You are on page 1of 21

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/332447323

Laponite-based Nanomaterials for Biomedical Applications: A Review

Article  in  Current Pharmaceutical Design · April 2019


DOI: 10.2174/1381612825666190402165845

CITATIONS READS
2 774

7 authors, including:

Sabya Sachi Das Sima Singh


Birla Institute of Technology, Mesra University of KwaZulu-Natal
18 PUBLICATIONS   14 CITATIONS    31 PUBLICATIONS   155 CITATIONS   

SEE PROFILE SEE PROFILE

Afzal Hussain
King Saud University
59 PUBLICATIONS   314 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Nanomedicine View project

Biomedical Approach For Nanoparticulate Drug Delivery Syetem View project

All content following this page was uploaded by Sima Singh on 16 April 2019.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.net
Current Pharmaceutical Design, 2019, 25, 1-20 1
REVIEW ARTICLE

Laponite-based Nanomaterials for Biomedical Applications: A Review

Sabya Sachi Das1, Neelam2, Kashif Hussain3, Sima Singh4,*, Afzal Hussain1, Abdul Faruk5 and
Tebyetekerwa Mike6

1
Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi-835215, Jharkhand, India;
2
Department of Pharmaceutical Sciences, NIMS University, Jaipur-303121, Rajasthan; 3Gyani Inder Singh Institute of Professional
Studies, Dehradun-248003, Uttarakhand, India 4School of Health Sciences, College of Health Sciences, University of KwaZulu-Natal,
Durban 4000, South Africa; 5Department of Pharmaceutical Sciences, Hemwati Nandan Bahuguna Garhwal University, Srinagar,
Uttarakhand, India; 6State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Material Science,
Donghua University, P. R. China

Abstract: Laponite based nanomaterials (LBNMs) are highly diverse regarding their mechanical, chemical, and
structural properties, coupled with shape, size, mass, biodegradability and biocompatibility. These ubiquitous
ARTICLEHISTORY properties of LBNMs make them appropriate materials for extensive applications. These have enormous potential
for effective and targeted drug delivery comprised of numerous biodegradable materials which results in en-
hanced bioavailability. Moreover, the clay material has been explored in tissue engineering and bioimaging for
Received: December 8, 2018
Accepted: March 20, 2019 the diagnosis and treatment of various diseases. The material has been profoundly explored for minimized toxic-
ity of nanomedicines. The present review compiled relevant and informative data to focus on the interactions of
laponite nanoparticles and application in drug delivery, tissue engineering, imaging, cell adhesion and prolifera-
DOI:
tion, and in biosensors. Eventually, concise conclusions are drawn concerning biomedical applications and identi-
10.2174/1381612825666190402165845   fication of new promising research directions.

Keywords: Laponite (LAP), Laponite-Based Nano-Materials (LBNMs), Applications, Composition and characteristics, Drug delivery and
biosensor.

1. INTRODUCTION leading to the formation of octahedral or tetrahedral nano-clay


Exhaustive research has been carried out for the detection, di- sheets [28]. They have recently provided new opportunities for
agnosis, and treatment of several diseases prevailing around us improved hybrid materials due to their proven capacity to interre-
today. However, trends and advances in science and technology, late with drugs and other biological molecules during synthesis and
keep availing new areas of research [1]. In the recent past, various use [29]. These silicates are used as fundamental units to assemble
researchers have witnessed extraordinary attention and innovations organic moieties in the nanometer array. The amassing approaches
to exploit nanomaterials and mitigate several issues in drug delivery depend on the clay features which comprise, i) coated clays (ver-
[2]. Currently, numerous nanomaterials of different sizes, structure, miculites, kaolinites and smectites) can easily get adsorbed in leni-
morphology and origin (natural and synthetic) have been synthe- ent experimental circumstances, altered ions and molecules; nano-
sized for prime application in various biomedical areas such as composites and polymers resilient interlinked complexes and nano-
biosensing, medical diagnosis, standardized catalysis, and other combinations, and ii) microfibrous clay minerals such as palygor-
usage [3] Some of these nanomaterials include multifunctional skite and sepiolite, can produce hybrid and complex materials via
liposomal nanoparticles, [4]functionalized fullerenes, [5,6] func- the interface with diverse kinds of organic moieties by the periph-
tionalized nanotubes,[7,8] iron oxide nanoparticles, [9,10] polym- eral planes and, in certain situations, the interior planes of these
eric micelles, [11,12] dendrimers,[13,14] nanoshells, [15] and po- nano-structured particulate solids [30].
lymeric microspheres [16]. Unquestionably, nanomaterials have Amongst the various clay mineral nanomaterials, synthetic clay
found ubiquitous uses in various areas of biomedical application material Laponite® (Table 1) has stood out as the most commonly
owing to innate biosafety and biocompatible concern [17,18]. This used nanoclay in biomedical applications. Laponite®, therefore,
may be due to the additional unique properties which include ad- forms the basis of our current review, given its various appreciable
justable size, increased drug/material loading, ease in adjusting properties, limitations, advantages and disadvantages along with
surface properties, stimuli-responsive drug release kinetics, and preparation methods. The review also gathered comparative data of
better pharmacokinetics [19-21]. A wide range of nanomaterials various clay nanomaterials (natural and synthetic) which would be
with some of the above properties have been investigated and em- very informative for scientists or researchers working in the same
ployed in various biomedical and pharmaceutical fields [22-25], domain.
such as clay-based nanoparticles [26,27].
2. LAPONITE
Clay minerals are typically layered silicate structures (phyl-
losilicates) of plate-like morphology. Their arrangements are estab- Laponite (LAP) is a synthetic nano clay smectite with a struc-
lished on coatings fabricated through cationic ligands octahedrally ture and composition closely related to the natural clay mineral
or tetrahedrally synchronized to hydroxyl and oxygen linkages hectorite (Fig. 1) [31,32]. The activities and utility of LAP for dif-
ferent purposes depend upon its grades (Table 2). It has a distinct
layered structure with a permanent negative surface charge on every
*Address correspondence to this author at the School of Health Sciences, single face and aspect of the molecules and positive charge distribu-
College of Health Sciences, University of KwaZulu-Natal, Durban 4000,
South Africa; E-mail: simasingh87@gmail.com
tions along the surface edges [32,33], enhancing LAP nanoparticle
properties such as high aqueous stability and shear thinning fea-

1381-6128/19 $58.00+.00 © 2019 Bentham Science Publishers


2 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

Table 1. Composition, characteristics and synthesis conditions of laponites.

Composition % (on dry weight basis)


1. Silicone dioxide ( SiO 2 ) : 60.4
2. Ferric oxide ( Fe 2 O 3 ) : 0.02
3. Magnesium oxide ( MgO ) : 26.0
4. Calcium oxide ( CaO ) : 0.20
5. Sodium oxide ( Na2 O) : 3.0
6. Water ( H 2 O) : 6.9
7. Carbon dioxide ( CO 2) : 0.29
8. Sulphur trioxide ( SO 3 ) : 0.10
9. Lithium oxide ( Li 2O) : 1.1
Sum of mass : 98.01
Physicochemical characteristics
a) Molecular f ormula : H 12 Li 2Mg 16 Na 2 O 72 Si 24
b) IUPAC Name : D ilithium;hexadecamagnesium;disodi um;1,3,5,7 -
tetraoxido - 2,4,6,8,9,10 -hexaoxa -1,3,5,7 -
tetrasilatricyclo[3.3.1.13,7]decane;dodecahydroxide
c) Molecular weight (g/mol ) 2286.804
:
d) Morphology : Fine white powder; swells to produce a clear
colorless thixotropic gel when dispersed in water
e) Cation exchange capacity ( ! /g) : 0.79
f) Size of average primary particle : 40 _ 10 _ 1
(nm 3)
g) Moisture ( % at 105 °C) : 8
h) pH ( 2 % aqueous dispersion ) : 8.5
-3)
i) Measured density ( g cm : 2.53
j) Refractive index : 1.54

! Starting Material Conditions Remarks


Mg salts, Na 2SiO 3 Na 2 CO 3 or NaOH 1 bar, boiling or reflux, 10 -2- h; 185 - Commercially available as
Li salt, HF or LiF 265°C at 10 -50 bar, " 8 h Laponite material

MgCl 2 , Na 2 SiO 3, HNO 3 , NH 4 OH, 125 -300 °C, P H2O , 1 -24 h Less structural Li with increasing
NaOH, LiOH temperature, layer charge due to
octahedral vacancies

Talc, Li 2 CO 3 (calcinated at 760 - 185°C, P H2O Laponite like material; talc is


980°C), Na 2 SiO 3 Na 2 CO 3 relatively cheap and readily
available source

K 2 SiF 6, LiF, Li 2CO 3 MgO, SiO 2 1420 °C, 2h Li-laponite; very small flakes as
byproduct

Established reaction and conditions for the synthesis of pure laponite material:

Na 0.7 Si 8 Mg 5.5 Li0.3 O 20 (OH) 4 +12H ++8H 2 O_ 0.7Na ++8Si(OH) 4 +5.3Mg 2+ +0.4Li +

(pH of dissolution medium - 6-9)

