You are on page 1of 14

Perspective

Therapeutic
Delivery
For reprint orders, please contact: reprints@futuremedicine.com

Clay-based drug-delivery systems: what


does the future hold?

Clays for drug delivery have been used from ancient time due to the large availability Giuseppe Lazzara†,1, Serena
of clay minerals and their unprecedented properties. The empirical use of nanoclays Riela†,2 & Rawil F Fakhrullin*,3
from the past is converted in a stimulating scientific task aimed at building up
1
Dipartimento di Fisica e Chimica,
Università degli Studi di Palermo, Viale
nanoarchitectonic vehicles for drug delivery in a targeted and stimuli-responsive
delle Scienze, pad. 17, 90128 Palermo,
fashion. Here the historical aspects are discussed; next the modern examples of Italy
applications of different clay-based materials are discussed. A special focus is given to 2
Dipartimento STEBICEF, sezione di
halloysite clay nanotubes, which are an emerging and very promising nanomaterial for Chimica, Università degli Studi di
drug-delivery purposes due to its special morphology and unique chemical properties. Palermo, Viale delle Scienze, pad. 17,
90128 Palermo, Italy
Advantages and limitations of these natural nanomaterials are critically discussed 3
Institute of Fundamental Medicine and
pointing out the future perspectives and directions for further research. Biology, Kazan Federal University, Kreml
uramı 18, Kazan, Republic of Tatarstan
Keywords:  drug delivery • halloysite • nanoclay • nanotubes 420008, Russian Federation
*Author for correspondence:
kazanbio@ gmail.com
Nanoclays for medical purposes: d­ifferent m­ inerals for medicinal uses and †
Authors contributed equally
from the past to modern regulations were listed, and this was also the
applications first mineralogical classifications.
Nowadays there is a simultaneous and grow- There are several naturally occurring min-
ing interest in the development of innovative erals with tubular morphologies (chrysotile,
process technologies and new materials that cylindrite, tochilinite, among others) whose
can reduce the environmental impact on the structure, occurrence and properties are
ecosystems  [1–3] . Specifically, the researchers extensively reported.
have focused their attention on renewable From the mineralogical viewpoint, nano-
and nontoxic natural resources to face key clays belong to the family of phyllosilicates
environmental problems such as the accumu- composed of tetrahedral and octahedral
lation of wastes and water pollution. sheets. The tetrahedral sheet consists of sili-
Pharmaceutical technology is not an con–oxygen tetrahedra linked to neighboring
exception and natural source excipients are tetrahedra by sharing three corners, which
recovering positions against the synthetic results in a hexagonal network; the remain-
materials  [4,5] . Due to their large availability, ing fourth oxygen of each tetrahedron also
clay minerals are known for medical appli- belongs to the adjacent octahedral sheet.
cation since a very long time [6] . Mixture The latter is usually composed of aluminum
of clay and iron hydroxides have been used or magnesium in sixfold coordination with
by prehistoric ancestors not only for paint oxygen from the tetrahedral sheet and with
caves but also to cure wounds. The clay use hydroxyl groups. The thickness of each layer
is documented in ancient Egypt as anti- is ca. 1 nm and the lateral dimensions may
inflammatory agents and antiseptics. Later change from tenth of nanometers to several
on, in the middle age, Ibn al-Baitar discussed microns. Several layers may be joined in a
eight kinds of medicinal earth. When Phar- clay crystallite by electrostatic force, Van der
macopoeia appeared in Renaissance, clays Waals force, interlayer cations or by hydrogen
have been classified among other drugs. The bonding. These layers organize themselves to part of

10.4155/tde-2017-0041 © 2017 Future Science Ltd Ther. Deliv. (2017) 8(8), 633–646 ISSN 2041-5990 633
Perspective  Lazzara, Riela & Fakhrullin

form stacks with a regular gap called the interlayer or Clays represents a class of old materials that has
the gallery. The different arrangement of the tetrahedral never been out of attracting scientific interest due to
and octahedral sheets allowed us to classify the clays in the chemical tunability and multiple possible uses in
three categories: 1:1, 2:1 and 2:1:1 phyllosilicates. The medical sciences.
1:1 phyllosilicates, such as kaolinite and halloysite, have
one tetrahedral and one octahedral sheet per clay layer; Nanoclay for adsorption & sustained release
as concerns the 2:1 clay minerals, each layer consists of of drugs
one octahedral sheet sandwiched between the two tet- Since last century, controlled drug-delivery systems
rahedral sheets. Examples are given by montmorillon- (DDS) are a recognized protocol to prepare materi-
ite (MTM), and illite. Finally, the 2:1:1 phyllosilicates, als that can efficiently encapsulate drug molecules,
such as cloisite, are composed of an octahedral sheet and release them at the target site for a defined period
adjacent to a 2:1 layer. Because of the isomorphous sub- of time and in a controlled manner. Usually release
stitutions, the clay may have a charge in the sheets. dosage forms are known to provide an initial burst
Drug molecules are encapsulated in clay minerals to release of the drug, without control upon the release
modify the rate, the time and to target the site of drug rate. In order to reach and maintain the therapeutic
release. Moreover, this strategy can be useful to protect plasmatic concentrations a control in the dosage con-
drugs against aging due to chemical and enzymatic trol is needed. Such a strategy is necessary to avoid
degradation. On this basis, a new concept of ‘excipient’ significant fluctuations in the drug on molecular
is generated for clay minerals as they are not inert fillers levels. Indeed, DDS can reduce the patient expenses
but, instead, they can have a functionality providing as well as the risks of toxicity, while it can improve
targeting release, prevention or reduction of side effects the drug efficacy, specificity, therapeutic index and
and increasing the product shelf-life [7] . tolerability of corresponding drugs [8] . Therefore,
Many polymeric materials have been widely inves- the development of both stimuli-responsive and con-
tigated with the same aim, but frequently their prop- trolled-release systems is crucial for the development
erties need to be improved by addition of inorganic of both clinical medicine and fundamental science
f­illers, as, again, natural mineral clays. (Figure 1) .

Immediate release
Prolonged zero order release
100
Drug release (%)

Time

Adverse effects
Toxic concentration
Therapeutic interval
Plasma concentration

Minimum effective concentration


no effects

Time

Figure 1. Correlation between drug-release profiles and plasma concentration.

