You are on page 1of 22

International Journal of Pharmaceutics 614 (2022) 121439

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Skin interaction, permeation, and toxicity of silica nanoparticles:


Challenges and recent therapeutic and cosmetic advances
Renata Pinho Morais a, *, Sabrina Hochheim b, Carolina C. de Oliveira c, Izabel C. Riegel-Vidotti b,
Cláudia E.B. Marino a
a
Department of Mechanical Engineering, Universidade Federal do Paraná, Curitiba, Brazil
b
Department of Chemistry, Universidade Federal do Paraná, Curitiba, Brazil
c
Department of Cell Biology, Universidade Federal do Paraná, Curitiba, Brazil

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

Keywords: Silica nanoparticles (SNPs) received more attention with the emergence of nanotechnology with the aim and
Silica nanoparticles promise of becoming innovative drug delivery systems. They have been fulfilling this objective with excellence
Topical drug delivery and nowadays they play a central role in biomedical applications. New SNPs application routes are being
Skin permeation
explored such as the epidermal, dermal, and transdermal routes. With that, novel models of synthesis, func­
Skin toxicity
tionalization, and applications constantly appear. However, it is essential that such innovations are accompanied
Topical formulations
by in-depth studies on permeation, biodistribution, metabolization, and elimination of the generated by-
products. Such studies are still incipient, if not rare. This article reviews significant findings on SNPs and
their skin interactions. An extensive literature review on SNPs synthesis and functionalization methodologies was
performed, as well as on the skin characteristics, skin permeation mechanisms, and in vivo toxicity assessments.
Furthermore, studies of the past 5 years on the main therapeutic and cosmetic products employing SNPs, with
greater emphasis on in vivo and ex vivo studies were included.

1. Introduction and motivation purpose must be very clear, thus from the beginning of development the
right direction can be taken. Skin administration of active compounds
There is a long history of substances application on the skin for either generally has three different goals: non-absorption, local absorption
cosmetic or skin treatments, disclosed in the oldest medical records and (epidermis and dermis), and transdermal absorption. Cosmetics, insect
in all periods of human evolution (Khavkin and Ellis, 2011; Kolarsick repellents, antifungals or disinfectants are examples of common for­
et al., 2011). Today, the market for transepidermal products is on the mulations designed to maintain the active compound on the skin sur­
rise, as it is a comfortable, safe, and non-invasive route for patients face. Topical formulations allow the active compounds to penetrate into
(Jepps et al., 2013; Zsikó et al., 2019; Bakshi et al., 2020). In addition, the skin deepest regions, and transdermal formulations aim to supply
the skin is a social organ, as it reflects our contact with society. In­ active compounds to systemic circulation (Lane, 2011).
dividuals who suffer from skin disorders can develop mental disorders Different strategies have been developed to overcome the skin
and poor quality of life due to low self-esteem (Cortés et al., 2021; Dreno outermost layer limiting barrier, the stratum corneum, to improve drugs
et al., 2021). In this way, cosmetic care increasingly co-emerges with permeation, which include chemical enhancers (ethanol, fatty acids, and
dermatocosmetic technology as another treatment option. (Khavkin and polysorbates) and physical methods (iontophoresis, electroporation,
Ellis, 2011). sonophoresis, and microneedles). These penetration enhancers, in gen­
However, due to a complex structure of multiple layers, the skin acts eral, promote drug diffusion by disturbing stratum corneum and/or
as a barrier between the body and the external environment, regulating deeper layers structure, which can attack skin causing irritation and
the transit of substances in a highly selective manner (Kolarsick et al., even toxic adverse effects (Alexander et al., 2012; Foldvari, 2000; Goyal
2011; Wong et al., 2016). When planning a dermatological product, its et al., 2016; Medi et al., 2017). In addition, drugs classified by BCS

* Corresponding author at: Grupo de Biomateriais, Eletroquímica e Corrosão, Universidade Federal do Paraná, ZIP Code: 81531-980, Curitiba, Brazil.
E-mail addresses: renata.morais@ufpr.br (R.P. Morais), sabrina.hochheim@ufpr.br (S. Hochheim), krokoli@ufpr.br (C.C. de Oliveira), izabel.riegel@ufpr.br
(I.C. Riegel-Vidotti), claudiamarino@ufpr.br (C.E.B. Marino).

https://doi.org/10.1016/j.ijpharm.2021.121439
Received 23 September 2021; Received in revised form 16 December 2021; Accepted 28 December 2021
Available online 3 January 2022
0378-5173/© 2021 Elsevier B.V. All rights reserved.
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

(Biopharmaceutical Classification System) as belonging to class II (low biomedical applications. There are two basic silica structures: crystalline
solubility and high permeability) and class IV (low solubility and low and amorphous. Both forms have the same molecular formula, but their
permeability) are considered the most problematic drug types in the structural arrangements are different (F. Chen et al., 2018; L. Chen et al.,
pharmaceutical formulation (Teixeira et al., 2017). This is because 2018). However, amorphous silica is generally considered less harmful
taking into account the natural skin selectivity, drugs must be lipophilic to health than crystalline silica (Arts et al., 2007). Despite of this,
enough to cross the stratum corneum, but also have the necessary hy­ opinions about this issue are still very contradictory. Amorphous silica is
drophilicity to be able to cross the viable epidermis, dermis (more hy­ accepted by regulatory agencies such as the Food and Drug adminis­
drophilic regions), and reach the bloodstream (in the case of tration (FDA) and is used as an ingredient in oral administration in an
transdermal systems) (Lane et al., 2012). Another problem is that amount not to exceed 2 percent by weight of the food (Food & Drug
insufficiently solubilized drugs can undergo crystallization and cause Administration, 2020). However, the FDA does not have an official
acute toxicity (Zhu et al., 2018). Thus, an alternative found by re­ document that prohibits or express opinion about silica in topical and
searchers to overcome these difficulties was to load these drugs in drug transdermal products. On the other hand, the Scientific Committee on
release nanosystems, which are applicable both in cosmetic and thera­ Consumer Safety (SCCS) from European Commission, presented an
peutic areas. extensive document analyzing studies about many types of silica forms
In this context, nanoparticles have emerged to improve drug indicating that due the lack of research methods standardization, the
apparent solubility, permeability, and decrease toxicity to skin. They not unique characteristics of each material must be well evaluated to
only achieve a deeper drug penetration, but also are also able to create guarantee safety and biocompatibility (Scientific Committee on Con­
reservoirs, which increase stability and permanence on the target site. sumer Safety, 2015).
These nanoparticulate systems promote the delivery and sustained Since the 1980s, silica has been used in controlled drug delivery
release of drugs, increasing their effectiveness and reducing their systems (Unger et al., 1983), but it was after first report (Vallet-Regi
adverse effects (Carazo et al., 2018; Goyal et al., 2016). et al., 2001) on mesoporous silica nanoparticles (MCM-41 - mesoporous
Among the various nanoparticles used as drug carriers are silica silica with two-dimensional straight channel pore) as drug delivery
nanoparticles (SNPs). Their excellent properties, such as large surface systems, that SNPs began to be extensively explored within biomedical
area, biocompatibility, porosity, versatility for surface modification, research (Arriagada et al., 2019). Researchers have successfully worked
control of parameters such as size and shape, ease of large-scale syn­ with SNPs as carriers to deliver a variety of therapeutic agents, ranging
thesis and low production costs make them promising tools for appli­ from small molecules (e.g., drugs) to macromolecules (e.g., proteins,
cation in dermatology and cosmetology (Esim et al., 2019; Gonçalves, DNA and RNA) (Lio et al., 2019; Ren et al., 2020; Sun et al., 2013; Zhang
2018; Jeelani et al., 2020). The porosity of SNPs allows the loading of et al., 2014).
therapeutic agents in large quantities. In addition, the size and volume SNPs have been gaining prominence because of their differential
of the pores can be adjustable. Therefore, optimizing the pore size ac­ properties, which include stability, hydrophilicity, biocompatibility,
cording to the size of the load and limit of controlled release is highly ease of functionalization/surface modification, large specific surface
essential (Bharti et al., 2015; Selvarajan et al., 2020). Many researchers area, high pore volume, controlled particle size, and high loading ca­
have reported the successful application of SNPs in topical drug pacity for drugs/therapeutic molecules associated with ease of synthesis
administration (Jadhav et al., 2017; Mo et al., 2020; Montazeri et al., and processing (Esim et al., 2019; Gonçalves, 2018; Jeelani et al., 2020).
2019; Nafisi et al., 2018; Sapino et al., 2017, 2015). For example, in the In addition, due to the strong Si-O bond, SNPs are more stable to external
research carried out by Berlier et al. (2013), rutin immobilization, a response, such as biological degradation and mechanical stress,
highly unstable flavonoid that is poorly soluble in aqueous medium, in compared to other nanosystems, such as liposomes, nanoemulsions and
mesoporous SNPs functionalized with aminopropyl groups resulted in dendrimers, which excludes the need for any external SNPs synthesis
active compound better performance in terms of photostability and skin stabilization (Bharti et al., 2015; Karimi et al., 2016). Other advantages
accumulation. However, despite the advances, the mechanisms involved are that SNPs pore size and porosity can be adjusted, as well as SNPs
in SNPs interaction, transport, and fate in the skin have not yet been surface functionalization can allow the control of drug loading and
fully understood. release, a process that is necessary to properly load various drug mole­
Therefore, this article aims to review significant findings related to cules, whether hydrophobic, hydrophilic, or having a positive or nega­
SNPs preparation and functionalization and their skin interactions. First, tive charge (Bharti et al., 2015).
this review addresses SNPs synthesis and superficial modification stra­ SNPs used for drug delivery can be classified into two main types:
tegies, their skin permeation capacity, and findings on these systems porous or non-porous (solid) SNPs (Tang and Cheng, 2013). Mesoporous
toxicity. Finally, the most relevant advances and evidence in the cuta­ SNPs are the most common form of porous SNPs. Mesoporous SNPs are
neous use of SNPs for therapeutic and cosmetic applications are pre­ characterized by their mesopores (pore size 2–50 nm), which can be
sented, focusing on in vivo and ex vivo studies. For this, electronic filled with drugs through physical or chemical adsorption. In contrast,
searches were conducted through the PubMed, ScienceDirect and Web non-porous SNPs do not have a porous structure, thus drugs are
of Science databases and studies from the last 5 years were selected encapsulated or conjugated through various chemical ligands. Drug
combining the term ’silica nanoparticle’ with the terms ‘drug delivery’, release in mesoporous SNPs can be controlled using the “gatekeepers”
’skin’ and ‘cosmetic’. strategy, for example, metallic macromolecules or nanoparticles can be
attached to SNPs mesoporous surface to block and prevent premature
2. Silica nanoparticles as drug carriers release, or by modifying the inner pores surface to control drug binding
affinity. While drug release in non-porous SNPs is controlled by chem­
Worldwide, nanotechnology is being used in drug delivery systems ical binders or by silica matrix degradation (Souris et al., 2014; Tang and
development to prevent, control, and treat diseases. Thanks to their Cheng, 2013). As reported below, silica versatility allows different SNP
unique nanoscale properties (usually in the range from 1 up to 100 nm) structures construction with wide possibilities of shape, size, function­
and specific biological functions, nanosystems are able to transport alization, and encapsulated active principles.
drugs more effectively and to release them in a controlled, targeted or
on-demand manner at the target site, thus promoting an increased 2.1. Methods for silica nanoparticles synthesis
therapeutic efficacy and reducing side effects (Esim et al., 2019).
Silicon (Si) is one of the inorganic chemical elements found in the Chemical synthesis is the main route used to obtain different SNPs
earth crust. Its oxide forms are silicate (SiO4) and silica (silicon dioxide, types. It includes microemulsion method, Stöber’s method, and sol–gel
SiO2). Si is often used in industry, while its oxide forms are often used for method (Jeelani et al., 2020; Souris et al., 2014).

2
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

SNPs synthesis by the microemulsion method has been used since the through changes in water/silane ratio, type of catalyst or solvent, and
90′ s (Arriagada and Osseo-Asare, 1994). In this method, the alkyl sili­ temperature (Jeelani et al., 2020; Mebert et al., 2017; Souris et al.,
cate molecules solubilized in the oil phase diffuse in a surfactant layer, 2014). Many contemporary researches describe SNPs synthesis using the
forming micelles in the presence of water (Jeelani et al., 2020; Mebert Stöber method or its optimizations (Barrera et al., 2016; Gholami et al.,
et al., 2017). The micelles stabilized by surfactants (tweens or pluronics) 2013; Y.C. Lin et al., 2018; Z. Lin et al., 2018; Ren et al., 2020). Fig. 2
act as nanoreactors for SNPs synthesis. It is inside these nanoreactors shows the experimental conditions most used to synthesize SNPs with
that silica precursors undergo hydrolysis and condensation reactions to different structures and morphologies through the sol–gel process using
form SNPs (Fig. 1). This method involves oil-in-water (O/W) micelles or TEOS as silica precursor.
water-in-oil (W/O) reverse micelles formation (Jeelani et al., 2020; Novel methods to synthesize SNPs were described. One such method
Selvarajan et al., 2020; Yoo and Pak, 2013). Microemulsion is a ther­ is the microwave-assisted method, where mesoporous SNPs synthesis
modynamically stable, isotropic, highly tunable system, in which the occurs in an electromagnetic field along with a fast heating speed
micelles size and consequently SNPs can be adjusted by changing water (Farjadian et al., 2019). Microwaves can provide high localized heating
to surfactant proportion. Porous and non-porous SNPs can be prepared that may be higher than the reaction vessel recorded temperature
by this method (Souris et al., 2014). The ease of reaction control is an (Kumar et al., 2017). As compared to hydrothermal process that utilizes
advantage of this method. However, the use of organic solvents and high pressure and temperatures using autoclave heating, there are many
surfactants is a disadvantage due to the high cost, the need for purifi­ advantages of this technique such as rapid heating to crystallization
cation, and recovery of nanoparticles for large-scale synthesis (Mebert temperature, faster super saturation by the rapid dissolution of precip­
et al., 2017). itated gels, and a shorter crystallization time, besides being a low-cost
The sol–gel method was first used in 1845 by M. Ebelman (Dimitriev approach (Kumar et al., 2017; Narayan et al., 2018). Various reports
et al., 2008) to obtain glass. Since then, due to its ability to form pure found that the result by microwave heating can produce high ordered
and homogeneous products in mild conditions, the method still con­ mesoporous SNPs within a very short time and with high purity
tinues to be used in glass production, which has been perfected to pro­ (Abdelaal and Shaikjee, 2020; Peres et al., 2018).
duce other ceramic materials, such as silica (Jeelani et al., 2020; Another method that is rapid, cost effective, and capable of being
Rahman and Padavettan, 2012). The sol–gel method involves hydrolysis operated under ambient conditions for SNPs production is sonochemical
and condensation of metallic alkoxides such as tetraethylorthosilicate synthesis. Ultrasonic irradiation causes cavitation in a liquid medium
(TEOS, Si(OC2H5)4) or inorganic salts such as sodium silicate (Na2SiO3) where the formation, growth, and implosive collapse of bubbles occurs,
in the presence of mineral acid (for example, HCl) or base (for example, resulting the generation of high-speed microjets, which can generate
NH3) acting as catalysts (Jeelani et al., 2020). Hydrolysis of silica pre­ SNPs (Masjedi-Arani et al., 2014). The use of ultrasound during the
cursor molecules forms silanol groups. The condensation/polymeriza­ synthesis can generate uniformly shaped, monodisperse, and size-
tion between the silanol groups creates siloxane bridges (Si – O – Si) controlled spherical SNPs with relatively short reaction times (Kim
forming a three-dimensional structure of silica covered with –OH et al., 2016). A recent trend is biogenic synthesis that uses bacteria,
groups (Arriagada et al., 2019; Rahman and Padavettan, 2012). Some of fungi, algae (biomass), and plant compounds (extract and metabolites)
this method advantages are direct, scalable, and controllable synthesis. in the production of SNPs (Jeelani et al., 2020). SNPs can be synthesized
In addition, the particles size, distribution, and morphology can be using agro-industrial by-products considered as waste material such as
controlled by changing the reaction parameters, such as temperature, sugarcane bagasse (Falk et al., 2019), rice husk (Athinarayanan et al.,
pH, surfactant concentration, silica source, and catalyst (Mebert et al., 2015), rice straw (Nandiyanto et al., 2016), and olive residue (Rezaeian
2017). et al., 2021). The biogenic synthesis is an economically viable, energy
The Stöber’s method developed in 1968 by Stöber et al. (Stöber et al., saving, and green approach.
1968) from a silica alkoxide alcohol aqueous solution in the presence of
ammonia as catalyst, is a sol–gel process that generates spherical and
monodisperse silica particles with uniform size (50–2000 nm in diam­ 2.2. Strategies for silica nanoparticles surface modification
eter). This technique is based on controlled hydrolysis and silica pre­
cursor condensation, in most cases TEOS, occurring in the presence of SNPs surface can be modified through chemical (by covalent
excess water and a low molar alcohol such as ethanol. The particle size bonding) or physical (by physical adsorption) functionalization. Phys­
and morphology (to a lesser extent) can be adjusted during synthesis ical modification is generally reversible and is not as common as
chemical modification due to non-covalent bond instability (F. Chen

Fig. 1. Schematic illustration of the microemulsion method to produce silica nanoparticles. Adapted from ref. (Selvarajan et al., 2020).

3
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Fig. 2. The experimental conditions commonly used in silica nanoparticles synthesis by the sol–gel process from TEOS. Adapted from ref. (Liberman et al., 2014).

et al., 2018; L. Chen et al., 2018). (Fig. 3) (Jeelani et al., 2020; Souris et al., 2014; Vallet-Regí, 2014). In
SNPs chemical modification is made possible by the presence of high co-condensation, the organofunctional silane is added during the syn­
density of silanol and siloxane groups on SNPs surface. The number of thesis together with a source of silica. The functionalization occurs when
available silanol groups and the SNPśshape are dependent on the syn­ surface silanol groups react with organofunctional silane. This strategie
thetic route. For example, these groups can react with a silane (abundant resultes in uniform incorporation of functional groups (Idris et al., 2020;
and commercially available) by introducing other functional groups on Jeelani et al., 2020; Souris et al., 2014; Vallet-Regí, 2014). These
SNPs surface. The most frequently used silanes have amine or sulfur organofunctional silane agents have the structure R’-Si-X3, where X
groups at the end of an alkylsilane (for example, 3-aminopropyl trie­ represents alkoxy (OR), and R’ is an organofunctional group. In the
thoxysilane (APTS) and 3-mercaptopropyl trimethoxysilane (MPTS)), presence of water, hydrolysis occurs to produce alkoxysilanols and,
along with several PEG-silanes. APTS and MPTS can act as coupling occasionally, silane triols, as well as the condensation of silanols into
agents between SNPs and other chemical moieties (F. Chen et al., 2018; siloxanes (Idris et al., 2020). The hydrolysis and condensation reactions
L. Chen et al., 2018; Liberman et al., 2014). As an example, APTS was of co-condensation method are as follows:
used to graft amino group (NH2) into hollow mesoporous SNPs to load
the anti-inflammatory drug sulfasalazine (Ghasemi et al., 2017). In Hydrolysis: R Si(OR)3 + 3H2 O⇄R Si(OH)3 + 3R OH
′ ′ ′

addition, through PEG-silane, PEG molecules are grafted onto SNPs Condensation: 2R Si(OH)3 ⇄R Si(OH)2 OSi(OH)2 R + H2 O
′ ′ ′

surface (Cauda et al., 2010; He et al., 2011).