!
tures, with improved drug loading capacity and drug interactions into single nanosheets, which is a simple procedure that signifi-
[34,35]. In LAP, the metal cationic inter-layer is intercalated be- cantly enhances the surface area and interactions with different!
tween two tetrahedral silica layers, giving a 2:1 smectite layered biomolecules [36].
structure resemblance. The LAP-nanocomposite plane has a poor At suitable concentration, LAP forms gels in aqueous systems
net negative charge due to the cationic exchange, and the conse- by electrostatic interactions amongst negatively charged surfaces
quential unstable charges generate a net positive charge distribution and positively charged edges of the nanodiscs of radius 12.5 nm
on the surface end of each LAP nanoparticle. Therefore, the feeble- and a width of 1 nm, giving outstanding suspension thixotropy to
ness of charges on the surface end of the nanoparticles associated systems, in addition to other unique properties [37-39] (Fig. 2).
with alike clays makes LAP as a significant clay for biomedical Special characteristics of LAP include the potential interactions
applications. Minor surface charges on LAP provide delamination with organic molecules on LAP modified planes, inter-film
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 3

Fig. (1). a) Schematic and b) TEM images of LAP nanoparticles showing shape and size. c) Molecular structure of LAP, smectite 2:1 (TOT where T stands for
Tetrahedral and O for Octahedral) phyllosilicate (Image reproduced with permission from RSC in ref. 36)

Fig. (2). a) LAP, [(Si 8 Mg 5.5 Li 0.3 ) O 20 (OH) 4] − 0.7) has a permanent negative surface charge due to the isomorphic substitutions in the crystal structure and a
pH dependent edge charge from unsatisfied valences in the disrupted crystal lattice. b) Potential interactions between organic molecules and the clay particle
surfaces, inter-layer pores, and inter-particle spaces give many mechanisms. (Image reproduced with permission from John Wiley and Sons in ref. 29)
4 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

Table 2. Different grades of laponite and their benefits/limitations.

Laponite Temporary Permanent sol


Gel forming Features Benefits and limitations
grade name sol forming forming

Universal application and high efficiency in water based sys-


RD X General purpose grade tems; rheology control in surface coatings, household products
and general and industrial fields

As for LAPONITE RD, universal application and high effi-


ciency in water based systems, recommended for use in formu-
RDS X General purpose sol grade
lations that have low levels of free water. Suitable for use in soft
and hard water up to 20 °dH/ 20 °E/ 350ppm CaCO3

Aqueous dispersions of this grade will remain as stable liquids


for very long periods of time. Recommended for use in highly
filled surface coatings that have low levels of free water.
S482 X Very high sol stability grade
Also for use in non-rheology applications, such as electrically
conductive, antistatic and barrier films.
Suitable for use in all levels of water hardness

Ready-for-use, simply mix into a readymade formulation. Rec-


ommended for use in highly filled surface coatings. Also for use
A long term stable aqueous dis-
SL25 X in non-rheology applications, such as electrically conductive,
persion of Laponite antistatic and barrier films.
Suitable for use in all levels of water hardness.

Functions well in formulas with very high or very low pH level.


Organic modification for extra
Recommended for systems containing higher levels of dissolved
EP X performance in “difficult” sys-
solids. Very efficient stabilization of emulsions and suspended
tems
solids.

Optimized for use in non-rheology applications such as electri-


JS X High sol stability grade
cally conductive, antistatic and barrier films

Personal care grades:


High purity, certified low heavy
For rheology control in personal care and cosmetic applications,
XLG X metal and low microbiological
used to stabilize emulsions, lotions and creams.
content.

High purity, certified low heavy


For rheology control in personal care and cosmetic applications,
XLS X metal and low microbiological
often used in rinse-off products containing surfactants.
content.

High purity, certified low heavy For rheology control in personal care and cosmetic applications,
XL21 X metal and low microbiological optimized for use in skincare formulations stabilized at pH 5.5
content. or lower.

Optimized for rapid dispersion in


D X For rheology control in toothpaste formulations.
sorbitol solution.

apertures and inter-molecule spaces. These interactions show viable immobilization of DNA or proteins for bio-sensing [50-54] and
mechanisms such as hydrogen bonding, cation linking, cation synthesis of inorganic nanoparticles with biomedical functionalities
altercation, hydrophobic interactions, anion interchange and proton [55-58]. This review discusses the application of LBNMs in differ-
transference; all dependant on pH of the medium, electrostatic ent areas of biomedicine; including drug delivery, tissue engineer-
characteristics and size of the adjoining molecule [28,36,40,41]. ing, imaging, biosensors and cell adhesion and proliferation. Lastly,
The characteristic précised surface area property coupled with this we advance prospects conclusion on the adoption of LBNMs in
unique charge distribution provides numerous excellent interaction various medical fields. LBNMs have had an explosion of
behaviors between LAP nanodisks and other biomolecules [42,43]. publications in recent years, and therefore our review could not
The high edge area to surface ratio of LAP is suitable for edge cover all these excellent published works.
modifications. Apart from grafting, surface modifications can also
be performed by ion exchange, leading to the production of LAP 3. APPLICATIONS OF HYBRID LAPONITE-BASED
with a wide range of properties. Furthermore, other properties of NANOMATERIALS
LAP such as biocompatibility and inertness have strongly enabled Nanoscale LAP materials can be treated with lipophilic mole-
its study and further applications in drug delivery [44-49], cules (Fig. 2 a,b) and hence, provide a broad array of hybrid mate-
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 5

Table 3. Laponite-drug hybrids used for biomedical applicationsa.

Drug Polymer/s Application Form References

Doxorubicin Enhanced solubility, sustain release and therapeutic efficacy. Nanodisk [44]

Doxorubicin (DOX) Complex inhibited tumor growth more significantly than free Nanodisk [45]
DOX

Doxorubicin PEG, PLA pH-enhanced drug release behavior in a sustained manner, Nanohybrid [46]
DOX-loaded nanocarriers effectively internalized by CAL-72
cells (an osteosarcoma cell line).

Doxorubicin (DOX) PAMAM Effectively inhibited the proliferation of KB cells (a human Nanodisk-dedrimer [47]
epithelial carcinoma cell line) than free DOX complex

Folic acid (FA)-DOX APMES Specifically target cancer cells overexpressing high-affinity FA Nanodisk [48]
receptors

Doxorubicin (DOX) PEG-LA Targeted carrier for efficient loading and specific delivery of Nanodisk [49]
different anticancer drugs to liver cancer cells.

Dexamethasone Sodium hyaluro- Enhanced controlled release Gel [70]


nate

Bone Morphogenic Safely harnessed BMP2 for bone induction. Gel [71]
Protein 2 (BMP2)

Silicate nanoparticles PVA, alginate Wound healing Interpenetrating network [72]


(IPN) hydrogel

Tetrazine Alginate Enhanced controlled release Cryogel [73]

Silicate nanoparticles Si-HPMC Cartilage tissue engineering Interpenetrating network [74]


(IPN) hydrogel

Bovine serum albumin Chitosan, PVA Improved the adsorption capacity of BSA hydrogel nanocomposite [75]
(BSA) beads

Donepezil Eudragit® E-100 Enhanced encapsulation efficiency and controlled release Nanoclay hybrid [76]

Doxorubicin (DOX) Alginate Diminished the effect of DOX ion-trapping in the acidic ex- Hydrogels [85]
tracellular environment of the tumor.

Itraconazole Eudragit®E-100, Release of effect of drug was effected by the type of cation and Nanohybrid [86]
aminoal- pH of system.
kylmethacrylate

Doxorubicin PAH-PSS Polyelectrolyte multilayers improved the sustained release Multicoated nanohybrids [89]
properties.

Doxorubicin PLGA) Enhanced the sustained release characteristic with reduced Nanocomposite [91]
cytotoxicity at targeted cells.

Dexamethasone PEG Enhanced physiological stability and sustained pH-responsive Nanoplates [92]
release properties.