634 Ther. Deliv. (2017) 8(8) future science group


Clay-based drug-delivery systems: what does the future hold?  Perspective

Nanomaterials are considered to be important in


refining drug delivery, and they may be pharmaceuti-
Natura
cals as well as diagnostics. In the years several nanoma- l
terials, both natural and synthetic, have been consid-
ered and tested as system for synthetize controlled drug Bio
compa
carrier and delivery. Among the plethora of nanomate- tibility
rials, clay minerals are an important, widely abundant,
low-cost and nontoxic class of materials with unique Increa
se
properties such as intercalation, swelling, ion exchange solubil d
No ity
and adsorption properties [9] . Normally, in the USA, injecta
ble
clay minerals are used as excipients in commercial- High
absorp
ized pharmaceutical products [6,10–12] and indicated No stu tion
die
as ‘Inactive Ingredient Database’ [13] . Clay minerals in hum s
ans
used as ingredients can have some advantages [14–17] . Sustain
ed
releas
For example, MTM was used: by Choy et al. for taste e
masking of sildenafil improving its organoleptic quali-
ties  [18] ; by Choy et al. for both the taste masking and
solubility enhancement of aripiprazole [19] ; and by
Ambrogi et al. for the photostabilization of photolabile
piroxicam [20] . Furthermore, clay minerals are promis-
ing materials that could be employed as new drug car- Figure 2. Comparison between advantages and
rier and delivery systems in fundamental science and disadvantage of clays used as drug carrier.
clinical medicine [21,22] . Indeed, they could be used with respect to the total % on halloysite publications.
to administer drugs to reach the site of action and to Indeed, this interest is confirmed by the highest % of
minimize temporal variation in drug concentration, patent respect to journal publications (Figure 4C), con-
maintaining a constant release, during the treatment firming the interest to use it in industrial scale.
(Figure 2) [23] . The preparation of clay–drug complex is usually
Until the 1965s, Sorby et al. noted that oral admin- carried out mixing, in a proper volume ratio, an aque-
istration of promazine was reduced by co-administra- ous dispersion of clay with a solution (water/organic
tion of attapulgite clay; indeed, it was observed that solvent) of drug. The resulting mixture is allowed to
the presence of the clay slowed the appearance of drug equilibrate for a suitable time under favorable pH con-
in the urine, confirming the interaction between the ditions, usually at room temperature; afterward the
drug and the clay, and they concluded that the adsorp- solid phase is filtered off, washed several times with the
tion interaction is important to the effect obtained appropriate solvent to ensure the removal of the physi-
in vivo  [24,25] . Up to now the safety proof data of clay cally adsorbed drug and, finally, dried under vacuum.
minerals clearly suggest them to be nontoxic for trans- There are many mechanisms that can be involved
dermal application, oral administration and so on [26] . in the interaction between clay and drug molecules.
Most of the research concerning the applications of They depend on the clay mineral involved as well as
clay minerals for medical purpose is focused on kaolin on the physical–chemical and functional groups of
(Si/Al oxide, 1:1), MTM and sepiolite (SPT) (Si/Al the drugs, which can generate different interactions
oxide, 2:1), where ca. 1 nm-thick clay alumino-silicate such as hydrogen bonding, hydrophilic/hydrophobic
sheets are stacked in bulky platelets or processed with interaction, ion exchange and so on. Clay minerals
exfoliation to bilayer sheets. Halloysite (HNT) (Si/Al are either negatively/positively charged, which is the
oxide, 1:1) is another nanoclay with great potentials, main reason for their ion exchange capacity. The so-
where the most common morphology is constituted by called cationic clay minerals (e.g., kaolin, MTM, hal-
elongated tubule (Figure 3) . Indeed, these clays have loysite) possess an overall mean negative charge due to
high surface areas, colloidal dimensions of their par- the external surface contribution, but also a positive
ticles and an empty cavity, in the case of HNT, that charge in their interlayer space where interstitial water
can encapsulated drug molecules. molecules are also located. These clays are able to ion
Figure 4 reflects the increasing interest of the scien- exchange with basic drugs in solution. Furthermore, in
tific community toward the use of halloysite nanotubes order to develop clay mineral systems for controlled-
as drug carrier and delivery. It is interesting to note release of drugs, a simple procedure is related to the
as the percentage (%) of the number of publications intercalation of drugs into the clay minerals interlayers.
on halloysite as drug carrier (Figure 4B) is duplicate The possibility of intercalate drugs in the interlayers of

future science group www.future-science.com 635


Perspective  Lazzara, Riela & Fakhrullin

clay–DOX showed a faster DOX release rate, with


A B
cumulative release of approximately 30% over 30 h
respect to neutral conditions were only a 5% of drug
released was observed [28,29] .
The kinetics of drug release from clay are complex
mechanisms. Generally, the active molecules release
profiles can be described by several kinetics models, as
summarized in Table 1.
Ft indicates the drug release fraction at time t, k is
C
the kinetic release constant of the respective equations,
t is the release time and n is the characteristic diffusion
exponent, depending on the release mechanism and the
geometry of the device. If Fickian diffusion occurs, n
decreased to 0.5 for slab/cylinder/sphere. If non-Fick-
ian (anomalous) diffusion dominates, the n value is
between the above value corresponding to the polymer
chain relaxation (n < 1) for slab/cylinder/sphere. Thus,
Figure 3. Structure of halloysite nanotubes. (A) Schematic representation the drugs release mechanism can be identified through
of the rolled structure of halloysite; (B) transmission electron microscopy determination of the n value. It should be noted that
and atomic foce microscopy images of halloysite nanotube edges; clays have at least two different chemistry surfaces pro-
Reproduced with permission from [26] © Wiley (2015). (C) Field emission viding at least two different binding mechanisms that
scanning electron microscopy image of halloysite on Si-wafer (left)
and schematic illustration of crystalline structure of halloysite (right).
make the release kinetics complex.
Reproduced with permission from [27] © The American Chemical Society With regard to DEM model, it describes a mecha-
(2012). nism consisting of two parallel reactions involving spe-
cies adsorbed into two different sites [37,38] . Finally, the
silicate minerals allows a good opportunity to prepare Hill model assumes that adsorption is a cooperative
inorganic and organic hybrids that possess the typical phenomenon where the ligand binding ability at one
features of both the organic guest and inorganic host site on the substrate may influence different binding
in a single material. The drug encapsulation efficacy sites on the same substrate [34] .
(%) and drug content (%) in the clay–drug hybrid are Sciascia et al. evaluated the potential applicability as
obtained according to equations (1) and (2) from spectro- DDS of MTM and Tween 20-MTM with respect to the
photometric investigations and/or t­hermogravimetric cinnamic acid drug. The adsorption isotherms of cin-
analysis. namic acid onto both the MTM and organoclay follow
Mass of the drug loaded in the clay
a sigmoidal shape where the affinity of the drug toward
Drug encapsulation efficiency (%) = ×100
Mass of the total drug added the clays increases with the amount of adsorbed drug.
The data were analyzed by several models, and it turned
Drug content (%) =
Mass of the drug loaded in the clay
×100 out that the Hill equation represents the most reliable
Mass of the clay − drug composite synthesized model. Differently, under conditions that mimic oral
The anticancer drug doxorubicin (DOX) was intro- drug administration and the consequent physiological
duced in both kaolin and HNT, reducing toxicity and release (pH 1 and 6.8, at 37.0°C) the system followed a
increasing the efficiency of release of the drug. A very DEM release profile [39] . Turco Liveri et al. immobilized
high loading capacity, of the drug, was observed in commercial Vitamin A (VitA) in MTM and SPT by
the two clays (∼54 and 80 wt% in kaolin and HNT, impregnation. The studies of kinetic release showed that
respectively) at neutral pH. The driving forces, of the the release of VitA depends on the nature of the support
clay–DOX interaction, are predominantly electrostat- as well as on the pH. Besides the controlled release, it
ics between the negative charge present on the siloxane was demonstrated that the SPT is able to prevent the
face of clays and positive DOX. The highest value of oxidative degradation of the VitA [40] . An improvement
the loading in HNT with respect to that for kaolin in the solubility, and in the control/release of ibuprofen,
is probably due to the possibility of encapsulate DOX with high permeability and low solubility, was archived
in the nanotube lumen. The acidity of solutions, in by loading it with several clays such as kaolin [41] ,
both cases, shows significant effect on DOX release MTM  [42] , HNT and covalently modified HNT with
from clay-DOX hybrid confirming that the electro- 3-aminopropyltriethoxysilane  [43–45] . It is worth not-
static interactions occur in both hybrids; indeed, under ing that organosilane modification of external face of
imulated tumor intracellular conditions (pH 5.5),
­ HNT promotes the loading of ibuprofen by creating

