extracellular matrix systems, it has been used in the preparation
undesirable local or systemic responses in the body. The accumulation of scaffolds, films micro/nanoparticles, and fabrication of new matrix
of the polymers at the tissue site and slow degradation is detrimental. systems, and now as MNs [188].
Thus, if the biopolymers used in MNs fabrication are degrading in the Protein-based polymers such as collagen, gelatin, and zein are cur-
body, their byproduct should be non-toxic. As the MNs penetrate bio- rently used in pharmaceutical industry as drug delivery carriers. Gelatin
logical barriers and come in contact with viable tissue, the appraisal of is a favorable material for MNs fabrication due to its moderate to low an-
biocompatibility of these MNs is essential. tigenicity and non-carcinogenic nature. Also, gelatin has been widely
Majority of the polysaccharides or carbohydrate polymers are ob- used in the preparation of soft gelatin capsules. Collagen is biodegrad-
tained from natural sources. Moreover, the structural resemblance of able and biocompatible. It swells upon contact with water, but can
glycosaminoglycans (GAGs) with skin components has led to the only be digested by specific collagenases and pepsin-cleaving enzymes

614
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

[189]. Similar behavior and degradation were observed with gelatin, as Russian inventors developed a microstructure for transdermal ad-
it is an irreversible hydrolyzed form of collagen. A report demonstrated ministration and its method of production (RU2701361C1). The appli-
that collagen exhibits poor stability and nonspecific immune reactions cation was granted in September 2019. They claimed a microstructure
in vivo. But in the case of gelatin, the absence of an aromatic ring containing a biocompatible polymer, selected from HA, CMC, alginic
makes it non-immunogenic. Thus, gelatin has been used to fabricate acid, pectin, carrageenan, chondroitin (sulfate), chitosan, etc. The adhe-
MNs in combination with other biopolymers [190,191]. In recent days, sive material was selected from ethyl cellulose, hydroxymethyl cellu-
zein has gained significance in bone tissue engineering and MNs fabrica- lose, etc. The aspect ratio of the microstructure was 1:5 to 1:2. They
tion. Zein has been recognized as GRAS, and possesses low immunoge- used a microform to fabricate the microstructures. The microstructures
nicity [192,193]. Silk is an attractive scaffolding material suitable for increased the rate of absorption of therapeutic agents into the skin. Ac-
tissue regeneration due to its biocompatibility. Seri® Surgical Scaffold cording to the invention, the D-type microstructure maximally in-
(Allergan, Inc. MA) is a silk-based biomaterial approved by FDA. The creased the mechanical strength of the microstructure due to the use
preclinical biological testing of the scaffold confirmed its overall bio- of triple structure [202]. A Japanese inventor developed a hyaluronic
compatibility, non-toxicity, pyrogenicity, and allergenicity [194]. All acid microstructure with excellent solubility characteristics
these instances confirm the biocompatibility and biodegradation of (JP2019523680A). The inventor claimed that the microstructure had
these natural polymers, and their use in the fabrication of MNs. an excellent dissolution rate in the skin which was regulated by
adjusting the ratio of crosslinked HA and therapeutic agent. The content
6. Clinical applications of the crosslinked HA and non-crosslinked HA was 5 to 100% (w/w) and
0 to 95% (w/w), respectively. When the crosslinked HA was 15–30% of
Currently, little is known of the long-term effects caused due to re- the total weight of HA, the dissolution rate was 50–60%, 65–75%, and
peated insertion of MNs in the skin. It is important to know the dimen- 70–85% after 10 min, 30 min, and 60 min, respectively after MN applica-
sions of micropores created by MNs and the time required for skin tion. The delivery system can be used as a transdermal transmitter
barrier recovery to successfully commercialize MNs [195]. For this, it is [203]. A patent was filed for a microneedle applicator by a Russian in-
necessary to conduct studies in human subjects [196]. Only a few clini- ventor (RU2016144039A). The application was granted in April 2018.
cal studies have been conducted using MNs. The clinical studies that The invention claimed a microneedle applicator consisting of a
have employed MNs have been compiled in Table 6 describing microneedle template, containing biodegradable microneedles loaded
their phase and current status of the trial [197]. Other clinical drug. The microneedle applicator can be used for loading therapeutic
studies involving biodegradable MNs have been listed in Table 7 drugs, vaccines, or cosmetic preparations [204]. Zhongli Ding and co-
[85,119,198–199,200–201]. workers developed a microarray for delivery of therapeutic agent and
method of preparation of the same (EP2934660B1). The application
7. Patent status was granted in July 2019. The microstructure comprised of a backing,
and microstructure array. The microstructures consisted of a distal
The use of natural polymers has gained momentum since past few layer composed of a biodegradable polymer and a proximal layer. The
years. The patents for biodegradable MNs involving the use of natural therapeutic agent was loaded on the distal layer. The biodegradable
polymers are few. Among these, the majority of patents have been ap- polymer was selected from about 50–100% of a hydrophobic polymer
plied for hyaluronic acid. with a glass transition temperature of about 20–25 °C [205].

Table 6
Microneedles currently in clinical trials.

Clinical Trials. Type of MN Drug molecule Primary outcome/description of study Phase Status
gov Identifier

NCT02955576 Microneedle HA patch Calcipotriol - The improvement of psoriasis condition in the time frame of two weeks NA Recruiting
betamethasone
dipropionate
NCT02682056 Biocompatible polymers – A comparative study for blood glucose level testing in patients with diabetes NA Completed
using microneedle patches, intravenous (IV) catheter draw, and lancet.
NCT03207763 Solid conical shape Placebo Vaccine delivery targeting the antigen-presenting cells present in the skin NA Completed
Microneedles made of
water-soluble excipients
NCT03203174 Microneedle; Type not Botulinum Toxin The primary purpose is to treat palmar hyperhidrosis Phase 1 Completed
available Type A
NCT00837512 Hollow Insulin To treat Type 1 Diabetes III Completed
NCT02594644 Stainless steel MNs (MR200, Aminolevulinic To treat Keratosis, Actinic. The results revealed that after the 20-min NA Completed
Clinical Resolutions Acid incubation arm, average actinic keratosis clearance was 76% vs 58% on the
Laboratory, Inc.) sham side (P< 0.01), which is not significant
NCT02995057 Gold- or silver-coated, or – The purpose of this study is the safety demonstration of microneedle, i.e., NA Completed
uncoated nickel microneedles biocompatibility and inertness. Participants have proven nickel allergy.
NCT00539084 MicronJet from nanopass Local anesthesia The primary purpose is to achieve painless delivery of drugs directly into the NA Completed
i.e., Lidocaine skin's superficial layers using the MicronJet microneedle device compared to
insertions of intravenous catheters.
NCT03607903 MicronJet600 microneedle Adalimumab A Randomized, Double-Blind, Placebo-controlled, Double-dummy Study to Adalimumab Active, not
from NanoPass Assess Microneedle Delivery in Comparison to Subcutaneous Injection of ID - II recruiting
Adalimumab in Healthy Volunteers.
Adalimumab
SC - III
NCT03472235 Dermapen – To treat Melasma. The primary outcome measures exhibited that no NA Not yet
improved efficacy recruiting
NCT02596750 Microneedle Roller (MR2 4% lidocaine To evaluate the role of microneedle pretreatment in the speed at which NA Completed
roller, Clinical Resolution anesthesia develops after applying topical 4% lidocaine.
Laboratories, Inc.)

615
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

Table 7
Clinical studies and outcomes of biodegradable MNs fabricated from natural polymers.

Year Natural Application About the study MN treatment (number of Other/control Major outcome of the study References
polymer human subjects) treatment (s) group
used to (number of human
fabricate subjects)
MNs

2014 Hyaluronic Skin Double-blind, Twice daily for 12 weeks (24) None (0) Both the retinyl retinoate- and ascorbic [198]
acid wrinkles randomized controlled acid-loaded dissolving MN patches
trial demonstrated statistically significant
differences in all Visiometer R-values (P <
0.05). In particular, highly significant
differences were shown at R1 (skin
roughness) and R5 (arithmetic average
roughness) (P < 0.001).
2017 Hyaluronic Psoriatic Ethics Committee of the Once daily at night for 1 week None (0) The mPASI (mean Psoriasis Area Severity [199]
acid plaques Catholic Medical Center (10) Index) was 5.4 (± 1.2) before treatment
Office of Human and 1.8 (± 1.1) after treatment (P < 0.001).
Research Protection The median decrease in mPASI was 60%
(VC17RESI0010) (range: 25–100%) after 1 week application
of the MN patch.
2017 Hyaluronic Crow's feet IRB of P&K Skin Research 20 min twice a week for HA essence group, Before the treatment, the global visual [119]
acid wrinkles Center P1512–55 8 weeks (31) twice a week for wrinkle score was 2.3±0.56 on the HA MN
8 weeks (31) group and 2.35±0.49 on the HA essence
group. At 8 weeks, the score of the HA MN
group was 2.32±0.54 and of the HA
essence group was 2.35±0.55.
2018 Hyaluronic Prurigo IRB No. 4–2015-1086 Mometasone furoate 0.1% cream Mometasone Investigator global assessment (IGA) score [85]
acid nodularis was applied two times per day furoate 0.1% cream demonstrated that weekly application of
for four weeks; followed by the was applied two MN after topical steroids was more
MN patch once per week for times per day for effective than topical steroids alone (2.33
12 h (24) four weeks (24) ± 0.23 for MN; 1.33 ± 0.24 for control)
(paired t-test, p<0.05)
2019 Hyaluronic Skin aging Approved by Twice a week onto the face in None (0) The averaged wrinkle depths before and [200]
acid International Medical the left and right corner of the after application of the product were 363 ±
and Dental Ethics eye and on a defined area on the 146 μm and 287 ± 166 μm, respectively.
Commission GmbH right volar forearm for 12 weeks After the treatment, an average increase of
(Freiburg, Germany) (20) 26% overall improvement of the wrinkle
depths was observed.
2020 Hyaluronic Crow's feet Randomized Once every 3 days, in the Once every 3 days, After 8 weeks of use, the maximum depth [201]
acid wrinkles double-blind clinical evening, for 8 weeks (23) in the evening, for of biggest wrinkles improvement in the
trial 8 weeks (23) Ad-HMN (high molecular weight HA
dissolving MN patch) group and (low
molecular weight HA dissolving MN patch)
group was 23.34% (P < 0.001) and 13.32%
(P < 0.01), respectively.