SNPs surface modification for subsequent molecules of interest fix­ Organoalkoxysilanes can be co-condensed into growing nano­
ation is generally carried out by two main strategies: co-condensation particles to produce an inorganic–organic hybrid for further additional
(direct incorporation) during synthesis and post-synthesis graft

4
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Fig. 3. Schematic representation of surface modification strategies: co-condensation (1) and post-synthesis grafting (2). CTAB is the cetyltrimethylammonium
bromide surfactant, F127 is the triblock copolymer targeting agent, and NH4OH is ammonium hydroxide catalyst. Adapted from ref. (Selvarajan et al., 2020).

functionalization (Idris et al., 2020; Souris et al., 2014). The organo­ administered in a biological environment, since surface modification
alkoxysilanes commonly used to functionalize SNPs are APTS, methyl­ plays a fundamental role in altering the surface reactivity improving
triethoxysilane (MTES), vinyltriethoxysilane (VTES), 3- SNPs biocompatibility. In addition, without surface modification, the
aminopropyltrimethoxysilane (APTMS), 3-glycidoxypropyl-trimethoxy­ silanol (Si–OH) groups present on SNPs surface can interact with bio­
silane (GPTMS), and 3-acryloxypropyltrimethoxysilane (AOPTMS). The logical molecules, such as lipids and cell membrane proteins, altering
co-condensation of two organoalkoxysilanes can insert basic and acidic these molecules’ structure (Tang et al., 2012).
functional groups into SNPs structure, resulting in new properties and Functionalization allows modifying specific SNP locations (wall and
chemical applications (Souris et al., 2014). In addition, certain orga­ pore entry and the outer/inner surface) with functional groups and
nosilanes play a dual role, they can functionalize SNPs surface and molecules (F. Chen et al., 2018; L. Chen et al., 2018; Jeelani et al., 2020).
transform their shape. SNPs morphological variants can be synthesized These groups and functional molecules include spacers for colloidal
by the co-condensation method by incorporating organosilanes, and stability (Loiola et al., 2019), therapeutic agents (Xie et al., 2014),
nanoparticles morphology will depend on type and quantity of intro­ agents for active targeting (Ricci et al., 2018), blocking molecules to
duced organoalkoxysilane precursor (Narayan et al., 2018). Huh et al. stimulate drug release (Moorthy et al., 2019) among others (Fig. 4).
(2003) found that organoalkoxysilane precursors presence during
condensation influenced particle final shape, and changing the precur­ 2.3.1. Lipid coating
sor or its concentration, SNPs morphology were adjusted to various The presence of an outermost lipid layer around SNPs allows for a
shapes, including spheres, tubes, and rods of various dimensions. In better interaction with the stratum corneum and other skin layers, in
addition, it is through this method that functional groups can be addition to providing increased biocompatibility and ability to prevent
incorporated without significantly affecting SNPs pore topology (Souris nanoparticles aggregation (Iannuccelli et al., 2014; Tada et al., 2014). In
et al., 2014). an in vivo skin permeation study, lipid-coated SNPs have already been
The post-synthesis graft in SNPs explores the presence of surface shown to permeate the skin to a greater extent (about 42%) than un­
silanol groups (Si–OH) that act as convenient anchor points for organic coated SNPs (about 10%) (Iannuccelli et al., 2014). The lipophilic nature
functionalization. The graft is commonly performed by silylation on the of lipid-coated SNPs was able to increase their permeation in the stratum
free (– – Si–OH)) and geminal silinal (Si(OH) ) groups, although
– 2 corneum, although they had a larger size (363 ± 74 nm in size) than the
hydrogen-linked silanol groups are less accessible to modification, as uncoated SNPs (162 ± 51 nm in size). Furthermore, lipid-coated SNPs
they quickly form hydrophilic networks among themselves. The original exhibited a more marked tendency to reach deeper layers stratum
nanoparticles morphology and pore structure remain intact with the
graft, although the technique often results in non-uniform distributions
of organic functional groups (Souris et al., 2014). Zaharudin et al.
(2020) observed morphology preservation, after functionalizing meso­
porous SNPs with amine, carboxyl, and thiol groups using the post-
synthesis graft technique. The authors observed in TEM images that
the original nanoparticles morphology was preserved. In addition, the
post-synthesis graft method provides numerous opportunities to cova­
lently attach diverse labile functional groups, such as ester group, for
example, that is prone to hydrolyze (Mirzaei et al., 2020; Vallet-Regí,
2014) on SNPs surface. A problematic issue with post-synthesis graft
technique is the possible blocking of pores opening by the functional
groups, which can inevitably limit the access of drug molecules to sites
deep inside the pores (Vallet-Regí, 2014).

2.3. Surface modification of silica nanoparticles for topical and


transdermal administration

It is recommended to functionalize SNPs that are intended to be Fig. 4. Multifunctionality of porous silica nanoparticles.

5
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

corneum when applied on the volar forearm (region having low hair tissue bond. In addition to acting as a targeting agent, PEGs form a
follicles density) compared with the dorsal (region having high hair hydrophilic layer around SNPs, which increases the dispersion and half-
follicles density) forearm application, suggesting a preferential pathway life of these nanoparticles by delaying opsonization, that is, proteins
through the lipid domain among corneocytes rather than along the adsorption to nanoparticles, thus allowing immune response escape
follicle orifices. (Rosenholm et al., 2010; Tang et al., 2012).

2.3.2. Positively charged functional groups 2.3.4. Stimulus-responsive agents


It is assumed that the first permeation stage is the attachment of Surface functionalization offers numerous possibilities to develop
nanoparticles to skin cells membrane. It is well known that the epithelial stimuli responsive systems in order to delivery therapeutic molecules on
cell membranes on skin surface are negatively charged. In this way, demand (Möller and Bein, 2017). Smart nanocarriers are being designed
functionalizing SNPs surface with positively charged functional groups owning characteristics that respond to stimuli and can trigger the asset
(for example, NH2 group) is a strategy to improve their permeation release only when and where needed. These responsive elements are
through the skin (Baspinar and Borchert, 2012; Nafisi et al., 2018; Yil­ added to nanoparticles by means of surface modification strategies
maz and Borchert, 2006). The positively charged nanoformulations are (Karimi et al., 2016). In general, these intelligent nanocarriers incor­
absorbed intensely by the negatively charged corneocytes, which in­ porate one or two elements, namely, a sensitive linker and/or a capping
crease encapsulated drug retention time and bioavailability, especially agent (Fig. 5). The responsive linker is capable of breaking, degrading or
those of poorly soluble drugs (Baspinar and Borchert, 2012; Yilmaz and undergoing a conformational change in the provided stimulus presence.
Borchert, 2006). Examples of research that used this type of function­ Capping agents, such as inorganic nanoparticles (INPs), polymers or
alization strategy to increase drugs permeability are described in the last macromolecules, block the pores entry and hinder premature drug exit
sessions. (Vallet-Regí et al., 2018).
SNPs can be modified to respond to a variety of different stimuli.
2.3.3. Targeting agents These can be endogenous factors such as enzyme (Liu et al., 2015), pH
The drugs targeted delivery to a specific location on skin has been (Yan et al., 2020) and reactive oxygen species (ROS) (Xu et al., 2019) or
receiving attention in dermatology (Chen et al., 2021; Schneider-Rauber exogenous factors such as light (Li et al., 2020), ultrasound (Paris et al.,
et al., 2020). However, high precision and accuracy drug delivery to skin 2015), magnetic field (Chen et al., 2016), and temperature (J.T. Sun
target sites (for example, hair follicle, pilosebaceous gland, and dermis et al., 2012; M. Sun et al., 2012).
layers) is challenging, due to the protective skin structure. Therefore, the In the development of topical and transdermal stimuli-responsive
superficial nanosystems modification is an essential factor to achieve systems, it is necessary to take into account during the design of these
this goal (Chen et al., 2021). Recently, they investigated the role of systems the layer in the skin where the stimulus manifests (in the case of
PEGylation in targeted SNPs delivery to hair follicles (Al Mahrooqi et al., endogenous stimuli) or which skin layer it reaches (in the case of
2021). It was observed that SNPs functionalized with polyethylene exogenous stimuli). Depending on which skin layer it is intended to
glycols (PEGs) penetrated the cutaneous appendages, and nanoparticles release, on demand, the encapsulated drug, one must think about which
functionalized with PEG 5000 Da penetrated more deeply into hair synthesis materials and surface modification strategies to use in order to
follicles compared to those functionalized with PEG 750 Da. Because the the system be able to penetrate and permeate or to stay on the skin
longer PEG chains are more flexible than the shorter PEG chains, they surface, in order to achieve the target location for release. For example,
allowed greater protection for the remaining thiol groups on nano­ polymers such as chitosan, which in addition to being a pH-responsive
particle surface, providing deeper penetration by reducing the particle- material, has been shown to be a permeation enhancing agent (He

Fig. 5. Schematic illustration of representative stimuli-responsive drug delivery silica nanoparticles (SNPs). SNPs incorporating capping agents (1 and 2) and
sensitive linkers (3).

6
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

et al., 2009). In this way, chitosan can be used in the development of pH- (Xu et al., 2017). The authors developed a mesoporous SNPs modified
responsive systems that intend on permeation (Alruwaili et al., 2020). In with 4-(imidazoyl carbamate) phenylboronic acid pinacol ester (ICBE)
addition, stimulus-responsive systems can be modified, for example, that contained glucose oxidase and insulin by a host–guest complexation
with positively charged functional groups or lipids that are strategies between ICBE and α-cyclodextrin. Glucose oxidase was encapsulated to
used to increase skin permeation, which were discussed previously. convert glucose into gluconic acid and generate H2O2. The phenyl­
Besides, these systems can be incorporated into formulations containing boronic ester on nanoparticle surface can be oxidized in H2O2 presence,
permeation enhancers, in order to increase their skin permeation ca­ which resulted in host–guest complexation destruction, drug-loaded
pacity (Dhal et al., 2020). mesoporous SNPs disassembly and, consequently, preloaded insulin
To the best of our knowledge, the stimuli that have been investigated release. After transdermal microneedle administration in diabetic rats,
for delivering skin medications on demand based on SNPs include the insulin exhibited glucose-mediated and H2O2-responsive release
following parameters. behaviors.

2.3.4.1. Enzymes. The deregulation of certain enzymes, being down or 2.3.4.4. Temperature. Changes in skin temperature are related to local
overexpressed in certain pathological states, can be used to trigger drug diseases involving inflammatory conditions. The thermal difference
release from SNPs (Karimi et al., 2016; Manzano and Vallet-Regí, 2020). between healthy and diseased skin can be used as a trigger to initiate the
The skin has a metabolic capacity, where numerous enzymes facilitate a drugs release only at intended locations. Among the materials, thermo-
wide range of biotransformations, but intrinsic enzymes are generally responsive polymers have attracted the most interest to build
scarce in skin metabolism, and their exploration in the development of temperature-responsive systems, as are capable of responding to local
stimulus-responsive systems for drug release is rarely reported. How­ temperature variation, changing naturally their hydration profile (Chen
ever, a regulated or specific production of certain enzymes can occur et al., 2021). Thermosensitive mesoporous SNPs have been designed for
under pathological conditions. In this context, bacterial enzymes topical delivery of the antioxidant quercetin (Ugazio et al., 2016). SNPs
resulting from infectious processes in skin wounds are the most inves­ mesopores were functionalized by copolymerizing the thermo-
tigated as a triggering stimulus (Chen et al., 2021). Wu et al. (2015) responsive polymer N-isopropylacrylamide (NIPAM) with 3- (meth­
developed mesoporous SNPs biohybrids capable of releasing antimi­ acryloxypropyl) trimethoxysilane (MPS). To verify the thermo-
crobial agents in the presence of hyaluronidase, an enzyme produced by responsive behavior, quercetin profiles release from mesoporous SNPs
the pathogenic bacteria Staphylococcus aureus. Mesoporous SNPs were with larger mesoporous size (5.0 nm) and smaller pore size (3.5 nm)
loaded with the antibiotic amoxicillin and later coated with lysozyme, were monitored at 20 and 40 ◦ C, in other words, below and above the
hyaluronic acid, and 1,2-ethanediamine (EDA) -modified polyglycerol lower critical solution temperature (LCST). At temperatures above LCST,
methacrylate (PGMA) using the layer-by-layer (LBL) method. In vitro the copolymer chains collapse, allowing the release of immobilized drug
tests showed that hyaluronidase triggered the specific nanosystem molecules within mesopores. The results showed that the thermo-
cleavage. In addition, the nanosystem antimicrobial activity was eval­ responsive behavior was more evident in mesoporous SNPs with larger
uated in vivo in a mouse model of S. aureus-infected mice, resulting in an mesoporous size (20.0% difference in quercetin release between the two
almost complete inhibition of bacterial growth, with a bacteriostatic rate temperatures) compared to mesoporous SNPs with smaller pore size
of 99.9%, after epidermal administration. (11.8% difference between the two temperatures). Regarding the ex vivo
permeation capacity of mesoporous SNPs with larger pore sizes, they
2.3.4.2. pH. The skin has pH variations. Between its compartments, the showed no transepidermal penetration of quercetin and only 2.17 μg/
pH gradient varies from 4.5 to 7.5, with normal skin surface being more cm2 of quercetin accumulated in the skin, which was desired because it
acidic due to acidic constituents and exocrine secretions from the skin is a formulation for photoprotection.
glands and less acidic in deep areas. Changes in skin pH can also be
related to pathological conditions, such as atopic dermatitis, acne, 2.3.4.5. Light. Light is a particularly attractive tool for activating drugs
wounds, and psoriasis (Chen et al., 2021). In this way, the inequalities of release from stimulus-responsive systems. The wavelengths of ultravi­
pH conditions in the skin can be used as a trigger to promote bioactive olet (UV, 200–400 nm), visible light (400–750 nm) or near infrared
agents release in target locations. Mesoporous SNPs loaded with antigen (NIR, 750–2000 nm) can be used to activate responsiveness to light (Tao
(ovalbumin) and covered with lipid bilayer were used to coat micro­ et al., 2020). When using an external light source, the chemical structure
needle arrays in order to build a pH sensitive system for delivering in­ can be disturbed by inducing the delivery system photocleavage or
tradermal antigen (Tu et al., 2017). Hence, silicon microneedle arrays photoisomerization and adjusting the drug release profile to obtain
were chemically modified to obtain a pH sensitive surface (positively better control over treatment dose and duration. Different light pene­
charged at pH 5.8) using pyridine groups. Ex vivo system application to tration depths can be reached in skin tissues. The greater the wavelength
human skin resulted in successful nanoparticles release due to a change the greater the depth reached, as skin components spread light more
in pH from 5.8 to 7.4. The authors pointed out that the method is not efficiently with shorter wavelengths, which reduces penetration depth
restricted to antigens delivery for intradermal vaccination but can also (Chen et al., 2021; Costa et al., 2019). NIR and visible lights have a
be used to deliver any compound that can be encapsulated in meso­ better safety profile for humans than UV. The absorption of UV light,
porous SNPs such as drugs, RNA, DNA, and proteins. especially in the UVB range (280–320 nm), causes progressive damage
to human skin, generating free radicals, destroying vitamins A and C,
2.3.4.3. Reactive oxigen species. Reactive oxygen species (ROS) play a damaging collagen and DNA (Costa et al., 2019; Martínez-Carmona
vital role in several biological processes. Generally, endogenous ROS are et al., 2017). Recently, biodegradable mesoporous SNPs responsive to
maintained at a moderate level, but pathological conditions trigger visible light have been developed for topical drugs administration in
oxidative stress causing its overproduction. Many dermatological dis­ osteoarthritis treatment (Zhao et al., 2021). The nanosystem was built
orders, such as photosensitivity diseases, certain types of skin malig­ by supramolecular interaction between mesoporous SNPs modified with
nancy, and some inflammatory conditions have been shown to be azobenzene and β-cyclodextrin-modified poly (2-methacryloyloxyethyl
associated with ROS overproduction (Chen et al., 2021). This biological phosphorylcholine). The authors observed that visible light was able to
change aroused researchers’ interest in ROS exploration as a trigger for trigger azobenzene isomerization and, thus, to induce the drug release
stimulus-responsive delivery systems. Hydrogen peroxide (H2O2) is one after passing through the dermal tissue (nude mouse skin).
of typical ROS.H2O2-Responsive mesoporous SNPs were integrated with
microneedle patches for glucose-monitored transdermal insulin delivery

7
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

2.3.5. Loading of active ingredientes differentiated in a cyclic and dynamic process starting from the ‘stratum
The efficiency of loading and releasing drugs from SNPs depends basale’ cells. This differentiation gives rise to different layers within the
strongly on the chemical characteristics of surface functionalization. The epidermis, leading to different cells maturation degree. The last differ­
Si-OH groups present on SNPs surface are important to control the drugs entiation stage occurs when cells undergo cornification and become
loading through physical adsorption (Karimi et al., 2016). In addition, flattened, lose their nucleus and intracellular content, turning into cor­
thanks to the high surface area and large pore volume (porous and neocytes (Angelova-Fischer et al., 2018; Jepps et al., 2013; Khavkin and
mesoporous SNPs), surface functionalization (both external and inter­ Ellis, 2011; Kolarsick et al., 2011; Koppes et al., 2017; Rawlings and
nal) provides a high loading capacity for a variety of bioactive com­ Harding, 2004; Wong et al., 2016; Yosipovitch et al., 1998; Zsikó et al.,
pounds, whether hydrophobic or hydrophilic drugs. Hydrophobic drugs, 2019). Corneocytes are surrounded by an organized hydrolipidic mantle
for example, can be loaded onto SNPs through functional groups con­ formed by hydrophilic molecules - mostly amino acids from filaggrin
taining aromatic rings, which induce hydrophobicity SNPs surface, thus protein degradation (Koppes et al., 2017) - and lipids such as ceramides,
preventing polar substances loading (Karimi et al., 2016; Narayan et al., cholesterol, and fatty acids resulting from cells maturation (Jepps et al.,
2018). On the other hand, functional groups molecular dimensions on 2013; Wong et al., 2016). This layer defines the permeation kinetics
pore surface can directly influence its diameter, which can affect drug limiting step of dermatological products (Ghasemiyeh and Mohammadi-
loading and quantity within the pore. Another important point to note is Samani, 2020; Jepps et al., 2013; Rawlings and Harding, 2004; Zsikó
drug release prevention before SNPs reach the target site. For this pur­ et al., 2019). Products such as moisturizers, repellents, and sunscreens
pose, drugs can be linked to SNPs structure through a cleavable covalent are designed to stay on that stratum corneum with no need of deep
bond, or pore entrances can be sealed by adding removable caps permeation.
responsive to stimuli after drug loading (Karimi et al., 2016). In the innermost epidermis layers, cells present a three-dimensional
voluminous shape, are abundantly joined by structures called desmo­
3. Skin & silica nanoparticles interaction somes, which give it adhesion and mechanical strength to remain united
(Rawlings and Harding, 2004; Simpson et al., 2011). In contrast to the
Skin is considered the largest human organ. It is one of our first stratum corneum, which has lipophilic characteristics, viable epidermis
defenses against the outside body environment (Khavkin and Ellis, contains a large amount of water, which determines its hydrophilicity
2011), preventing chemical and microbiological agents to enter, the (Lemos et al., 2018). Closer to the epidermis basal layer, other cell types
excessive water loss, and controling the body thermoregulation (Kolar­ are also found such as melanocytes, Langerhans cells and Merkel cells
sick et al., 2011). For any dermatological application, whether cosmetic (mechanoreceptors) (Khavkin and Ellis, 2011; Kolarsick et al., 2011).
or medicinal, the skin physiological functions cannot be neglected and Melanocytes are located on the basal epidermis layer, being
in-depth studies on skin-product interface complexity, permeation, responsible for melanin production and deliver to keratinocytes through
distribution, and elimination are required. In this section, a brief their membrane projections (Videira et al., 2013). Factors such as UV
description about the structures able to serve as therapeutic targets is radiation, inflammation, hormonal dysregulation, and stress (Eves et al.,
presented and permeation routes are presented. 2006; Lotti et al., 2002; Videira et al., 2013) can stimulate melanocytes,
Anatomically, the skin is a heterogeneous organ that can be divided which end up producing more melanin causing pigmentation and
into two layers, epidermis and dermis, attached to an underlying layer eventually, spots on the skin. Melanocytes are usually targets for der­
called hypodermis (Fig. 6). The epidermis is constituted by approxi­ mocosmetics with a lightening purpose, which means that to reach
mately 95% of keratinocytes – keratin-producing cells - which are them, it is necessary to cross an efficient keratinocyte barrier (D’Mello

Fig. 6. The layers of human skin (epidermis and dermis) and subcutaneous tissue (hypodermis). Extended: epidermis and its strata (1) and dermis (2), hypodermis
(3) and their structures and components.