636 Ther. Deliv. (2017) 8(8) future science group


Clay-based drug-delivery systems: what does the future hold?  Perspective

A
Halloysite
Kaoline
Montmorilonite
Sepiolite
Other

B C

6% 11%
27%
Halloysite
Kaoline Journal
Montmorilonite Patent
ite

e
Sepiolite
ys

lin

e
nit

lite
llo

o
Ka

ilo
Ha

pio
or
56%

Se
ntm
Mo

Figure 4. Increasing interest of the scientific community toward the use of halloysite nanotubes as drug carrier
and delivery. Comparison of the number of scientific publications on the (A) ‘halloysite’, ‘kaolin’, ‘montmorillonite’
and ‘sepiolite’ terms, and (B) ‘halloysite’, ‘kaolin’, ‘montmorillonite’ and ‘sepiolite’ refined each with the term
‘drug’; (C) distribution (%) of scientific publications in ‘patent’ and ‘journal’ for each clay investigated. Data
analysis of publications, as on February 2017, was done using the SciFinder Scholar search system using as
‘Document type’ the ‘Journal’ and ‘Patent’, only.

an electrostatic attraction between the carboxyl groups inclusions. The interaction of halloysite nanotubes
present in the drug molecule and the grafted aminopro- with microscopic algae Chlorella pyrenoidosa was inves-
pyl groups in the clay. The electrostatic attraction was tigated and no penetration of the nanomaterials into
stronger than the hydrogen bonding interactions in the cell interior was observed [53] . It was also reported that
ibuprofen–HNT hybrid (Figure 5). halloysite nanotubes were safe for one of the most
common fresh water ciliate protist Paramecium cauda-
Halloysite nanotubes tum  [54] . Similarly, HNTs exhibits no toxicity toward
Halloysite as nanocontainer Escherichia coli bacteria [55] and yeast cells [56] .
Toxicity studies on halloysite nanotubes toward living Toxicity in vivo tests were reported for free living
organisms have been widely reported and the general nematodes (worms) Caenorhabditis elegance [46] , proved
outcome is that HNTs can be considered a safe materi- that HNTs did not enter the worms as they accumu-
als in a very wide concentration range [46,47] . Halloysite lated within the intestines, inflicting only mechanical
has an advantage over other clays because it does not stress onto the alimentary system (Figure 6) .
need exfoliation, which is typically time consuming Based on the safety concerns of bare HNTs, research
and expensive, and can be easily dispersed in pharma was devoted to their use as carrier for drugs. The most
formulations. Phytotoxic study on Raphanus sativus attractive feature of halloysite is its inner lumen with a
L proved a low impact on plant life by halloysite [48] . diameter capable of entrapping chemical agents such
HNT is proved to be a biocompatible material also as macromolecules, including drugs, DNA, proteins
for human cell cultures such as breast cancer cells [49] , and other chemically active agents, for example, anti-
thyroid cancer cells [17,38,50] , hepatic cancer cells [51,52] corrosion for protective coating [57–59] . In this context
and epithelial adenocarcinoma cells [49] . It was dem- the empty lumen of halloysite acts as a nanocontainer
onstrated that concentration up to 0.2 g/dm3 can for processes that benefit from suitable molecules
be considered safe, this value is typical for i­norganic s­ustained release  [60–65] .

future science group www.future-science.com 637


Perspective  Lazzara, Riela & Fakhrullin

Table 1. Kinetic and isotherm equations.


  Equation Ref.
First-order kinetic [30]
Ft = M ∞ (1 − e − kt )
Higuchi square kinetic [31]
Ft = kt1/ 2
Korsmeyer–Peppas model [32]
Ft = kt n
DEM [33]
Ft = Fe′ ⋅ (1 − e − k′t ) + Fe′′⋅ (1 − e − k′′t )
Hill
Q m ⋅ ( K H ⋅ Ce ) ⋅ n [34]
Qe =
1 + ( K H ⋅ Ce ⋅ n )
Langmuir [35]
K L Q m Ce
Qe =
1 + K L Ce
Freundlich [36]
Q e = K F Ce
1/ n

DEM: Double exponential model.