IRB: Institutional Review Board.

David L. Kaplan, and teammates filed a patent for silk fibroin based for all subunit vaccines compared to control groups [24]. Considering
microneedles and methods of fabricating the same (CA2815285C). The the applications of biodegradable MNs in vaccination [22,23,144], it
application was granted in December 2019. They claimed that the can only be said that the future of vaccination lies in MN-assisted deliv-
microneedle device, extending from the base to the tip, composed of ery systems.
silk fibroin was highly biocompatible and easy to fabricate. The silk fi- Owing to the wide range of functional groups present in biopoly-
broin remains stable at room temperature and is implantable. They mers, they allow the fabrication of MNs with tunable functionalities
stated that the rate of delivery of active agent can be controlled by reg- and properties. Thus, these biopolymer-based MNs offer great potential
ulating the structure of silk fibroin. These microneedles can be used to not only in drug administration but also in biosensing and physiological
deliver active agents into or across biological barriers such as cell mem- stimuli-responsive systems. The future of MNs lies in the use of combi-
brane, tissue, and skin. The method for fabrication of the microneedles natorial strategies. The combination of MNs and external stimuli such as
used micromolds [206]. ultrasound and techniques like iontophoresis has enhanced transder-
mal drug delivery [207,208]. The research in the field of MN-based sen-
8. Future prospects sor devices will grow rapidly in the next few years. Novel strategies of
MN drug-eluting balloon for enhanced delivery of drug to the vascular
The fabrication of biodegradable MNs using biopolymers has tissue are being developed and can cause a stir in MN delivery systems
sparked the imagination of formulation scientists because of their rec- in the future [209]. Smart, wearable theranostic MN devices can be used
ognized biodegradability, biocompatibility, ease of fabrication, and sus- for personalized medication delivery. These non-invasive, cost-effective
tainable character [129]. Natural polymers are being exploited to their devices will facilitate the dynamic dosing of therapeutics and bring
use in the fabrication of MNs including silk fibroin [28,206], Bletilla stri- about a paradigm shift in the management of chronic diseases such as
ata polysaccharide [132], and fish scale biopolymer [60,162–164]. The cancer, diabetes, Alzheimer's disease, and infectious diseases in the fu-
whole humankind is a spectator of the havoc that coronavirus has ture [210].
caused. Recently, biodegradable MNs composed of CMC were used to Since the inception of 3D printing or additive manufacturing in the
fabricate the recombinant coronavirus vaccine. It was found that signif- 1980s, it has revolutionized the pharmaceutical and biomedical fields.
icantly high SARS-CoV-2 IgG responses were detected as early as week 2 3D printing-assisted fabrication of biodegradable MNs has opened a

616
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

novel avenue for enhanced drug delivery. These devices will revolution- [9] X. He, J. Sun, J. Zhuang, H. Xu, Y. Liu, D. Wu, Microneedle system for transdermal
drug and vaccine delivery: devices, safety, and prospects, Dose-Response,
ize macromolecular delivery, especially that of insulin in the future 2019https://doi.org/10.1177/1559325819878585.
[211,212]. Adoption of technologies such as stereolithography, fused [10] C.J. Martin, C.J. Allender, K.R. Brain, A. Morrissey, J.C. Birchall, Low temperature fab-
deposition modeling, and two-photon polymerization, will enable to rication of biodegradable sugar glass microneedles for transdermal drug delivery
applications, J. Control. Release (2012)https://doi.org/10.1016/j.jconrel.2011.10.
regulate MNs geometry without compromising the resolution and the
024.
aspect ratio. Moreover, 3D printing can facilitate facile customization [11] M. Sharma, Transdermal and intravenous nano drug delivery systems, in: Appl,
of TDDS to accommodate factors such as differences in skin hydration Target. Nano Drugs Deliv. Syst. 2019https://doi.org/10.1016/b978-0-12-814029-
and thickness, that influence drug delivery [213]. Recently, a 4D printed 1.00018-1.
[12] A.H. Sabri, Y. Kim, M. Marlow, D.J. Scurr, J. Segal, A.K. Banga, L. Kagan, J.B. Lee, Intra-
MN array with backward-facing barbs has been reported by Han et al. It dermal and transdermal drug delivery using microneedles – fabrication, perfor-
was demonstrated that barbed MN arrays enhanced tissue adhesion by mance evaluation and application to lymphatic delivery, Adv. Drug Deliv. Rev.
18 folds compared to barbless MN arrays. Enhanced tissue adhesion re- (2019)https://doi.org/10.1016/j.addr.2019.10.004.
[13] C.H. Chen, V.B.H. Shyu, C.T. Chen, Dissolving microneedle patches for transdermal
sults in a more stable and robust performance for drug delivery, bio-
insulin delivery in diabetic mice: potential for clinical applications, Materials
sensing, and biofluid collection. Han et al. fabricated the MNs using (Basel), 2018https://doi.org/10.3390/ma11091625.
poly(ethylene glycol) diacrylate. The use of biopolymers in this regard [14] D.P. Wermeling, S.L. Banks, D.A. Hudson, H.S. Gill, J. Gupta, M.R. Prausnitz, A.L.
should be investigated in the future [214]. Stinchcomb, Microneedles permit transdermal delivery of a skin-impermeant
medication to humans, Proc. Natl. Acad. Sci. U. S. A. (2008)https://doi.org/10.
1073/pnas.0710355105.
9. Conclusion [15] K. Cheung, G. West, D.B. Das, Delivery of large molecular protein using flat and
short microneedles prepared using focused ion beam (FIB) as a skin ablation
tool, Drug Deliv. Transl, Res, 2015https://doi.org/10.1007/s13346-015-0252-0.
Biodegradable and dissolving microneedles have gained significant
[16] T. Han, D.B. Das, Permeability enhancement for transdermal delivery of large mol-
attraction in the field of transdermal drug delivery and have been ecule using low-frequency sonophoresis combined with microneedles, J. Pharm.
researched extensively by formulation scientists because of their recog- Sci. (2013)https://doi.org/10.1002/jps.23662.
nized biodegradability, biocompatibility, faster onset of action, ease of [17] R.F. Donnelly, T.R.R. Singh, M.M. Tunney, D.I.J. Morrow, P.A. McCarron, C.
O’Mahony, A.D. Woolfson, Microneedle arrays allow lower microbial penetration
fabrication, and sustainable character. Apart from the various natural than hypodermic needles in vitro, Pharm. Res. (2009)https://doi.org/10.1007/
polymers used to fabricate biodegradable MNs like alginate, cellulose s11095-009-9967-2.
and its derivatives, chitosan, chondroitin sulfate, hyaluronic acid, sugars, [18] B. Gualeni, S.A. Coulman, D. Shah, P.F. Eng, H. Ashraf, P. Vescovo, G.J. Blayney, L.D.
Piveteau, O.J. Guy, J.C. Birchall, Minimally invasive and targeted therapeutic cell de-
and xanthan gum, polymers like silk fibroin and fish scale biopolymers livery to the skin using microneedle devices, Br. J. Dermatol. (2018)https://doi.org/
are some of the newest additions to these natural polymers. Patents in- 10.1111/bjd.15923.
volving the use of natural polymers for MN fabrication are few. How- [19] A. Tröls, M.A. Hintermüller, M. Saeedipour, S. Pirker, B. Jakoby, Drug dosage for
ever, biodegradable MNs have gained entry into the preclinical and microneedle-based transdermal drug delivery systems utilizing evaporation-
induced droplet transport, Microfluid, Nanofluidics, 2019https://doi.org/10.1007/
clinical stages. With the advent of 3D printing, smart, wearable s10404-019-2257-3.
theranostics, stimuli-responsive delivery systems, and biosensing de- [20] Z. Zhao, Y. Chen, Y, Shi, Microneedles, a potential strategy in transdermal delivery
vices, MN technology holds enormous potential for clinical application and application in the management of psoriasis, RSC Adv, 2020https://doi.org/10.
1039/d0ra00735h.
and commercialization. The future appears to be very bright for novel
[21] S.H. Bariya, M.C. Gohel, T.A. Mehta, O.P. Sharma, Microneedles: an emerging trans-
delivery and monitoring systems based on MN technologies. However, dermal drug delivery system, J. Pharm. Pharmacol. (2012)https://doi.org/10.1111/
the use of natural polymers in this regard must be investigated further j.2042-7158.2011.01369.x.
to utilize the advantages associated with their biodegradability and [22] M.J. Mistilis, A.S. Bommarius, M.R. Prausnitz, Development of a thermostable
microneedle patch for influenza vaccination, J. Pharm. Sci. (2015)https://doi.org/
biocompatibility. 10.1002/jps.24283.
[23] E. Kim, G. Erdos, S. Huang, T. Kenniston, L.D. Falo, A. Gambotto, Preventative vac-
cines for zika virus outbreak: preliminary evaluation, EBioMedicine, 2016https://
doi.org/10.1016/j.ebiom.2016.09.028.
Declaration of competing interest [24] E. Kim, G. Erdos, S. Huang, T.W. Kenniston, S.C. Balmert, C.D. Carey, V.S. Raj, M.W.
Epperly, W.B. Klimstra, B.L. Haagmans, E. Korkmaz, L.D. Falo, A. Gambotto,
The authors declared no conflicts of interest. Microneedle array delivered recombinant coronavirus vaccines: immunogenicity
and rapid translational development, EBioMedicine, 2020https://doi.org/10.1016/
j.ebiom.2020.102743.
References [25] K. Fukushima, T. Yamazaki, R. Hasegawa, Y. Ito, N. Sugioka, K. Takada, Pharmacoki-
netic and pharmacodynamic evaluation of insulin dissolving microneedles in dogs.,
[1] P. Mukhopadhyay, K. Sarkar, M. Chakraborty, S. Bhattacharya, R. Mishra, P.P. Diabetes Technol. Ther. (2010). doi:https://doi.org/10.1089/dia.2009.0176.
Kundu, Oral insulin delivery by self-assembled chitosan nanoparticles: in vitro [26] S. Liu, M.N. Jin, Y.S. Quan, F. Kamiyama, H. Katsumi, T. Sakane, A. Yamamoto, The
and in vivo studies in diabetic animal model, Mater. Sci. Eng. C. (2013)https:// development and characteristics of novel microneedle arrays fabricated from
doi.org/10.1016/j.msec.2012.09.001. hyaluronic acid, and their application in the transdermal delivery of insulin, J. Con-
[2] D.K. Mishra, V. Dhote, P.K. Mishra, Transdermal immunization: biological frame- trol. Release (2012)https://doi.org/10.1016/j.jconrel.2012.05.030.
work and translational perspectives, Expert Opin. Drug Deliv. (2013)https://doi. [27] J.W. Lee, S.O. Choi, E.I. Felner, M.R. Prausnitz, Dissolving microneedle patch for
org/10.1517/17425247.2013.746660. transdermal delivery of human growth hormone, Small, 2011https://doi.org/10.
[3] H. Tanwar, R. Sachdeva, Transdermal drug delivery system: a review | International 1002/smll.201001091.
Journal of Pharmaceutical Sciences and Research, Int. J. Pharm. Sci. Res. 7 (2016) [28] K. Tsioris, W.K. Raja, E.M. Pritchard, B. Panilaitis, D.L. Kaplan, F.G. Omenetto, Fabri-
2274–90. doi:10.13040/IJPSR.0975-8232.7(6).2274-90. cation of silk microneedles for controlled-release drug delivery, Adv. Funct. Mater.
[4] MARKETSANDMARKETS, Transdermal Drug Delivery System Market by Type (2012)https://doi.org/10.1002/adfm.201102012.
(Patches and Semisolid Formulations), Applications (Pain Management, Central [29] W. Li, R.N. Terry, J. Tang, M.R. Feng, S.P. Schwendeman, M.R. Prausnitz, Rapidly sep-
Nervous System Disorders, Hormonal Applications, Cardiovascular Diseases), End arable microneedle patch for the sustained release of a contraceptive, Nat. Biomed,
User, and Region - Global Forecast to 2023, 2020. Eng, 2019https://doi.org/10.1038/s41551-018-0337-4.
[5] M.B. Brown, G.P. Martin, S.A. Jones, F.K. Akomeah, Dermal and transdermal drug [30] S. Bhatnagar, P. Kumari, S.P. Pattarabhiran, V.V.K. Venuganti, Zein microneedles for
delivery systems: current and future prospects, Drug Deliv. 13 (2006) 175–187, localized delivery of chemotherapeutic agents to treat breast cancer: drug loading,
https://doi.org/10.1080/10717540500455975. release behavior, and skin permeation studies, AAPS PharmSciTech, 2018https://
[6] D.K. Mishra, V. Pandey, R. Maheshwari, P. Ghode, R.K. Tekade, Cutaneous and doi.org/10.1208/s12249-018-1004-5.
Transdermal Drug Delivery: Techniques and Delivery Systems (2018)https://doi. [31] Y. Ito, H. Murano, N. Hamasaki, K. Fukushima, K. Takada, Incidence of low bioavail-
org/10.1016/B978-0-12-817909-3.00015-7. ability of leuprolide acetate after percutaneous administration to rats by dissolving
[7] T. Waghule, G. Singhvi, S.K. Dubey, M.M. Pandey, G. Gupta, M. Singh, K. Dua, microneedles, Int. J. Pharm. (2011)https://doi.org/10.1016/j.ijpharm.2011.01.039.
Microneedles: a smart approach and increasing potential for transdermal drug de- [32] L. Xie, H. Zeng, J. Sun, W. Qian, Engineering microneedles for therapy and diagno-
livery system, Biomed. Pharmacother. 109 (2019) 1249–1258, https://doi.org/10. sis: a survey, Micromachines, 2020https://doi.org/10.3390/mi11030271.
1016/j.biopha.2018.10.078. [33] A.Z. Alkilani, M.T.C. McCrudden, R.F. Donnelly, Transdermal drug delivery: Innova-
[8] M.S. Giri Nandagopal, R. Antony, S. Rangabhashiyam, N. Sreekumar, N. Selvaraju, tive pharmaceutical developments based on disruption of the barrier properties of
Overview of microneedle system: a third generation transdermal drug delivery ap- the stratum corneum, Pharmaceutics, 2015https://doi.org/10.3390/
proach, Microsyst. Technol. (2014)https://doi.org/10.1007/s00542-014-2233-5. pharmaceutics7040438.