Doxorubicin Alginate Enhanced the anticancer activity Nanohybrid [93]


a
Abbreviations: Polyethylene oxide (PEO); Four-armed poly(ethylene glycol) (PEG-D4); Poly(N-isopropylacrylamide) (PNIPAM); Polyvinyl alcohol,
(PVA)-alginate (LAP:PVA-Alginate); Silated hydroxypropylmethyl cellulose (Si-HPMC); Polylactic acid (PLA); poly(amidoamine) (PAMAM); polyethylene
glycol-linked lactobionic acid (PEG-LA); 3-aminopropyldimethylethoxysilane (APMES); poly(allylamine) hydrochloride, poly(sodium styrene sulfonate)
(PAH/PSS); Poly(lactide-co-glycolide) (PLGA)

rials with distinctive properties for several potential applications sustained release of poorly aqueous soluble materials and also for
[35,59-63]. The treated nanoscale LAP materials can be used in refining their bioavailability.
several applications which include drug delivery, tissue engineer-
ing, biosensors and cell adhesion and proliferation. In biomedical 3.1. Drug Delivery
applications (Table 3), particularly in nanomedicine, this material LAP has numerous vital technical applications, which go be-
embraces great potential since it acts as a relevant drug carrier for yond the conventional usage of clays in pharmaceutics and cosmet-
6 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

ics. The high chances of probable interactions amongst LAP and wound curative applications at ideal concentration of LAP (0.5%).
biological molecules induce exciting opportunities for LAP loaded Furthermore, these LAP complexes show appropriate swelling,
biomolecule or drug delivery systems. In the perspective of oral improved blood coagulation activity and mechanical properties.
drug/biomolecule delivery, distinctive pharmacological formula- Koshy et al. [73] prepared an injectable nanocomposite hydrogel
tions include diffusing LAP molecules in aqueous or oil mediums system by incorporating LAP into hydrogel containing encapsulated
entailing drug moiety and, succeeding stabilization, and then re- protein. This strategy of simple and versatile controlled release of
gaining of LAP-drug hybrid as a compact segment, further embed- recombinant proteins prominently simplifies the design of hydrogel
ded into tablet form. Thus, the intercalation of itraconazole [64], systems for sustained drug delivery and therapeutic protein release
donepezil [76] and amoxicillin [44] into LAP composites has been applications. Boyer et al. [74] developed a hydrogel system by
considered for controlled drug distribution applications [60,64-76]. mixing LAP clay with silated hydroxypropylmethyl cellulose (Si-
Jung et al. [64] prepared an Itraconazole-Laponite (ITA-LAP) HPMC) for procuring hybrid IPN of Si-HPMC-LAP. The use of Si-
hybrid by intercalating ITA in between interlayer space of LAP and HPMC-LAP hydrogels, combined with chondrocytes in subcutis of
de-intercalating in the ionized state. The LAP clay was used as a nude mice, exhibited their impending usage for cartilage tissue
solubility controller and the intercalation of drug molecules into engineering, prominently in the treatment of articular cartilage de-
interlayer space in LAP conjugates improved the dissolution rate of fects. Mahdavinia et al. [75] developed Chitosan-PVA-LAP mag-
ITA. Gaharwar et al. [65] investigated the capability of LAP cross- netic hydrogel beads by the solution-mixing method and were used
linked polyethylene oxide (PEO) nanocomposites as bioactive scaf- for the adsorption of bovine serum albumin. Magnetic Fe3O4 nano-
folds by studying the in vitro cytocompatibility of LAP-PEO cross- composites were initially restrained on LAP sheets with decent
linked nanocomposite layers using MC3T3-E1 mouse preosteoblast dispersing aptitude in order to evade any accumulation of nanopar-
cells. Calabrese et al. [66] designed a surfactant-modified organo- ticles in chitosan-PVA matrices. Furthermore, the addition of LAP
clay (OC) based delivery system for the oral intake of cinnamic nanoparticles to the nanocomposite substrates enhanced diversity
acid (CA). The existence of LAP (bio-surfactant) improved the and propagation of cells.
drug loading efficacy by about 2 times. The drug-LAP hybridiza- The most important property is the two discrete positions on the
tion method can be responsible for a novel way of formulating nano LAP nanodisk surfaces and the edges of the LAP nanoclays which
drug delivery devices with controlled release and solubility utilities. contributes to their multi-functionality as markers and transporters.
It also acts as an innovative drug delivery carrier with controlled Functional groups are essential for further bio-conjugation or cova-
release features. The half-release time (t1/2) and drug encapsulation lent add-on of functional moieties that can be familiarized via cova-
efficiency of the drug depend directly upon the LAP content in the lent bonding of sensitive molecules like the propylsiloxanes shown
formulation. Liu et al. [67] prepared a novel injectable nano- in Fig. (2a), at the edge –OH groups [37,80,81]. Co-adsorption of
complex tissue adhesive hydrogel entailing of dopamine (DOPA) polyethylene glycol (PEG) and alkylammonium ions in the clay
impregnated with (Polyethylene glycol- LAP) PEG/LAP matrix. layers or on the clay surfaces can modify the rheological properties
Incorporation of 2 wt % LAP favorably amplified the bulk me- and solubility [82,83] and similarly, the pyrophosphate ions (Fig.
chanical assets, the rate of curing, and adhesive property of the 2c) are used as supplementary stabilizers in the arrangement of high
adhesive hydrogel due to its robust interfacial binding with DOPA, ionic strength dispersions [84]. Furthermore, the capacity of LAP to
ascertaining it as an effective method for endorsing bioactivity in a perform as a DOX drug moiety carrier was established as a speci-
bioinert. Stempfle et al. [68] examined the dispersion of LAP men of a diagnostically pertinent application [44,85] and this is
nanoparticles, that aided as a cross-linking agent to attain enhanced because the LAP interlayer voids can be used for effective drug
particle properties in poly-N-isopropylacrylamide (PNIPAM) LAP encapsulation with enhanced retaining capacity. In particular, nano-
complex hydrogels. The main tactic of the study was to envisage clays have demonstrated to be excellent candidates for drug deliv-
the gesture of nanoparticles in hydrogels which would allow re- ery carriers as they show the ability to accommodate bulky particles
searchers for an improved consideration of the dynamics within or functional groups into their interlayer voids of 2-D alumino-
these complex systems. Ghadiri et al. [69] formulated Laponite- silicate sheets and finally release the drug complexes by ion ex-
Mafenide (LAP-Maf) hydrogel by solution an improved intercala- change system [27].
tion method and LAP-Maf-Alginate (LAP-Maf-Alg) complex was A cutting-edge hybrid drug carrier system employing LBNMs
formulated by incorporating LAP-Maf hydrogel complex into stable was developed by Takahashi et al. [83] using porous and spongy
alginate base. Cytotoxicity tests showed that the diffusion of mag- nanocrystals of a bulge LAP, conjugated to a di-block copolymer
nesium (Mg2+) ions from the surface of LAP-Maf hydrogel reduced enclosing PEG and polyamine segment. LAP altered with complex
the cytotoxic effects of Maf, which can further enhance the wound of (ɑ-acetal-poly(ethylene glycol)-block-[poly(2-(N, N-
curative process. Studies showed that the LAP-Maf-Alg film could dimethylamino) ethyl methacrylate)] formed a standardized or-
be used to absorb wound exudates and LAP-Maf hydrogel could be ganic-inorganic hybrid dispersion in the water medium. Due to the
used as a potential wound dressing material. Fraile et al. [70] re- high affinity between the drug moiety and LAP surface, LAP-PEG
ported that dexamethasone (DEX) could be retained on LAP, pri- mixture was used as a carrier for sustained release of poorly water-
marily through hydrogen bonding formation and complexation with soluble compounds. Surface modification with PEG remarkably
sodium counter ions. LAP could be used as a carrier for controlled enhanced the dispersion steadiness of high ionic strength. However,
release and delivery of DEX for the treatment of ophthalmic disor- their consequences led to only a few pharmaceutical requirements
ders. Gibbs et al. [71] confirmed that the adsorptive properties of which included, a drug carrier mode for sustained release of drug
LAP nanoparticle gels were capable to confine bone morphogenic and the enhancement of bioavailability when pyrene, a typical
protein 2 (BMP2) activities that persuaded ectopic bone and signifi- molecule for a poorly water-soluble drug was incorporated for tar-
cantly reduced the effective therapeutic dose in the concentration geted drug delivery. To improve their findings, hydrophilic drugs
range of sub-microgram per ml that was able to intervene os- together with the smectite nanoclays were instead employed to form
teogenesis compared to current methods. Golafshan et al. [72] de- new effective delivery systems.
veloped an interpenetrating polymer network (IPN) complex hy-
drogel comprising LAP-polyvinyl alcohol-alginate (LAP-PVA-Ag). Park et al. [76] successfully used donepezil (DNZ) and different
The varying concentration (0, 0.5, 1 and 2 wt. %) of LAP signifi- clay nanoparticles (LAP, saponite, and montmorillonite) as organic
cantly effected the physical, mechanical and biological characteris- guest molecules and inorganic matrices, respectively and formed
tics of complex IPN hydrogels and the mechanical strength of LAP- DNZ-nano clay hybrids. Their results indicate that DNZ molecule
PVA-Ag considerably improved (upon 2 times) compared to PVA- was effectively interlinked by cation exchange reaction deprived of
Ag. LAP: PVA-Ag hybrid could act as an ultimate hydrogel for any weakening of their conjugated functional groups. The cation
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 7