Electrostatic interactions can be used to target the and/or its chemical modification is needed. Fakhrul-
adsorption site of specific molecules. In this context, lin et al.  [72] fabricated a novel DDS based on HNTs
Lvov  et al. reported the immobilization of laccase, loaded with brilliant green, as a drug model, and coated
glucose oxidase, lipase and pepsin at inner or outer with dextrin to clog the tube opening until the cell
n­anotube surface  [66] . absorbs these nanocarriers where sugar can be cleavable
Besides drug immobilization in HNT inner by intercellular glycosyl hydrolases. Such a strategy
lumen, some inorganic salts can be loaded and release allowed the accumulation and enzymatically induced
from halloysite lumen [67] . Complete release of the release of drug in cells with higher proliferation rates,
inorganic compounds from halloysite nanotubes is which is a characteristic of tumoral cells (Figure 7) .
achieved within 1–2 h. The introduction of anionic The curcumin (Cur) selective release was achieved by
surfactant in the inner lumen is a possible strategy covalent linking on halloysite external surface through
to obtain hydrophobic cavity [68,69] . Loading dioctyl glutathione or pH responsive bonds (HNT‑Cur
sulfosuccinate sodium salt allowed to use HNT as p­rodrug)  [51] .
cargo to stabilize oil-in-water emulsions [70] . Reverse Under normal physiological conditions, the
micelles for water-in-oil emulsion were prepared by h­a lloysite prodrug showed relatively good stability, and
using cationic surfactant adsorbed selectively onto as a consequence of the dual stimuli-responsive nature
the external surface [71] . of this nanomaterial, enhanced release of Cur was
The introduction of targeting moieties on halloysite observed upon exposure of the prodrug to acidic or
surface can generate an accumulation of halloysite into glutathione-rich conditions similar to the cellular
the cell, with specific interaction including high pro- microenvironment of hepatic cancer cells (Figure 8) .
pensity to cross-cell membranes penetrating even into Stimuli responsive polymers are another class of
the cell nucleus. This aspect will be discussed in the compounds whose conformation varies upon an
next paragraph. external stimulus such as temperature, change in
light, pH and so on. Therefore, covering halloysite
Halloysite-based smart nanocomposites with surface with these types of polymers could generate
stimuli responsive features novel materials with lots of applications in the field of
More recently, the research was devoted to find more scaffolds for tissue engineering, biosensors and DDS.
complex nanoarchitectures to approach a stimuli In particular, thermo-responsive system was
responsive drug release, compatibilization of the drug obtained by introducing a thermo-responsive poly-
carrier, multiple drug delivery or a combination of such mer such as poly(N-isopropylacrylamide) on halloy-
features. With this aim, HNT-based n­anocomposites site surface [52] . This polymer was attached, by amide

638 Ther. Deliv. (2017) 8(8) future science group


Clay-based drug-delivery systems: what does the future hold?  Perspective

IBU loading (mass %)


Hydrogen bonding
0
11.7% Halloysite

ln (%release)
-1
11.8% Heated halloysite
In vitro release
IBU curve of IBU -2
12.7%

APTE8-modified halloysite Modified Korameyer-Pappas model


f = af + b -3

14.8%
-1 0 1 2 3 4
APTE8-modified and Electrostatic attraction
ln (time)
heated halloysite

Figure 5. Schematic representation of ibuprofen loading and release on halloysite and modified halloysite.
Reproduced with permission from [44] © Elsevier (2014).


ondensation, on external HNT surface previously undergoes a coil-to-globule transition at the lower crit-
modified with 3-aminopropyltrimethoxysilane. A ical solution temperature, around 32°C, temperature-
large surface density of polymer onto the nanotubes responsive features were observed. This material was
was endowed by the synthetic route adopted in this used for Cur delivery; the drug was loaded onto the car-
work. As the poly(N-isopropylacrylamide) corona rier at 25°C, whereas the drug release was investigated

Mouth
A B C D
Intestine Uterus Intestine
Procorpus
Vulva
Eggs Rectum
Metacorpus
Eggs
Isthmus

E F G
Cumulative number of worms (%)

Cumulative survival of worms


% of eggs refer to control

Body weight (µm) HNTs (mg/ml) Time (days)

Figure 6. Dark-field microscopy imaging of distribution of halloysite in Caenorhabditis elegance worms. (A) Inside the foregut;
(B and C) in the midgut (no nanotubes in embryos, uterus and vulva were detected); (D) inside the hindgut. (Bottom images)
In vivo effects on C. elegance growth and fertility: (A) the cumulative curves of body length of nematodes treated with increasing
concentrations of halloysite (mg ml-1); (B) the influence of tube concentration on fertility in adult hermaphrodites; (C) cumulative
survival curve for increasing concentrations of halloysites (mg ml-1).
Reproduced with permission from [46] © The Royal Society of Chemistry (2015).

future science group www.future-science.com 639


Perspective  Lazzara, Riela & Fakhrullin

A
BG Dextrin
Dextrin
stoppers

Loaded BG

B C D E

Figure 7. Halloysite-based prototype nanocontainers for drug delivery. (A) Fabrication of BG-loaded HNTs and the subsequent
coating with dextrin stoppers. (B) Transmission electron microscopy (TEM) image of BG-loaded halloysite; (C) scanning electron
microscopy image of a dextrin cap on the end of the functionalized nanotube; (D) TEM images of A549 cell incubated with dextran-
coated clay nanotubes; (E) resazurin assay results demonstrating the medial lethal dose (LD50 ) value of BG-loaded halloysite for A549
cells. The insets show atomic force microscopy images of the distribution of doxorubicin-halloysite in the A549 cells.
BG: Brilliant green.
Reproduced from [72] under Creative Commons license (2015).

at 37°C in a media that simulate the gastrointestinal This novel nanomaterial represents an example of a
transit (Figure 9) . The in vitro release tests showed a prolonged antioxidant synergistic protection where the
targeted and sustained release of the active species into antioxidant function was attributed to Trolox, which is
the intestine. grafted on the HNT external surface, whereas querce-
A synergic nanoantioxidant was proposed using tin, supramolecular loaded in the inner lumen, acts as
Trolox covalently linked on the external surface of a second co-antioxidant (Figure 10) .
h­a lloysite and quercetin loaded into HNT lumen [74] . Recently, the development of a dual drug delivery
is an important task of scientific community since it
MeO OH offers remarkable advantaged if compared with the
traditional single dosage. Co-delivery, indeed, may
enhance a kind of synergism between different drugs,
O increasing therapeutic target selectivity, reducing
1) HS NH3
Cl O Si N the drug resistance through distinct mechanisms of
O Si SH S S
O
OMe O O
O
OMe action.
2) MeO OMe
HO OH Recently Zhou et al. reported a dual-DDS compris-
O MeO OH ing poly(L-lactide)/HNTs electrospun mats capable of
O Si OH
O
Si OH
O co-delivering hydrophilic (polymixin B) and hydro-
phobic (dexamethasone) drugs [75] . Filling of electro-
HNT-SH HNT-Cur prodrug
HNT-SH HNT-Cur prodrug spun poly(L-lactide) mats with HNTs makes that the
nanocomposite obtained presents an increase in the
Figure 8. Schematic representation of the synthesis of the halloysite
nanotubes-curcumin prodrug.
breaking stress and, in addition, the mats degraded
HNT: Halloysite nanotubes. more rapidly if compared with pure electrospun mats,
Reproduced with permission from [73] © The Royal Society of Chemistry (2017). under the same conditions. Moreover, the presence

640 Ther. Deliv. (2017) 8(8) future science group


Clay-based drug-delivery systems: what does the future hold?  Perspective

PNIPAAM

Loading Release
Functionalization
25°C, pH 7.4 37°C, pH 6.8

Figure 9. Schematic representation of temperature-triggered loading and release of curcumin into halloysite
nanotubes modified with poly(N-isopropylacrylamide).
Reproduced with permission from [73] © The Royal Society of Chemistry (2017).