617
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

[34] Y.B. Schuetz, A. Naik, R.H. Guy, Y.N. Kalia, Emerging strategies for the transdermal [61] Y. Cao, Y. Tao, Y. Zhou, S. Gui, Development of sinomenine hydrochloride-loaded
delivery of peptide and protein drugs, Expert Opin. Drug Deliv. (2005)https://doi. polyvinylalcohol/maltose microneedle for transdermal delivery, J. Drug Deliv. Sci.
org/10.1517/17425247.2.3.533. Technol. (2016)https://doi.org/10.1016/j.jddst.2016.06.007.
[35] C.M. Schoellhammer, D. Blankschtein, R. Langer, Skin permeabilization for trans- [62] M.B. Perez Cuevas, M. Kodani, Y. Choi, J. Joyce, S.M. O’Connor, S. Kamili, M.R.
dermal drug delivery: recent advances and future prospects, Expert Opin. Drug Prausnitz, Hepatitis B vaccination using a dissolvable microneedle patch is immu-
Deliv. (2014)https://doi.org/10.1517/17425247.2014.875528. nogenic in mice and rhesus macaques, Bioeng. Transl, Med, 2018https://doi.org/
[36] Y. Shahzad, R. Louw, M. Gerber, J. Du Plessis, Breaching the skin barrier through 10.1002/btm2.10098.
temperature modulations, J. Control. Release (2015)https://doi.org/10.1016/j. [63] Y.C. Kim, F.S. Quan, J.M. Song, A. Vunnava, D.G. Yoo, K.M. Park, R.W. Compans, S.M.
jconrel.2015.01.019. Kang, M.R. Prausnitz, Influenza immunization with trehalose-stabilized virus-like
[37] S. Dharadhar, A. Majumdar, S. Dhoble, V. Patravale, Microneedles for transdermal particle vaccine using microneedles, Procedia Vaccinol, 2010https://doi.org/10.
drug delivery: a systematic review, Drug Dev. Ind. Pharm. (2019)https://doi.org/ 1016/j.provac.2010.03.004.
10.1080/03639045.2018.1539497. [64] G. Cole, A.A. Ali, C.M. McCrudden, J.W. McBride, J. McCaffrey, T. Robson, V.L. Kett,
[38] M. Pearton, V. Saller, S.A. Coulman, C. Gateley, A.V. Anstey, V. Zarnitsyn, J.C. Birchall, N.J. Dunne, R.F. Donnelly, H.O. McCarthy, DNA vaccination for cervical cancer: stra-
Microneedle delivery of plasmid DNA to living human skin: formulation coating, tegic optimisation of RALA mediated gene delivery from a biodegradable
skin insertion and gene expression, J. Control. Release 160 (2012) 561–569, microneedle system, Eur. J. Pharm. Biopharm. (2018)https://doi.org/10.1016/j.
https://doi.org/10.1016/j.jconrel.2012.04.005. ejpb.2018.02.029.
[65] M. He, G. Yang, X. Zhao, S. Zhang, Y. Gao, Intradermal implantable PLGA
[39] M. Sharma, Transdermal and intravenous nano drug delivery systems, in: Appl,
microneedles for etonogestrel sustained release, J. Pharm. Sci. (2020)https://doi.
Elsevier, Target. Nano Drugs Deliv. Syst., 2019 499–550, https://doi.org/10.1016/
org/10.1016/j.xphs.2020.02.009.
b978-0-12-814029-1.00018-1.
[66] M. Battisti, R. Vecchione, C. Casale, F.A. Pennacchio, V. Lettera, R. Jamaledin, M.
[40] E. Larrañeta, R.E.M. Lutton, A.D. Woolfson, R.F. Donnelly, Microneedle arrays as
Profeta, C. Di Natale, G. Imparato, F. Urciuolo, P.A. Netti, Non-invasive production
transdermal and intradermal drug delivery systems: materials science, manufac-
of multi-compartmental biodegradable polymer microneedles for controlled intra-
ture and commercial development, Mater. Sci. Eng. R Reports. (2016)https://doi.
dermal drug release of labile molecules, Front. Bioeng, Biotechnol, 2019https://doi.
org/10.1016/j.mser.2016.03.001.
org/10.3389/fbioe.2019.00296.
[41] K.J. Lee, S.S. Jeong, D.H. Roh, D.Y. Kim, H.K. Choi, E.H. Lee, A practical guide to the
[67] E. Marin, M.I. Briceño, C. Caballero-George, Critical evaluation of biodegradable
development of microneedle systems – in clinical trials or on the market, Int. J.
polymers used in nanodrugs, Int. J. Nanomedicine 8 (2013) 3071–3091, https://
Pharm. (2020)https://doi.org/10.1016/j.ijpharm.2019.118778.
doi.org/10.2147/IJN.S47186.
[42] L. Wei-Ze, H. Mei-Rong, Z. Jian-Ping, Z. Yong-Qiang, H. Bao-Hua, L. Ting, Z. Yong, [68] N.R. Nair, V.C. Sekhar, K.M. Nampoothiri, A. Pandey, Biodegradation of biopoly-
Super-short solid silicon microneedles for transdermal drug delivery applications, mers, in: Curr, Elsevier Inc., Dev. Biotechnol. Bioeng. Prod. Isol. Purif. Ind. Prod,
Int. J. Pharm. (2010)https://doi.org/10.1016/j.ijpharm.2010.01.024. 2016 739–755, https://doi.org/10.1016/B978-0-444-63662-1.00032-4.
[43] S. Pradeep Narayanan, S. Raghavan, Fabrication and characterization of gold-coated [69] K.I. Draget, Alginates, in: Handb, Hydrocoll. Second Ed. 2009https://doi.org/10.
solid silicon microneedles with improved biocompatibility, Int. J. Adv. Manuf. 1533/9781845695873.807.
Technol. (2019)https://doi.org/10.1007/s00170-018-2596-3. [70] A.R. Nesic, S.I. Seslija, The influence of nanofillers on physical–chemical properties
[44] W. Chen, H. Li, D. Shi, Z. Liu, W. Yuan, Microneedles as a delivery system for gene of polysaccharide-based film intended for food packaging, Food Packag,
therapy, Front, Pharmacol, 2016https://doi.org/10.3389/fphar.2016.00137. 2017https://doi.org/10.1016/b978-0-12-804302-8.00019-4.
[45] S. Doddaballapur, Microneedling with dermaroller, J. Cutan. Aesthet, Surg, [71] C.H. Goh, P.W.S. Heng, L.W. Chan, Alginates as a useful natural polymer for micro-
2009https://doi.org/10.4103/0974-2077.58529. encapsulation and therapeutic applications, Carbohydr. Polym. (2012)https://doi.
[46] Y. Kapoor, M. Milewski, L. Dick, J. Zhang, J.R. Bothe, M. Gehrt, K. Manser, B. Nissley, org/10.1016/j.carbpol.2011.11.012.
I. Petrescu, P. Johnson, S. Burton, J. Moseman, V. Hua, T. Grunewald, M. Tomai, R. [72] R.D. Koyani, Biopolymers for microneedle synthesis: from then to now,
Smith, Coated microneedles for transdermal delivery of a potent pharmaceutical Biomanufacturing Rev, 2019https://doi.org/10.1007/s40898-019-0006-8.
peptide, Biomed. Microdevices (2020)https://doi.org/10.1007/s10544-019-0462- [73] T. Heinze, O.A. El Seoud, A. Koschella, Production and Characteristics of Cellulose
1. From Different Sources, in, 2018https://doi.org/10.1007/978-3-319-73168-1_1.
[47] S. Li, W. Li, M. Prausnitz, Individually coated microneedles for co-delivery of multi- [74] J. Shokri, K. Adibki, Application of cellulose and cellulose derivatives in pharmaceu-
ple compounds with different properties, Drug Deliv. Transl, Res, 2018https://doi. tical industries, in: Cellul. - Medical, Pharm. Electron. Appl., 2013. doi:https://doi.
org/10.1007/s13346-018-0549-x. org/10.5772/55178.
[48] J.H. Park, M.G. Allen, M.R. Prausnitz, Polymer microneedles for controlled-release [75] J.E. Song, S.H. Jun, S.G. Park, N.G. Kang, A semi-dissolving microneedle patch incor-
drug delivery, Pharm. Res. (2006)https://doi.org/10.1007/s11095-006-0028-9. porating TEMPO-oxidized bacterial cellulose nanofibers for enhanced transdermal
[49] M. Wang, L. Hu, C. Xu, Recent advances in the design of polymeric microneedles for delivery, Polymers (Basel). 12 (2020) 1873. doi:https://doi.org/10.3390/
transdermal drug delivery and biosensing, Lab Chip, 2017https://doi.org/10.1039/ POLYM12091873.
C7LC00016B. [76] D.I. Sánchez-Machado, J. López-Cervantes, M.A. Correa-Murrieta, R.G. Sánchez-
[50] S. Fakhraei Lahiji, Y. Kim, G. Kang, S. Kim, S. Lee, H. Jung, Tissue interlocking dissolv- Duarte, P. Cruz-Flores, G.S. la Mora-López, Chitosan, in: Nonvitamin Nonmineral
ing microneedles for accurate and efficient transdermal delivery of biomolecules, Nutr, Suppl. 2018https://doi.org/10.1016/B978-0-12-812491-8.00064-3.
Sci. Rep. (2019)https://doi.org/10.1038/s41598-019-44418-6. [77] F. Shahidi, R. Abuzaytoun, Chitin, chitosan, and co-products : chemistry, produc-
[51] S. Lee, S.F. Lahiji, J. Jang, M. Jang, H. Jung, Micro-pillar integrated dissolving tion, applications, and health effects, 49 (2005).
microneedles for enhanced transdermal drug delivery, Pharmaceutics, [78] J. Lizardi-Mendoza, W.M. Argüelles Monal, F.M. Goycoolea Valencia, Chemical
2019https://doi.org/10.3390/pharmaceutics11080402. characteristics and functional properties of chitosan, in: Chitosan Preserv, Agric.
Commod. 2016https://doi.org/10.1016/B978-0-12-802735-6.00001-X.
[52] M.H. Ling, M.C. Chen, Dissolving polymer microneedle patches for rapid and effi-
cient transdermal delivery of insulin to diabetic rats, Acta Biomater. 9 (2013) [79] J. Chi, X. Zhang, C. Chen, C. Shao, Y. Zhao, Y. Wang, Antibacterial and angiogenic chi-
8952–8961, https://doi.org/10.1016/j.actbio.2013.06.029. tosan microneedle array patch for promoting wound healing, Bioact. Mater. 5
(2020) 253–259, https://doi.org/10.1016/j.bioactmat.2020.02.004.
[53] J.J. Norman, S.O. Choi, N.T. Tong, A.R. Aiyar, S.R. Patel, M.R. Prausnitz, M.G. Allen,
[80] M. Nurunnabi, V. Revuri, K.M. Huh, Y. kyu Lee, Polysaccharide based nano/
Hollow microneedles for intradermal injection fabricated by sacrificial
microformulation: an effective and versatile oral drug delivery system, in: Nano-
micromolding and selective electrodeposition, Biomed. Microdevices (2013)
structures Oral Med., 2017. doi:https://doi.org/10.1016/B978-0-323-47720-8.
https://doi.org/10.1007/s10544-012-9717-9.
00015-8.
[54] R. Mishra, T.K. Maiti, T.K. Bhattacharyya, Development of SU-8 hollow
[81] F.G. Huffman, Uronic acids, in: Encycl, Food Sci. Nutr. 2003https://doi.org/10.1016/
microneedles on a silicon substrate with microfluidic interconnects for transder-
b0-12-227055-x/01221-9.
mal drug delivery, J. Micromechanics Microengineering. (2018)https://doi.org/
[82] A. Schieber, D. Lopes-Lutz, Analytical methods - functional foods and dietary sup-
10.1088/1361-6439/aad301.
plements, in: Compr, Biotechnol. Second Ed. 2011https://doi.org/10.1016/B978-
[55] X. Hong, L. Wei, F. Wu, Z. Wu, L. Chen, Z. Liu, W. Yuan, Dissolving and biodegrad- 0-08-088504-9.00320-2.
able microneedle technologies for transdermal sustained delivery of drug and vac-
[83] C.G. Boeriu, J. Springer, F.K. Kooy, L.A.M. van den Broek, G. Eggink, Production
cine, Drug Des. Devel, Ther, 2013https://doi.org/10.2147/DDDT.S44401.
methods for hyaluronan, Int. J. Carbohydr, Chem, 2013https://doi.org/10.1155/
[56] J.H. Park, M.G. Allen, M.R. Prausnitz, Biodegradable polymer microneedles: fabrica- 2013/624967.
tion, mechanics and transdermal drug delivery, J. Control. Release (2005)https:// [84] J. Zhu, X. Tang, Y. Jia, C.T. Ho, Q. Huang, Applications and delivery mechanisms of
doi.org/10.1016/j.jconrel.2005.02.002. hyaluronic acid used for topical/transdermal delivery – a review, Int. J. Pharm.
[57] Q.Y. Li, J.N. Zhang, B.Z. Chen, Q.L. Wang, X.D. Guo, A solid polymer microneedle 578 (2020) 119127, https://doi.org/10.1016/j.ijpharm.2020.119127.
patch pretreatment enhances the permeation of drug molecules into the skin, [85] J.U. Shin, J.D. Kim, H.K. Kim, H.K. Kang, C. Joo, J.H. Lee, D.H. Jeong, S. Song, H. Chu, J.S.
RSC Adv, 2017https://doi.org/10.1039/c6ra26759a. Lee, H. Lee, K.H. Lee, The use of biodegradable microneedle patches to increase
[58] S.C. Park, M.J. Kim, S.K. Baek, J.H. Park, S.O. Choi, Spray-formed layered polymer penetration of topical steroid for prurigo nodularis, Eur. J, Dermatology,
microneedles for controlled biphasic drug delivery, Polymers (Basel), 2018https://doi.org/10.1684/ejd.2017.3164.
2019https://doi.org/10.3390/POLYM11020369. [86] Y. Kim, S.A. Bhattaccharjee, M. Beck-Broichsitter, A.K. Banga, Fabrication and char-
[59] Z. Luo, W. Sun, J. Fang, K.J. Lee, S. Li, Z. Gu, M.R. Dokmeci, A. Khademhosseini, Bio- acterization of hyaluronic acid microneedles to enhance delivery of magnesium
degradable gelatin methacryloyl microneedles for transdermal drug delivery, Adv. ascorbyl phosphate into skin, Biomed. Microdevices (2019)https://doi.org/10.
Healthc, Mater, 2019https://doi.org/10.1002/adhm.201801054. 1007/s10544-019-0455-0.
[60] P. Medhi, O. Olatunji, A. Nayak, C.T. Uppuluri, R.T. Olsson, B.N. Nalluri, D.B. Das, [87] G. Bonfante, H. Lee, L. Bao, J. Park, N. Takama, B. Kim, Comparison of polymers to
Lidocaine-loaded fish scale-nanocellulose biopolymer composite microneedles, enhance mechanical properties of microneedles for bio-medical applications,
AAPS PharmSciTech, 2017https://doi.org/10.1208/s12249-017-0758-5. Micro Nano Syst. Lett. 8 (2020) 13, https://doi.org/10.1186/s40486-020-00113-0.