8
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

et al., 2016; Kolarsick et al., 2011). Proksch, 2018). In this way, the compatibility between product and skin
Langerhans cells compose the skin immune system and are respon­ must be respected, in order not to cause skin changes and irritations
sible for antigens uptake and presentation to T lymphocytes (Khavkin (Prakash et al., 2017; Proksch, 2018). If the skin pH reaches values close
and Ellis, 2011; Kolarsick et al., 2011). Allergies development by to 7, lipid ionization is certain, and a repulsion between the hydrophilic
dermatological products are directly related to the ability to activate heads will cause irritation and flaking of the skin (Lieckfeldt et al.,
those cells (Callard and Harper, 2007; Malachin et al., 2017; Rancan 1995).
et al., 2012) which constitutes another challenge in dermatological To better plan products skin permeation, it is necessary to keep in
products, therefore, an ideal product must be inert to Langerhans cells. mind the targeted layer desired. Cosmetic products, by definition,
Just below the epidermis, the basal lamina is a porous membrane should not reach systemic circulation (CosmeticsInfo, 2021). On the
formed type IV collagen, laminin, and proteoglycans that acts as a bar­ other hand, therapeutic drugs are especially added to formulations
rier and control the exchange of fluids and cells between dermis and containing chemical enhancers to facilitate their entry into the systemic
epidermis (Kolarsick et al., 2011). Fibroblasts are the main cells found in circulation (Herman and Herman, 2015).
the dermis, being responsible to collagen (mainly type I and III) and As already mentioned, stratum corneum penetration is challenging
elastin synthesis which provide mechanical strength to this tissue for products applied on skin. To achieve this, molecules should pass to
(Khavkin and Ellis, 2011; Kolarsick et al., 2011; Wong et al., 2016). adjacent layers through intercellular pathway (between cells), trans­
During aging there is a progressive decrease in collagen, elastin, and cellular pathway (crossing cell membranes) or via cutaneous append­
proteoglycan fibers production, causing the skin to lose its elasticity and ages (Fig. 7). In general, due to hydrolipidic mantle nature, most of
vitality (Chaudhary et al., 2020). That is the reason of an increasing hydrophobic molecules reach their destinations by passing through the
number of dermocosmetic products that aim to delay aging by stimu­ intercellular pathway, while small compounds of neutral hydrophilic
lating collagen and elastin production, or by preventing its degradation nature preferentially pass through the transcellular pathway (Ghase­
(Chaudhary et al., 2020; Kwatra, 2020). miyeh and Mohammadi-Samani, 2020; Herman and Herman, 2015;
The cutaneous appendages, like pilous and sweat glands, sebaceous Sheshala et al., 2019).
glands and hair follicules originate in the dermis, and connect to the Many authors dispute that only a small range of molecules can
epidermis (Jepps et al., 2013). Such skin-associated structures are effectively permeate to the layers below epidermis, in contrast, others
specialized in maintaining the body homeostasis in different ways claim that substances applied to skin can be detected in the blood at
(Kolarsick et al., 2011). Local treatments that aimed do treat or beau­ defined times after application (Herman and Herman, 2015; Malachin
tifying these structures are possible as well, through cosmetics or cos­ et al., 2017; Matsuo et al., 2016; Ostrowski et al., 2014a; Wani et al.,
meceuticals use. It means that each of these parts constitutes a specific 2020), and therefore it is an illusion to think that a cosmetic product will
target for problems like hyperhidrosis, acne and hair loss or hirsutism. not reach the bloodstream. The easy access to dermatological products
At a deeper skin level, below the dermis is the hypodermis (subcu­ along with the lack of clear information about their permeation and
taneous adipose tissue). The main cells of hypodermis are adipocytes, accumulation effects, make the consumer blinded about the possible
which are arranged in lobes (Jepps et al., 2013; Khavkin and Ellis, 2011; undesirable effects. In face of the exposed, detailed studies on these
Kolarsick et al., 2011). This is the skin layer that cosmetics for cellulitis molecules’ distribution, accumulation, and excretion are necessary
and lipodystrophy treatment are destined for. It can be noted that a (Rancan et al., 2012, Desprez et al., 2018; López-Sánchez et al., 2021;
cosmetic that aims to reduce fat concentration within the adipocytes Steinmetz et al., 2021), otherwise, unexpected reactions can happen.
needs to permeate many layers until reaching its target and may be a Plenty of cosmetics and cosmeceuticals containing INPs are now
reason for great failure on the part of these products. available to consumers. They are mainly cosmetics containing gold (Au),
When describing the skin, it is important to consider its pH, which on silver (Ag), zinc oxide (ZiO), and titanium dioxide (TiO2) nanoparticles
epidermis surface is generally acidic, between 4 and 6, with small var­ that serve as antioxidants, antibacterial, and UV light reflectors,
iations throughout the body and internally is neutral, close to 7.4 respectively (Ferraris et al., 2021). In fact, nanoparticles penetration can
(Prakash et al., 2017; Proksch, 2018). Maintaining the acid mantle is be a desired goal, or not, depending on the effects wanted to achieve. For
essential to keep the skin functional barrier (Prakash et al., 2017) and example, in the case of photoprotection, if nanoparticles (NPs) used in a
altering this balance can induce diseases such as acne, bacterial in­ formulation penetrate the deeper layers of skin, it no longer exerts the
fections, dermatitis, and delayed wound healing (Bullock et al., 2020; intended effect, failing to achieve its objective (Y.C. Lin et al., 2018; Z.

Fig. 7. Potential penetration pathways through the skin. 1. Transepidermal pathway: intercellular (transport between corneocytes through the lipid matrix) and
transcellular (transport crossing corneocytes and intercellular lipids). 2. Transappendageal pathway: transfollicular (transport through hair follicles) and tran­
sudoriparous (transport through sweat glands).

9
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Lin et al., 2018; Ugazio et al., 2016). On the other hand, transdermal 4. Biocompatibility of silica nanoparticles – Toxicity
nanosystems for systemic drug delivery must be able to permeate the
skin layers and reach blood vessels, only in this way they can meet the Nanotechnology is a very suitable tool to solve problems regarding
objective of their construction. the apparent solubility and delivery of drugs. However, for some types of
Some studies comparing INPs with other types of organic and nanoparticles, especially for INPs, what is known so far about the
deformable NPs, such as liposomes or transferosomes (Ascenso et al., toxicity and accumulation of such products in the human body is often
2015; Cosco et al., 2015) confirm that the last one is able to translocate controversial (Chen et al., 2020; Mohammadpour et al., 2019). Two
through the stratum corneum, present greater skin permeation than main questions about nanoparticles biocompatibility need to be raised
INPs, such as silica, TiO2 and iron (Fe) NPs (Knorr et al., 2019). SNPs are concerning the possible material toxicity. The first one is about the raw
claimed to stay on the skin surface, without a deep permeation (Salvioni material itself, which must be non-toxic and biocompatible, in this
et al., 2021), but that is a very controversial topic. In turn, studies have context biopolymers have an advantage over inorganic materials, since
shown that Langerhans cells were able to internalize the SNPs that we do not have enzymes to degrade the latter one. The second concerns
ended up in the bloodstream and caused systemic effects (Matsuo et al., about the nanomaterial size, as it is known that smaller particles can
2016; Rancan et al., 2012). Matsuo et al. (2016) observed that SNPs not permeate the body more deeply, bypassing the immune system, reticu­
only do penetrate skin, but also described their presence in tissues such loendothelial system, and reaching more organs (Busch et al., 2021; L.
as lymph nodes, liver, and brain under normal conditions. Also, some Chen et al., 2018; Guo et al., 2021; Matsuo et al., 2016; Wills et al.,
authors have shown that despite their size, submicron inorganic parti­ 2016).
cles, including for example, iron oxide (FeO) and AuNPs can permeate Silica toxicity to the lungs, when inhaled, is already well established
the skin depending on the vehicle in which they are found (Cao et al., (Cao et al., 2017; Murugadoss et al., 2017; Shin et al., 2017), and the
2021; Musazzi et al., 2017). Still others claim that permeation to deeper same pulmonary effects were linked due the inhalation of others INPs
layers only occurs via cutaneous appendage (Bolzinger et al., 2011). like ZnO and TiO2 (Garcés et al., 2021) or even, AuNPs and AgNPs
Although it occurs through a smaller area (0.1% of total skin surface), (Talarska et al., 2021). However, the effects caused by other adminis­
recent investigations demonstrate their relevance in topically applied tration routes needs to be better explored. With the increasing passively
nanoparticles penetration. Hair follicles tend to form agglomeration consumption of INPs in food and medicine, an improved understanding
sites and allow a deeper penetration for a variety of particle types. Since of its effects is necessary (F. Chen et al., 2018; L. Chen et al., 2018; Guo
they extend towards the lower parts of the dermis, they act as an effi­ et al., 2021; Murugadoss et al., 2017; Shin et al., 2017). In a study by
cient reservoir for drugs and nanoparticles, where they can diffuse Deng et al. (2021), it was demonstrated that after 2 weeks of oral SNPs
continuously to surrounding spaces, cross the capillary walls, and reach administration to rats, the particles concentration in several organs was
the blood system (Busch et al., 2021; Knorr et al., 2019; Lemos et al., significantly higher than in the control group, showing the effect of
2018). biodistribution and bioaccumulation. In addition, a study carried out by
To better understand and take conclusions about permeation, dis­ Cao et al. (2017), have proved that after SiO2 administration to rats via
tribution, interaction with skin structures, content release, as well as the inhalation, the particles were able to cause significant damage to the
elimination of such nanoparticles, it is necessary to know their physi­ rodent’s liver, showing potential particles distribution throughout the
cochemical characteristics, size, shape, surface and charge (Iannuccelli organism. An extensive review by Murugadoss et al. (2017), performed
et al., 2014; Lemos et al., 2018; Rancan et al., 2012). Pathways under­ from 2010 to 2016, revealed that the main mechanisms of silica in vitro
taken and nanoparticles distribution can be monitored by analytical toxicity are associated with oxidative stress due to the generation of free
techniques and image analysis (e.g. inductively coupled plasma optical radicals, directly reflecting in genotoxicity, stimulating the innate im­
emission spectroscopy/mass spectroscopy (ICP-OES/MS), transmission mune system with increased pro-inflammatory cytokine production,
electron microscopy/energy-dispersive X-ray spectroscopy (TEM/EDX), autophagy, and apoptosis (Deng et al., 2021; Guo et al., 2021; Mur­
scanning electron microscopy/EDX (SEM/EDX) and confocal laser ugadoss et al., 2017; D.P. Wang et al., 2020, W.H. Wang et al., 2020).
scanning microscopy (CLSM)) that allow to obtain detailed information These results are not very different when compared to other INPs
of the skin layers after product application (Cao et al., 2021; Matsuo toxicity. Shrivastava et al. (2016) did a 2-week study of AuNPs oral
et al., 2016; Nigro et al., 2018; Rancan et al., 2012; Xu et al., 2020). administration that caused significant increase in free radicals and in­
Regarding particles charged surfaces and the way they affect flammatory interleukins in rats treated compared to the control group.
permeation, much remains to be clarified. Ostrowski et al. (2014a,b) and Confirming, once again, the need for detailed studies and dose adjust­
reported that functionalized SNPs with approximately 55 nm and posi­ ments. In vivo tests demonstrate that the main affected organs after oral,
tively charged are not able to penetrate murine skin, even if the skin intravenous or inhalation silica administration are lungs, liver, spleen,
barrier is compromised (Ostrowski et al., 2014a). In a more recent study, kidneys, heart, and brain (Hozayen et al., 2019; Liang et al., 2018;
Jung et al. (2019) observed that negative-charged 33 nm and 78 nm Murugadoss et al., 2017; Rawat et al., 2017). SNPs distribution route is
SNPs (functionalized with carboxylate groups) did not permeate the extremely dependent on the administration route (Guo et al., 2021),
stratum corneum of human cadaver skin. In contrast, Nafisi et al. (2018) however there are few studies regarding to transdermal application
demonstrated that SNPs functionalized with positively charged groups, route.
were more efficient to deliver lidocaine to human skin ex vivo (drug Most authors seem to agree that SNPs toxicity depends on their
detection at the receptor fluid). Iannuccelli et al. (2014) proved that physicochemical properties, in addition to other factors such as the dose,
SNPs polarity has an even more pronounced role than size itself, by the time of exposure and the formulation applied (F. Chen et al., 2018; L.
demonstrating that 360 nm particles coated with lipids were detected, Chen et al., 2018; Chen et al., 2020; Giovannini et al., 2018; Hadipour
by tape stripping method, at the stratum corneum in in vivo tests with Moghaddam et al., 2019; Mohammadpour et al., 2020, 2019). Some
humans. In addition, Sapino et al. (2017) added SNPs loaded with authors, for example, associate SNPs porosity, with greater toxicity
methotrexate in various vehicles, aqueous and oily, and concluded that when the pores are large. A larger surface area increases the likelihood
vegetable oils significantly increase the drug release to the receptor of adherence to biological structures, causing greater unwanted effects
fluid. (Chen et al., 2020; Mohammadpour et al., 2019). Therefore, modifica­
In view of the above, it is observed that cutaneous SNPs application tions to SNPs design, including size, composition, and surface chemistry
will remain in the emerging field until all the mechanisms by which it is are being thought and designed to adjust the toxicity profile, with a
governed are fully understood. focus on increasing biocompatibility and decreasing toxicity (F. Chen
et al., 2018; L. Chen et al., 2018; Li et al., 2019; Nafisi et al., 2017).
In the context of the necessary tests, it is known that in vitro assays

10
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

are important ways of conducting a great screening for new synthesized detect NPs in some organs such as regional lymph nodes, although
materials, for this, nanoparticulate materials toxicity is commonly without any evidence of toxicity (Ostrowski et al., 2014a).
investigated using different cell lines. However, due to the huge number Most studies focus their attention on the effects of in vitro toxicity and
of protocols and the great variability among nanoparticles many dis­ skin permeation on 3D models of skin, but it is evident the lack of studies
parities are found between in vitro and in vivo tests and, therefore, on distribution and elimination of SNPs in the body. In the case of
generalizations cannot be made (L. Chen et al., 2018; Guo et al., 2021; (copper oxide) CuO and ZnO nanoparticles, some of the toxicity is
Hozayen et al., 2019; Mohammadpour et al., 2020; Shin et al., 2017; Xu related to ion dissociation and deep permeation of Cu+ and Zn+ ions that
et al., 2020). Surprisingly, rare studies on in vivo systemic toxicity can cause skin irritation (Holmes et al., 2020, 2016). The same, is not
generated by topically applied SNPs are available. Many studies are valid to SNPs, thus, the synthesis of new silica frameworks requires
limited to in vitro particle-cell interaction and most focus only on skin intense investigation of their toxic properties, since the various struc­
permeation, not going beyond that. tural and surface changes that silica can suffer from, may reflect on
Ryu et al. (2018) carried out an extensive study of subchronic unknown impacts on biological organisms generating adverse effects not
toxicity with the continuous application, for 90 days, of 20 nm colloidal previously observed. Some examples of systemic in vivo studies are listed
silica NPs in an area corresponding to 10% of Sprague Dawley rats body in Table 1.
area. Lu et al. (2018) used Ag-funcionalized mesoporous nanoparticles
to treat infected wounds in rats for 5 days. In both studies, no significant 5. Silica nanoparticles in cutaneous and transdermal therapy
clinical, hematological, biochemical, and histological changes were
found. Although small changes were found in the analyzed parameters, With the advent of nanotechnology applied to cancer therapies, the
none of them can be directly correlated with skin silica exposure. Due to use of drug nanocarriers to target the drug to tumor without damaging
the widespread use of silica in dermatological products, some studies try normal body cells was a major achievement for medical science. Among
to predict whether the use of SNPs in people affected by atopic derma­ the nanotechnological advances for skin cancer treatment, the use of
titis or contact dermatitis could have their inflammatory conditions silica-based nanosystems has proved to be an attractive and promising
aggravated by using such products. Hirai et al. (2012, 2015) proved that therapeutic strategy due to the several superior characteristics already
in these individuals IgE production may be exacerbated, due to an in­ reported, in comparison with other nanocarriers. In this context, Sapino
crease in Th2 defense response mechanism, which may even lead to et al. (2015) proposed a nanoformulation based on mesoporous SNPs
anaphylaxis. Apparently, it is only when SNPs permeation is done in a (particle size of 250 ± 50 nm) for topical delivery of the phytocomposite
more aggressive way (e.g., subcutaneous injections) that it is possible to quercetin for melanoma therapy. Quercetin is a flavonoid with strong

Table 1
Studies concerning local or systemic SNPs toxicity in the last decade.
SNPs Type Purpose Permeation type Tecidual/Systemic toxicity References