of HNTs in the fibers facilitated tunable release of The final aspect to clarify in a possible future use
polymixin B and dexamethasone that is interesting in of halloysite as drug carrier and delivery systems is
p­roducing wound-dressing materials. related to the real possibility to use these nanoma-
Another dual-drug loaded HNT delivery system terials in pharmaceutical science. Since halloysite is
was obtained by the covalent linkage of β-cyclodextrin not biodegradable nanomaterial it cannot be injected
(β-CD) units on HNT external surface [76] . CDs are intravenously. The use of modified halloysite is
well known in drug delivery due to their capacity li­mited as an active ingredient in external medical
to entrap hydrophobic drugs. Thanks to the pres- treatment with slow release of encapsulated drugs
ence of two different cavities, it is possible to encap- (e.g., in implants, creams or wound treatment of
sulate two or more drug molecules s­imultaneously. t­issues)  [10,65] .
In this context different systems were developed In this context, the modification of HNT external
(Figure 11) . Such a multicavity nanomaterial was used surface with amino acid derivatives allows to obtain
for the simultaneous co-delivery of quercetin and nanohybrids that can be incorporated in a peptide
silibinin  [38] . Experimental data highlighted that the hydrogel to obtain novel drug carrier applicable in
quercetin molecules interact preferentially with the transdermal therapy [77] . HNT-NH 2 nanomaterial
hydrophobic CD cavity, while the silibinin only with was used as intermediate to covalent link Fmoc-
the HNT lumen. The interaction between cells and phenylalanine (f-HNT). This amino acid is able to
the carrier revealed that the materials were taken up give hydrogels featured by the presence of uniform
into cells surrounding the nuclei. entangled fibers. Unlike in the case of silane-mediate
The introduction of targeting functionalities as aminogroup tailoring [78] the presence of the amino
well as glycocluster effect on halloysite/CD systems acidic functionality on HNT surface allows specific
were achieved by attaching on the β-CD core several π–π interaction between modified HNT and Fmoc-
m­annose units  [50] . phenylalanine-generating hybrid supramolecular

A B
Stoichiometric coefficient

HNT-Trolox Quc

ROO• DPPH•
HNT-Trolox HNT-Trolox/Quc

Figure 10. (A) Schematic representation of quercetin loading into HNT–Trolox. (B) Synergic effect in HNT–Trolox/
Que: experimental stoichiometric coefficient in MeCN (blue bars, ±SD, N = 3) compared with theoretic one, as
calculated from the sum of contributions of HNT–Trolox (green bars) and HNT/Que (red bars) alone.
HNT: Halloysite nanotubes.
Reproduced with permission from [73] © The Royal Society of Chemistry (2017).

future science group www.future-science.com 641


Perspective  Lazzara, Riela & Fakhrullin

OTBDMS
7

O
O
MeO S
O
Si O
O O O
O O

OAc
AcO
O
AcO
1) AcO HNT-βCD
SH n SH
2) MeONa/MeOH
3) BF3OEt2

HO
HO O
HO
OH OTBDMS
HO
S 7 7

n S O
O O
O
MeO S
MeO S O
O Si O
Si O S O O O O S
O O O O O n
O HO
HO O S
n
O S
HO S S
HO HO S
HO
O S S n S n
HO
HO O
HO O
HO HO HO OH

n
OH
OH HO HO OH
OH
OH
HNT multicavity
HNT-CD
Drugs Drugs
Drugs Drugs

Figure 11. Schematic representation of the synthesis of the halloysite nanotubes multicavity and halloysite
nanotubes-cyclodextrin.

h­ydrogel with peculiar physico-chemical properties. halloysite clay is concerned, another difficulty that is
Deep investigation of hybrid gel properties, indeed, worth of being investigated in the future is the pos-
showed that the introduction of HNT filler in the sibility to ‘clean’ HNT samples obtaining more uni-
hydrogel matrix induced decreased thermal stabil- form nearly monodisperse nanotubes. This is crucial
ity but, in general, a concomitant increase in the in applications as release properties are influenced
gel strength. The hybrid hydrogel was employed for by the HNT deposit and polydispersion distribution
camptothecin delivery. Release tests in physiologi- in sizes. On the other hand, Halloysite used as DDS
cal conditions showed a synergistic action of HNTs could have an advantage due to the biocompatibility,
and gel matrix, as accounted for by the best drug low cost and natural resources of the material and the
d­ispersion and the slower rate of release as a conse- simple methods used to prepare dosage forms. These
quence of the HNTs incorporation in the gel phase. characteristics should allow for quick industrial scale-
up. The goal should be to increase the use of halloysite
Conclusion & future perspective clay nanotubes in DDS suitable for different routes of
Clay minerals-based materials are very promising for administration. In the near future, we foresee its usage
the development of DDS. Although there are a number in medical cosmetic formulations, animal treatment,
of papers on this direction, there are still several issues in dental resins, for timed-release antimicrobial spray
to be solved and other opportunities to be investigated. and as a tableting excipient. The possibility to prepare
Due to their inorganic nature, the route of adminis- smart caps as well as the possibility to use HNT inter-
tration are somehow limited as the blood injection is layers beside its cavity are also opening bright perspec-
not yet considered safe unless further in vivo tests are tives for developing controlled/targeted delivery sys-
able to find a possible route for the elimination of this tems. In this context, HNTs could be used as a tablet
inorganic material from the blood flow. As far as the for delivery and controlled-release of several drugs.

642 Ther. Deliv. (2017) 8(8) future science group


Clay-based drug-delivery systems: what does the future hold?  Perspective

Financial & competing interests disclosure RB‑FR12ETL5). The authors have no other relevant affiliations
This work was funded by the subsidy allocated to Kazan Fed- or financial involvement with any organization or entity with a
eral University for the state assignment in the sphere of scien- financial interest in or financial conflict with the subject mat-
tific activities (16.2822.2017/4.6). The work was performed ter or materials discussed in the manuscript apart from those
according to the Russian Government Program of Competi- disclosed.
tive Growth of Kazan Federal University. The work was par- No writing assistance was utilized in the production of this
tially supported by the University of Palermo, FIRB 2012 (prot. manuscript.