618
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

[88] M. Zaitoun, M. Ghanem, S. Harphoush, Sugars : types and their functional proper- enterotoxin challenge, Vaccine, 2019https://doi.org/10.1016/j.vaccine.2019.05.
ties in food and human health, Int. J. Public Heal. Res. 6 (4) (2019) 93–99. 055.
[89] P. Serrano-Castañeda, J.J. Escobar-Chávez, I.M. Rodríguez-Cruz, L.M. Melgoza- [114] P. Snetkov, K. Zakharova, S. Morozkina, R. Olekhnovich, M. Uspenskaya, Hyaluronic
Contreras, J. Martínez-Hernández, Microneedles as enhancer of drug absorption acid: the influence of molecular weight on structural, physical, physico-chemical,
through the skin and applications in medicine and cosmetology, J. Pharm. and degradable properties of biopolymer, Polymers (Basel). 12 (2020). doi:
Pharm. Sci. (2018). doi:10.18433/jpps29610. https://doi.org/10.3390/polym12081800.
[90] R.F. Donnelly, D.I.J. Morrow, T.R.R. Singh, K. Migalska, P.A. McCarron, C. O’Mahony, [115] S. Vasvani, P. Kulkarni, D. Rawtani, Hyaluronic acid: a review on its biology, aspects
A.D. Woolfson, Processing difficulties and instability of carbohydrate microneedle of drug delivery, route of administrations and a special emphasis on its approved
arrays, Drug Dev. Ind. Pharm. (2009)https://doi.org/10.1080/ marketed products and recent clinical studies, Int. J. Biol. Macromol. (2019)
03639040902882280. https://doi.org/10.1016/j.ijbiomac.2019.11.066.
[91] E.P. Yalcintas, D.S. Ackerman, E. Korkmaz, C.A. Telmer, J.W. Jarvik, P.G. Campbell, [116] J.R.E. Fraser, T.C. Laurent, U.B.G. Laurent, Hyaluronan: its nature, distribution, func-
M.P. Bruchez, O.B. Ozdoganlar, Analysis of in vitro cytotoxicity of carbohydrate- tions and turnover, in: J, Intern. Med. (1997)https://doi.org/10.1046/j.1365-2796.
based materials used for dissolvable microneedle arrays, Pharm. Res. (2020) 1997.00170.x.
https://doi.org/10.1007/s11095-019-2748-7. [117] J.H. Sze, J.C. Brownlie, C.A. Love, Biotechnological production of hyaluronic acid: a
[92] E. Larrañeta, R.E.M. Lutton, A.D. Woolfson, R.F. Donnelly, Microneedle arrays as mini review, 3 Biotech, 2016https://doi.org/10.1007/s13205-016-0379-9.
transdermal and intradermal drug delivery systems: materials science, manufac- [118] S.G. Lee, J.H. Jeong, K.M. Lee, K.H. Jeong, H. Yang, M. Kim, H. Jung, S. Lee, Y.W. Choi,
ture and commercial development, Mater. Sci. Eng. R Reports. 104 (2016) 1–32, Nanostructured lipid carrier-loaded hyaluronic acid microneedles for controlled
https://doi.org/10.1016/j.mser.2016.03.001. dermal delivery of a lipophilic molecule, Int. J, Nanomedicine, 2013https://doi.
[93] G. Singhvi, N. Hans, N. Shiva, S. Kumar Dubey, Xanthan gum in drug delivery appli- org/10.2147/IJN.S54529.
cations, in: Nat, Elsevier, Polysaccharides Drug Deliv. Biomed. Appl., 2019 121–144, [119] S.Y. Choi, H.J. Kwon, G.R. Ahn, E.J. Ko, K.H. Yoo, B.J. Kim, C. Lee, D. Kim, Hyaluronic
https://doi.org/10.1016/b978-0-12-817055-7.00005-4. acid microneedle patch for the improvement of crow’s feet wrinkles, Dermatol.
[94] G. Sworn, Xanthan gum, in: Handb, Hydrocoll. Second Ed. 2009https://doi.org/10. Ther. (2017)https://doi.org/10.1111/dth.12546.
1533/9781845695873.186. [120] Y. Hao, Y. Chen, M. Lei, T. Zhang, Y. Cao, J. Peng, L. Chen, Z. Qian, Near-infrared re-
[95] R.S.J. Ingrole, H.S. Gill, Microneedle coating methods: a review with a perspective, J. sponsive PEGylated gold nanorod and doxorubicin loaded dissolvable hyaluronic
Pharmacol. Exp. Ther. (2019)https://doi.org/10.1124/jpet.119.258707. acid microneedles for human epidermoid cancer therapy, Adv. Ther. (2018)
[96] Y.K. Demir, Z. Akan, O. Kerimoglu, Characterization of polymeric microneedle ar- https://doi.org/10.1002/adtp.201800008.
rays for transdermal drug delivery, PLoS One, 2013https://doi.org/10.1371/ [121] H. Yang, X. Wu, Z. Zhou, X. Chen, M. Kong, Enhanced transdermal lymphatic deliv-
journal.pone.0077289. ery of doxorubicin via hyaluronic acid based transfersomes/microneedle complex
[97] Y.K. Demir, Z. Akan, O. Kerimoglu, Sodium alginate microneedle arrays mediate the for tumor metastasis therapy, Int. J. Biol. Macromol. 125 (2019) 9–16, https://doi.
transdermal delivery of bovine serum albumin, PLoS One, 2013https://doi.org/10. org/10.1016/j.ijbiomac.2018.11.230.
1371/journal.pone.0063819. [122] S. Selwyn, J. Durodie, The antimicrobial activity of sugar against pathogens of
wounds and other infections of man, in: Prop, Water Foods, Springer
[98] Y. Zhang, G. Jiang, W. Yu, D. Liu, B. Xu, Microneedles fabricated from alginate and
Netherlands, 1985 293–308, https://doi.org/10.1007/978-94-009-5103-7_18.
maltose for transdermal delivery of insulin on diabetic rats, Mater. Sci. Eng. C.
(2018)https://doi.org/10.1016/j.msec.2017.12.006. [123] M. Milewski, Y. Kapoor, Z. Ding, J. Zhang, E. Ghartey-Tagoe, K. Manser, B. Nissley, I.
Petrescu, L. Xu, B. Duffield, G. Chen, Q. Yang, P. Singh, R. Smith, Stabilization and
[99] F. Dias, C. Duarte, Cellulose and its derivatives use in the pharmaceutical
transdermal delivery of an investigational peptide using MicroCor® solid-state dis-
compounding practice, Cellul. - Medical, Pharm. Electron. Appl. (2013). doi:
solving microstructure arrays, J. Pharm. Sci. (2020)https://doi.org/10.1016/j.xphs.
https://doi.org/10.5772/56637.
2019.11.006.
[100] R. Ergun, J. Guo, B. Huebner-Keese, Cellulose, in: Encycl, Food Heal. 2015https://
[124] T. Ilić, S. Savić, B. Batinić, B. Marković, M. Schmidberger, D. Lunter, M. Savić, S. Savić,
doi.org/10.1016/B978-0-12-384947-2.00127-6.
Combined use of biocompatible nanoemulsions and solid microneedles to improve
[101] Y.H. Park, S.K. Ha, I. Choi, K.S. Kim, J. Park, N. Choi, B. Kim, J.H. Sung, Fabrication of
transport of a model NSAID across the skin: in vitro and in vivo studies, Eur. J.
degradable carboxymethyl cellulose (CMC) microneedle with laser writing and
Pharm. Sci. (2018)https://doi.org/10.1016/j.ejps.2018.09.023.
replica molding process for enhancement of transdermal drug delivery, Biotechnol.
[125] S. Gopi, A. Amalraj, N.P. Sukumaran, J.T. Haponiuk, S. Thomas, Biopolymers and