exchangeability of the LAP regulated the captivation extent and In their reported results, the drug release mechanisms of ITA–LAP
molecular prearrangement of drug particles in the interlayers of were explained; depending upon the kind and variety of cations in
matrices, and the release of sequences of DNZ. The thermal con- the dissolution media, the entire quantity of ITA release varied
stancy of drug fragments was also enhanced after the hybridization during the first 72 hours. So, such an LBNM, when incorporated
with clay (LAP) particles. The release rate was improved by using with suitable cations, could be employed as a drug delivery system
Eudragit® E-100 (bulky cationic polymer); nanocomposites layered with a wide range of consequences.
with this polymer showed a quicker drug release in a shorter time With an improved drug release profile, Wang et al. [44] em-
period. Consequently, the LAP molecules were recommended as an ployed and studied LAP induced nano-carrier system for anticancer
innovative drug delivery carrier moiety with controlled release drug delivery and cancer treatment applications. LAP nanodisks
features. were employed to carry DOX molecule to the cancer cells for can-
Similar to Park et al. [76], Jung et al. [86] followed the works cer treatment applications as shown in Fig. (3). The resultant LAP-
of Takahashi et al. [83] but focused on the enhanced release rate of DOX complex showed a high loading of DOX and exhibited a sus-
the low-water-soluble drug, itraconazole (ITA) in a controlled man- tained release profile of DOX molecules (Fig. 4). Significantly, the
ner. The former had worked on changing from the use of hydro- LAP-DOX complex enhanced the ability to deliver DOX efficiently
phobic drugs to the hydrophilic based drug. Jung's research group to the cancer cells with considerably improved therapeutic efficacy.
employed LAP as a drug carrier, with which ITA was conjugated to Following the work of Wang et al. [44], Xiao et al. [89] reported a
study its drug release patterns. Janssen Pharmaceuticals developed simple method to modify DOX-loaded LAP nanoparticles by alter-
ITA as an antifungal agent with a wide spectrum of treatments, and nate deposition of cationic polyallylamine hydrochloride and ani-
now with pervasive acceptance [87]. ITA is also used for the treat- onic polysodiumstyrene sulfonate (PAH/PSS) polyelectrolytes over
ment of Aspergillus sp. and Candida sp., the most common fungal the surface of DOX-loaded LAP nanoparticles. The multilayer-
pathogens [88]. It is known that water-insoluble drugs, such as ITA coated DOX-LAP nanohybrid effectively inhibited the growth of
show complications of low bioavailability due to their poor rate of MCF-7 cancer cell lines and could be used in the treatment of
release in aqueous media. Numerous types of cations were involved breast cancer. Similar work with DOX was followed through Nair
to control the release rates so as to accelerate the ion exchange reac- et al. [90], who developed a noble method for the controlled release
tion and to curtail the recrystallization of released drug molecules. of DOX using LAP-polylactide-co-glycolide (PLGA) complex

Fig. (3). Schematic illustration of the intercalation of DOX into LAP (Image reproduced with permission from RSC in ref. 44)
8 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

Fig. (4). Confocal laser scanning microscope images of human epithelial carcinoma cells treated with phosphate buffered saline (PBS), free DOX, and
LAP/DOX nanodisks with a DOX concentration of 0.6 µM for 2 h at 37 °C. (Image reproduced with permission from John Wiley and Sons in ref. 44)

(LPC) vesicles, through a single step double-emulsion method NPs makes it an apt carrier for bone-tissue engineering application.
which showed exfoliated morphology and improved the thermal Goncalves et al. [93] prepared DOX-loaded LAP-Alginate (LAP-
stability. The LPC vesicles could be used for controlled drug deliv- AG-DOX) hybrid hydrogels with enhanced stability by integrating
ery with enhanced encapsulation of other hydrophilic and hydro- nanodisks of biocompatible LAP into AG hydrogels using calcium
phobic drugs. Li et al. [91] explored a unique anticancer drug deliv- as a crosslinker. Results showed that the LAP-AG-DOX hybrid
ery system by means of LAP-DOX nanodisks. The in vivo antitu- hydrogels enhanced the antitumor activity of DOX with good cyto-
mor efficiency and systemic toxicity of LAP-DOX nanodisks was compatibility.
illustrated using a tumor-bearing mouse model. Outcomes of the
study revealed that at similar DOX concentration, LAP-DOX nano- 3.2. Tissue Engineering
disks were able to constrain the tumor growth more expressively The major concern of tissue engineering is to advance numer-
than free DOX as well as the survival time of mice treated with ous artificial 3-dimensional shells or scaffolds with suitable chemi-
LAP-DOX complex was more than the mice treated with free DOX. cal and physical properties that can diligently imitate the natural
Roozbahani et al. [92] developed DOX-LAP nanoplates (DL-NPs) extracellular medium [94]. These shells should be biodegradable,
by encapsulating DOX into the interlayer surfaces or voids of LAP wholly blend after implantation, and be absorptive in nature to pro-
nanoplates through a modified intercalation technique. The con- vide a greater surface area to volume ratio for cell proliferation,
trolled release of DOX from LAP nanoplates exhibited pH-sensitive attachment, and differentiation (Table 4). New tissue should
behavior. Cytocompatibility with enhanced control release of DL- quickly form, and no additional surgical procedures should be per-
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 9

Table 4. Laponite-drug/polymer hybrids used for tissue engineering, bioimaging and other significant applicationsa.

Drug/ polymer Application Form References

Tetracycline; PLGA Reduced burst release of the drug and significantly improved the tensile Nanofibres [94]
strength of the polymer nanofibrous mats.

PEG Viability of encapsulated hMSCs was improved Hydrogel [97]

PEG-DTT Adjusted polymer quantity tailored both the degradation rate and modulus of Hydrogel [99]
hydrogels

5-aminovaleric acid; ChiPgA Enhanced the porosity of composite, biocompatible. Scaffold [100]

Gelatin Significantly improved mechanical properties Foam [101]

PLA Supported neovascularization (integral for nutrient transport). Nanocomposite film [102]

BAAm Enhanced the rheological properties of hydrogel. Hydrogel [105]

PEG-DA Significantly enhanced both the compressive and tensile properties Hydrogel/ [107]
Scaffold

PNIPAM Enhanced the mechanical and biological properties. Hydrogel [108]

Strontium ranelate; PCL Enhanced alkaline phosphatase (ALP) activity and maintained cell viability. Scaffold [110]

Sodium alginate, Sodium exhibited excellent adsorption capacity on heavy metal ions, which enhanced Hydrogel [111]
acrylate the adsorption of Ni2+, Cu2+, Zn 2+ and Cd2+ ions by 10.4, 8.0, 23.0,and 14.3 fold
compared to active carbon (AC).

Gellan gum methacrylate; Enhanced gel mechanical properties, formed gels withstand steam sterilization. Nanocomposite hy- [113]
polyethylene glycol di- drogel
methacrylate

PEG; iron oxide (maghemite – Magnetic iron oxide nanoparticles embedded in the laponite matrix exhibited Ferrogel [129]
γ-Fe2O3) strong T2-weighted MRI (magnetic resonance imaging) contrast.

PNIPAM Tissue engineering and bioimaging applications. Hydrogel [130]

PNIPAM; polystyrene Tissue engineering, wound healing and bioimaging applications. Hydrogel [132]

Chitosan; PEO Enhanced tuning of cellular adhesion and control biomineralization. Nanocomposite [133]

Aequorin; PEG Sol–gel encapsulated aequorin protein offers potential as a one shot biolumi- Sol-gel matrix [139]
nescence based biosensor for the determination of Ca2+.
a
Abbreviations: Poly(lactide-co-glycolide) (PLGA); Poly (ethylene glycol) (PEG); Dithiothreitol (DTT); chitosan/polygalacturonic acid (ChiPgA); Polylactic
acid (PLA); N,N‘-methylenebis(acrylamide) (BAAm); Poly(ethylene glycol) diacrylate (PEGDA); Poly(N-isopropylacrylamide) (PNIPAM); Polycaprolactone
(PCL); polyethylene oxide (PEO).

formed after implantation for them to be removed out of the body ous materials with extraordinary mechanical properties. These dis-
[95]. Exceptional contenders for soft-tissue engineering scaffolds tinct properties were found suitable and useful for application in
include hydrogels, which are polymer networks extensively swollen biomedical tissue engineering. In addition to PNIPA, LAP-based
with water [96]. However, extensive application of hydrogels as nanocomposite hydrogels have been synthesized using other classes
ideal scaffolds is limited owing to their bio-inertness [81], reduced of monomers such as acrylamide [105] and acrylic acid [106].
mechanical properties [82] and their fragile behavior after getting To further improve the mechanical properties of the nanocom-
dried [83]. To create mechanically robust hydrogels, nanoclays posite hydrogels for use in tissue engineering, Chien et al. [107]
have been employed as cross-linkers to make nanocomposite hy- synthesized biocompatible poly(ethylene glycol) diacrylate
drogels with improved mechanical properties such as compressive (PEGDA)-LAP hybrid hydrogels which supported both two-
strength and stiffness (modulus) [100-103]. dimensional and three-dimensional (2D and 3D) cell cultures of
Owing to the high interfacial area, nanoclays offer enhanced human mesenchymal stem cell. Incorporation of LAP nanoparticles
mechanical properties due to strengthening when used in a polymer successfully enhanced the mechanical properties (flexible and com-
matrix. Along this line, Haraguchi et al. [104] prepared a modified pressive) of PEGDA hydrogels. The improved mechanical proper-
elastic nanocomposite hydrogel by in situ polymerizations of N- ties of PEGDA/LAP hydrogels highlighted their possible applica-
isopropyl acrylamide (NIPA) monomer with existing LAP nano- tions in tissue engineering.
clays (Fig. 5). The resulting hydrogel was capable of overcoming In spite of these noticeable advances in flexible modulus, the
the limitations of conventional chemically cross-linked polymer toughness and durability attained still had an extent less than that of
networks. The incorporation of LAP nanoclays into poly(NIPA) lenient biological tissues. In 2012, Wang et al. [108] improved the
(PNIPA) hydrogels gave highly stable and structurally homogene- mechanics of PNIPA-LAP nanocomposites by making gel com-
10 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