Executive summary
Nanoclays for medical purposes: from the past to modern applications
• Clay minerals have been known for medical applications since a very long time from ancient Egypt to
Renaissance.
• Drug molecules are encapsulated in clay minerals to modify the rate, the time and to target the site of drug
release.
• Clay minerals as they are not inert fillers but, instead, they can have a functionality providing targeting
release, prevention or reduction of side effects and increasing the product shelf-life.
Nanoclay for adsorption & sustained release of drugs
• Drug-delivery system is a recognized protocol to increase the drug specificity, efficacy, tolerability and
therapeutic index.
• Clay minerals are used as excipients in commercialized pharmaceutical products and indicated as ‘Inactive
Ingredient Database’.
• Most of the research concerning the applications of clay minerals for medical purpose is focused on kaolin,
montmorillonite, sepiolite and, more recently, halloysite nanotubes.
• Interaction between clay and drug molecules depend on functional groups of the drugs, which can generate
different interactions such as hydrogen bonding, hydrophilic/hydrophobic interaction, ion exchange and so on.
• The kinetics of drug release from clay are complex mechanisms as clays have at least two different chemistry
surfaces providing at least two different binding mechanisms.
Halloysite nanotubes
• Halloysite as a nanocontainer.
• Halloysite nanotubes can be considered a safe materials toward living organisms such as plant, human cell,
microscopic algae Chlorella pyrenoidosa, ciliate protist Paramecium caudatum, Escherichia coli bacteria, yeast
cells and free living nematodes (worms) Caenorhabditis elegance.
• The most attractive feature of halloysite is its inner lumen with a diameter capable of entrapping chemical
agents. Electrostatic interactions can be used to target the adsorption site of specific molecules.
Halloysite-based smart nanocomposites with stimuli-responsive features
• Complex nanoarchitectures can be achieved to approach a stimuli responsive drug release, compatibilization
of the drug carrier, multiple drug delivery or a combination of such features.
• Halloysite nanotubes coated with dextrin was a strategy to clog the tube opening until the cell absorbs these
nanocarriers where sugar can be cleavable by intercellular glycosyl hydrolases.
• The drug selective release was achieved by covalent linking on halloysite external surface through glutathione
or pH responsive bonds.
• Covering halloysite surface with these types of polymers could generate novel materials with lots of
applications in the fields of scaffolds for tissue engineering, biosensors and drug-delivery systems.
• A synergic nanoantioxidant was proposed using Trolox covalently linked on the external surface of halloysite
and quercetin loaded into HNT lumen to regenerate the main antioxidant and afford a prolonged synergistic
protection.
• Dual-drug loaded halloysite nanotubes delivery system was obtained by the covalent linkage of β-cyclodextrin
units on nanotubes external surface. The presence of two cavities offers the remarkable possibility for a
simultaneous encapsulation of two or more drug molecules with different physico-chemical properties,
followed by a different release path.

2 Abe H, Liu J, Ariga K. Catalytic nanoarchitectonics for


References environmentally compatible energy generation. Mater.
Papers of special note have been highlighted as: • of interest
Today 19(1), 12–18 (2016).
1 Kazuhiko M, Kazunari D. Development of novel
3 Kuwahara Y, Tamagawa S, Fujitani T, Yamashita H.
photocatalyst and cocatalyst materials for water splitting
Removal of phosphate from aqueous solution using layered
under visible light. Bull. Chem. Soc. Jpn 89(6), 627–648
double hydroxide prepared from waste iron-making slag.
(2016).
Bull. Chem. Soc. Jpn 89(4), 472–480 (2016).

future science group www.future-science.com 643


Perspective  Lazzara, Riela & Fakhrullin

4 Ariga K, Ji Q, McShane MJ, Lvov YM, Vinu A, Hill JP. material for biomedical applications. Chemistry 19(15),
Inorganic nanoarchitectonics for biological applications. 4869–4875 (2013).
Chem. Mater. 24(5), 728–737 (2012). 20 Ambrogi V, Nocchetti M, Latterini L. Promethazine–
5 Ruiz-Hitzky E, Aranda P, Darder M, Rytwo G. Hybrid montmorillonite inclusion complex to enhance drug
materials based on clays for environmental and biomedical photostability. Langmuir 30(48), 14612–14620 (2014).
applications. J. Mater. Chem. 20(42), 9306–9321 (2010). 21 Viseras C, Cerezo P, Sanchez R, Salcedo I, Aguzzi C.
6 Carretero MI. Clay minerals and their beneficial effects upon Current challenges in clay minerals for drug delivery. App.
human health. A review. App. Clay Sci. 21(3–4), 155–163 Clay Sci. 48(3), 291–295 (2010).
(2002). 22 Ariga K, Abe H, Ji Q, Lvov YM. Halloysite and
7 Aguzzi C, Cerezo P, Viseras C, Caramella C. Use of clays as related mesoporous carriers for advanced catalysis and
drug-delivery systems: possibilities and limitations. App. Clay drug delivery. In: Functional Polymer Composites with
Sci. 36(1–3), 22–36 (2007). Nanoclays. RSC Smart Materials Lvov Y, Guo B, Fakhrullin
8 75/18/Eec D. Quality of prolonged release oral solid dosage RF (Eds). 207–222 (2017).
forms. In: Dictionary of Pharmaceutical Medicine.34 (1992). 23 75/318/Eec D. Quality of prolonged release oral solid dosage
9 Naumenko EA, Fakhrullin RF. Toxicological evaluation of forms. (1992).
clay nanomaterials and polymer-clay nanocomposites. In: 24 Sorby DL, Liu G. Effects of adsorbents on drug absorption
Functional Polymer Composites with Nanoclays.RSC Smart II: effect of an antidiarrhea mixture on promazine
Materials Lvov Y, Guo B, Fakhrullin RF(Eds). 399– absorption. J. Pharm. Sci. 55(5), 504–510 (1966).
419 (2017). 25 Sorby DL. Effect of adsorbents on drug absorption
10 Yendluri R, Otto DP, De Villiers MM, Vinokurov V, I. Modification of promazine absorption by activated
Lvov YM. Application of halloysite clay nanotubes as a attapulgite and activated charcoal. J. Pharm. Sci. 54(5),
pharmaceutical excipient. Int. J. Pharm. 521(1–2), 267–273 677–683 (1965).
(2017). 26 Lvov Y, Wang W, Zhang L, Fakhrullin R. Halloysite clay
• Demonstrates the fabrication of a versatile halloysite-based nanotubes for loading and sustained release of functional
pharmaceutical delivery platform. compounds. Adv. Mater. 28(6), 1227–1250 (2015).
11 Bergaya F, Lagaly G. Handbook of Clay Science, Volume 5 2nd 27 Yah WO, Takahara A, Lvov YM. Selective modification
Edition.Bergaya F, Lagaly G (Eds). (2013). of halloysite lumen with octadecylphosphonic acid: new
inorganic tubular micelle. J. Am. Chem. Soc. 134(3),
12 Park J-H, Shin H-J, Kim MH et al. Application of
1853–1859 (2012).
montmorillonite in bentonite as a pharmaceutical excipient
in drug-delivery systems. J. Pharm. Invest. 46(4), 363–375 28 Zhang Y, Long M, Huang P et al. Emerging integrated
(2016). nanoclay-facilitated drug-delivery system for papillary
thyroid cancer therapy. Sci. Rep. 6, 33335 (2016).
13 FDA. Inactive ingredients database for approved drug
products. FDA/Center for Drug Evaluation and Research. 29 Li L, Fan H, Wang L, Jin Z. Does halloysite behave like an
Office of Generic Drug. Division of Labelling and Program inert carrier for doxorubicin? RSC Adv. 6(59), 54193–54201
Support (2009). (2016).
14 Mahmoudi M, Adib-Hajbaghery M, Mashaiekhi M. • A facile and effective method to fabricate anticancer drug-
Comparing the effects of bentonite & calendula on the carrier halloysite platforms using halloysite nanoclay.
improvement of infantile diaper dermatitis: a randomized 30 Gibaldi M, Feldman S. Establishment of sink conditions in
controlled trial. Indian J. Med. Res. 142(6), 742–746 (2015). dissolution rate determinations. Theoretical considerations
15 Li S, Jiang C, Chen X, Wang H, Lin J. Lactobacillus and application to nondisintegrating dosage forms. J. Pharm.
casei immobilized onto montmorillonite: survivability in Sci. 56(10), 1238–1242 (1967).
simulated gastrointestinal conditions, refrigeration and 31 Higuchi T. Mechanism of sustained-action medication.
yogurt. Food Res. Int. 64, 822–830 (2014). Theoretical analysis of rate of release of solid drugs dispersed
16 Khediri F, Mrad AI, Azzouz M et al. Efficacy of diosmectite in solid matrices. J. Pharm. Sci. 52(12), 1145–1149 (1963).
(Smecta) ® in the treatment of acute watery diarrhoea in 32 Ritger PL, Peppas NA. A simple equation for description of
adults: a multicentre, randomized, double-blind, placebo- solute release I. Fickian and non-Fickian release from non-
controlled, parallel group study. Gastroenterol. Res. swellable devices in the form of slabs, spheres, cylinders or
Pract. 2011, 783196 (2011). discs. J. Control. Release 5(1), 23–36 (1987).
17 Riela S, Massaro M, Colletti CG et al. Development and 33 Wilczak A, Keinath TM. Kinetics of sorption and desorption
characterization of co-loaded curcumin/triazole-halloysite of copper (II) and lead (II) on activated carbon. Water
systems and evaluation of their potential anticancer activity. Environ. Res. 65(3), 238–244 (1993).
Int. J. Pharm. 475(1–2), 613–623 (2014).
34 Ringot D, Lerzy B, Chaplain K, Bonhoure JP, Auclair E,
18 Lee JH, Choi G, Oh YJ et al. A nanohybrid system for taste Larondelle Y. In vitro biosorption of ochratoxin A on the
masking of sildenafil. Int. J. Nanomedicine 7, 1635–1649 yeast industry by-products: comparison of isotherm models.
(2012). Bioresour. Technol. 98(9), 1812–1821 (2007).
19 Oh Y-J, Choi G, Choy YB et al. Aripiprazole- 35 Langmuir I. The constitution and fundamental properties
montmorillonite: a new organic-inorganic nanohybrid