Bioprocess Eng. (2016)https://doi.org/10.1007/s12257-015-0634-7.
their composites for drug delivery: a brief review, Macromol. Symp. (2018)
[102] L.E. González García, M.N. MacGregor, R.M. Visalakshan, N. Ninan, A.A. Cavallaro,
https://doi.org/10.1002/masy.201800114.
A.D. Trinidad, Y. Zhao, A.J.D. Hayball, K. Vasilev, Self-sterilizing antibacterial
[126] H.S. Gill, M.R. Prausnitz, Coating formulations for microneedles, Pharm. Res. (2007)
silver-loaded microneedles, Chem. Commun. (2019)https://doi.org/10.1039/
https://doi.org/10.1007/s11095-007-9286-4.
c8cc06035e.
[127] Y.C. Kim, F.S. Quan, R.W. Compans, S.M. Kang, M.R. Prausnitz, Formulation of
[103] M.C. Chen, S.F. Huang, K.Y. Lai, M.H. Ling, Fully embeddable chitosan microneedles microneedles coated with influenza virus-like particle vaccine, AAPS
as a sustained release depot for intradermal vaccination, Biomaterials, 2013https:// PharmSciTech, 2010https://doi.org/10.1208/s12249-010-9471-3.
doi.org/10.1016/j.biomaterials.2012.12.041.
[128] H.J. Choi, J.M. Song, B.J. Bondy, R.W. Compans, S.M. Kang, M.R. Prausnitz, Effect of
[104] Z. Ahmad, M.I. Khan, M.I. Siddique, H.S. Sarwar, G. Shahnaz, S.Z. Hussain, N.I. osmotic pressure on the stability of whole inactivated influenza vaccine for coating
Bukhari, I. Hussain, M.F. Sohail, Fabrication and characterization of thiolated chito- on microneedles, PLoS One, 2015https://doi.org/10.1371/journal.pone.0134431.
san microneedle patch for transdermal delivery of tacrolimus, AAPS PharmSciTech, [129] D.F.S. Fonseca, P.C. Costa, I.F. Almeida, P. Dias-Pereira, I. Correia-Sá, V. Bastos, H.
2020https://doi.org/10.1208/s12249-019-1611-9. Oliveira, M. Duarte-Araújo, M. Morato, C. Vilela, A.J.D. Silvestre, C.S.R. Freire,
[105] N. ‘Izzah Ibrahim, S.K. Wong, I.N. Mohamed, N. Mohamed, K.Y. Chin, S. Ima- Pullulan microneedle patches for the efficient transdermal administration of insu-
Nirwana, A.N. Shuid, Wound healing properties of selected natural products, Int. lin envisioning diabetes treatment, Carbohydr. Polym. (2020)https://doi.org/10.
J. Environ. Res. Public Health. 15 (2018). doi:https://doi.org/10.3390/ 1016/j.carbpol.2020.116314.
ijerph15112360. [130] L.K. Vora, A.J. Courtenay, I.A. Tekko, E. Larrañeta, R.F. Donnelly, Pullulan-based dis-
[106] M.C. Chen, M.H. Ling, K.Y. Lai, E. Pramudityo, Chitosan microneedle patches for solving microneedle arrays for enhanced transdermal delivery of small and large
sustained transdermal delivery of macromolecules, Biomacromolecules, biomolecules, Int. J. Biol. Macromol. (2020)https://doi.org/10.1016/j.ijbiomac.
2012https://doi.org/10.1021/bm301293d. 2019.12.184.
[107] A. Sadeqi, H.R. Nejad, G. Kiaee, S. Sonkusale, Cost-effective fabrication of chitosan [131] S. Luo, S. Song, C. Zheng, Y. Wang, X. Xia, B. Liang, G. Feng, Biocompatibility of
microneedles for transdermal drug delivery, Conf. Proc. ... Annu. Int. Conf. IEEE Bletilla striata Microspheres as a Novel Embolic Agent (2015)https://doi.org/10.
Eng. Med. Biol. Soc. IEEE Eng. Med. Biol. Soc. Annu. Conf. (2018). doi:https://doi. 1155/2015/840896.
org/10.1109/EMBC.2018.8513691. [132] L. Hu, Z. Liao, Q. Hu, K.G. Maffucci, Y. Qu, Novel Bletilla striata polysaccharide
[108] A. Chandrasekharan, Y.J. Hwang, K.Y. Seong, S. Park, S. Kim, S.Y. Yang, Acid-treated microneedles: fabrication, characterization, and in vitro transcutaneous drug de-
water-soluble chitosan suitable for microneedle-assisted intracutaneous drug de- livery, Int. J. Biol. Macromol. (2018)https://doi.org/10.1016/j.ijbiomac.2018.05.
livery, Pharmaceutics, 2019https://doi.org/10.3390/pharmaceutics11050209. 097.
[109] J. Chi, X. Zhang, C. Chen, C. Shao, Y. Zhao, Y. Wang, Antibacterial and angiogenic chi- [133] W. Yu, G. Jiang, Y. Zhang, D. Liu, B. Xu, J. Zhou, Polymer microneedles fabricated
tosan microneedle array patch for promoting wound healing, Bioact, Mater, from alginate and hyaluronate for transdermal delivery of insulin, Mater. Sci.
2020https://doi.org/10.1016/j.bioactmat.2020.02.004. Eng. C. (2017)https://doi.org/10.1016/j.msec.2017.05.143.
[110] Y. gang Shi, Y. cheng Meng, J. rong Li, J. Chen, Y. hua Liu, X. Bai, Chondroitin sulfate: [134] S. Kim, J. Eum, H. Yang, H. Jung, Transdermal finasteride delivery via powder-
extraction, purification, microbial and chemical synthesis, J. Chem. Technol. carrying microneedles with a diffusion enhancer to treat androgenetic alopecia,
Biotechnol. (2014). doi:https://doi.org/10.1002/jctb.4454. J. Control. Release (2019)https://doi.org/10.1016/j.jconrel.2019.11.002.
[111] A. Aravamudhan, D.M. Ramos, A.A. Nada, S.G. Kumbar, Natural polymers, in: Nat, [135] S. Fakhraei Lahiji, S.H. Seo, S. Kim, M. Dangol, J. Shim, C.G. Li, Y. Ma, C. Lee, G. Kang,
Synth. Biomed. Polym. 2014https://doi.org/10.1016/b978-0-12-396983-5.00004- H. Yang, K.Y. Choi, H. Jung, Transcutaneous implantation of valproic acid-
1. encapsulated dissolving microneedles induces hair regrowth, Biomaterials,
[112] D. Poirier, F. Renaud, V. Dewar, L. Strodiot, F. Wauters, J. Janimak, T. Shimada, T. 2018https://doi.org/10.1016/j.biomaterials.2018.03.019.
Nomura, K. Kabata, K. Kuruma, T. Kusano, M. Sakai, H. Nagasaki, T. Oyamada, Hep- [136] H.R. Jeong, J.Y. Kim, S.N. Kim, J.H. Park, Local dermal delivery of cyclosporin a, a hy-
atitis B surface antigen incorporated in dissolvable microneedle array patch is an- drophobic and high molecular weight drug, using dissolving microneedles, Eur. J.
tigenic and thermostable, Biomaterials, 2017https://doi.org/10.1016/j. Pharm. Biopharm. (2018)https://doi.org/10.1016/j.ejpb.2018.02.014.
biomaterials.2017.08.038. [137] J.Y. Kim, M.R. Han, Y.H. Kim, S.W. Shin, S.Y. Nam, J.H. Park, Tip-loaded dissolving
[113] S. Liu, S. Zhang, Y. Duan, Y. Niu, H. Gu, Z. Zhao, S. Zhang, Y. Yang, X. Wang, Y. Gao, P. microneedles for transdermal delivery of donepezil hydrochloride for treatment
Yang, Transcutaneous immunization of recombinant Staphylococcal enterotoxin B of Alzheimer’s disease, Eur. J. Pharm. Biopharm. 105 (2016) 148–155, https://doi.
protein using a dissolving microneedle provides potent protection against lethal org/10.1016/j.ejpb.2016.06.006.