Fig. (5). Nanocomposite hydrogels for enhanced mechanical properties. In contrast to a) organically cross-linked (OR) PNIPA polymer gels b,c) gels cross-
linked with clay nanoparticles (NC) display marked improvements in strength, elongation, and toughness (numbers represent relative crosslink density). d) The
large degree of spacing between clay cross-linkers allows for long and flexible polymer chains. (Image reproduced with permission from John Wiley and Sons
in ref. 88)

plexes with a suitable coated structure imitating control release of Numerous polysaccharides such as chitosan, alginate and meth-
the drug. The well-arranged nanoscale complex permitted the inte- ylcellulose have been blended with LAP to overcome the brittleness
gration of LAP composites of up to 30 wt%, accomplished a 360- of their single-network hydrogels, hence yielding biomaterials with
fold upsurge in elastic modulus (collectively with a six-fold growth enhanced properties [111,112]. Attempting to form a novel
in hardness and five-fold rise in strength) in contrast to the ortho- nanocomposite hydrogel, recently, Pacelli et al. [113] mixed a
doxly produced PNIPA-LAP nanocomposite gel complexes. The functionalized gellan gum (GG-MA), with LAP. A biocompatible
modulus value of 1.5–43 MPa of these PNIPA-LAP nanocomposite polysaccharide, GG-MA has received increased consideration due
gels signifies the utmost testified worth for a polymeric hydrogel to to its extensive applicability, ranging from drug delivery
date, and, analytically, attains mechanical characteristics similar to applications to cartilage tissue engineering applications [114]. The
that of hard biological tissues such as cornea and cartilage [108]. amalgamation of LAP in GG-MA network was able to form robust
Wang et al. [109] studied the applicability of LAP bioceramic and stable hydrogels with practically unvaried or integral mechani-
for potential and enhanced bone-tissue engineering applications. In cal features after thermal treatment. The thermal behavior was reli-
their work, LAP bioceramic with a smooth surface and somewhat ant on the quantity of nano-clay presented in the polymeric net-
systematic holey arrangements was formed. The LAP bioceramic work, with respect to the solo matrices of GG-MA, which cannot
showed an admirable surface hydrophilicity and protein adsorption sustain decontamination deprived of degradation. This property
activities. With the decent bone healing effect revealed by in vivo signifies an important fact towards the design of biomaterials poten-
experiments, and also excellent biosafety, the synthesized LAP tially beneficial as wound dressing covering or any other significant
bioceramic held an abundant potential for applications in bone tis- biomedical application where a sterile system is required.
sue engineering (Fig. 6). Similarly, Nair et al. [110] recently, were LAP can be established as tissue matrices for engineering a
able to synthesize a complex of strontium ranelate (SRA) with LAP diverse class of tissues, perhaps for craniofacial (dental) and ortho-
in the presence of polycaprolactone (PCL) for bone tissue pedic (soft–hard interfaces) applications. In this context, Gaharwar
engineering applications. et al. [115] explored the structural and mechanical properties of
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 11

Fig. (6). In vitro osteogenic differentiation of rMSCs and in vivo bone repair evaluation.
a) ALP activity (normalized for the DNA content, nmol of transformed substrate per unit of time and per mass of DNA) of rMSCs cultured onto different
substrates in growth medium after 14 days culture. Insert of (a) shows the picture of alizarin red staining of rMSCs cultured onto TCP (left) and LAP ceramic
(right) in growth medium without any inducing factors on day 14. (b–f) show the macroscopic appearance of bone defects (A 2-mm bone defect was created in
the middle of the tibia, which was implanted with LAP ceramic as shown in (d)). (b) and (e) show the macroscopic appearance of defects without and with
implantation for 24 weeks, a trace of LAP ceramic residual is observed in (e) as pointed by an arrow. (c) and (f) show the radiographic images of bone defects
without and with LAP implantation after 24 weeks. (Image reproduced with permission from Elsevier in ref. 93)

covalently as well as physically cross-linked nanocomposite hydro- method comprising a self-assembled, polymer-centered conformal
gels comprising of silicate/LAP nanoparticles and polyethylene coating, assembled by an aqueous-based layer-by-layer (LbL)
glycol (PEG). Nair et al. [116] prepared polycaprolactone (PCL) method, as a dual-purpose biomimetic implant surface which pro-
composite scaffolds comprising LAP-strontium ranelate (LAP-SR) vided sustained release of an antibiotic agent trailed by an active
complex for the controlled release of strontium ranelate. Results growth factor for orthopedic implant applications. The multi-
from the in vitro study using human osteosarcoma cells showed that layered coating entails two parts: a base osteoinductive agent con-
the complex PCL-LAP-SR was used in the treatment of osteoporo- taining a bone morphogenetic protein-2, recombinant human bone
sis and for bone tissue engineering applications. Wu et al. [117] morphogenic protein (rhBMP-2), below an antibacterial constituent,
prepared polyethylene glycol-silicate (PEG-Si) based hydrogels that enclosing gentamicin (GEM). LAP barrier layers improved the
were tangibly cross-linked by silicate nanoparticles, LAP and cross- sequential separation amongst the release of the two drugs and ex-
linked by either Pluronic F127-diacrylate or PEG-diacrylate. The tended the release of the core rhBMP-2 growth factor, ensuring
ensuing networks resisted uniaxial extensions up to 2000%, signify- more physiologically significant dosing of rhBMP-2. Wang et al.
ing that the hydrogel networks can absorb considerable amounts of [120] prepared LAP bioceramics by sintering LAP powder compos-
energy during alteration and so it could be used as a matrix for tis- ites. These LAP bioceramics persuaded the osteoblast differentia-
sue engineering application, pressure sensitive adhesives and tion of rat mesenchymal stem cells (rMSCs) in growth medium
wound dressing uses. Ordikhani et al. [118] prepared chitosan– deprived of inducing factors and hence could be used for the treat-
Laponite (CS-LAP) nanocomposite films by an electrophoretic ment of bone defects and bone-tissue engineering applications.
deposition technique. The intercalation of the CS-LAP and drug
macromolecules, vancomycin (VCM), into silicate matrix enhanced 3.3. Bioimaging
the controlled release of VCM. The cellular outcomes showed that The principle of all imaging techniques, single-photon-emission
the presence of LAP affected cell viability, attachment and alkaline computed tomography (SPECT) and optical imaging), (X-ray or
phosphate activity of MG-63 osteoblast-like cells, applicable in computed tomography (CT), echography, positron emission tomo-
bone-tissue engineering applications. Paula et al. [119] developed a graphy (PET), magnetic resonance imaging (MRI), is to build a 2D
12 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

Fig. (7). T2-Weighted MR images (a) and linear fitting of 1/T2 (b) of LAPFe3O4 and Fe3 O4 NPs at different Fe concentrations (0.005, 0.01, 0.02, 0.04, and 0.08
mM). 1 and 2 represent LAP-Fe3O4 and Fe3O4 NPs, respectively. MR imaging of cancer cells in vitro. T2-Weighted MR images (c) and MR signal intensity
analysis (d) of HeLa cells after treated with PBS, LAP-Fe3O 4 NPs at different Fe concentrations for 6 h. In vivo T2-weighted images (a) of tumor (red circle),
liver (red arrow) and kidney (red star) after intravenous injection of LAP-Fe3O4 NPs (930 µg mL−1 Fe, in 0.1 mL PBS) for 0 h, 2 h, 4 h, and 6 h. In vivo MR
signal intensity (b) of liver, kidney and tumor after intravenous injection of LAP-Fe3O4 NPs at different times. (Image reproduced with permission from RSC in
ref. 33)

Fig. (8). Improved adhesion and proliferation of MSCs on PEO gel films with increasing incorporation of LAPNPs. (Image reproduced with permission from
Elsevier in ref. 143)
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 13

Fig. (9). Protein interaction with the nanoclay hybrids. Fluorescence enhancement (F/F0) of Nile Red on LAP in aqueous dispersion (left) and SternVolmer
quenching plot (F0/F) of Eu(ttfa) 3(topo)2 on LAP in aqueous dispersion (right) after addition of BSA (solid squares) and β-lactoglobulin (open circles), meas-
ured after 16 h of equilibration time. (Image reproduced with permission from RSC in ref. 122)