N-(6-aminohexyl)- Analyze the AHAPS-SiO2-NP Only when injected into Not detected in the analyzed organs. No (Ostrowski
aminopropyltrimethoxysilane (AHAPS) penetration on skin with normal subcutaneous tissues NPs distribution beyond the regional lymph et al., 2014a)
functionalized SiO2-NP 55 nm, positively and altered barrier function were detected in regional node was observed.
charged lymph node
MSNs functionalized with 3 - Anti-melanoma activity of Subcutaneous injections No organ toxicity; biochemical analyzes did (Ferreira
aminopropyltriethoxysilane alkoxide Indomethacin not show kidney damage; absence of et al., 2020)
(APTES) and incorporated with genotoxicity prevented by NPs use
Indomethacin by sol–gel method
MSN decorated with PTHPMA-co-AEMA Develop a novel stimuli-responsive Crossed the skin when the No harmful effects on skin surface. (Anirudhan
(tetrahydropyranyl Methacrylate and amino anticancer device patch containing NPs was and Nair,
ethyl methacrylate stimulated with 2018)
Ultrasound
Amorphous silica from 30 to 1000 nm, To investigate the relationship Intradermal injections nSP70 can penetrate the skin barrier and (Hirai et al.,
negatively charged, combined or not with between the SNPs size and enter keratinocytes and Langerhans cells. 2012)
Dermatophagoides pteronyssinus (Dp) adjuvant activity using a model for Enhanced IL-18 and thymic stromal
antigens atopic dermatitis (AD) lymphopoietin production, leading to
systemic Th2 response and aggravation of
AD-like skin lesions as induced by Dp-
antigen treatment.
Amorphous silica of 30 nm combined or not Investigate the effects of topical Applied to the skin for 13 Low-level IgG production with little change (Hirai et al.,
with Dermatophagoides pteronyssinus (Dp) application of mite extract and weeks in IgE production (IgE-biased immune 2015)
antigens amorphous silica nanoparticles, on response) and increased sensitivity to
allergic sensitization and Atopic anaphylaxis.
Dermatites
MSNs-NH2 Ag-decorated, 61.8 nm, and Wound healing Applied over incisions for No toxicity was found: no change in weight; (Lu et al.,
negatively charged produced by Sol-gel 5 days no hematological changes; no biochemical 2018)
method changes in AST, ALT, alkaline phosphatase,
albumin, blood urea nitrogen, creatinine,
cholesterol and triglyceride. No histological
alterations heart, liver, spleen, lung and
kidney of the rats
MSNs negatively charged, 300–500 nm by salicylic acid and ketoconazole for Formulations containing Histopathological tests around the wound (Masood
sol–gel method antifungal treatment and wound SNPs were applied on skin showed zero erythema score as compared to et al., 2021)
healing for 14 days drug suspension which showed an erythema
score of four
Spherical SiO2-NP with a diameter of 55 ± 6 To modulate allergic contact The formula was applied No effects were observed in all clinical, (Ostrowski
nm Surface functionalization with AHAPS dermatitis when SNPs were during 5 consecutive days histologic, morphometric, and molecular et al., 2014b)
applied with axazolone on lesional skin parameters investigated (IgE and skin)
Colloidal SiO2, 20 nm in size treated with L- Toxicological effects only 90 days of exposure to A complete clinical, hematological, (Ryu et al.,
arginine and negatively charged rats’ skin biochemical, and histopathological analysis 2014)
showed no toxicity

11
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

antioxidant activity that can reverse UV-induced oxidative skin damage, photodegraded. SNPs also increased skin quercetin penetration, the
in addition, it has anti-inflammatory and anticancer properties, however nanoformulation, containing 0.27% w/w of flavonoid, showed higher
low water solubility, low stability and short half-life may restrict its use amounts of quercetin retained in the skin (10.89 μg/cm2) compared to
in skin products and therapies (Sapino et al., 2015). In order to over­ formulation containing free quercetin. Besides, quercetin-encapsulated
come these difficulties, the authors encapsulated quercetin in SNPs SNPs inhibited JR8 melanoma cells proliferation to a greater extent
functionalized with aminopropyl and then evaluated this nanosystem than quercetin alone. At a concentration of 60 μM, the nanosystem
potential to act as a topical transporter for quercetin release through caused about 50% inhibition of cell proliferation and in lower concen­
photodegradation, ex vivo permeation in swine skin, and cell prolifera­ trations (1, 10, 30 μM), the nanosystem did not influence cell
tion tests in vitro. For carrying out photodegradation and ex vivo proliferation.
permeation tests in porcine skin, SNPs were incorporated into an O/W SNPs are also being integrated into photodynamic therapy (PDT) to
emulsion. The results showed that functionalized SNPs improved quer­ treat skin cancer. It is a minimally invasive therapy, which has shown
cetin photostability, among 0.0084% w/w of encapsulated quercetin promising results in several malignancies treatment, including mela­
contained in the O/W emulsion, 98.9 ± 1.8 of its was not noma and non-melanoma skin cancer (Calixto et al., 2016; Zielińska

Fig. 8. A) Effect of Verteporfin-mesoporous silica nanoparticles (Ver-MSNs) on B16F10 cells in dark conditions and under red light exposure. MTT assay of B16F10
cells was carried out after 4 h incubations with Ver-MSNs. Results are expressed as mean values ± standard deviation. *** p < 0.001 referred to non-irradiated (0 s)
plain mesoporous silica nanoparticles MSNs. ◦◦◦ p < 0.001 referred to irradiated mesoporous silica nanoparticles MSNs. B) Ver-MSNs nanoparticles inhibit mouse
melanoma tumor growth. Results are expressed as mean values ± standard deviation. *** p < 0.001 referred to the control. p < 0.05 referred to MSNs. C)
Representative photos of tumor masses obtained after sacrifice of animals treated with glycerol (Vehicle), MSNs and Ver-MSNs. D) Immunofluorescent images of
CD31 staining (green) as an indication of tumor angiogenesis and nuclear DAPI counterstain (blue). Magnification = 20 × . Reprinted with permission from ref.
(Clemente et al., 2019), Copyright 2019 Elsevier. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of
this article.)

12
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

et al., 2018). PDT combines a light with a specific wavelength and a rats), the nanocomposite considerably reduced bacteria number (2 log
photosensitizer. When these two elements interact, in the presence of steps) on the infected skin.
oxygen, ROS are generated leading to selective destruction of tumor Still based on this subject, among plants extracted compounds that
cells (Arriagada et al., 2019; Calixto et al., 2016; Zielińska et al., 2018). have antibacterial and wound healing properties, curcumin, a yellow
Therefore, Clemente et al. (2019) investigated in vitro and in vivo PDT polyphenol extracted from Curcuma longa roots, has been highlighted in
efficacy with mesoporous SNPs functionalized with aminopropyl loaded many scientific research (Hamam and Nasr, 2020; Mirzahosseinipour
with verteporfin, an FDA approved hydrophobic photosensitizer (Zie­ et al., 2020). Despite its excellent properties, its use in therapeutic for­
lińska et al., 2018), as a potential treatment for skin melanoma. The mulations is limited due to its low water solubility, physicochemical
results of mouse melanoma cell line (B16-F10) proliferation assay instability, rapid metabolism, and reduced bioavailability (J.T. Sun
showed that the combination of mesoporous SNPs functionalized with et al., 2012; M. Sun et al., 2012). In order to overcome these limitations
aminopropyl/verteporfin and red-light irradiation (693 nm) was able to and increase therapeutic efficacy, nanosystems have been designed to
significantly reduce cells proliferation in a concentration ranging from protect, deliver and target curcumin to specific sites of interest. Faced
0.1 to 10 μg/ml. Besides, the effect was already detectable at 0.1 μg/ml, with these challenges, Hamam and Nasr (2020) proposed a topical
with 68.26% (0.157 ± 0.008 SD, p < 0.001) cell viability compared to formulation (1% curcumin) based on curcumin-loaded mesoporous
simple SNPs irradiated with red light (Fig. 8A). When the prepared SNPs (SNPs with size of 363.0 ± 11.30 nm loading 98.72% of curcumin)
nanoparticles (5 μg/ml in glycerol) were administered topically to as a wound-healing agent. The in vivo wound healing study carried out in
melanoma-bearing mice and stimulated by red light (applied four times Wistar rats for 21 days showed that the percentage of wound contraction
in 16 days), tumor mass was reduced by 50.2 ± 6.6% compared to un­ in curcumin-nanoformulation treated rats was higher compared to that
treated mice (glycerol only) (Fig. 8B and 8C). In addition, the authors of rats treated with sulfadiazine (control formulation), however this
also observed a decrease in tumor vascularization (Fig. 8D), suggesting difference was not statistically significant (P > 0.05). On the last day of
that the in vivo nanosystem antitumor effect can be mediated not only by measurement (day 21), both formulations stimulated complete wound
direct cytotoxicity to tumor cells, but also by inhibition of tumor neo­ closure. In addition, excision lesion cross-sections of skin specimens
angiogenesis, a fact that was not detected in the treatment only with were obtained for histological evaluation of inflammation, angiogenesis,
mesoporous SNPs. fibroblast proliferation, presence of collagen, and reepithelization. For
The chronic nature and complications associated with wounds that both formulations, inflammatory reactions reduced considerably on day
do not heal have led to the emergence of nanotechnology-based thera­ 21 of treatment, the angiogenesis process was almost complete on day 7,
pies that aim to accelerate the healing process and, consequently, repair fibroblast proliferation increased visibly on day 14, and a high degree of
damaged skin. Nanodevices can not only act in regeneration and repair, wound reepithelization was achieved on day 21, without significant
but also allow the delivery on demand of specific therapeutic molecules differences between the test and control formulation. However, on day
to the injured area. Quignard et al. (2017) reported that ultra-thin SNPs 21, the collagen level increased significantly in rats treated with cur­
with positive surface charge (~10 nm) as sources of silicic acid favored cumin nanoformulation compared to those treated with sulfadiazine.
wound healing in vitro. Suspensions of SNPs with different degrees of In another study, SNPs loaded with curcumin were used in antimi­
surface charge (positive and negative) and under sub-saturated condi­ crobial photodynamic therapy (aPDT) for wound healing (Mirza­
tions (below 2 mM Si) were evaluated. In a wound healing model with hosseinipour et al., 2020). Antimicrobial and antibiofilm efficacy of
human skin fibroblast cells (CCD-25SK) amine-functionalized SNPs curcumin-loaded SNPs (particle sizes ranging from 36 to 40 nm) against
(positively charged) were able to release silicic acid (the bioactive silica Pseudomonas aeruginosa and Staphylococcus aureus bacteria and aPDT
form) in the culture media, and to promote wound closure more effec­ effect on wound healing were investigated through in vitro tests. In
tively than negatively charged SNPs at a dose of 50 μM. According to the aPDT, curcumin and curcumin-loaded SNPs were used as photosensi­
authors, this effect may be related to these nanoparticles easy cell tizers and the light source employed was blue (465 nm), corresponding
internalization, followed by their intracellular dissolution, releasing si­ to the maximum absorption of curcumin. In the photodynamic inacti­
licic acid at a faster rate (50–70% after 76 h of incubation) than its direct vation test of planktonic cells, 10 min irradiated curcumin-loaded SNPs
uptake from the media (20–25% after 6 h of incubation). (1 mg/mL) showed superior antimicrobial activity, showing a signifi­
Another study demonstrated dendritic mesoporous SNPs (with hy­ cant reduction in S. aureus planktonic cells growth (>6. log10 CFU, p <
drodynamic particle size of ~150 nm and pore size of ~13.78 nm) ad­ 0.05) and P. aeruginosa planktonic cells (>6.log10 CFU, p < 0.05)
hesive ability to promote and accelerate wound healing (Pan et al., compared to the control group (Fig. 9A). Similar behavior was observed
2020). The authors proposed an innovative adhesive strategy based on using biofilms, illustrating the efficacy of irradiated curcumin-loaded
SNPs for closing skin wounds. After application, SNPs formed nano­ SNPs (1 mg/mL), exhibiting significant photoinactivation against
composites with body fluid from the wounds, and subsequently elicited S. aureus biofilms (P < 0.01,> 3 log10 CFU reduction) and P. aeruginosa
an acute inflammatory response that initiated skin wound repair pro­ (P < 0.01,> 5 log10 CFU reduction) (Fig. 9B). In addition, curcumin-
cess. Meanwhile, rapid SNPs biodegradation and elimination prevented loaded SNPs have been shown to have wound healing properties, as
persistent inflammation, ensuring accelerated healing and skin tissue confirmed by in vitro scratch assay using HDF fibroblast cell (Fig. 9C and
restoration. The results of in vivo wound closure and wound healing tests D).
showed that the SNPs accelerated the healing process (approximately 3 Silica-based nanosystems have also been designed to treat skin
days) and increased wound rupture resistance on day 10 by 143% fungal infections. Econazole (ECO) is an imidazole antifungal widely
compared to the control group. used in fungal skin infections treatment (Verma and Pathak, 2012),
On the other hand, skin lesions are often accompanied by bacterial however the efficacy and bioavailability of ECO have been limited by
infections that hinder the repair process. Mebert et al. (2018) reported a their poor aqueous solubility and dissolution rate (Al-Marzouqi et al.,
new wound healing dressing incorporating core–shell SNPs (with 2008). In order to overcome this limitation, Montazeri et al. (2019)
diameter of 336 ± 43 nm) loaded with two topical antibiotics (genta­ loaded ECO into amine-functionalized mesoporous SNPs to improve
micin sulfate and sodium rifamycin) in type I collagen hydrogels. Sul­ topical antifungal delivery. The nanosystem was tested in vitro for
fonate groups surface modified SNPs allowed significant gentamicin antifungal activity against Candida albicans, as well as in vivo through
(5950 μg/g) adsorption while silica shell layer grafted with mercapto­ skin irritation test to assess the possibility of skin irritation. The cream
propyl groups, allowed rifamycin (670 μg/g) adsorption. Antibiotics (with 1% w/w of ECO) containing amine functionalized SNPs loaded
release from collagen-silica nanocomposite, containing 30 mg/mL SNPs, with ECO (in 1/1 proportion of ECO and SNPs) showed greater anti­
allowed a sustained in vitro antibacterial effect against Staphylococcus fungal activity against Candida albicans in relation to the other pro­
aureus over 10 days. In addition, in a skin infection in vivo model (Wistar portions (1/2 and 1/3) and the control, a cream with free ECO. The

13
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Fig. 9. A) Bacterial survival of S. aureus and P. aeruginosa planktonic following aPDT with curcumin (50 μg/mL) and (50 μg/mL and 1 mg/mL). B) Effect curcumin
(50 μg/mL) and curcumin-SiNp (50 μg/mL and 1 mg/mL) exposure - aPDT on killing of biofilm -grown strain of S. aureus and P. aeruginosa. C) In vitro scratch wound
healing assay of HDF fibroblast cells in presence of curcumin and curcumin-SiNP monitored by light microscopy. D) Bar graph illustrating percentage wound closure
at indicated time points (0, 2, 6, and 24 h after scratching when the cells were treated with the 50 μg/mL concentration of curcumin and curcumin-SiNP) during the
scratch wound assay. * P < 0.05 represents significant difference versus the control. Reprinted with permission from ref. (Mirzahosseinipour et al., 2020), Copyright
2020 Elsevier.

authors attributed the nanosystem superior antifungal activity to posi­ Another dermatological disorder is psoriasis, characterized by
tive surface charge (functionalization with NH2 groups), a superficial hyperproliferation and aberrant keratinocytes differentiation. Prolifer­
modification that helped improve fungi penetration. In addition, no ation inhibition is one of the effective options for psoriasis treatment
irritation was observed after application of the nanosystem-containing (Mo et al., 2020). Mo et al. (2019) observed that erianin, a natural
cream on rabbit skin after 72 h. compound extracted from the plant Dendrobium chrysotoxum Lindl, has a

14
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

potent inhibitory effect on keratinocyte proliferation. In order to capsaicinoids action site, the authors examined nonivamide concentra­
improve the low water compound solubility, increase the anti­ tion that penetrated in this skin region. Since substantially higher stra­
proliferation activity and improve its skin distribution, Mo et al. (2020) tum corneum drug levels might have a negative impact on the
developed dendritic mesoporous silica nanospheres loaded with erianin formulation safety, the amount of oil-loaded powder was tenfold
as a promising proposal for psoriasis treatment. Therefore, the nano­ reduced consequently decreasing nonivamide content (0.09% non­
system (erianin concentration of 30 nM) cells viability and anti­ ivamide). In the ex vivo penetration assay, test formulation containing
proliferative effect on human keratinocyte (HaCaT) cells were accessed. 0.09% nonivamide and standard formulation (0.05% nonivamide hy­
The results showed that erianine-loaded dendritic mesoporous silica drophilic cream), resulted in the same order of nonivamide concentra­
nanospheres (particle mean size 161 nm/pore size 4.6 nm) presented an tions magnitude present in the stratum corneum. The tested formulation
antiproliferative and pro-apoptotic effect superior to the erianine-loaded had a sustained penetration profile, showing within 1 to 12 h incubation
dendritic mesoporous silica nanospheres (particle mean size 164 nm/ time a continuous increase in penetrated nonivamide, which was still
pore size 3.5 nm) and to free erianin. Cell viabilities were 61.8% (eria­ present at the action site after 24 h incubation. In addition, the in vivo
nin), 49.3% (nanospheres loaded with erianin showing smaller pore skin irritation test showed that the formulation (0.09% nonivamide) was
size) and 48.3% (nanospheres loaded with erianin showing larger pore not irritating to skin after an incubation period of 1 h.
size). Besides, initial or late apoptosis percentages were respectively The drug lidocaine is commonly used as a topical anesthetic, how­
8.5% and 3.0% (erianin), 9.5% and 5.5% (nanospheres loaded with ever its low water solubility and negligible tissue absorption limit its
erianin with smaller pore size) and 11.7% and 4.0% (nanospheres bioavailability when applied to the skin (Nafisi et al., 2018). To over­
loaded with erianin having a larger pore size). In the ex vivo skin come these limitations Nafisi et al. (2018) incorporated this anesthetic
permeation test with pigskin, gel formulation (Carbopol gel) containing into amine functionalized mesoporous SNPs (particle size 95 nm). In
erianin-loaded 4.6 nm pore size nanospheres showed greater drug vitro release using cellulose dialysis membrane (conditions: 37 ◦ C and
accumulation than the gel containing 3.5 nm pore size nanospheres and pH 7.4) and ex vivo drug permeation in human skin by using vertical
the gel loaded with free erianin. The authors attributed this result to diffusion Franz cells were investigated. The authors observed a greater
greater surface area of nanospheres with a larger pore size, which would lidocaine release for amine-functionalized mesoporous SNPs (61%) than
facilitate drug retention and release on the skin. non-functionalized SNPs (20%) in the first 3 h. Also, the amine-
Fibrosis is another disease that can affect skin. It is commonly functionalized mesoporous SNPs allowed for better skin lidocaine
characterized by excessive skin collagen deposition. In response to tissue permeation compared to non-functionalized SNPs. Ex vivo permeation
damage, collagen fibers act by preventing the rest of the tissue from data of nanosystems were compared to the same control concentration
being harmed, creating a thick border that causes fibrosis. An example of (lidocaine hydroalcoholic solution, 0.4 mg/mL). After 24 h, almost 64%
such a disease is scleroderma (Sng et al., 2018). In this context, Morry of the amine-functionalized mesoporous SNPs drug content was
et al. (2015) proposed a new platform for skin fibrosis treatment based permeated, while the control solution showed 36%, and the non-
on mesoporous SNPs coated with polyethyleneimine (PEI) and poly­ functionalized mesoporous SNPs exhibited 13%. The results found
ethylene glycol (PEG) (hydrodynamic size of 104 ± 1.7 nm), loaded regarding high permeation of nanosystem lidocaine released was
HSP47-targeted small interfering RNA (siRNA). HSP47 is a protein that attributed to electrostatic interaction between positive surface charges
plays an important role in collagen homeostasis, by inducing excessive of amine functionalized SNPs and negative charge of the skin cells
collagen synthesis, leading to fibrotic diseases. The authors strategy to membrane.
combat fibrogenesis was to take advantage of intrinsic antioxidant An attractive alternative to minimize the challenges associated with
properties of mesoporous SNPs and the promising capacity of siRNAs in other routes of drug administration, such as oral and parenteral, is the
gene silencing (gene neutralization through interfering RNA) in order to transdermal route. Advanced devices based on SNPs are being designed
reduce HSP47 levels, particularly superexpressed in fibrotic tissues. An for transdermal drug delivery. For example, a recent study developed
effective treatment to skin fibrosis was obtained using the described cotton patches based on chitosan-pullulan hydrogel and mesoporous
nanocarrier. Treatment of primary murine dermal fibroblast cells with SNPs loaded with the alkaloid colchicine (particle size 167,1 ± 51,36
nanocarrier (17.5 μg/mL of nanoparticles) under conditions of nm) to treat osteoarthritis (Mohamed et al., 2020). First, colchicine was
menadione-induced oxidative stress reduced ROS. After 1 h menadione encapsulated in mesoporous SNPs (drug-entrapment efficiency 67.4 ±
exposure cellular ROS increased 7.6 times, and cells nanocarrier pre- 3%), which were incorporated into a chitosan-pullulan hydrogel, and
treatment decreased ROS production to non-menadione level and loaded in cotton gauze forming an easy-to-apply transdermal patch. The
similar to that obtained by an established antioxidant, N-acetylcysteine. final colchicine amount in the prepared patch was 0.5 mg/cm2. The
In addition, intradermal nanomaterial injection (dose of 220 μg nano­ developed system combined the virtues of nanometer size and huge
particles as 0.65 nmol siRNA) in mice with bleomycin-induced sclero­ SNPs surface area to chitosan/pullulan hydrogel matrix sustained
derma (in vivo skin fibrosis model) effectively decreased skin HSP47 release effect. The nanodevice permeation performance was investi­
protein expression to normal level, which resulted in a reduction in pro- gated using isolated rat skin (ex vivo), laser confocal microscopy using
fibrotic gene expressions and skin thickness (19%, p ≤ 0.01 vs. bleo­ fluorescein using excised rat skin and its efficacy as osteoarthritis
mycin alone). treatment in rat model (in vivo). The ex vivo permeation test demon­
In order to improve chronic itching treatment, research (Heck et al., strated that colchicine-containing transdermal patches (free and
2017) has proposed a film-forming formulation containing porous silica entrapped in mesoporous SNPs) permeated five and seven times,
particles (Syloid® XDP 3050) loaded with the antipruritic drug non­ respectively, deeper in skin than aqueous colchicine solution. Increased
ivamide (capsaicin synthetic analogue) for skin sustained delivery of this penetration promoted by the hydrogel patch incorporating fluorescein
active ingredient. The authors tested the formulations on the following encapsulated in mesoporous SNPs was demonstrated by a broader and
criteria: skin permeation (ex vivo test through post-auricular porcine more intense distribution of fluorescein in all skin layers. In addition, in
skin), skin penetration (ex vivo test by skin surface biopsy followed by a therapeutic investigation using in vivo model of monoiodine-induced
cyanoacrylate cryosection) and skin irritation (in vivo test with healthy osteoarthritis (MIA) the mesoporous SNPs treated animals displayed
volunteers). Although nonivamide concentration in the test formulation improved locomotor activity (by 74%), blood glutathione level (23%)
(0.9% nonivamide) was approximately twenty times greater than in and notable decline in malondialdehyde (34%), nitric oxide (28%), TNF-
conventional clinically proven therapy (hydrophilic nonivamide cream α (61%), and COX-2 (74%) levels, which are stress biomarkers oxidative.
at 0.025% and 0.05%), nonivamide amount and fluxpermeated by skin
area were similar. This proved that the formulation permeation profile is
at a therapeutically safe and efficient rate. As viable epidermis is the