644 Ther. Deliv. (2017) 8(8) future science group


Clay-based drug-delivery systems: what does the future hold?  Perspective

of solids and liquids. part i. solids. J. Am. Chem. Soc. 38(11), 50 Massaro M, Riela S, Baiamonte C et al. Dual drug-loaded
2221–2295 (1916). halloysite hybrid-based glycocluster for sustained release
36 Wu XL, Zhao D, Yang ST. Impact of solution chemistry of hydrophobic molecules. RSC Adv. 6(91), 87935–87944
conditions on the sorption behavior of Cu(II) on Lin’an (2016).
montmorillonite. Desalination 269(1–3), 84–91 (2011). • The effective nuclear uptake of drug-loaded polymer-coated
37 Sciascia L, Turco Liveri ML, Merli M. Kinetic and equilibrium halloysite into cellular nuclei.
studies for the adsorption of acid nucleic bases onto K10 51 Massaro M, Amorati R, Cavallaro G et al. Direct chemical
montmorillonite. App. Clay Sci. 53(4), 657–668 (2011). grafted curcumin on halloysite nanotubes as dual-responsive
38 Massaro M, Piana S, Colletti CG et al. Multicavity prodrug for pharmacological applications. Colloids Surf. B.
halloysite-amphiphilic cyclodextrin hybrids for co-delivery Biointerfaces 140, 505–513 (2016).
of natural drugs into thyroid cancer cells. J. Mater. Chem. • Delivery of macromolecular prodrug (curcumin) into
B 3(19), 4074–4081 (2015). human cells.
39 Calabrese I, Gelardi G, Merli M, Liveri MLT, Sciascia L. 52 Cavallaro G, Lazzara G, Massaro M et al. Biocompatible
Clay-biosurfactant materials as functional drug-delivery poly(N-isopropylacrylamide)-halloysite nanotubes
systems: slowing down effect in the in vitro release of for thermoresponsive curcumin release. J. Phys. Chem.
cinnamic acid. App. Clay Sci. 135, 567–574 (2017). C 119(16), 8944–8951 (2015).
40 Calabrese I, Turco Liveri ML, Ferreira MJ et al. Porous 53 Lvov Y, Aerov A, Fakhrullin R. Clay nanotube encapsulation
materials as delivery and protective agents for Vitamin A. for functional biocomposites. Adv. Colloid Interface Sci. 207,
RSC Adv. 6(71), 66495–66504 (2016). 189–198 (2014).
41 Mallick S, Pattnaik S, Swain K et al. Formation of physically 54 Kryuchkova M, Danilushkina AA, Lvov YM, Fakhrullin RF.
stable amorphous phase of ibuprofen by solid state milling Evaluation of toxicity of nanoclays and graphene oxide in
with kaolin. Eur. J. Pharm. Biopharm. 68(2), 346–351 vivo: a Paramecium caudatum study. Environm. Sci. Nano 3,
(2008). 442-452 doi:10.1039/C5EN00201J (2016) (Epub ahead of
42 Dziadkowiec J, Mansa R, Quintela A, Rocha F, Detellier C. print). 
Preparation, characterization and application in controlled 55 Zhang Y, Chen Y, Zhang H, Zhang B, Liu J. Potent
release of ibuprofen-loaded guar gum/montmorillonite antibacterial activity of a novel silver nanoparticle-halloysite
bionanocomposites. App. Clay Sci. 135, 52–63 (2017). nanotube nanocomposite powder. J. Inorg. Biochem. 118,
43 Li H, Zhu X, Xu J, Peng W, Zhong S, Wang Y. The 59–64 (2013).
combination of adsorption by functionalized halloysite 56 Konnova SA, Sharipova IR, Demina TA et al.
nanotubes and encapsulation by polyelectrolyte coatings Biomimetic cell-mediated three-dimensional assembly of
for sustained drug delivery. RSC Adv. 6(59), 54463–54470 halloysite nanotubes. Chem. Commun. 49(39), 4208–4210
(2016). (2013).
44 Tan D, Yuan P, Annabi-Bergaya F et al. Loading and in vitro 57 Sanchez-Ballester NM, Ramesh GV, Tanabe T et al.
release of ibuprofen in tubular halloysite. App. Clay Sci. 96, Activated interiors of clay nanotubes for agglomeration-
50–55 (2014). tolerant automotive exhaust remediation. J. Mater. Chem.
45 Tan D, Yuan P, Annabi-Bergaya F et al. Natural halloysite A 3(12), 6614–6619 (2015).
nanotubes as mesoporous carriers for the loading of 58 Vinokurov VA, Stavitskaya AV, Chudakov YA et al.
ibuprofen. Micropor. Mesopor. Mater. 179, 89–98 (2013). Formation of metal clusters in halloysite clay nanotubes. Sci.
46 Fakhrullina GI, Akhatova FS, Lvov YM, Fakhrullin RF. Technol. Adv. Mater. 18(1), 147–151 (2017).
Toxicity of halloysite clay nanotubes in vivo: a Caenorhabditis 59 Abdullayev E, Sakakibara K, Okamoto K, Wei W,
elegans study. Environm. Sci. Nano 2(1), 54–59 (2015). Ariga K, Lvov Y. Natural tubule clay template synthesis of
47 Kryuchkova M, Danilushkina A, Lvov Y, Fakhrullin R. silver nanorods for antibacterial composite coating. ACS
Evaluation of toxicity of nanoclays and graphene oxide Appl. Mater. Interfaces 3(10), 4040–4046 (2011).
in vivo: a Paramecium caudatum study. Environm. Sci. 60 Price R, Gaber BP, Lvov Y. In-vitro release characteristics
Nano 3(2), 442–452 (2016). of tetracycline HCl, khellin and nicotinamide adenine
• The toxicity of various nanoclays is compared with that of dineculeotide from halloysite; a cylindrical mineral. J.
graphene oxide nanosheets. Microencaps. 18(6), 713–722 (2001).
48 Bellani L, Giorgetti L, Riela S, Lazzara G, Scialabba A, • Demonstrates the encapsulation and release of biologically
Massaro M. Ecotoxicity of halloysite nanotube-supported active molecules tailored to halloysite nanocontainers.
palladium nanoparticles in Raphanus sativus L. Environ. 61 Veerabadran NG, Price RR, Lvov YM. Clay nanotubes
Toxicol. Chem. 35(10), 2503–2510 (2016). for encapsulation and sustained release of drugs. ACS
49 Vergaro V, Abdullayev E, Lvov YM et al. Nano 02(02), 115–120 (2007).
Cytocompatibility and uptake of halloysite clay nanotubes. 62 Dionisi C, Hanafy N, Nobile C et al. Halloysite clay
Biomacromolecules 11(3), 820–826 (2010). nanotubes as carriers for curcumin: characterization and
• This study has pioneered the investigation on uptake and application. IEEE Trans. Nanotechnol. 15(5), 720–724
biodistribution of halloysite nanoclay by human cells in (2016).
culture.