or 3D image comprehending information that can be used to under- sessed both in vivo and in vitro by means of a human cervical carci-
stand the physiology or metabolism of the organisms. However, noma cell line, the HeLa xeno-grafted tumor model and the HeLa
sometimes contrast is naturally present in the obtained images. To cells, respectively. In both, in vitro tumor cell MRI and in vivo tu-
improve this, contrast agents are administered and their dynamic mor grafted nude mice MRI, there was a noticeable signal intensity
behavior in the body facilitate the observation of the anatomic or alteration in the presence and with the administration of LAP-Fe3O4
pathologic structure, invisible without contrast agent. Recently, NPs nanoparticles (Fig. 7) respectively owing to the cellular uptake of
have increasingly been considered as promising contrast agents the nanoparticles. This exhibits the potential of using LAP-Fe3O4
because of their unique physical, chemical, and biological proper- nanoparticles as a T2-weighted divergence agent for MRI.
ties [121-123] (Fig. 9), particularly in MRI imaging. This is because
currently, MRI is one of the most influential imaging tools for envi- 3.4. Cell Adhesion and Proliferation
sioning the anatomical arrangements of soft tissues with cellular In response, primarily, to the opportunities afforded by LBNMs
and even sub-cellular 3D resolutions [124,125]. MRI is extensively in biomedicine, an evolving cluster of studies has also explored the
used for the diagnosis and identification of diseases such as tumors, interface of cells with these nanomaterials. The application of
cerebrovascular and cardiovascular diseases [126,127]. LAPNPs and composites in cell adhesion and proliferation was
Despite wide use, the sensitivity of MRI in distinguishing the previously coined more into in vitro categorization of cellular reac-
healthy tissues and malignant tissues is lagging. Therefore, en- tions and responses [29]. Notwithstanding lingering questions re-
hanced efficacy of MRI diagnosis is a current [31] urgent task in garding cytocompatibility, numerous studies have revealed not only
the currently designed advanced MRI contrast agents. LAP based usual cellular signals and responses but also on clay-reliant aug-
composite nanomaterials have recently been employed in an at- mentations, predominantly on cell adhesion and propagation upon
tempt to study and expound a possible solution to this problem. gel substrates and polymeric matrices.
Modified LAP technology could act as improved recognition sensi- Haraguchi et al. [130] studied the consequences of the amount
tivities in bioimaging, bioanalysis and biosensing. Felbeck et al. of clay in LAP nanoparticle merged with PNIPA hydrogels on cel-
[128] investigated different approaches for the superficial function- lular feasibility, linkage and propagation. With earlier investiga-
alization of LAP and formulated 25 nm core-sized nanoparticles tions [131,132], organically polymer based PNIPA hydrogels alone
(disk-shaped), as nano-carriers for altered fluorophores. The Si−OH were incompetent to keep up the linkage of the multiplying cells.
functional groups at the edges of these nanodisks were significantly Furthermore, LAP-clay associated PNIPA hybrids were competent
embedded with 3-aminopropyldimethylethoxysilane (APES). The to sustain both the bonding and propagation of HepG2 (immortal-
robust effect of charge on the dye and accumulation affinity on the ized cell line comprising of human liver carcinoma cells), human
illumination of the inevitable dyes at the edges of nanodisks was umbilical vein endothelial cells (HUVECs) and skin fibroblast that
noticed from the consequences during formulation. are intensely related with the concentration of LAP in the formu-
Tzitzios et al. [129], by a single step precipitation method, lated hydrogel. Haraguchi’s group tagged the sharp dependence of
synthesized laponite-gamma-iron oxide (LAP/γ-Fe2O3) composite cellular linkage on LAP quantity in the aforementioned system, to
nanomaterials. The blended nano-composites were investigated by the i) anionic surface charges consulted by the LAP, ii) hydropho-
a sequence of corresponding methods to further explain the struc- bicity of the fine PNIPA substrate by the hydrophilic LAP nano-
tural arrangement and configuration, and also to reveal the mag- composites and iii) amended protein adsorption. A similar LAP
netic properties of the final material. Fe2O3 nanoparticles interca- dependent effect on cell adhesion and propagation was studied.
lated in the LAP matrix exhibited strong T2-weighted MRI diver- Chang et al. [107] found it sufficient to add 2.5 wt% LAP to endure
gence, which was practically 2-fold higher as compared to mono- the linkage and propagation of human mesenchymal stromal/stem
dispersed Fe2O3 nanoparticles (core sizes of ~13 nm). However, no cells (hMSCs) on below par cell linked PEG hydrogel films. The
in vivo MRI divergence augmentation effect of the attained LAP-γ- researchers still recommended conceivable importance of LAP
Fe2O3 was present until recently when Ling et al. [31] exploited a nanocomposites in establishing adhesive sites for cellular linkage,
simplistic co-precipitation method to formulate a novel water- attachment and in allowing protein site-specific adsorption.
dispersible LAP-Fe3O4 nanoparticles with enhanced control release
effect. The MRI performance of LAP-Fe3O4 nanoparticles was as-
14 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

The series of studies by Gaharwar et al. [59, 132-133] and Table 5. Pharmaceutical and non-pharmaceutical applications
Schexnailder et al. [134] on LAP-PEO nanocomposite films fo- of laponite in various fields.
cused on the utility and potential of LAP nanoclays in enabling
adhesion and proliferation of cells on polymeric gels. Like many
biopolymers, PEO is non-fouling and hydrophilic moiety; and is Personal care • Skincare and sunscreen emulsions
thus resilient to the cellular linkage. Yet still, PEO is frequently products • “Emulsifier-free” emulsion systems
used to synthesize physically cross-linked PEO–LAP hydrogels and
• Alphahydroxy acid creams
(PNIPA)-based polymers employed to amalgamate mostly cova-
lently cross-linked LAP-polymer films or hydrogels [135-137]. The • Toothpaste
adhesion of NIH3T3 fibroblasts [133, 134], mesenchymal stem • Color cosmetics
cells (MSCs) [110] and MC3T3-E1 preosteoblasts [59] was found
to be extremely reliant on the ratio of PEO added to LAP. At no- • Gelled skin cleansers and Depilatory creams
ticeable ratios of LAP to PEO of 40% and beyond, fibroblasts were • Exfoliant and astringent cleansers
noticed to attach accurately and multiply significantly (Fig. 8). In-
• Nail lacquers and Shampoos
creasing the nanoclay amount up to 70% further improved cell
proliferation. Prominently, these results were constantly perceived Paper and • Static dissipative (antistatic) coatings
in the growth phase and also in the overall amount of cells polymer films
delivered at the plateau phase, signifying the presence of bonding • Electrographic paper and film
edges and sites afforded by the LAP, signifying LAP nano- • Inert barrier films
composites as a governing factor [134,135].
• Anti-blocking coatings
3.5. Biosensors • Paper coating colours
Most results in biology are complex, nonlinear, transient and • Ink jet coatings
very indistinct. Conversely, improved speed, quantity and sensitiv-
• Industrial specialty papers
ity are a prerequisite in biomedicine [138]. Extensively optical sen-
sors that use hybrid NPs center on the recognition of analytes and • Microparticle for retention and drainage systems
molecules that either absorb or radiate light. These sensors signifi-
cantly depend on the involvement of biomolecules that comprises Surface coat- • Decorative & architectural finishes
fluorescent or chromophoric groups (i.e. hemeproteins, flavopro- ings • Textured coatings
teins, and fluorescently-labeled proteins). These biomolecules can
• Water-in-water multicolor paint
also be primed by co-entangling a biomolecule along with a fluoro-
phore or chromophore that reacts specifically to the protein- • Automotive OEM and refinish
reconciled reaction (i.e. variation in O2 levels, pH change,). On the • Clearcoats and varnishes
other hand, bioluminescence can be utilized as a signal to specify
the existence of species that modify or change the function of lumi- • Industrial and protective coatings
nescent proteins such as aequorin [139]. LAP hybrid NPs together • Rust conversion coatings
with the already established technologies can act as analytical
• Water reducible alkyds
probes to improve efficiency.
• Wood stains
The protein corona [140-142] and nanoparticle cytotoxicity are
among the most important factors considered while establishing • Decking stains
hybrid nanoparticles in biophotonics. Felbeck et al. [143] recently • Wood varnishes
carried out optical studies on the interaction of LAP nanocompo-
sites or hybrids with β-lactoglobulin and bovine serum albumin • Printing inks
(BSA). In their study, LAP hybrids were loaded with Eu-complex- • Artist’s & children’s paints
Eu (ttfa)3(topo)2 and with Nile Red (fluorescent solvatochromic
• Pigment suspensions
dye). Fluorescent solvatochromic dye Nile Red (NR) was already
previously studied with proteins [144-146], and Eu-complex had House prod- • Oven cleaners and degreasers
also been successfully incorporated into LAP hybrids earlier [147]. ucts
Their investigation suitably followed the behavior of proteins by • Gelled bleach cleaners
using LAP-based nanohybrids. The LAP complex of both the dyes • Sprayable cleaners and spray-with-cling formu-
optically responded to the existence of β-lactoglobulin and BSA. lations
Conversely, while Nile Red manages to express a noticeable emis-
sion enhancement, the Eu-complex somewhat lost emission • Tablet disintegranting agent
strength. Moreover, the emission strength of the NR-LAP-BSA • Carpet shampoos
hybrid was more than for the analogous LAP-β-lactoglobulin com- • Acidic and alkali toilet cleaners
plex. This was attributed to the higher quantity of hydrophobic
positions in BSA, which lead NR molecules to prolix into. These • Hard surface cleaners and air fresheners
results suggest that the performance of proteins can appropriately • Liquid automatic dishwashing detergents
be trailed using dye-LBNMs.
• Antistatic products and anti-redeposition agent
Further, there are lots of general applications of laponite as
multifunctional material including personal care products, paper Agricultural • Seed germination gels
and polymer films, surface coatings, house products, agricultural • Plant rooting gels
usages (Table 5). In the present review, brief compilations of bene-
fits and limitations including general information of various clay • Agrochemical flowables – herbicides, pesticides
nanomaterials have been illustrated in Table 6. Moreover, Table 7 • Essential element suspensions
summarized various preparation techniques followed with
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 15

Table 6. Classification of different types of clay nanomaterials: Benefits and Limitations.