15
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

6. Silica nanoparticles in cosmetology effects (Burnett and Wang, 2011). Thus, to reduce inherent sunscreens
deficiencies, many studies have sought to find new and better forms of
Silica is a component of many cosmetic products. SNPs can be found photoprotection, focusing on methods related to encapsulation and
in leave-in cosmetic products for hair, skin, lips, face and nails. SNPs are incorporation to form more stable, aesthetically acceptable (more
used to enhance the effectiveness, texture, and shelf-life of cosmetic pleasant to the skin) formulations, with high UVR protection capacity
products. It adds absorbency and acts as an anti-caking agent (Fytianos and low health risks. SNPs have been reported to have high photo­
et al., 2020). stability and UVR protection effects (Knežević et al., 2018; D.P. Wang
It is known that sunscreens are considered a valuable tool in et al., 2020, W.H. Wang et al., 2020). In this context, Y.C. Lin et al.
providing photoprotection against ultraviolet radiation (UVR) harmful (2018) and Z. Lin et al. (2018) produced two versions of hydrogel

Fig. 10. A) (a) Ex vivo two-photon microscopic images of porcine skins obtained after 4 h of rhodamine 6G-labeled bp-SNPs (yellow) topical application. The blue
signal represents collagen second harmonic generation in the dermal layer. (b) Fluorescence images of porcine skin tissue histological section described in (a).
Control image of skin without treatment. The red signal represents rhodamine-6G and the blue signal represents DAPI. (c) Behavior of different sunscreens types
when applied to skin. B) (a) Schematic of in vivo anti-UV assessment and (b) photoimages of murine dorsal skin obtained after 3 days of topical application of various
samples and treatment with high-dose UVB. (c) H & E-stained histological sections of murine dorsal skin tissues described in (b) and (d) normalized epidermal
thicknesses of the histological sections described in (c) (n = 3, * P < 0.05 and ** P < 0.01). Reprinted with permission from ref. (Yoo et al., 2020), Copyright 2020
American Chemical Society. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

16
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Carbopol 940-based sunscreen, one containing avobenzone-loaded had a significant impact on formulation viscosity profile. Of the three
mesoporous silica microparticles (single MS), that were synthesized diameters examined (Aerosil R 202/14 nm, Aerosil R 812/6.5 nm and
with a structure-directing agent (Pluronic P123) and the other con­ Aerosil R 812S/7 nm), the largest presented the highest viscosity, at the
taining avobenzone-loaded mesoporous silica microparticles (hybrid same time that it generated yield stress due to an effective network
MS), that were developed with the combination of two structure-guiding formation. The silica particles concentration also influenced formulation
agents (Pluronic P123 and Pluronic F68). The hybrid MS exhibited a viscosity, with an increasing concentration showing an increase in vis­
greater specific surface area (853 m2/g) than the single MS (764 m2/g) cosity. In terms of biosurfactants, rhamnolipids and sophorolipids
and particle sizes of about 1.5 and 1 μm, respectively. These nano­ showed very similar behavior, which meaned that they act as an
formulations effectiveness (0.67% avobenzone) has been evaluated in ex emulsifier and does not affect the system rheological behavior. Silicone
vivo and in vivo tests. The ex vivo skin permeation assay on pigskin oil replacement with other natural oils was only possible with canola
revealed that the absorption of avobenzone contained in the nano­ and castor oil, since jojoba, squalene, and mineral oil did not stabilize it.
formulations was one-third to one-half of the free control, thus trapping The formulation rheology provided a stable emulsion, the shear-
avobenzone in microparticles reduced its skin absorption, where its thinning effect ensured lip gloss easy applicability, while viscosity and
action would be undesirable. Specifically, the hydrogel containing elasticity provided a good product fixation to lips. After applying the
hybrid MS demonstrated lower avobenzone deposition compared to final formulation to the lips, pigments and the oil shine effect were
single MS (p < 0.05). In addition, nanoformulations UVA protection was visible. In addition, the application proved to be easy and not too sticky,
investigated in vivo in nude mouse. Topical application of hydrogel due to the silica particles present.
containing hybrid MS on mice skin before UVA irradiation prevented the
increase of transepidermal water loss (TEWL), formation of furrows in 7. Conclusions
the skin, keratinocyte apoptosis, and neutrophils infiltration that are
indications of skin photodegradation. Knowing skin anatomy and physiology is of fundamental importance
Recently, Yoo et al. (2020) proposed sunscreens based on biocom­ to understand SNPs behavior, interaction, and mechanisms of bio­
patible and biodegradable organosilica nanoparticles with self- distribution, metabolization, and elimination when they are adminis­
encapsulated phenyl motifs using phenylsilane precursors (bp-SNPs) tered on the skin. Despite efforts to evaluate these points, there is still no
for simultaneous UVR absorption/reflection. The bp-SNPs size (75–95 clear understanding of skin SNPs permeability and toxicity, and the
nm) was designed to be large enough to reflect the UVA, but small research so far does not allow reaching a consensus on the subject.
enough to be imperceptible when applied, thus preventing the formu­ Therefore, more investigative studies are needed, especially studies
lation from leaving a white layer on the skin. This study objective was involving long-exposure tests. However, the use of functionalization
achieved, and the bp-SNPs were clearly transparent with low opacity strategies to modify SNPs surface, in addition to making them more
when compared with inorganic filters. The bp-SNPs ex vivo penetration biocompatible, can be useful tools in silica nanosystems intelligent
was evaluated by applying to pig skin a fluorophore rhodamine-6G- construction with high skin penetration and permeation capacity.
labeled nanoparticles suspension and analyzed by means of two- Among the functionalization strategies, the insertion of positive groups
photon and fluorescence microscopy. Microscopy images obtained on SNPs surface, such as amine groups, sets up the most frequently used
from swine skins after 4 h of rhodamine-6G-labeled bp-SNPs topical strategy by the selected studies to promote SNPs skin permeation. In
application revealed that the nanoparticles tended to adhere to skin addition, these studies demonstrate high SNPs potential to improve
outer without penetrating the dermis (Fig. 10Aa). In addition, the drugs cutaneous penetration and consequently the therapeutic perfor­
fluorescence images of cryo-sectioned skin tissues showed that most of mance. Therapeutic SNPs cited were able to deliver the bioactive com­
the bp-SNPs were accumulated on the skin tissues surface (Fig. 10Ab). pounds to the skin (local delivery) and act as transdermal formulations,
The bp-SNPs in vivo protective effect was evaluated against sunburn on releasing the encapsulated actives in a controlled manner or even on
the skin of nude mice. After applying the bp-SNPs (0.5 mg/mL) and the demand. The nanoencapsulation allowed increasing the physicochem­
positive and negative controls to the mice dorsal skin and exposing them ical stability and apparent solubility of drugs with low stability and low
to a UVB lamp for three days, it was demonstrated that the non-treated aqueous solubility. Such limitations imply an insignificant cutaneous
skin tissue was significantly damaged, and erythema (redness of the absorption of the drug and a reduction in its bioavailability. SNPs are
skin) was observed. However, the skin tissue that was treated with bp- already employed in cosmetic industry, thanks to their excellent prop­
SNPs and the positive control (0.1 mg/mL of TiO2 nanoparticles and erties and their low production cost. New uses for SNPs in cosmetics and
0.3 mg/mL of ZnO nanoparticles) exhibited the stratum corneum cosmeceuticals products has also been proposed being mostly studies
outermost layer flaking of but without visible erythema or edema that exploit photoprotection, show promising results.
(Fig. 10Bb). Histological skin tissues sections were prepared to assess the
epidermal and dermal layers damage caused by sunburn (Fig. 10Bc). In 8. Future perspectives
unprotected skin, significant epidermal thickening and epidermal hy­
pertrophy were observed. As shown in Fig. 10Bd, the epidermal layer Although it is a research field still little explored, silica nanosystems
thicknesses of UV exposed unprotected skin was twice thicker than development for topical use is moving in the right direction, that is, in
normal skin. However, bp-SNPs treated skin did not present severely search of more studies. However, there is still much to be explored, so
epidermal thickness under UR exposure and its anti-UV effect was we can highlight some gaps that need to be filled: i) lack of standardi­
comparable to that of commercially available positive control zation methods for toxicological and in vitro, ex vivo, and in vivo efficacy
nanoparticles. experiments on skin, allowing a proper comparison of results between
Silica particles also acted as a stabilizing agent in a lip gloss formu­ different research groups; ii) insufficient data on SNPs safety/toxicity
lation (Drakontis and Amin, 2020). In this study, a combination of profile; iii) lack of evidence-based randomized clinical studies exploring
biosurfactants (rhamnolipids and sophorolipids) with silica particles SNPs therapeutic efficacy and safety for topical use and iv) lack of
(Aerosil R 202, R 812 and R 812S) were used to formulate a sustainable knowledge about SNPs industrial scale production. Therefore, the use of
lip gloss emulsion. The formulations viscosity profile was evaluated SNPs in clinical practice and in consumer cosmetic/dermocosmetic
using the following parameters: silica particles diameter size, formula­ products will largely depend on the progress and success of future
tion particles concentration, together with the different types of bio­ research.
surfactant and used oils. In addition, pigments were added to samples to The pharmaceutical and cosmetic markets are in fact attentive to
form the final lip gloss formulation so its performance after application new technological discoveries stemming from scientific research that
could be investigated. It was found that the silica particle diameter size prove efficacy and safety. A new class of products that was born from the

17
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

cosmetic industry desire to go beyond simply adorning and beautifying Arriagada, F.J., Osseo-Asare, K., 1994. Synthesis of Nanometer-Sized Silica by Controlled
Hydrolysis in Reverse Micellar Systems, in: The Colloid Chemistry of Silica, p.
the skin, cosmeceuticals, are considered functional cosmetic products, as
113–128. https://doi.org/10.1021/ba-1994-0234.ch005.
their bioactive formulations exert a beneficial therapeutic effect on skin Arts, J.H.E., Muijser, H., Duistermaat, E., Junker, K., Kuper, C.F., 2007. Five-day
appearance. Finally, the future challenge of topical silica nano­ inhalation toxicity study of three types of synthetic amorphous silicas in Wistar rats
formulations will probably be to improve skin well-being, through and post-exposure evaluations for up to 3 months. Food Chem. Toxicol. 45,
1856–1867. https://doi.org/10.1016/J.FCT.2007.04.001.
cosmeceutical formulations development, which address important Ascenso, A., Raposo, S., Batista, C., Cardoso, P., Mendes, T., Praça, F.G., Bentley, M.V.L.
functional issues to meet consumer demands, especially biodegradable B., Simões, S., 2015. Development, characterization, and skin delivery studies of
nanoformulations based on organic silicon. Organic silicon is essential related ultradeformable vesicles: transfersomes, ethosomes, and transethosomes. Int.
J. Nanomedicine 10, 5837. https://doi.org/10.2147/IJN.S86186.
for skin, as it acts as an antioxidant, fighting free radicals and stimu­ Athinarayanan, J., Periasamy, V.S., Alhazmi, M., Alatiah, K.A., Alshatwi, A.A., 2015.
lating collagen synthesis. Synthesis of biogenic silica nanoparticles from rice husks for biomedical
applications. Ceram. Int. 41, 275–281. https://doi.org/10.1016/J.
CERAMINT.2014.08.069.
CRediT authorship contribution statement Bakshi, P., Vora, D., Hemmady, K., Banga, A.K., 2020. Iontophoretic skin delivery
systems: Success and failures. Int. J. Pharm. 586 https://doi.org/10.1016/j.
Renata Pinho Morais: Investigation, Conceptualization, Method­ ijpharm.2020.119584.
Barrera, E.G., Livotto, P.R., do Santos, J.H.Z., 2016. Hybrid silica bearing different
ology, Writing – original draft. Sabrina Hochheim: Investigation, organosilanes produced by the modified Stöber method. Powder Technol. 301,
Conceptualization, Writing – original draft. Carolina C. de Oliveira: 486–492. https://doi.org/10.1016/J.POWTEC.2016.04.025.
Supervision, Writing – review & editing, Methodology, Conceptualiza­ Baspinar, Y., Borchert, H.H., 2012. Penetration and release studies of positively and
negatively charged nanoemulsions - Is there a benefit of the positive charge? Int. J.
tion. Izabel C. Riegel-Vidotti: Supervision, Writing – review & editing,
Pharm. 430, 247–252. https://doi.org/10.1016/j.ijpharm.2012.03.040.
Funding acquisition, Methodology, Conceptualization. Cláudia E.B. Berlier, G., Gastaldi, L., Sapino, S., Miletto, I., Bottinelli, E., Chirio, D., Ugazio, E., 2013.
Marino: Supervision, Writing – review & editing, Funding acquisition, MCM-41 as a useful vector for rutin topical formulations: synthesis, characterization
and testing. Int. J. Pharm. 457, 177–186. https://doi.org/10.1016/j.
Methodology, Conceptualization.
ijpharm.2013.09.018.
Bharti, C., Gulati, N., Nagaich, U., Pal, A., 2015. Mesoporous silica nanoparticles in
Declaration of Competing Interest target drug delivery system: a review. Int. J. Pharm. Investig. 5, 124. https://doi.
org/10.4103/2230-973x.160844.
Bolzinger, M.A., Briançon, S., Chevalier, Y., 2011. Nanoparticles through the skin:
The authors declare the following financial interests/personal re­ Managing conflicting results of inorganic and organic particles in cosmetics and
lationships which may be considered as potential competing interests: pharmaceutics. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 3, 463–478.
https://doi.org/10.1002/wnan.146.
Renata Pinho Morais reports financial support was provided by Coor­
Bullock, A.J., Garcia, M., Shepherd, J., Rehman, I., Sheila, M., 2020. Bacteria induced pH
dination of Higher Education Personnel Improvement. Sabrina Hoch­ changes in tissue-engineered human skin detected non-invasively using Raman
heim reports financial support was provided by Coordination of Higher confocal spectroscopy. Appl. Spectrosc. Rev. 55, 158–171. https://doi.org/10.1080/
Education Personnel Improvement. Claudia Eliana Bruno Marino re­ 05704928.2018.1558232.
Burnett, M.E., Wang, S.Q., 2011. Current sunscreen controversies: A critical review.
ports financial support was provided by National Council for Scientific Photodermatol. Photoimmunol. Photomed. 27, 58–67. https://doi.org/10.1111/
and Technological Development. j.1600-0781.2011.00557.x.
Busch, L., Keziban, Y., Dähne, L., Keck, C.M., Meinke, M.C., Lademann, J., Patzelt, A.,
2021. The impact of skin massage frequency on the intrafollicular transport of silica
Acknowledgements nanoparticles: Validation of the ratchet effect on an ex vivo porcine skin model. Eur.
J. Pharm. Biopharm. 158, 266–272. https://doi.org/10.1016/j.ejpb.2020.11.018.
The authors acknowledge National Council for Scientific and Tech­ Calixto, G.M.F., Bernegossi, J., De Freitas, L.M., Fontana, C.R., Chorilli, M.,
Grumezescu, A.M., 2016. Nanotechnology-based drug delivery systems for
nological Development (CNPq), Brazil (grant C.E.B. Marino - 303126/ photodynamic therapy of cancer: A review. Molecules 21, 342. https://doi.org/
2019-1 and grant I.C. Riegel-Vidotti - 306038/2017-0) and Coordina­ 10.3390/molecules21030342.
tion for the Improvement of Higher Education Personnel (CAPES), Brazil Callard, R.E., Harper, J.I., 2007. The skin barrier, atopic dermatitis and allergy: a role for
Langerhans cells? Trends Immunol. 28, 294–298. https://doi.org/10.1016/j.
(Finance code 001) for financial support. it.2007.05.003.
Cao, M., Li, B., Guo, M., Liu, Y., Zhang, L., Wang, Y., Hu, B., Li, J., Sutherland, D.S.,
References Wang, L., Chen, C., 2021. In vivo percutaneous permeation of gold nanomaterials in
consumer cosmetics: implication in dermal safety assessment of consumer
nanoproducts. Nanotoxicology 15, 131–144. https://doi.org/10.1080/
Abdelaal, H.M., Shaikjee, A., 2020. Microwave-based fast synthesis of clear-cut hollow
17435390.2020.1860264.
spheres with mesoporous wall of silica nanoparticles as excellent drug delivery
Cao, W., Zhou, Y., Niu, Y., Zhu, X., Song, Y., Guo, R., 2017. Quantitative analysis of
vehicles. J. Nanoparticle Res. 22, 1–9. https://doi.org/10.1007/S11051-020-04918-
hepatic toxicity in rats induced by inhalable silica nanoparticles using acoustic
3/FIGURES/11.
radiation force imaging. J. Ultrasound Med. 36, 1829–1839. https://doi.org/
Al Mahrooqi, J.H., Khutoryanskiy, V.V., Williams, A.C., 2021. Thiolated and PEGylated
10.1002/jum.14219.
silica nanoparticle delivery to hair follicles. Int. J. Pharm. 593, 120130 https://doi.
Carazo, E., Borrego-Sánchez, A., García-Villén, F., Sánchez-Espejo, R., Cerezo, P.,
org/10.1016/j.ijpharm.2020.120130.
Aguzzi, C., Viseras, C., 2018. Advanced inorganic nanosystems for skin drug
Alexander, A., Dwivedi, S., Ajazuddin, Giri, T.K., Saraf, Swarnlata, Saraf, Shailendra,
delivery. Chem. Rec. 18, 891–899. https://doi.org/10.1002/tcr.201700061.
Tripathi, D.K., 2012. Approaches for breaking the barriers of drug permeation
Cauda, V., Argyo, C., Bein, T., 2010. Impact of different PEGylation patterns on the long-
through transdermal drug delivery. J. Control. Release 164, 26–40. https://doi.org/
term bio-stability of colloidal mesoporous silica nanoparticles. J. Mater. Chem. 20,
10.1016/j.jconrel.2012.09.017.
8693–8699. https://doi.org/10.1039/c0jm01390k.
Al-Marzouqi, A.H., Solieman, A., Shehadi, I., Adem, A., 2008. Influence of the
Chaudhary, M., Khan, A., Gupta, M., 2020. Skin ageing: pathophysiology and current
preparation method on the physicochemical properties of econazole-β-cyclodextrin
market treatment approaches. Curr. Aging Sci. 13, 22–30. https://doi.org/10.2174/
complexes. J. Incl. Phenom. Macrocycl. Chem. 60, 85–93. https://doi.org/10.1007/
1567205016666190809161115.
s10847-007-9356-6.
Chen, F., Hableel, G., Zhao, E.R., Jokerst, J.V., 2018a. Multifunctional nanomedicine
Alruwaili, N.K., Zafar, A., Imam, S.S., Alharbi, K.S., Alotaibi, N.H., Alshehri, S.,
with silica: Role of silica in nanoparticles for theranostic, imaging, and drug
Alhakamy, N.A., Alzarea, A.I., Afzal, M., Elmowafy, M., 2020. Stimulus Responsive
monitoring. J. Colloid Interface Sci. 521, 261–279. https://doi.org/10.1016/j.
Ocular Gentamycin-Ferrying Chitosan Nanoparticles Hydrogel: Formulation
jcis.2018.02.053.
Optimization, Ocular Safety and Antibacterial Assessment. Int. J. Nanomedicine 15,
Chen, L., Liu, J., Zhang, Y., Zhang, G., Kang, Y., Chen, A., Feng, X., Shao, L., 2018b. The
4717. https://doi.org/10.2147/IJN.S254763.
toxicity of silica nanoparticles to the immune system. Nanomedicine 13, 1939–1962.
Angelova-Fischer, I., Fischer, T.W., Abels, C., Zillikens, D., 2018. Accelerated barrier
https://doi.org/10.2217/nnm-2018-0076.
recovery and enhancement of the barrier integrity and properties by topical
Chen, W.H., Luo, G.F., Lei, Q., Cao, F.Y., Fan, J.X., Qiu, W.X., Jia, H.Z., Hong, S., Fang, F.,
application of a pH 4 vs. a pH 5.8 water-in-oil emulsion in aged skin. Br. J. Dermatol.
Zeng, X., Zhuo, R.X., Zhang, X.Z., 2016. Rational design of multifunctional magnetic
179, 471–477. https://doi.org/10.1111/bjd.16591.
mesoporous silica nanoparticle for tumor-targeted magnetic resonance imaging and
Anirudhan, T.S., Nair, A.S., 2018. Temperature and ultrasound sensitive gatekeepers for
precise therapy. Biomaterials 76, 87–101. https://doi.org/10.1016/j.
the controlled release of chemotherapeutic drugs from mesoporous silica
biomaterials.2015.10.053.
nanoparticles. J. Mater. Chem. B 6, 428–439. https://doi.org/10.1039/c7tb02292a.
Chen, X., Zhu, S., Hu, X., Sun, D., Yang, J., Yang, C., Wu, W., Li, Y., Gu, X., Li, M., Liu, B.,
Arriagada, F., Nonell, S., Morales, J., 2019. Silica-based nanosystems for therapeutic
Ge, L., Gu, Z., Xu, H., 2020. Toxicity and mechanism of mesoporous silica
applications in the skin. Nanomedicine 14, 2243–2267. https://doi.org/10.2217/
nnm-2019-0052.