619
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

[138] K. Siddhapura, H. Harde, S. Jain, Immunostimulatory effect of tetanus toxoid loaded [161] W. Yu, G. Jiang, D. Liu, L. Li, H. Chen, Y. Liu, Q. Huang, Z. Tong, J. Yao, X. Kong, Fab-
chitosan nanoparticles following microneedles assisted immunization, rication of biodegradable composite microneedles based on calcium sulfate and
Nanomedicine Nanotechnology, Biol. Med, 2016https://doi.org/10.1016/j.nano. gelatin for transdermal delivery of insulin, Mater. Sci. Eng. C. (2017)https://doi.
2015.10.009. org/10.1016/j.msec.2016.10.063.
[139] Y.-S. Tsai, M.-Y. Chen, S.-K. Lan, H.-T. Tsai, M.-C. Chen, T.-S. Tzai, Transdermal deliv- [162] O. Olatunji, C.C. Igwe, A.S. Ahmed, D.O.A. Alhassan, G.O. Asieba, B. Das Diganta,
ery of leuprolide acetate with chitosan microneedles: a promising tool for andro- Microneedles from fish scale biopolymer, J. Appl. Polym. Sci. 131 (2014) 1–10,
gen deprivation therapy, Eur. Urol. Suppl. (2017)https://doi.org/10.1016/s1569- https://doi.org/10.1002/app.40377.
9056(17)30808-4. [163] O. Olatunji, A. Denloye, Production of hydrogel microneedles from fish scale bio-
[140] P. Schipper, K. van der Maaden, V. Groeneveld, M. Ruigrok, S. Romeijn, S. Uleman, polymer, J. Polym. Environ. (2019)https://doi.org/10.1007/s10924-019-01426-x.
C. Oomens, G. Kersten, W. Jiskoot, J. Bouwstra, Diphtheria toxoid and N-trimethyl [164] O. Olatunji, R.T. Olsson, Microneedles from fishscale-nanocellulose blends using
chitosan layer-by-layer coated pH-sensitive microneedles induce potent immune low temperature mechanical press method, Pharmaceutics, 2015https://doi.org/
responses upon dermal vaccination in mice, J. Control. Release (2017)https://doi. 10.3390/pharmaceutics7040363.
org/10.1016/j.jconrel.2017.07.017. [165] X. You, J.H. Chang, B.K. Ju, J.J. Pak, Rapidly dissolving fibroin microneedles for trans-
[141] M.C. Chen, K.Y. Lai, M.H. Ling, C.W. Lin, Enhancing immunogenicity of antigens dermal drug delivery, Mater. Sci. Eng. C. (2011)https://doi.org/10.1016/j.msec.
through sustained intradermal delivery using chitosan microneedles with a 2011.06.010.
patch-dissolvable design, Acta Biomater, 2018https://doi.org/10.1016/j.actbio. [166] S. Wang, M. Zhu, L. Zhao, D. Kuang, S.C. Kundu, S. Lu, Insulin-loaded silk fibroin
2017.11.004.
microneedles as sustained release system, ACS Biomater. Sci, Eng, 2019https://
[142] T. Tomono, A new way to control the internal structure of microneedles: a case of doi.org/10.1021/acsbiomaterials.9b00229.
chitosan lactate, Mater, Today Chem, 2019https://doi.org/10.1016/j.mtchem.2019.
[167] Z. Yin, D. Kuang, S. Wang, Z. Zheng, V.K. Yadavalli, S. Lu, Swellable silk fibroin
04.009.
microneedles for transdermal drug delivery, Int. J. Biol. Macromol. (2018)https://
[143] A.F. Moreira, C.F. Rodrigues, T.A. Jacinto, S.P. Miguel, E.C. Costa, I.J. Correia, Poly
doi.org/10.1016/j.ijbiomac.2017.07.178.
(vinyl alcohol)/chitosan layer-by-layer microneedles for cancer chemo-
[168] J.Y. Lee, S.H. Park, I.H. Seo, K.J. Lee, W.H. Ryu, Rapid and repeatable fabrication of
photothermal therapy, Int. J. Pharm. (2020)https://doi.org/10.1016/j.ijpharm.
high A/R silk fibroin microneedles using thermally-drawn micromolds, Eur. J.
2019.118907.
Pharm. Biopharm. (2015)https://doi.org/10.1016/j.ejpb.2015.04.024.
[144] A. Nakatsukasa, K. Kuruma, M. Okamatsu, T. Hiono, M. Suzuki, K. Matsuno, H. Kida,
T. Oyamada, Y. Sakoda, Potency of whole virus particle and split virion vaccines [169] N. Elharfaoui, M. Djabourov, W. Babel, Molecular weight influence on gelatin gels:
using dissolving microneedle against challenges of H1N1 and H5N1 influenza vi- structure, enthalpy and rheology, Macromol. Symp. 256 (2007) 149–157, https://
ruses in mice, Vaccine, 2017https://doi.org/10.1016/j.vaccine.2017.04.009. doi.org/10.1002/masy.200751017.
[145] W. Yao, C. Tao, J. Zou, H. Zheng, J. Zhu, Z. Zhu, J. Zhu, L. Liu, F. Li, X. Song, Flexible [170] Q. Xing, K. Yates, C. Vogt, Z. Qian, M.C. Frost, F. Zhao, Increasing mechanical
two-layer dissolving and safing microneedle transdermal of neurotoxin: a strength of gelatin hydrogels by divalent metal ion removal, Sci. Rep. 4 (2014)
biocomfortable attempt to treat rheumatoid arthritis, Int. J. Pharm. 563 (2019) 1–10, https://doi.org/10.1038/srep04706.
91–100, https://doi.org/10.1016/j.ijpharm.2019.03.033. [171] C. Vepari, D.L. Kaplan, Silk as a biomaterial, Prog. Polym. Sci. 32 (2007) 991–1007,
[146] S. Naito, Y. Ito, T. Kiyohara, M. Kataoka, M. Ochiai, K. Takada, Antigen-loaded dis- https://doi.org/10.1016/j.progpolymsci.2007.05.013.
solving microneedle array as a novel tool for percutaneous vaccination, Vaccine, [172] P. Song, W. Zhang, C. Xu, H. Guan, Y. Tu, G. Wu, Effects of silkworm variety on the
2012https://doi.org/10.1016/j.vaccine.2011.11.111. mechanical and structural properties of silk, J. Chinese Adv. Mater. Soc. 6 (2018)
[147] K. Fukushima, A. Ise, H. Morita, R. Hasegawa, Y. Ito, N. Sugioka, K. Takada, Two- 827–837, https://doi.org/10.1080/22243682.2018.1556334.
layered dissolving microneedles for percutaneous delivery of peptide/protein [173] D.C. Aduba, H. Yang, Polysaccharide fabrication platforms and biocompatibility as-
drugs in rats, Pharm. Res. (2011)https://doi.org/10.1007/s11095-010-0097-7. sessment as candidate wound dressing materials, Bioengineering. 4 (2017). doi:
[148] Z. Gui, X. Wu, S. Wang, Y. Cao, J. Wan, Q. Shan, Z. Yang, J. Zhang, S. Gui, Dissolving https://doi.org/10.3390/bioengineering4010001.
microneedles integrated with liquid crystals facilitate transdermal delivery of [174] G. Bonfante, H. Lee, L. Bao, J. Park, N. Takama, B. Kim, Comparison of polymers to
sinomenine hydrochloride, J. Pharm. Sci. (2017)https://doi.org/10.1016/j.xphs. enhance mechanical properties of microneedles for bio-medical applications,
2017.07.027. Micro Nano Syst. Lett. 8 (2020) 13, https://doi.org/10.1186/s40486-020-00113-0.
[149] Y. Ito, M. Taniguchi, A. Hayashi, M. Anai, S. Morita, E. Ko, N. Yoshimoto, Y. Yoshii, S. [175] Y. Zhang, G. Jiang, W. Yu, D. Liu, B. Xu, Microneedles fabricated from alginate and