Type of clay Formula Origin Substitution Layer Routes of ad- Dosage form Benefits and Limitations
mineral charge ministration

Montmorillo- Mx(Al2−xMgx) Natural Octahedral Negative Oral Solid (tablets, cap- Low pH yielded; accompa-
nite Si4O10(OH)2 sules, powders); nying minerals depend
·nH2O Liquid (suspensions, mainly on pressure and
emulsions) temperature conditions;
additional minerals depend
Topical Creams, ointments
upon starting compositions.
and powders

Transdermal Films and pathches

Rectal Suspensions, tablets

Vaginal Ointments, creams


and gels

Hectorite Mx(Mg3−xLix) Natural Octahedral Negative Oral Solid (tablets, cap- It is commercially available
Si4O 10(OH) 2 sules, powders); as laponite and talc; talc is
·nH2O Liquid (suspensions, relatively cheap and readily
emulsions) available source.

Saponite Mx Mg3(Si4−xAlx ) Natural Tetrahedral Negative Oral Solid (tablets, cap- Poor crystallinity at ≤ 200
O10(OH) 2·nH2O sules, powders); °C; no complex formation
Liquid (suspensions, during gel preparation;
emulsions) amount of amorphous mate-
rial depends on time and
amount of NH4OH; addition
of Mg improves the forma-
tion of saponite.

Flurohectorite Mx(Mg3−xLix) Synthetic Octahedral Negative Oral Solid (tablets, cap- Very small flakes as by-
Si4O 10 F2·nH2O sules, powders); products; excess LiF (start-
Liquid (suspensions, ing material) needed to
emulsions) form flurohectorite.

Laponite Mx(Mg3−x Lix) Synthetic Octahedral Negative Oral Solid (tablets, cap- Very much useful in phar-
Si4O 10(OH) 2 sules, powders); maceutical, tissue engineer-
·nH2O Liquid (suspensions, ing and biomedical applica-
emulsions) tion due to its smooth deg-
radation. Used as rheology
Topical Creams, ointments
modifier. Thermal and
and powders
pressure sensitive.
Transdermal Films and pathches

Fluromica NaMg2.5 Si4O 10 F2 Synthetic Octahedral Negative Topical Film and patches Impurities from starting
material zeolite stuff re-
mains; < 10% amorphous.

Kaolinite Al2Si2O5 (OH)4 Natural - Neutral Oral Solid (tablets, cap- Na-gel resulted in degrada-
sules, powders); tion at 350 °C to byprod-
Liquid (suspensions, ucts.; many metal substitu-
emulsions) tions are shown to be possi-
ble.
Topical Film and patches

Local Powder and polymer


mixtures

(Table 6) Contd....
16 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

Type of clay Formula Origin Substitution Layer Routes of ad- Dosage form Benefits and Limitations
mineral charge ministration

Halloysite Al2Si2O5 (OH)4 Natural - Neutral Oral Solid (tablets, cap- Forms dilute solutions;
·2H2O sules, powders); better result with NH 4 and
Liquid (suspensions, F.
emulsions)

Topical Film and patches

Local Powder and polymer


mixtures

Kanemite Na2Si4 O9 Natural/ Tetrahedral Negative Oral Solid (tablets, cap- Identical to montmorillo-
·5H2O Synthetic sules, powders); nite.
Liquid (suspensions,
emulsions)

Makatite NaHSi2O 5 Natural/ Tetrahedral Negative Rectal Suspensions, tablets Not specific.
·7H2O Synthetic

Octasilicate Na2Si8 O17 Synthetic Tetrahedral Negative Local Powder and polymer Exact structural elucidations
·9H2O mixtures could not be identified.

Magadiite Na2Si14O29 Natural/ Tetrahedral Negative Topical Film and patches Used for electrochemical
·10H2O Synthetic studies specifically.

Kenyaite Na2Si20O4 Synthetic Tetrahedral Negative Local Powder and polymer Not specific.
·10H2O mixtures

Table 7. Various preparation techniques of clay nanocomposites: advantages and disadvantages.

Preparation Technique Driving Force Advantages Disadvantages

In-situ polymerization Interaction strength between mono- Suitable for low or non-soluble poly- Clay exfoliation depends on the extent of
mer and clay surface; Enthalpic mers; A conventional process for ther- clay swelling and diffusion rate of
evolvement during the interlayer moset nanocomposites. monomers in the gallery; Oligomer may
polymerization. be formed upon incompletely polymeri-
zation.

Solution exfoliation Entropy gained by desorption of Prefer to water-soluble polymers. Compatible polymer-clay solvent system
solvent, which compensates for the is not always available; Use of large
decrease in conformational entropy quantities of solvent; Co-intercalation
of intercalated polymers. may occur for solvent and polymer.

Melt intercalation Enthalpic contribution of the poly- Environmental benign approach; No Slow penetration (transport) of polymer
mer-organoclay interactions. solvent is required; Nanocomposites can within the confined gallery.
be processed with conventional plastic
extrusion and molding technology

advantages and disadvantages of the various clay nanomaterials. reaction of the drug with LAP, whatsoever the physical adsorption
Thus, these clay nanomaterials need to be explored further as safe or intercalation, results not only in improved solubility and phys-
drug delivery carriers for challenging molecules with obvious ad- icochemical stability of drug molecules, but also offers a controlled
vantages. Natural and synthetic need much attention of the research and sustained drug-release rate, and eventually achieves improved
scientists working on nanomedicine as drug carrier for the recent bioavailability due to efficient drug delivery. In various drug deliv-
era. ery systems, LAP-drug hybrid systems or LBNM’s show admirable
biocompatibility, greater anion exchange ability, and pH-sensitive
4. FUTURE PROSPECTIVE solubility interrelated with drug load content. Furthermore, it is
LAP as formulation additives and drug carriers are applied by a established that the significant inorganic materials such as LAP and
variety of routes including oral, transdermal, and local administra- their intercalated hybrids or nanocomposite hybrids would allow for
tions. The amount of LAP, depending on the routes of administra- the development of novel polymer hybrids or composite materials
tion for biomedical applications, can be selected in order to control with advanced mechanical properties, applicable for biomedical
key parameters including size, swellability, mucoadhesiveness, applications. The advanced developments of LAP-drug hybrid sys-
dispersibility, and drug-release rate. In addition, the hybridization tems or LBNM, via various routes of administration, will allow the
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 17