18
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

nanoparticles in eyes. Nanoscale 12, 13637–13653. https://doi.org/10.1039/ Ghasemiyeh, P., Mohammadi-Samani, S., 2020. Potential of nanoparticles as permeation
d0nr03208e. enhancers and targeted delivery options for skin: Advantages and disadvantages.
Chen, Y., Chen, N., Feng, X., 2021. The role of internal and external stimuli in the Drug Des. Devel. Ther. 14, 3271–3289. https://doi.org/10.2147/DDDT.S264648.
rational design of skin-specific drug delivery systems. Int. J. Pharm. 592, 120081 Gholami, T., Salavati-Niasari, M., Bazarganipour, M., Noori, E., 2013. Synthesis and
https://doi.org/10.1016/j.ijpharm.2020.120081. characterization of spherical silica nanoparticles by modified Stöber process assisted
Clemente, N., Miletto, I., Gianotti, E., Invernizzi, M., Marchese, L., Dianzani, U., Renò, F., by organic ligand. Superlattices Microstruct. 61, 33–41. https://doi.org/10.1016/J.
2019. Verteporfin-loaded mesoporous silica nanoparticles inhibit mouse melanoma SPMI.2013.06.004.
proliferation in vitro and in vivo. J. Photochem. Photobiol. B Biol. 197, 111533 Giovannini, G., Warncke, P., Fischer, D., Stranik, O., Hall, A.J., Gubala, V., 2018.
https://doi.org/10.1016/j.jphotobiol.2019.111533. Improving colloidal stability of silica nanoparticles when stored in responsive gel:
Cortés, H., Rojas-Márquez, M., Del Prado-Audelo, M.L., Reyes-Hernández, O.D., application and toxicity study. Nanotoxicology 12, 407–422. https://doi.org/
González-Del Carmen, M., Leyva-Gómez, G., 2021. Alterations in mental health and 10.1080/17435390.2018.1457729.
quality of life in patients with skin disorders: a narrative review. Int. J. Dermatol. Gonçalves, M.C., 2018. Sol-gel silica nanoparticles in medicine: A natural choice. design,
1–9 https://doi.org/10.1111/IJD.15852. synthesis and products. Molecules 23. https://doi.org/10.3390/
Cosco, D., Paolino, D., Maiuolo, J., Marzio, L.D., Carafa, M., Ventura, C.A., Fresta, M., molecules23082021.
2015. Ultradeformable liposomes as multidrug carrier of resveratrol and 5-fluoro­ Goyal, R., Macri, L.K., Kaplan, H.M., Kohn, J., 2016. Nanoparticles and nanofibers for
uracil for their topical delivery. Int. J. Pharm. 489, 1–10. https://doi.org/10.1016/J. topical drug delivery. J. Control. Release 240, 77–92. https://doi.org/10.1016/j.
IJPHARM.2015.04.056. jconrel.2015.10.049.
CosmeticsInfo, 2021. Cosmetic Regulation in the European Union [WWW Document]. Guo, C., Liu, Y., Li, Y., 2021. Adverse effects of amorphous silica nanoparticles: Focus on
Cosmet. Regul. Eur. Union. URL https://cosmeticsinfo.org/ (accessed 12.14.21). human cardiovascular health. J. Hazard. Mater. 406 https://doi.org/10.1016/j.
Costa, D.F., Mendes, L.P., Torchilin, V.P., 2019. The effect of low- and high-penetration jhazmat.2020.124626.
light on localized cancer therapy. Adv. Drug Deliv. Rev. 138, 105–116. https://doi. Hadipour Moghaddam, S.P., Mohammadpour, R., Ghandehari, H., 2019. In vitro and in
org/10.1016/j.addr.2018.09.004. vivo evaluation of degradation, toxicity, biodistribution, and clearance of silica
D’Mello, S.A.N., Finlay, G.J., Baguley, B.C., Askarian-Amiri, M.E., 2016. Signaling nanoparticles as a function of size, porosity, density, and composition. J. Control.
pathways in melanogenesis. Int. J. Mol. Sci. 17, 1–18. https://doi.org/10.3390/ Release 311–312, 1–15. https://doi.org/10.1016/j.jconrel.2019.08.028.
ijms17071144. Hamam, F., Nasr, A., 2020. Curcumin-loaded mesoporous silica particles as wound-
Deng, Y.D., Zhang, X.D., Yang, X.S., Huang, Z.L., Wei, X., Yang, X.F., Liao, W.Z., 2021. healing agent: An In vivo study. Saudi J. Med. Med. Sci. 8, 17. https://doi.org/
Subacute toxicity of mesoporous silica nanoparticles to the intestinal tract and the 10.4103/sjmms.sjmms_2_19.
underlying mechanism. J. Hazard. Mater. 409 https://doi.org/10.1016/j. He, Q., Zhang, Z., Gao, F., Li, Y., Shi, J., 2011. In vivo biodistribution and urinary
jhazmat.2020.124502. excretion of mesoporous silica nanoparticles: effects of particle size and PEGylation.
Desprez, B., Dent, M., Keller, D., Klaric, M., Ouédraogo, G., Cubberley, R., Duplan, H., Small 7, 271–280. https://doi.org/10.1002/smll.201001459.
Eilstein, J., Ellison, C., Grégoire, S., Hewitt, N.J., Jacques-Jamin, C., Lange, D., He, W., Guo, X., Xiao, L., Feng, M., 2009. Study on the mechanisms of chitosan and its
Roe, A., Rothe, H., Blaauboer, B.J., Schepky, A., Mahony, C., 2018. A strategy for derivatives used as transdermal penetration enhancers. Int. J. Pharm. 382, 234–243.
systemic toxicity assessment based on non-animal approaches: The Cosmetics https://doi.org/10.1016/J.IJPHARM.2009.07.038.
Europe Long Range Science Strategy programme. Toxicol. Vitr. 50, 137–146. Heck, R., Lukić, M., Savić, S.D., Daniels, R., Lunter, D.J., 2017. Ex vivo skin permeation
https://doi.org/10.1016/j.tiv.2018.02.017. and penetration of nonivamide from and in vivo skin tolerability of film-forming
Dhal, S., Gavara, R.R., Pal, K., Banerjee, I., Mishra, M., Giri, S., 2020. Facile transdermal formulations containing porous silica. Eur. J. Pharm. Sci. 106, 34–40. https://doi.
delivery of upconversion nanoparticle by iontophoresis-responsive magneto- org/10.1016/j.ejps.2017.05.045.
upconversion oleogel. Nano Express 1, 010012. https://doi.org/10.1088/2632- Herman, A., Herman, A.P., 2015. Essential oils and their constituents as skin penetration
959X/AB81E1. enhancer for transdermal drug delivery: A review. J. Pharm. Pharmacol. 67,
Dimitriev, Y., Ivanova, Y., Iordanova, R., 2008. History of sol-gel science and technology 473–485. https://doi.org/10.1111/jphp.12334.
(review). J. Univ. Chem. Technol. Metall. 43, 181–192. Hirai, T., Yoshikawa, T., Nabeshi, H., Yoshida, T., Tochigi, S., Ichihashi, K., Uji, M.,
Drakontis, C.E., Amin, S., 2020. Design of sustainable lip gloss formulation with Akase, T., Nagano, K., Abe, Y., Kamada, H., Itoh, N., Tsunoda, S., Yoshioka, Y.,
biosurfactants and silica particles. Int. J. Cosmet. Sci. 42, 573–580. https://doi.org/ Tsutsumi, Y., 2012. Amorphous silica nanoparticles size-dependently aggravate
10.1111/ics.12642. atopic dermatitis-like skin lesions following an intradermal injection. Part. Fibre
Dreno, B., Amici, J.M., Demessantflavigny, A.L., Wright, C., Taieb, C., Desai, S.R., Toxicol. 9 https://doi.org/10.1186/1743-8977-9-3.
Alexis, A., 2021. The impact of acne, atopic dermatitis, skin toxicities and scars on Hirai, T., Yoshioka, Y., Takahashi, H., Ichihashi, K., Udaka, A., Mori, T., Nishijima, N.,
quality of life and the importance of a holistic treatment approach. Clin. Cosmet. Yoshida, T., Nagano, K., Kamada, H., Tsunoda, S.I., Takagi, T., Ishii, K.J.,
Investig. Dermatol. 14, 623–632. https://doi.org/10.2147/CCID.S315846. Nabeshi, H., Yoshikawa, T., Higashisaka, K., Tsutsumi, Y., 2015. Cutaneous exposure
Esim, O., Kurbanoglu, S., Savaser, A., Ozkan, S.A., Ozkan, Y., 2019. Nanomaterials for to agglomerates of silica nanoparticles and allergen results in IgE-biased immune
drug delivery systems, in: New Developments in Nanosensors for Pharmaceutical response and increased sensitivity to anaphylaxis in mice. Part. Fibre Toxicol. 12,
Analysis. Elsevier, pp. 273–301. https://doi.org/10.1016/B978-0-12-816144- 1–6. https://doi.org/10.1186/s12989-015-0095-3.
9.00009-2. Holmes, A.M., Kempson, I., Turnbull, T., Paterson, D., Roberts, M.S., Roberts, M.S., 2020.
Eves, P.C., MacNeil, S., Haycock, J.W., 2006. α-Melanocyte stimulating hormone, Penetration of zinc into human skin after topical application of nano zinc oxide used
inflammation and human melanoma. Peptides 27, 444–452. https://doi.org/ in commercial sunscreen formulations. ACS Appl. Bio Mater. 3, 3640–3647. https://
10.1016/j.peptides.2005.01.027. doi.org/10.1021/acsabm.0c00280.
Falk, G., Shinhe, G.P., Teixeira, L.B., Moraes, E.G., de Oliveira, A.P.N., 2019. Synthesis of Holmes, A.M., Song, Z., Moghimi, H.R., Roberts, M.S., 2016. Relative penetration of zinc
silica nanoparticles from sugarcane bagasse ash and nano-silicon via oxide and zinc ions into human skin after application of different zinc oxide
magnesiothermic reactions. Ceram. Int. 45, 21618–21624. https://doi.org/10.1016/ formulations. ACS Nano 10, 1810–1819. https://doi.org/10.1021/
J.CERAMINT.2019.07.157. acsnano.5b04148.
Farjadian, F., Roointan, A., Mohammadi-Samani, S., Hosseini, M., 2019. Mesoporous Hozayen, W.G., Mahmoud, A.M., Desouky, E.M., El-Nahass, E.S., Soliman, H.A.,
silica nanoparticles: synthesis, pharmaceutical applications, biodistribution, and Farghali, A.A., 2019. Cardiac and pulmonary toxicity of mesoporous silica
biosafety assessment. Chem. Eng. J. 359, 684–705. https://doi.org/10.1016/J. nanoparticles is associated with excessive ROS production and redox imbalance in
CEJ.2018.11.156. Wistar rats. Biomed. Pharmacother. 109, 2527–2538. https://doi.org/10.1016/j.
Ferraris, C., Rimicci, C., Garelli, S., Ugazio, E., Battaglia, L., 2021. Nanosystems in biopha.2018.11.093.
cosmetic products: A brief overview of functional, market, regulatory and safety Huh, S., Wiench, J.W., Yoo, J.C., Pruski, M., Lin, V.S.Y., 2003. Organic functionalization
concerns. Pharmaceutics 13. https://doi.org/10.3390/pharmaceutics13091408. and morphology control of mesoporous silicas via a co-condensation synthesis
Ferreira, N.H., Ribeiro, A.B., Rinaldi-Neto, F., Fernandes, F.S., do Nascimento, S., method. Chem. Mater. 15, 4247–4256. https://doi.org/10.1021/cm0210041.
Braz, W.R., Nassar, E.J., Tavares, D.C., 2020. Anti-melanoma activity of Iannuccelli, V., Bertelli, D., Romagnoli, M., Scalia, S., Maretti, E., Sacchetti, F., Leo, E.,
indomethacin incorporated into mesoporous silica nanoparticles. Pharm. Res. 37, 2014. In vivo penetration of bare and lipid-coated silica nanoparticles across the
1–11. https://doi.org/10.1007/s11095-020-02903-y. human stratum corneum. Colloids Surf. B Biointerfaces 122, 653–661. https://doi.
Foldvari, M., 2000. Non-invasive administration of drugs through the skin: Challenges in org/10.1016/j.colsurfb.2014.07.046.
delivery system design. Pharm. Sci. Technol. Today 3, 417–425. https://doi.org/ Idris, A., Man, Z., Maulud, A.S., Bustam, M.A., Mannan, H.A., Ahmed, I., 2020.
10.1016/S1461-5347(00)00317-5. Investigation on particle properties and extent of functionalization of silica
Food & Drug Administration, 2020. CFR—Code of Federal Regulations Title 21 [WWW nanoparticles. Appl. Surf. Sci. 506, 144978 https://doi.org/10.1016/j.
Document]. Dep. Heal. Hum. Serv. URL https://www.accessdata.fda.gov/scripts/cd apsusc.2019.144978.
rh/cfdocs/cfcfr/CFRSearch.cfm?fr=182.1711&SearchTerm=silica (acessado Jadhav, S.A., Scalarone, D., Brunella, V., Ugazio, E., Sapino, S., Berlier, G., 2017.
11.14.21). Thermoresponsive copolymer-grafted SBA-15 porous silica particles for temperature-
Fytianos, G., Rahdar, A., Kyzas, G.Z., 2020. Nanomaterials in cosmetics: recent updates. triggered topical delivery systems. Express Polym. Lett. 11, 96–105. https://doi.org/
Nanomaterials 10, 979. https://doi.org/10.3390/NANO10050979. 10.3144/expresspolymlett.2017.11.
Garcés, M., Cáceres, L., Chiappetta, D., Magnani, N., Evelson, P., 2021. Current Jeelani, P.G., Mulay, P., Venkat, R., Ramalingam, C., 2020. Multifaceted application of
understanding of nanoparticle toxicity mechanisms and interactions with biological silica nanoparticles. A review. Silicon 12, 1337–1354. https://doi.org/10.1007/
systems. New J. Chem. 45, 14328–14344. https://doi.org/10.1039/d1nj01415c. s12633-019-00229-y.
Ghasemi, S., Farsangi, Z.J., Beitollahi, A., Mirkazemi, M., Rezayat, S.M., Sarkar, S., 2017. Jepps, O.G., Dancik, Y., Anissimov, Y.G., Roberts, M.S., 2013. Modeling the human skin
Synthesis of hollow mesoporous silica (HMS) nanoparticles as a candidate for barrier - Towards a better understanding of dermal absorption. Adv. Drug Deliv. Rev.
sulfasalazine drug loading. Ceram. Int. 43, 11225–11232. https://doi.org/10.1016/ 65, 152–168. https://doi.org/10.1016/j.addr.2012.04.003.
j.ceramint.2017.05.172.