Kobuchi, T. Sakaeda, K. Takada, Application of dissolving microneedles to glucose maltose for transdermal delivery of insulin on diabetic rats, Mater. Sci. Eng. C. 85
monitoring through dermal interstitial fluid, Biol. Pharm. Bull. (2014)https://doi. (2018) 18–26, https://doi.org/10.1016/j.msec.2017.12.006.
org/10.1248/bpb.b14-00406. [176] Z. Cheng, H. Lin, Z. Wang, X. Yang, M. Zhang, X. Liu, B. Wang, Z. Wu, D. Chen, Prep-
[150] H. Kim, K.Y. Seong, J.H. Lee, W. Park, S.Y. Yang, S.K. Hahn, Biodegradable aration and characterization of dissolving hyaluronic acid composite microneedles
microneedle patch delivering antigenic peptide-hyaluronate conjugate for cancer loaded micelles for delivery of curcumin, Drug Deliv. Transl. Res. 10 (2020)
immunotherapy, ACS Biomater. Sci, Eng, 2019https://doi.org/10.1021/ 1520–1530, https://doi.org/10.1007/s13346-020-00735-2.
acsbiomaterials.9b00961. [177] Y.H. Chiu, M.C. Chen, S.W. Wan, Sodium hyaluronate/chitosan composite
[151] Y. Qiu, C. Li, S. Zhang, G. Yang, M. He, Y. Gao, Systemic delivery of artemether by microneedles as a single-dose intradermal immunization system,
dissolving microneedles, Int. J. Pharm. (2016)https://doi.org/10.1016/j.ijpharm. Biomacromolecules. 19 (2018) 2278–2285, https://doi.org/10.1021/acs.biomac.
2016.05.006. 8b00441.
[152] Z. Chen, B. Han, L. Liao, X. Hu, Q. Hu, Y. Gao, Y. Qiu, Enhanced transdermal delivery [178] M.H. Ling, M.C. Chen, Dissolving polymer microneedle patches for rapid and effi-
of polydatin via a combination of inclusion complexes and dissolving microneedles cient transdermal delivery of insulin to diabetic rats, Acta Biomater. 9 (2013)
for treatment of acute gout arthritis, J. Drug Deliv. Sci. Technol. (2020)https://doi. 8952–8961, https://doi.org/10.1016/j.actbio.2013.06.029.
org/10.1016/j.jddst.2019.101487. [179] A.N. Cadinoiu, D.M. Rata, L.I. Atanase, Biocompatible injectable polysaccharide ma-
[153] J. Zhu, L. Dong, H. Du, J. Mao, Y. Xie, H. Wang, J. Lan, Y. Lou, Y. Fu, J. Wen, B. Jiang, Y. terials for drug delivery, in: Polysacch, Elsevier, Carriers Drug Deliv, 2019 127–154,
Li, J. Zhu, J. Tao, 5-Aminolevulinic acid-loaded hyaluronic acid dissolving https://doi.org/10.1016/B978-0-08-102553-6.00006-4.
microneedles for effective photodynamic therapy of superficial tumors with en-
[180] E. Papakonstantinou, M. Roth, G. Karakiulakis, Hyaluronic acid: a key molecule in
hanced long-term stability, Adv. Healthc, Mater, 2019https://doi.org/10.1002/
skin aging, Dermatoendocrinol. 4 (2012) 253, https://doi.org/10.4161/derm.
adhm.201900896.
21923.
[154] H. Du, P. Liu, J. Zhu, J. Lan, Y. Li, L. Zhang, J. Zhu, J. Tao, Hyaluronic acid-based dis-
[181] G. Kogan, L. Šoltés, R. Stern, J. Schiller, R. Mendichi, Hyaluronic acid: its function
solving microneedle patch loaded with methotrexate for improved treatment of
and degradation in in vivo systems, Stud. Nat. Prod. Chem. 34 (2008) 789–882,
psoriasis, ACS Appl. Mater. Interfaces (2019)https://doi.org/10.1021/acsami.
https://doi.org/10.1016/S1572-5995(08)80035-X.
9b15668.
[182] G.G. D’Ayala, M. Malinconico, P. Laurienzo, Marine derived polysaccharides for bio-
[155] G. Li, A. Badkar, S. Nema, C.S. Kolli, A.K. Banga, In vitro transdermal delivery of ther-
medical applications: chemical modification approaches, Molecules. 13 (2008)
apeutic antibodies using maltose microneedles, Int. J. Pharm. (2009)https://doi.
2069–2106, https://doi.org/10.3390/molecules13092069.
org/10.1016/j.ijpharm.2008.10.008.
[156] C. Dillon, H. Hughes, N.J. O’Reilly, C.J. Allender, D.A. Barrow, P. McLoughlin, Dissolv- [183] T. Sato, T. Ishii, Y. Okahata, In vitro gene delivery mediated by chitosan. Effect of
ing microneedle based transdermal delivery of therapeutic peptide analogues, Int. pH, serum, and molecular mass of chitosan on the transfection efficiency, Biomate-
J. Pharm. (2019)https://doi.org/10.1016/j.ijpharm.2019.04.075. rials. 22 (2001) 2075–2080. doi:https://doi.org/10.1016/S0142-9612(00)00385-9.
[157] A. Donadei, H. Kraan, O. Ophorst, O. Flynn, C. O’Mahony, P.C. Soema, A.C. Moore, [184] S. Bhatnagar, P.R. Gadeela, P. Thathireddy, V.V.K. Venuganti, Microneedle-based
Skin delivery of trivalent sabin inactivated poliovirus vaccine using dissolvable drug delivery: materials of construction, J. Chem. Sci. 131 (2019) 1–28, https://
microneedle patches induces neutralizing antibodies, J. Control. Release (2019) doi.org/10.1007/s12039-019-1666-x.
https://doi.org/10.1016/j.jconrel.2019.08.039. [185] Jawaid Marichelvam, Asim, corn and rice starch-based bio-plastics as alternative
[158] S. Bhatnagar, S.R. Chawla, O.P. Kulkarni, V.V.K. Venuganti, Zein Microneedles for packaging materials, Fibers. 7 (2019) 32, https://doi.org/10.3390/fib7040032.
Transcutaneous Vaccine Delivery: Fabrication, Characterization, and In Vivo Evalu- [186] Xanthan Gums | Cargill Pharmaceutical | Cargill, (n.d.). https://www.cargill.com/
ation Using Ovalbumin as the Model Antigen, ACS Omega, 2017https://doi.org/10. pharmaceutical/hydrocolloids-for-pharma/xanthan-gums-for-pharma (accessed
1021/acsomega.7b00343. December 15, 2020).
[159] A. Aditya, B. Kim, R.D. Koyani, B. Oropeza, M. Furth, J. Kim, N.P. Kim, Kinetics of col- [187] D.F.S. Petri, Xanthan gum: a versatile biopolymer for biomedical and technological
lagen microneedle drug delivery system, J. Drug Deliv. Sci. Technol. (2019)https:// applications, J. Appl. Polym. Sci. 132 (2015). doi:https://doi.org/10.1002/app.
doi.org/10.1016/j.jddst.2019.03.007. 42035.
[160] W. Yu, G. Jiang, D. Liu, L. Li, Z. Tong, J. Yao, X. Kong, Transdermal delivery of insulin [188] M.-B. Coltelli, S. Danti, K. De Clerck, A. Lazzeri, P. Morganti, Pullulan for advanced
with bioceramic composite microneedles fabricated by gelatin and hydroxyapatite, sustainable body- and skin-contact applications, J. Funct. Biomater. 11 (2020) 20,
Mater. Sci. Eng. C. (2017)https://doi.org/10.1016/j.msec.2016.12.111. https://doi.org/10.3390/jfb11010020.