future science group www.future-science.com 645


Perspective  Lazzara, Riela & Fakhrullin

63 Vergaro V, Lvov YM, Leporatti S. Halloysite clay 72 Dzamukova MR, Naumenko EA, Lvov YM, Fakhrullin RF.
nanotubes for resveratrol delivery to cancer cells. Macromol. Enzyme-activated intracellular drug delivery with tubule clay
Biosci. 12(9), 1265–1271 (2012). nanoformulation. Sci. Rep. 5, 10560 (2015).
64 Vergara D, Bellomo C, Zhang X et al. Lapatinib/paclitaxel • Fabrication and in vitro demonstration of anticancer
polyelectrolyte nanocapsules for overcoming multidrug effect of drug-loaded polymer functionalized halloysite
resistance in ovarian cancer. Nanomed. Nanotechnol. Biol. nanocontainers.
Med. 8(6), 891–899 (2012).
73 Massaro M, Lazzara G, Milioto S, Noto R, Riela S. Covalently
65 Lvov YM, Devilliers MM, Fakhrullin RF. The application modified halloysite clay nanotubes: synthesis, properties,
of halloysite tubule nanoclay in drug delivery. Expert Opin. biological and medical applications. J. Mater. Chem.
Drug Deliv. 13(7), 977–986 (2016). B doi:10.1039/C7TB00316A (2017) (Epub ahead of print).
66 Tully J, Yendluri R, Lvov Y. Halloysite clay nanotubes for 74 Massaro M, Riela S, Guernelli S et al. A synergic
enzyme immobilization. Biomacromolecules 17(2), 615–621 nanoantioxidant based on covalently modified halloysite-
(2016). trolox nanotubes with intra-lumen loaded quercetin. J.
67 Abdullayev E, Lvov Y. Halloysite clay nanotubes for Mater. Chem. B 4(13), 2229–2241 (2016).
controlled release of protective agents. J. Nanosci. 75 Zhang X, Guo R, Xu J et al. Poly(l-lactide)/halloysite
Nanotech. 11(11), 10007–10026 (2011). nanotube electrospun mats as dual-drug-delivery systems and
68 Cavallaro G, Lazzara G, Milioto S, Palmisano G, Parisi F. their therapeutic efficacy in infected full-thickness burns. J.
Halloysite nanotube with fluorinated lumen: non-foaming Biomater. Appl. 30(5), 512–525 (2015).
nanocontainer for storage and controlled release of oxygen in • Polymer/halloysite-based 2D nano-to-micro drug delivery
aqueous media. J. Colloid Interface Sci. 417, 66–71 (2014). platforms.
69 Cavallaro G, Lazzara G, Milioto S. Exploiting the colloidal 76 Massaro M, Riela S, Lo Meo P et al. Functionalized
stability and solubilization ability of clay nanotubes/ionic halloysite multivalent glycocluster as a new drug-delivery
surfactant hybrid nanomaterials. J. Phys. Chem. C 116(41), system. J. Mater. Chem. B 2(44), 7732–7738 (2014).
21932–21938 (2012).
77 Riela S, Rizzo C, Arrigo R et al. Hybrid supramolecular
70 Owoseni O, Nyankson E, Zhang Y et al. Release of gels of Fmoc-F/halloysite nanotubes: systems for sustained
surfactant cargo from interfacially-active halloysite clay release of camptothecin. J. Mater. Chem. B doi:10.1039/
nanotubes for oil spill remediation. Langmuir 30(45), C7TB00297A (2017) (Epub ahead of print).
13533–13541 (2014).
78 Yuan P, Southon PD, Liu Z et al. Functionalization
71 Cavallaro G, Lazzara G, Milioto S, Parisi F. Hydrophobically of halloysite clay nanotubes by grafting with
modified halloysite nanotubes as reverse micelles for water- γ-aminopropyltriethoxysilane. J. Phys. Chem. C 112(40),
in-oil emulsion. Langmuir 31(27), 7472–7478 (2015). 15742–15751 (2008).

646 Ther. Deliv. (2017) 8(8) future science group

You might also like