19
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Jung, E., Hui, X., Zhu, H., Zhang, A., Wang, W., Buchholz, B., Maibach, H., 2019. Effect adhesives for rapid wound closure. Int. J. Nanomedicine 13, 5849–5863. https://doi.
of iron and silica nanoparticles’ size on in vitro human skin binding and penetration. org/10.2147/IJN.S177109.
Toxicol. Res. Appl. 3, 1–10. https://doi.org/10.1177/2397847319893054. Malachin, G., Lubian, E., Mancin, F., Papini, E., Tavano, R., 2017. Combined action of
Karimi, M., Mirshekari, H., Aliakbari, M., Sahandi-Zangabad, P., Hamblin, M.R., 2016. human commensal bacteria and amorphous silica nanoparticles on the viability and
Smart mesoporous silica nanoparticles for controlled-release drug delivery. immune responses of dendritic cells. Clin. Vaccine Immunol. 24, 1–19. https://doi.
Nanotechnol. Rev. 5, 195–207. https://doi.org/10.1515/ntrev-2015-0057. org/10.1128/CVI.00178-17.
Khavkin, J., Ellis, D.A.F., 2011. Aging skin: histology, physiology, and pathology. Facial Manzano, M., Vallet-Regí, M., 2020. Mesoporous Silica Nanoparticles for Drug Delivery.
Plast. Surg. Clin. North Am. 19, 229–234. https://doi.org/10.1016/j. Adv. Funct. Mater. 30, 1902634. https://doi.org/10.1002/adfm.201902634.
fsc.2011.04.003. Martínez-Carmona, M., Lozano, D., Baeza, A., Colilla, M., Vallet-Regí, M., 2017. A novel
Kim, H.M., Lee, C.H., Kim, B., 2016. Sonochemical synthesis of silica particles and their visible light responsive nanosystem for cancer treatment. Nanoscale 9,
size control. Appl. Surf. Sci. 380, 305–308. https://doi.org/10.1016/J. 15967–15973. https://doi.org/10.1039/c7nr05050j.
APSUSC.2015.12.048. Masjedi-Arani, M., Salavati-Niasari, M., Ghanbari, D., Nabiyouni, G., 2014.
Knežević, N., Ilić, N., Dokić, V., Petrović, R., Janaćković, D., 2018. Mesoporous silica and A sonochemical-assisted synthesis of spherical silica nanostructures by using a new
organosilica nanomaterials as UV-blocking agents. ACS Appl. Mater. Interfaces 10, capping agent. Ceram. Int. 40, 495–499. https://doi.org/10.1016/J.
20231–20236. https://doi.org/10.1021/acsami.8b04635. CERAMINT.2013.06.029.
Knorr, F., Patzelt, A., Meinke, M.C., Vogt, A., Blume-Peytavi, U., Rühl, E., Lademann, J., Masood, A., Maheen, S., Khan, H.U., Shafqat, S.S., Irshad, M., Aslam, I., Rasul, A.,
2019. Interactions of nanoparticles with skin. In: NanoScience and Technology. Bashir, S., Zafar, M.N., 2021. Pharmaco-technical evaluation of statistically
Springer Verlag, pp. 329–339. https://doi.org/10.1007/978-3-030-12461-8_13. formulated and optimized dual drug-loaded silica nanoparticles for improved
Kolarsick, P.A.J., Kolarsick, M.A., Goodwin, C., 2011. Anatomy and physiology of the antifungal efficacy and wound healing. ACS Omega. https://doi.org/10.1021/
skin. J. Dermatol. Nurses. Assoc. 3, 203–213. https://doi.org/10.1097/ acsomega.0c06242.
JDN.0b013e3182274a98. Matsuo, K., Hirobe, S., Okada, N., Nakagawa, S., 2016. Analysis of skin permeability and
Koppes, S.A., Ljubojević Hadžavdić, S., Jakasa, I., Franceschi, N., Riethmüller, C., Jurakić toxicological properties of amorphous silica particles. Biol. Pharm. Bull. 39,
Tončic, R., Marinovic, B., Raj, N., Rawlings, A.V., Voegeli, R., Lane, M.E., Haftek, M., 1201–1205. https://doi.org/10.1248/bpb.b16-00258.
Frings-Dresen, M.H.W., Rustemeyer, T., Kezic, S., 2017. Effect of allergens and Mebert, A.M., Alvarez, G.S., Peroni, R., Illoul, C., Hélary, C., Coradin, T., Desimone, M.F.,
irritants on levels of natural moisturizing factor and corneocyte morphology. Contact 2018. Collagen-silica nanocomposites as dermal dressings preventing infection in
Dermatitis 76, 287–295. https://doi.org/10.1111/cod.12770. vivo. Mater. Sci. Eng. C 93, 170–177. https://doi.org/10.1016/j.msec.2018.07.078.
Kumar, S., Malik, M.M., Purohit, R., 2017. Synthesis methods of mesoporous silica Mebert, A.M., Baglole, C.J., Desimone, M.F., Maysinger, D., 2017. Nanoengineered silica:
materials. Mater. Today Proc. 4, 350–357. https://doi.org/10.1016/J. properties, applications and toxicity. Food Chem. Toxicol. 109, 753–770. https://
MATPR.2017.01.032. doi.org/10.1016/j.fct.2017.05.054.
Kwatra, B., 2020. Collagen supplementation: therapy for skin disorders: a review. World Medi, B.M., Layek, B., Singh, J., 2017. Electroporation for dermal and transdermal drug
J. Pharm. Res. 9, 2504–2518. https://doi.org/10.20959/wjpr20205-17513. delivery. In: Percutaneous Penetration Enhancers Physical Methods in Penetration
Lane, M.E., 2011. Nanoparticles and the skin applications and limitations. Enhancement. Springer Berlin Heidelberg, pp. 105–122. https://doi.org/10.1007/
J. Microencapsul. 28, 709–716. https://doi.org/10.3109/02652048.2011.599440. 978-3-662-53273-7_7.
Lane, M.E., Santos, P., Watkinson, A.C., Hadgraft, J., 2012. Passive Skin Permeation Mirzaei, M., Zarch, M.B., Darroudi, M., Sayyadi, K., Keshavarz, S.T., Sayyadi, J.,
Enhancement, in: Transdermal and Topical Drug Delivery: Principles and Practice. Fallah, A., Maleki, H., 2020. Silica mesoporous structures: effective nanocarriers in
John Wiley & Sons, Ltd, pp. 23–42. https://doi.org/10.1002/9781118140505.CH2. drug delivery and nanocatalysts. Appl. Sci. 10, 7533. https://doi.org/10.3390/
Lemos, C.N., Pereira, F., Dalmolin, L.F., Cubayachi, C., Ramos, D.N., Lopez, R.F.V., 2018. app10217533.
Nanoparticles influence in skin penetration of drugs: In vitro and in vivo Mirzahosseinipour, M., Khorsandi, K., Hosseinzadeh, R., Ghazaeian, M., Shahidi, F.K.,
characterization. In: Nanostructures for the Engineering of Cells, Tissues and Organs: 2020. Antimicrobial photodynamic and wound healing activity of curcumin
From Design to Applications. Elsevier, pp. 187–248. https://doi.org/10.10 encapsulated in silica nanoparticles. Photodiagnosis Photodyn. Ther. 29, 101639
16/B978-0-12-813665-2.00006-5. https://doi.org/10.1016/j.pdpdt.2019.101639.
Li, M., Cheng, F., Xue, C., Wang, H., Chen, C., Du, Q., Ge, D., Sun, B., 2019. Surface Mo, C., Lu, L., Liu, D., Wei, K., 2020. Development of erianin-loaded dendritic
modification of Stöber silica nanoparticles with controlled moiety densities mesoporous silica nanospheres with pro-apoptotic effects and enhanced topical
determines their cytotoxicity profiles in macrophages. Langmuir 35, 14688–14695. delivery. J. Nanobiotechnology 18, 55. https://doi.org/10.1186/s12951-020-00608-
https://doi.org/10.1021/acs.langmuir.9b02578. 3.
Li, S., Wang, F., Yang, Z., Xu, J., Liu, H., Zhang, L., Xu, W., 2020. Emulsifying Mo, C., Shetti, D., Wei, K., 2019. Erianin inhibits proliferation and induces apoptosis of
performance of near-infrared light responsive polydopamine-based silica particles to HaCaT cells via ROS-mediated JNK/c-Jun and AKT/mTOR signaling pathways.
control drug release. Powder Technol. 359, 17–26. https://doi.org/10.1016/j. Molecules 24, 2727. https://doi.org/10.3390/molecules24152727.
powtec.2019.09.064. Mohamed, A.L., Elmotasem, H., Salama, A.A.A., 2020. Colchicine mesoporous silica
Liang, C.L., Xiang, Q., Cui, W.M., Fang, J., Sun, N.N., Zhang, X.P., Li, Y.N., Yang, H., nanoparticles/hydrogel composite loaded cotton patches as a new encapsulator
Yu, Z., Jia, X.D., 2018. Subchronic oral toxicity of silica nanoparticles and silica system for transdermal osteoarthritis management. Int. J. Biol. Macromol. 164,
microparticles in rats. Biomed. Environ. Sci. 31, 197–207. https://doi.org/10.3967/ 1149–1163. https://doi.org/10.1016/j.ijbiomac.2020.07.133.
bes2018.025. Mohammadpour, R., Cheney, D.L., Grunberger, J.W., Yazdimamaghani, M.,
Liberman, A., Mendez, N., Trogler, W.C., Kummel, A.C., 2014. Synthesis and surface Jedrzkiewicz, J., Isaacson, K.J., Dobrovolskaia, M.A., Ghandehari, H., 2020. One-
functionalization of silica nanoparticles for nanomedicine. Surf. Sci. Rep. 69, year chronic toxicity evaluation of single dose intravenously administered silica
132–158. https://doi.org/10.1016/j.surfrep.2014.07.001. nanoparticles in mice and their Ex vivo human hemocompatibility. J. Control.
Lieckfeldt, R., Villalaín, J., Gómez-Fernández, J.C., Lee, G., 1995. Apparent pKa of the Release 324, 471–481. https://doi.org/10.1016/j.jconrel.2020.05.027.
fatty acids within ordered mixtures of model human stratum corneum lipids. Pharm. Mohammadpour, R., Yazdimamaghani, M., Cheney, D.L., Jedrzkiewicz, J.,
Res. https://doi.org/10.1023/A:1016280714593. Ghandehari, H., 2019. Subchronic toxicity of silica nanoparticles as a function of size
Lin, Y.C., Lin, C.F., Alalaiwe, A., Wang, P.W., Fang, Y.P., Fang, J.Y., 2018a. UV filter and porosity. J. Control. Release 304, 216–232. https://doi.org/10.1016/j.
entrapment in mesoporous silica hydrogel for skin protection against UVA with jconrel.2019.04.041.
minimization of percutaneous absorption. Eur. J. Pharm. Sci. 122, 185–194. https:// Möller, K., Bein, T., 2017. Talented mesoporous silica nanoparticles. Chem. Mater. 29,
doi.org/10.1016/j.ejps.2018.07.013. 371–388. https://doi.org/10.1021/acs.chemmater.6b03629.
Lin, Z., Wu, Y., Bi, Y., 2018b. Rapid synthesis of SiO 2 by ultrasonic-assisted Stober Montazeri, M., Razzaghi-Abyaneh, M., Nasrollahi, S.A., Maibach, H., Nafisi, S., 2019.
method as controlled and pH-sensitive drug delivery. J. Nanoparticle Res. 20, 1–13. Enhanced topical econazole antifungal efficacy by amine-functionalized silica
https://doi.org/10.1007/S11051-018-4411-3. nanoparticles. Bull. Mater. Sci. 43, 1–9. https://doi.org/10.1007/S12034-019-1974-
Lio, D.C.S., Liu, C., Oo, M.M.S., Wiraja, C., Teo, M.H.Y., Zheng, M., Chew, S.W.T., 2.
Wang, X., Xu, C., 2019. Transdermal delivery of small interfering RNAs with Moorthy, M.S., Hoang, G., Manivasagan, P., Mondal, S., Vy Phan, T.T., Kim, H., Oh, J.,
topically applied mesoporous silica nanoparticles for facile skin cancer treatment. 2019. Chitosan oligosaccharide coated mesoporous silica nanoparticles for pH-
Nanoscale 11, 17041–17051. https://doi.org/10.1039/c9nr06303j. stimuli responsive drug delivery applications. J. Porous Mater. 26, 217–226. https://
Liu, J., Zhang, B., Luo, Z., Ding, X., Li, J., Dai, L., Zhou, J., Zhao, X., Ye, J., Cai, K., 2015. doi.org/10.1007/s10934-018-0646-8.
Enzyme responsive mesoporous silica nanoparticles for targeted tumor therapy in Morry, J., Ngamcherdtrakul, W., Gu, S., Goodyear, S.M., Castro, D.J., Reda, M.M.,
vitro and in vivo. Nanoscale 7, 3614–3626. https://doi.org/10.1039/c5nr00072f. Sangvanich, T., Yantasee, W., 2015. Dermal delivery of HSP47 siRNA with NOX4-
Loiola, L.M.D., Batista, M., Capeletti, L.B., Mondo, G.B., Rosa, R.S.M., Marques, R.E., modulating mesoporous silica-based nanoparticles for treating fibrosis. Biomaterials
Bajgelman, M.C., Cardoso, M.B., 2019. Shielding and stealth effects of zwitterion 66, 41–52. https://doi.org/10.1016/j.biomaterials.2015.07.005.
moieties in double-functionalized silica nanoparticles. J. Colloid Interface Sci. 553, Murugadoss, S., Lison, D., Godderis, L., Van Den Brule, S., Mast, J., Brassinne, F.,
540–548. https://doi.org/10.1016/j.jcis.2019.06.044. Sebaihi, N., Hoet, P.H., 2017. Toxicology of silica nanoparticles: an update. Arch.
López-Sánchez, L., Miralles, P., Salvador, A., Merino-Sanjuán, M., Merino, V., 2021. In Toxicol. 91, 2967–3010. https://doi.org/10.1007/s00204-017-1993-y.
vitro skin penetration of bronidox, bronopol and formaldehyde from cosmetics. Musazzi, U.M., Santini, B., Selmin, F., Marini, V., Corsi, F., Allevi, R., Ferretti, A.M.,
Regul. Toxicol. Pharmacol. 122 https://doi.org/10.1016/j.yrtph.2021.104888. Prosperi, D., Cilurzo, F., Colombo, M., Minghetti, P., 2017. Impact of semi-solid
Lotti, T., Bianchi, B., Ghersetich, I., Brazzini, B., Hercogova, J., 2002. Can the brain formulations on skin penetration of iron oxide nanoparticles. J. Nanobiotechnology
inhibit inflammation generated in the skin? The lesson of α-melanocyte-stimulating 15, 1–10. https://doi.org/10.1186/s12951-017-0249-6.
hormone. Int. J. Dermatol. 41, 311–318. https://doi.org/10.1046/j.1365- Nafisi, S., Samadi, N., Houshiar, M., Maibach, H.I., 2018. Mesoporous silica
4362.2002.01408.x. nanoparticles for enhanced lidocaine skin delivery. Int. J. Pharm. 550, 325–332.
Lu, M.M., Bai, J., Shao, D., Qiu, J., Li, M., Zheng, X., Xiao, Y., Wang, Z., Chang, Z.M., https://doi.org/10.1016/j.ijpharm.2018.08.004.
Chen, L., Dong, W.F., Tang, C.B., 2018. Antibacterial and biodegradable tissue nano-