620
N. Dabholkar, S. Gorantla, T. Waghule et al. International Journal of Biological Macromolecules 170 (2021) 602–621

[189] A.K. Lynn, I.V. Yannas, W. Bonfield, Antigenicity and immunogenicity of collagen, J. [202] E. Peschl, K.W. Bauer, E. Peschl, K.W. Bauer, RU2701361C1, Über Nichtlineare Dif-
Biomed. Mater. Res. - Part B Appl. Biomater. 71 (2004) 343–354, https://doi.org/10. fer. 2. Ordnung, Die Für Eine Abschätzungsmethode Bei Partiellen Differ. Vom
1002/jbm.b.30096. Elliptischen Typus Bes. Wicht. Sind. 361 (1964) 18–30. doi:https://doi.org/10.
[190] W. Yu, G. Jiang, D. Liu, L. Li, H. Chen, Y. Liu, Q. Huang, Z. Tong, J. Yao, X. Kong, Fab- 1007/978-3-322-98502-6_4.
rication of biodegradable composite microneedles based on calcium sulfate and [203] Japanese Inventor, JP2019523680A, JP2019523680A, 2017.
gelatin for transdermal delivery of insulin, Mater. Sci. Eng. C. 71 (2017) 725–734, [204] R.F. Donnelly, D. Deliv, RU2016144039A, (2016) 1–21.
https://doi.org/10.1016/j.msec.2016.10.063. [205] T. Tepzz, H. Davon, EP2934660B1, 1 (2019) 1–28.
[191] W. Yu, G. Jiang, D. Liu, L. Li, Z. Tong, J. Yao, X. Kong, Transdermal delivery of insulin [206] E.M.P. David L. Kaplan, Konstantinos Tsioris, Fiorenzo G. Omenetto, CA2815285C,
with bioceramic composite microneedles fabricated by gelatin and hydroxyapatite, CA2815285C, 2020.
Mater. Sci. Eng. C. 73 (2017) 425–428, https://doi.org/10.1016/j.msec.2016.12.111. [207] M. Bok, Z.J. Zhao, S. Jeon, J.H. Jeong, E. Lim, Ultrasonically and iontophoretically en-
[192] A.A. El-Rashidy, G. Waly, A. Gad, J.A. Roether, J. Hum, Y. Yang, R. Detsch, A.A. hanced drug-delivery system based on dissolving microneedle patches, Sci. Rep.
Hashem, I. Sami, W.H. Goldmann, A.R. Boccaccini, Antibacterial activity and bio- (2020)https://doi.org/10.1038/s41598-020-58822-w.
compatibility of zein scaffolds containing silver-doped bioactive glass, Biomed.
[208] J. Madden, C. O’Mahony, M. Thompson, A. O’Riordan, P. Galvin, Biosensing in der-
Mater. 13 (2018). doi:https://doi.org/10.1088/1748-605X/aad8cf.
mal interstitial fluid using microneedle based electrochemical devices, Sens, Bio-
[193] J. Dong, Q. Sun, J.Y. Wang, Basic study of corn protein, zein, as a biomaterial in tis-
Sensing Res, 2020https://doi.org/10.1016/j.sbsr.2020.100348.
sue engineering, surface morphology and biocompatibility, Biomaterials. 25
[209] K.J. Lee, J.Y. Lee, S.G. Lee, S.H. Park, D.S. Yang, J.J. Lee, A. Khademhosseini, J.S. Kim,
(2004) 4691–4697, https://doi.org/10.1016/j.biomaterials.2003.10.084.
W.H. Ryu, Microneedle drug eluting balloon for enhanced drug delivery to vascular
[194] T. Yucel, M.L. Lovett, D.L. Kaplan, Silk-based biomaterials for sustained drug deliv-
tissue, J. Control. Release (2020)https://doi.org/10.1016/j.jconrel.2020.02.012.
ery, J. Control. Release 190 (2014) 381–397, https://doi.org/10.1016/j.jconrel.2014.
05.059. [210] O. Howells, N. Rajendran, S. Mcintyre, S. Amini-Asl, P. Henri, Y. Liu, O. Guy, A.E.G.
[195] R.F. Donnelly, of Microneedle-based Products, (2018) 307–322. Cass, M.C. Morris, S. Sharma, Microneedle array-based platforms for future
[196] Micro needle array-doxorubicin (MNA-D) in patients with cutaneous t-cell lym- theranostic applications, ChemBioChem, 2019https://doi.org/10.1002/cbic.
phoma (CTCL) - full text view - ClinicalTrials.gov, (n.d.). https://clinicaltrials.gov/ 201900112.
ct2/show/NCT02192021 (accessed July 7, 2020). [211] C.P.P. Pere, S.N. Economidou, G. Lall, C. Ziraud, J.S. Boateng, B.D. Alexander, D.A.
[197] Home - ClinicalTrials.gov, (n.d.). https://clinicaltrials.gov/ (accessed February 8, Lamprou, D. Douroumis, 3D printed microneedles for insulin skin delivery, Int. J.
2019). Pharm. (2018)https://doi.org/10.1016/j.ijpharm.2018.03.031.
[198] M. Kim, H. Yang, H. Kim, H. Jung, H. Jung, Novel cosmetic patches for wrinkle im- [212] S.N. Economidou, C.P.P. Pere, A. Reid, M.J. Uddin, J.F.C. Windmill, D.A. Lamprou, D.
provement: retinyl retinoate- and ascorbic acid-loaded dissolving microneedles, Douroumis, 3D printed microneedle patches using stereolithography (SLA)for in-
Int. J. Cosmet. Sci. (2014)https://doi.org/10.1111/ics.12115. tradermal insulin delivery, Mater. Sci. Eng. C. (2019)https://doi.org/10.1016/j.
[199] J.H. Lee, Y.S. Jung, G.M. Kim, J.M. Bae, A hyaluronic acid-based microneedle patch to msec.2019.04.063.
treat psoriatic plaques: a pilot open trial, Br. J. Dermatol. (2018)https://doi.org/10. [213] K. Moussi, A. Bukhamsin, T. Hidalgo, J. Kosel, Biocompatible 3D printed
1111/bjd.15779. microneedles for transdermal, intradermal, and percutaneous applications, Adv.
[200] M. Avcil, G. Akman, J. Klokkers, D. Jeong, A. Çelik, Efficacy of bioactive peptides Eng. Mater. (2020)https://doi.org/10.1002/adem.201901358.
loaded on hyaluronic acid microneedle patches: a monocentric clinical study, J. [214] D. Han, R.S. Morde, S. Mariani, A.A. La Mattina, E. Vignali, C. Yang, G. Barillaro, H.
Cosmet. Dermatol. (2020)https://doi.org/10.1111/jocd.13009. Lee, 4D printing of a bioinspired microneedle array with backward-facing barbs
[201] M. Jang, S. Baek, G. Kang, H. Yang, S. Kim, H. Jung, Dissolving microneedle with high for enhanced tissue adhesion, Adv. Funct. Mater. (2020)https://doi.org/10.1002/
molecular weight hyaluronic acid to improve skin wrinkles, dermal density and adfm.201909197.
elasticity, Int. J. Cosmet. Sci. 42 (2020) 302–309, https://doi.org/10.1111/ics.12617.

621

You might also like