20
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Nafisi, S., Schäfer-Korting, M., Maibach, H.I., 2017. Measuring silica nanoparticles in the in vitro studies. Eur. J. Pharm. Biopharm. 89, 116–125. https://doi.org/10.1016/j.
skin. In: Agache’s Measuring the Skin: Non-invasive Investigations, Physiology, ejpb.2014.11.022.
Normal Constants, Second Edition. Springer International Publishing, Schneider-Rauber, G., Argenta, D.F., Caon, T., 2020. Emerging technologies to target
pp. 1141–1164. https://doi.org/10.1007/978-3-319-32383-1_44. drug delivery to the skin – the role of crystals and carrier-based systems in the case
Nandiyanto, A.B.D., Rahman, T., Fadhlulloh, M.A., Abdullah, A.G., Hamidah, I., study of dapsone. Pharm. Res. 37, 1–21. https://doi.org/10.1007/s11095-020-
Mulyanti, B., 2016. Synthesis of silica particles from rice straw waste using a simple 02951-4.
extraction method. IOP Conf. Ser. Mater. Sci. Eng. 128, 012040 https://doi.org/ Scientific Committee on Consumer Safety, 2015. Safety Silica, Hydrated Silica, and Silica
10.1088/1757-899X/128/1/012040. Surface Modified with Alkyl Silylates (nano form). 10.2772/52532.
Narayan, R., Nayak, U.Y., Raichur, A.M., Garg, S., 2018. Mesoporous silica nanoparticles: Selvarajan, V., Obuobi, S., Ee, P.L.R., 2020. Silica Nanoparticles—A Versatile Tool for the
A comprehensive review on synthesis and recent advances. Pharmaceutics 10, 118. Treatment of Bacterial Infections. Front. Chem. 602. https://doi.org/10.3389/
https://doi.org/10.3390/pharmaceutics10030118. FCHEM.2020.00602.
Nigro, A., Pellegrino, M., Greco, M., Comandè, A., Sisci, D., Pasqua, L., Leggio, A., Sheshala, R., Anuar, N.K., Abu Samah, N.H., Wong, T.W., 2019. In vitro drug dissolution/
Morelli, C., 2018. Dealing with skin and blood-brain barriers: The unconventional permeation testing of nanocarriers for skin application: a comprehensive review.
challenges of mesoporous silica nanoparticles. Pharmaceutics 10. https://doi.org/ AAPS PharmSciTech 20. https://doi.org/10.1208/s12249-019-1362-7.
10.3390/pharmaceutics10040250. Shin, J.H., Jeon, K.S., Kim, J.K., Kim, Y., Jo, M.S., Lee, J.S., Baek, J.E., Park, H.S., An, H.
Ostrowski, A., Nordmeyer, D., Boreham, A., Brodwolf, R., Mundhenk, L., Fluhr, J.W., J., Park, J.D., Ahn, K., Oh, S.M., Yu, I.J., 2017. Subacute inhalation toxicity study of
Lademann, J., Graf, C., Rühl, E., Alexiev, U., Gruber, A.D., 2014a. Skin barrier synthetic amorphous silica nanoparticles in Sprague-Dawley rats. Inhal. Toxicol. 29,
disruptions in tape stripped and allergic dermatitis models have no effect on dermal 567–576. https://doi.org/10.1080/08958378.2018.1426661.
penetration and systemic distribution of AHAPS-functionalized silica nanoparticles. Shrivastava, R., Kushwaha, P., Bhutia, Y.C., Flora, S.J.S., 2016. Oxidative stress following
Nanomedicine Nanotechnology Biol. Med. 10, 1571–1581. https://doi.org/10.1016/ exposure to silver and gold nanoparticles in mice. Toxicol. Ind. Health 32,
j.nano.2014.04.004. 1391–1404. https://doi.org/10.1177/0748233714562623.
Ostrowski, A., Nordmeyer, D., Mundhenk, L., Fluhr, J.W., Lademann, J., Graf, C., Simpson, C.L., Patel, D.M., Green, K.J., 2011. Deconstructing the skin: cytoarchitectural
Rühl, E., Gruber, A.D., 2014b. AHAPS-functionalized silica nanoparticles do not determinants of epidermal morphogenesis. Nat. Rev. Mol. Cell Biol. 12, 565–580.
modulate allergic contact dermatitis in mice. Nanoscale Res. Lett. 9, 1–7. https:// https://doi.org/10.1038/nrm3175.
doi.org/10.1186/1556-276X-9-524. Sng, M.K., Chan, J.S.K., Teo, Z., Phua, T., Tan, E.H.P., Wee, J.W.K., Koh, N.J.N., Tan, C.
Pan, Z., Zhang, K.R., Gao, H.L., Zhou, Y., Yan, B.B., Yang, C., Zhang, Z., Dong, L., Chen, S. K., Chen, J.P., Pal, M., Tong, B.M.K., Tnay, Y.L., Ng, X.R., Zhu, P., Chiba, S.,
M., Xu, R., Zou, D.H., Yu, S.H., 2020. Activating proper inflammation for wound- Wang, X., Wahli, W., Tan, N.S., 2018. Selective deletion of PPARβ/δ in fibroblasts
healing acceleration via mesoporous silica nanoparticle tissue adhesive. Nano Res. causes dermal fibrosis by attenuated LRG1 expression. Cell Discov. 4, 15. https://
13, 373–379. https://doi.org/10.1007/s12274-020-2619-x. doi.org/10.1038/s41421-018-0014-5.
Paris, J.L., Cabanas, M.V., Manzano, M., Vallet-Regí, M., 2015. Polymer-grafted Souris, J.S., Chen, N.T., Cheng, S.H., Chen, C.T., Lo, L.W., 2014. Silica Nanoparticle
mesoporous silica nanoparticles as ultrasound-responsive drug carriers. ACS Nano 9, Platform. In: Cancer Theranostics. Elsevier Inc., pp. 363–391. https://doi.org/10.10
11023–11033. https://doi.org/10.1021/acsnano.5b04378. 16/B978-0-12-407722-5.00020-7.
Peres, E.C., Slaviero, J.C., Cunha, A.M., Hosseini-Bandegharaei, A., Dotto, G.L., 2018. Steinmetz, F.P., Wakefield, J.C., Boughton, R.M., 2021. Fractions of concern: Challenges
Microwave synthesis of silica nanoparticles and its application for methylene blue and strategies for the safety assessment of biological matter in cosmetics. Cosmetics
adsorption. J. Environ. Chem. Eng. 6, 649–659. https://doi.org/10.1016/J. 8, 4–9. https://doi.org/10.3390/cosmetics8020034.
JECE.2017.12.062. Stöber, W., Fink, A., Bohn, E., 1968. Controlled growth of monodisperse silica spheres in
Prakash, C., Bhargave, P., Tiwari, S., Majumdar, B., Bhargava, R.K., 2017. Skin surface the micron size range. J. Colloid Interface Sci. 26, 62–69. https://doi.org/10.1016/
pH in acne vulgaris: Insights from an Observational Study and Review of the 0021-9797(68)90272-5.
Literature. J. Clin. Aesthet. Dermatol. 10, 33–39. Sun, J.T., Yu, Z.Q., Hong, C.Y., Pan, C.Y., 2012a. Biocompatible zwitterionic sulfobetaine
Proksch, E., 2018. pH in nature, humans and skin. J. Dermatol. 45, 1044–1052. https:// copolymer-coated mesoporous silica nanoparticles for temperature-responsive drug
doi.org/10.1111/1346-8138.14489. release. Macromol. Rapid Commun. 33, 811–818. https://doi.org/10.1002/
Quignard, S., Coradin, T., Powell, J.J., Jugdaohsingh, R., 2017. Silica nanoparticles as marc.201100876.
sources of silicic acid favoring wound healing in vitro. Colloids Surf. B Biointerfaces Sun, L., Zhang, X., Zheng, C., Wu, Z., Li, C., 2013. A pH gated, glucose-sensitive
155, 530–537. https://doi.org/10.1016/j.colsurfb.2017.04.049. nanoparticle based on worm-like mesoporous silica for controlled insulin release.
Rahman, I.A., Padavettan, V., 2012. Synthesis of silica nanoparticles by sol-gel: size- J. Phys. Chem. B 117, 3852–3860. https://doi.org/10.1021/jp400442x.
dependent properties, surface modification, and applications in silica-polymer Sun, M., Su, X., Ding, B., He, X., Liu, X., Yu, A., Lou, H., Zhai, G., 2012b. Advances in
nanocomposites-A review. J. Nanomater. 2012, 1–15. https://doi.org/10.1155/ nanotechnology-based delivery systems for curcumin. Nanomedicine 7, 1085–1100.
2012/132424. https://doi.org/10.2217/nnm.12.80.
Rancan, F., Gao, Q., Graf, C., Troppens, S., Hadam, S., Hackbarth, S., Kembuan, C., Tada, D.B., Suraniti, E., Rossi, L.M., Leite, C.A.P., Oliveira, C.S., Tumolo, T.C.,
Blume-Peytavi, U., Rühl, E., Lademann, J., Vogt, A., 2012. Skin penetration and Calemczuk, R., Livache, T., Baptista, M.S., 2014. Effect of lipid coating on the
cellular uptake of amorphous silica nanoparticles with variable size, surface interaction between silica nanoparticles and membranes. J. Biomed. Nanotechnol.
functionalization, and colloidal stability. ACS Nano 6, 6829–6842. https://doi.org/ 10, 519–528. https://doi.org/10.1166/jbn.2014.1723.
10.1021/nn301622h. Talarska, P., Boruczkowski, M., Żurawski, J., 2021. Current knowledge of silver and gold
Rawat, N., Sandhya, Subaharan, K., Eswaramoorthy, M., Kaul, G., 2017. Comparative in nanoparticles in laboratory research—application, toxicity, cellular uptake.
vivo toxicity assessment places multiwalled carbon nanotubes at a higher level than Nanomaterials 11. https://doi.org/10.3390/nano11092454.
mesoporous silica nanoparticles. Toxicol. Ind. Health 33, 182–192. https://doi.org/1 Tang, F., Li, L., Chen, D., 2012. Mesoporous silica nanoparticles: synthesis,
0.1177/0748233715622307. biocompatibility and drug delivery. Adv. Mater. 24, 1504–1534. https://doi.org/
Rawlings, A.V., Harding, C.R., 2004. Moisturization and skin barrier function. Dermatol. 10.1002/adma.201104763.
Ther. 17, 43–48. https://doi.org/10.1111/j.1396-0296.2004.04s1005.x. Tang, L., Cheng, J., 2013. Nonporous silica nanoparticles for nanomedicine application.
Ren, G., Su, H., Wang, S., 2020. The combined method to synthesis silica nanoparticle by Nano Today 8, 290. https://doi.org/10.1016/J.NANTOD.2013.04.007.
Stöber process. J. Sol-Gel Sci. Technol. 96, 108–120. https://doi.org/10.1007/ Tao, Y., Chan, H.F., Shi, B., Li, M., Leong, K.W., 2020. Light: A magical tool for controlled
S10971-020-05322-Y. drug delivery. Adv. Funct. Mater. 30, 2005029. https://doi.org/10.1002/
Rezaeian, M., Afjoul, H., Shamloo, A., Maleki, A., Afjoul, N., 2021. Green synthesis of adfm.202005029.
silica nanoparticles from olive residue and investigation of their anticancer Teixeira, M.C., Carbone, C., Souto, E.B., 2017. Beyond liposomes: Recent advances on
potential. Nanomedicine (Lond). 16, 1581–1593. https://doi.org/10.2217/NNM- lipid based nanostructures for poorly soluble/poorly permeable drug delivery. Prog.
2021-0040. Lipid Res. 68, 1–11. https://doi.org/10.1016/j.plipres.2017.07.001.
Ricci, V., Zonari, D., Cannito, S., Marengo, A., Scupoli, M.T., Malatesta, M., Carton, F., Tu, J., Du, G., Reza Nejadnik, M., Mönkäre, J., van der Maaden, K., Bomans, P.H.H.,
Boschi, F., Berlier, G., Arpicco, S., 2018. Hyaluronated mesoporous silica Sommerdijk, N.A.J.M., Slütter, B., Jiskoot, W., Bouwstra, J.A., Kros, A., 2017.
nanoparticles for active targeting: influence of conjugation method and hyaluronic Mesoporous silica nanoparticle-coated microneedle arrays for intradermal antigen
acid molecular weight on the nanovector properties. J. Colloid Interface Sci. 516, delivery. Pharm. Res. 34, 1693–1706. https://doi.org/10.1007/s11095-017-2177-4.
484–497. https://doi.org/10.1016/j.jcis.2018.01.072. Ugazio, E., Gastaldi, L., Brunella, V., Scalarone, D., Jadhav, S.A., Oliaro-Bosso, S.,
Rosenholm, J.M., Sahlgren, C., Lindén, M., 2010. Towards multifunctional, targeted drug Zonari, D., Berlier, G., Miletto, I., Sapino, S., 2016. Thermoresponsive mesoporous
delivery systems using mesoporous silica nanoparticles - Opportunities & challenges. silica nanoparticles as a carrier for skin delivery of quercetin. Int. J. Pharm. 511,
Nanoscale. https://doi.org/10.1039/c0nr00156b. 446–454. https://doi.org/10.1016/j.ijpharm.2016.07.024.
Ryu, H.J., Seong, N.W., So, B.J., Seo, H.S., Kim, J.H., Hong, J.S., Park, M.K., Kim, M.S., Unger, K., Rupprecht, H., Valentin, B., Kircher, W., 1983. The use of porous and surface
Kim, Y.R., Cho, K.B., Seo, M.Y., Kim, M.K., Maeng, E.H., Son, S.W., 2014. Evaluation modified silicas as drug delivery and stabilizing agents. Drug Dev. Ind. Pharm. 9,
of silica nanoparticle toxicity after topical exposure for 90 days. Int. J. Nanomedicine 69–91. https://doi.org/10.3109/03639048309048546.
9, 127–136. https://doi.org/10.2147/IJN.S57929. Vallet-Regí, M., 2014. Mesoporous silica nanoparticles: their projection in nanomedicine.
Salvioni, L., Morelli, L., Ochoa, E., Labra, M., Fiandra, L., Palugan, L., Prosperi, D., Int. Sch. Res. Not. 2012, 1–20. https://doi.org/10.5402/2012/608548.
Colombo, M., 2021. The emerging role of nanotechnology in skincare. Adv. Colloid Vallet-Regí, M., Colilla, M., Izquierdo-Barba, I., Manzano, M., 2018. Mesoporous silica
Interface Sci. 293, 102437 https://doi.org/10.1016/j.cis.2021.102437. nanoparticles for drug delivery: current insights. Molecules 23, 47. https://doi.org/
Sapino, S., Oliaro-Bosso, S., Zonari, D., Zattoni, A., Ugazio, E., 2017. Mesoporous silica 10.3390/molecules23010047.
nanoparticles as a promising skin delivery system for methotrexate. Int. J. Pharm. Vallet-Regi, M., Rámila, A., Del Real, R.P., Pérez-Pariente, J., 2001. A new property of
530, 239–248. https://doi.org/10.1016/j.ijpharm.2017.07.058. MCM-41: drug delivery system. Chem. Mater. 13, 308–311. https://doi.org/
Sapino, S., Ugazio, E., Gastaldi, L., Miletto, I., Berlier, G., Zonari, D., Oliaro-Bosso, S., 10.1021/cm0011559.
2015. Mesoporous silica as topical nanocarriers for quercetin: Characterization and

21
R.P. Morais et al. International Journal of Pharmaceutics 614 (2022) 121439

Verma, P., Pathak, K., 2012. Nanosized ethanolic vesicles loaded with econazole nitrate Synthesized Silane-Borate Precursor. ChemMedChem 14, 1079–1085. https://doi.
for the treatment of deep fungal infections through topical gel formulation. org/10.1002/cmdc.201900142.
Nanomedicine Nanotechnology Biol. Med. 8, 489–496. https://doi.org/10.1016/j. Yan, T., He, J., Liu, R., Liu, Z., Cheng, J., 2020. Chitosan capped pH-responsive hollow
nano.2011.07.004. mesoporous silica nanoparticles for targeted chemo-photo combination therapy.
Videira, I.F.S., Magina, S., Moura, D.F.L., 2013. Mechanisms regulating melanogenesis. Carbohydr. Polym. 231, 115706 https://doi.org/10.1016/j.carbpol.2019.115706.
An. Bras. Dermatol. 88, 76–83. https://doi.org/10.1007/978-3-642-45407-3_24. Yilmaz, E., Borchert, H.H., 2006. Effect of lipid-containing, positively charged
Wang, D.P., Wang, Z.J., Zhao, R., Lin, C.X., Sun, Q.Y., Yan, C.P., Zhou, X., Cao, J.M., nanoemulsions on skin hydration, elasticity and erythema - An in vivo study. Int. J.
2020a. Silica nanomaterials induce organ injuries by Ca2+-ROS-initiated disruption Pharm. 307, 232–238. https://doi.org/10.1016/j.ijpharm.2005.10.002.
of the endothelial barrier and triggering intravascular coagulation. Part. Fibre Yoo, H., Pak, J., 2013. Synthesis of highly fluorescent silica nanoparticles in a reverse
Toxicol. 17, 1–21. https://doi.org/10.1186/s12989-020-00340-8. microemulsion through double-layered doping of organic fluorophores.
Wang, W.H., Liang, H.T., Yang-Wang, Y.T., Shih, C.J., 2020b. Synthesis of hierarchically J. Nanoparticle Res. 15, 1–10. https://doi.org/10.1007/s11051-013-1609-2.
mesoporous silica with encapsulated avobenzone as a UV protection filter. RSC Adv. Yoo, J., Kim, H., Chang, H., Park, W., Hahn, S.K., Kwon, W., 2020. Biocompatible
10, 15846–15852. https://doi.org/10.1039/d0ra01837f. organosilica nanoparticles with self-encapsulated phenyl motifs for effective UV
Wani, T.U., Mohi-ud-Din, R., Majeed, A., Kawoosa, S., Pottoo, F.H., 2020. Skin protection. ACS Appl. Mater. Interfaces 12, 9062–9069. https://doi.org/10.1021/
Permeation of Nanoparticles: Mechanisms Involved and Critical Factors Governing acsami.9b21990.
Topical Drug Delivery. Curr. Pharm. Des. 26, 4601–4614. https://doi.org/10.2174/ Yosipovitch, G., Xiong, G.L., Haus, E., Sackett-Lundeen, L., Ashkenazi, I., Maibach, H.I.,
1381612826666200701204010. 1998. Time-dependent variations of the skin barrier function in humans:
Wills, J.W., Hondow, N., Thomas, A.D., Chapman, K.E., Fish, D., Maffeis, T.G., Penny, M. transepidermal water loss, stratum corneum hydration, skin surface pH, and skin
W., Brown, R.A., Jenkins, G.J.S., Brown, A.P., White, P.A., Doak, S.H., 2016. Genetic temperature. J. Invest. Dermatol. 110, 20–23. https://doi.org/10.1046/j.1523-
toxicity assessment of engineered nanoparticles using a 3D in vitro skin model 1747.1998.00069.x.
(EpiDermTM). Part. Fibre Toxicol. 13, 1–21. https://doi.org/10.1186/s12989-016- Zaharudin, N.S., Mohamed Isa, E.D., Ahmad, H., Abdul Rahman, M.B., Jumbri, K., 2020.
0161-5. Functionalized mesoporous silica nanoparticles templated by pyridinium ionic liquid
Wong, R., Geyer, S., Weninger, W., Guimberteau, J.-C., Wong, J.K., 2016. The dynamic for hydrophilic and hydrophobic drug release application. J. Saudi Chem. Soc. 24,
anatomy and patterning of skin. Exp. Dermatol. 25, 92–98. https://doi.org/10.1111/ 289–302. https://doi.org/10.1016/j.jscs.2020.01.003.
exd.12832. Zhang, J., Niemelä, M., Westermarck, J., Rosenholm, J.M., 2014. Mesoporous silica
Wu, Y., Long, Y., Li, Q.L., Han, S., Ma, J., Yang, Y.W., Gao, H., 2015. Layer-by-Layer nanoparticles with redox-responsive surface linkers for charge-reversible loading
(LBL) self-assembled biohybrid nanomaterials for efficient antibacterial applications. and release of short oligonucleotides. Dalt. Trans. 43, 4115–4126. https://doi.org/
ACS Appl. Mater. Interfaces 7, 17255–17263. https://doi.org/10.1021/ 10.1039/c3dt53071j.
acsami.5b04216. Zhao, W., Wang, H.a., Wang, H., Han, Y., Zheng, Z., Liu, X., Feng, B., Zhang, H., 2021.
Xie, M., Xu, Y., Shen, H., Shen, S., Ge, Y., Xie, J., 2014. Negative-charge-functionalized Light-responsive dual-functional biodegradable mesoporous silica nanoparticles
mesoporous silica nanoparticles as drug vehicles targeting hepatocellular carcinoma. with drug delivery and lubrication enhancement for the treatment of osteoarthritis.
Int. J. Pharm. 474, 223–231. https://doi.org/10.1016/j.ijpharm.2014.08.027. Nanoscale. https://doi.org/10.1039/D0NR08887K.
Xu, B., Jiang, G., Yu, W., Liu, D., Zhang, Y., Zhou, J., Sun, S., Liu, Y., 2017. H2O2- Zhu, S., Li, S., Escuin-Ordinas, H., Dimatteo, R., Xi, W., Ribas, A., Segura, T., 2018.
Responsive mesoporous silica nanoparticles integrated with microneedle patches for Accelerated wound healing by injectable star poly(ethylene glycol)-b-poly(propylene
the glucose-monitored transdermal delivery of insulin. J. Mater. Chem. B 5, sulfide) scaffolds loaded with poorly water-soluble drugs. J. Control. Release 282,
8200–8208. https://doi.org/10.1039/c7tb02082a. 156–165. https://doi.org/10.1016/j.jconrel.2018.05.006.
Xu, Q., Jalilian, E., Fakhoury, J.W., Manwar, R., Michniak-Kohn, B., Elkin, K.B., Zielińska, A., Pereira, I., Antunes, S., Veiga, F.J., Santos, A.C., Nowak, I., Silva, A.M.,
Avanaki, K., 2020. Monitoring the topical delivery of ultrasmall gold nanoparticles Souto, E.B., 2018. Mesoporous silica nanoparticles as drug delivery systems against
using optical coherence tomography. Ski. Res. Technol. 26, 263–268. https://doi. melanoma. In: Design of Nanostructures for Theranostics Applications. Elsevier, pp.
org/10.1111/srt.12789. 437–466. https://doi.org/10.1016/B978-0-12-813669-0.00010-5.
Xu, Y., Shi, W., Li, H., Li, X., Ma, H., 2019. H2O2-Responsive Organosilica-Doxorubicin Zsikó, S., Csányi, E., Kovács, A., Budai-Szűcs, M., Gácsi, A., Berkó, S., 2019. Methods to
Nanoparticles for Targeted Imaging and Killing of Cancer Cells Based on a evaluate skin penetration in vitro. Sci. Pharm. 87, 19. https://doi.org/10.3390/
scipharm87030019.

22

You might also like