medication to be lodged to specific targeted parts of the body (or- [2] Gómez-Romero P, Sanchez C. Hybrid Materials, Functional Appli-
gans or cells). Although the field of clay-drug hybrid systems has cations. An Introduction, in Functional Hybrid Materials. 2005,
attained significant attention over the past decade with many chal- Wiley-VCH Verlag GmbH & Co. KGaA. p. 1-14.
lenges remained. From general prospects, LAP-drug hybrid systems [3] Gutierrez MC, Ferrer ML, Tartaj P del Monte F. Biomedical Ap-
plications of Organic–Inorganic Hybrid Nanoparticles. Hybrid
or LBNM’s seemed to be very effective and to work well in vitro, Nanocomposites for Nanotechnology: Electronic, Optical, Mag-
but some failed in in vivo performance due to complications in- netic and Biomedical Applications 2009; 707-68.
volved in real physiological conditions. Furthermore, toxicological [4] Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev
studies are required to determine whether the LAP-drug hybrid 2006; 58(14): 1532-55.
systems or LBNM’s can be applied with judiciously rationalized [5] Burghardt S, Hirsch A, Schade B, Ludwig K, Böttcher C.
clinical outcomes. The gene delivery and programed DDS (tar- Switchable supramolecular organization of structurally defined mi-
getability and/or environmental stimuli responsiveness, with clay celles based on an amphiphilic fullerene. Angew Chem Int Ed Engl
minerals) are further challenges. The innovative design in terms of 2005; 44(19): 2976-9.
formulations with clay-drug hybrids advance clinical use and con- [6] Brettreich M, Burghardt S, Böttcher C, Bayerl T, Bayerl S, Hirsch
A. Globular Amphiphiles: Membrane-­‐Forming Hexaadducts of
sequently, open new drug-delivery markets. Finally, research on C60. Angewandte Chemie International Edition, 2000.39(10): p.
clay minerals will expand their applications in biomedical fields, 1845-48.
such as regenerative tissue engineering, theranaustics, personalized [7] Klumpp C, Kostarelos K, Prato M, Bianco A. Functionalized car-
medicine, and implants. bon nanotubes as emerging nanovectors for the delivery of thera-
peutics. Biochim Biophys Acta 2006; 1758(3): 404-12.
CONCLUSION [8] Kamalha E, Shi X, Mwasiagi JI, Zeng Y. Nanotechnology and
As documented by a large number of papers and books, carbon nanotubes; A review of potential in drug delivery. Macro-
HLBNMs have been widely studied and applied in the biomedical mol Res 2012; 20(9): 891-8.
[9] Shi X, Thomas TP, Myc LA, Kotlyar A, Baker JR Jr. Synthesis,
field. LAP is an excellent material for biomedical applications, characterization, and intracellular uptake of carboxyl-terminated
coupled with cost and excellent properties ranging from chemical to poly(amidoamine) dendrimer-stabilized iron oxide nanoparticles.
mechanical properties. There is immense potential for their study Phys Chem Chem Phys 2007; 9(42): 5712-20.
and use in biomedicine. Important still, is their use with polymeric [10] Gupta AK, Gupta M. Synthesis and surface engineering of iron
additives which presents new LAP–drug interaction, release oxide nanoparticles for biomedical applications. Biomaterials 2005;
mechanisms and strength characteristics. These are essential for the 26(18): 3995-4021.
formulation of drug delivery systems, tissue engineering materials, [11] Nishiyama N, Kataoka K. Current state, achievements, and future
cell imaging, adhesion and proliferation mechanisms, and as bio- prospects of polymeric micelles as nanocarriers for drug and gene
sensors. In each application, new opportunities are arising from the delivery. Pharmacol Ther 2006; 112(3): 630-48.
[12] Kwon GS, Okano T. Polymeric micelles as new drug carriers. Adv
need for ideal materials to further commercialization for use on a Drug Deliv Rev 1996; 21(2): 107-16.
large scale. Furthermore, the synthesis of these materials at [13] Villalonga-Barber C, Micha-Screttas M, Steele BR, Georgopoulos
nanoscale and incorporation with other materials, make LBNMs A, Demetzos C. Dendrimers as biopharmaceuticals: synthesis and
signals possible future breakthroughs in biomedicine. properties. Curr Top Med Chem 2008; 8(14): 1294-309.
However, in spite of the quick improvement in respect to [14] Svenson S, Tomalia DA. Dendrimers in biomedical applications—
LBNMs, these novel nanocomposites must be prudently and wholly reflections on the field. Adv Drug Deliv Rev 2012; 64: 102-15.
[15] Lal S, Clare SE, Halas NJ. Nanoshell-enabled photothermal cancer
assessed for biocompatibility to be applicable for biomedical therapy: impending clinical impact. Acc Chem Res 2008; 41(12):
applications. Their novelties in biomedicine are fairly well- 1842-51.
established and are peculiarly understood regarding their physio- [16] Barratt G. Colloidal drug carriers: achievements and perspectives.
logical interactions with different living tissues. Additionally, Cell Mol Life Sci 2003; 60(1): 21-37.
among the numerous research articles mentioning the adoption of [17] Wang X, Wang Y, Chen ZG, Shin DM. Advances of cancer ther-
LBNMs, no systematic estimation and evaluation of the biocom- apy by nanotechnology. Cancer Res Treat 2009; 41(1): 1-11.
patibility of any LBNMs have been accomplished or published. [18] Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an
With this done, in the future, we can anticipate further extensive emerging treatment modality for cancer. Nat Rev Drug Discov
translational research on LAP, mainly in the areas of immune 2008; 7(9): 771-82.
[19] Ferrari M. Cancer nanotechnology: opportunities and challenges.
modulation, imaging, and cancer research. The therapeutic advan- Nat Rev Cancer 2005; 5(3): 161-71.
tages, bioactive ness and surface properties of modified LBNMs in [20] Alexis F, Pridgen E, Molnar LK, Farokhzad OC. Factors affecting
biomedical applications are a requirement, and the question as to the clearance and biodistribution of polymeric nanoparticles. Mol
whether these materials can become a center of focus as a field of Pharm 2008; 5(4): 505-15.
investigation for high-priority development. [21] Li S-D, Huang L. Pharmacokinetics and biodistribution of nanopar-
ticles. Mol Pharm 2008; 5(4): 496-504.
CONSENT FOR PUBLICATION [22] Wu X, Zhu W. Stability enhancement of fluorophores for lighting
Not applicable. up practical application in bioimaging. Chem Soc Rev 2015;
44(13): 4179-84.
FUNDING   [23] Yu Z, Schmaltz RM, Bozeman TC, et al. Selective tumor cell tar-
geting by the disaccharide moiety of bleomycin. J Am Chem Soc
None.   2013; 135(8): 2883-6.
[24] Ji D-K, Zhang Y, Zhang Y, et al. Receptor-targeting fluorescence
CONFLICT OF INTEREST imaging and theranostics using a graphene oxide based su-
The authors declare no conflict of interest, financial or other- pramolecular glycocomposite. J Mater Chem B Mater Biol Med
wise. 2015; 3(47): 9182-5.
[25] Balducci A, Wen Y, Zhang Y, et al. A novel probe for the non-
ACKNOWLEDGEMENTS invasive detection of tumor-associated inflammation. OncoImmu-
nology 2013; 2(2): e23034.
Declared none. [26] Choy J-H, Jung JS, Oh JM, et al. Layered double hydroxide as an
efficient drug reservoir for folate derivatives. Biomaterials 2004;
REFERENCES 25(15): 3059-64.
[1] Taylor-Pashow KML, Della Rocca J, Huxford RC, Lin W. Hybrid [27] Park T-U, Jung H, Kim H-M, Choy J-H, Lee C-W. Hybrid of itra-
nanomaterials for biomedical applications. Chem Commun (Camb) conazole, cyclosporine or carvedilol with a layered silicate and a
2010; 46(32): 5832-49. process for preparing the same 2003. Patent no. WO2004009120A1
18 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

[28] Bergaya F, Lagaly G. General introduction: clays, clay minerals, cling. 3. Theoretical and experimental study of the phenol-
and clay science. Handbook of clay science, 2006. 1: p. 1-18. polyphenol oxidase system immobilized in Laponite hydrogels and
[29] Dawson JI, Oreffo RO. Clay: new opportunities for tissue regenera- layer-by-layer self-assembled structures. Anal Chem 2001; 73(14):
tion and biomaterial design. Adv Mater 2013; 25(30): 4069-86. 3206-18.
[30] Ruiz-Hitzky E. Molecular access to intracrystalline tunnels of [54] Shan D, Mousty C, Cosnier S, Mu S. A new polyphenol oxidase
sepioliteBasis of a presentation given at Materials Discussion No. biosensor mediated by Azure B in laponite clay matrix. Electroana-
3, 24–26 September 2000, University of Cambridge, UK. J Mater lysis 2003; 15(19): 1506-12.
Chem 2001; 11(1): 86-91. [55] Aihara N, Torigoe K, Esumi K. Preparation and Characterization of
[31] Ding L, Hu Y, Luo Y, et al. LAPONITE®-stabilized iron oxide Gold and Silver Nanoparticles in Layered Laponite Suspensions.
nanoparticles for in vivo MR imaging of tumors. Biomater Sci Langmuir 1998; 14(17): 4945-9.
2016; 4(3): 474-82. [56] Lambert Y, Le Dantec R, Mugnier Y, et al. Second-Harmonic
[32] Ruzicka B, Zaccarelli E. A fresh look at the Laponite phase dia- Generation Imaging of LiIO 3 /Laponite Nanocomposite
gram. Soft Matter 2011; 7(4): 1268-86. Waveguides. Jpn J Appl Phys 2006; 45(9S): 7525.
[33] Tawari SL, Koch DL, Cohen C. Electrical double-layer effects on [57] Szabó T, Bakandritsos A, Tzitzios V, et al. Magnetic iron ox-
the brownian diffusivity and aggregation rate of laponite clay parti- ide/clay composites: effect of the layer silicate support on the mi-
cles. J Colloid Interface Sci 2001; 240(1): 54-66. crostructure and phase formation of magnetic nanoparticles.
[34] Carrow JK, Gaharwar AK. Bioinspired polymeric nanocomposites Nanotechnology 2007; 18(28): 285602.
for regenerative medicine. Macromol Chem Phys 2015; 216(3): [58] Lambert Y, Le Dantec R, Mugnier Y, et al. Second-harmonic gen-
248-64. eration imaging of LiIO3/laponite nanocomposite waveguides. Jpn
[35] Gaharwar AK, Peppas NA, Khademhosseini A. Nanocomposite J Appl Phys 2006; 45(9S): 7525.
hydrogels for biomedical applications. Biotechnol Bioeng 2014; [59] Gaharwar AK, Schexnailder PJ, Kline BP, Schmidt G. Assessment
111(3): 441-53. of using laponite cross-linked poly(ethylene oxide) for controlled
[36] Chimene D, Alge DL, Gaharwar AK. Two-Dimensional Nanoma- cell adhesion and mineralization. Acta Biomater 2011; 7(2): 568-
terials for Biomedical Applications: Emerging Trends and Future 77.
Prospects. Adv Mater 2015; 27(45): 7261-84. [60] Viseras C, Cerezo P, Sanchez R, Salcedo I, Aguzzi C. Current
[37] Herrera NN, Letoffe J, Putaux J, David L, Bourgeat-Lami E. challenges in clay minerals for drug delivery. Appl Clay Sci 2010;
Aqueous dispersions of silane-functionalized laponite clay plate- 48(3): 291-5.
lets. A first step toward the elaboration of water-based poly- [61] Lotsch BV, Ozin GA. Clay Bragg Stack Optical Sensors. Adv
mer/clay nanocomposites. Langmuir 2004; 20(5): 1564-71. Mater 2008; 20(21): 4079-84.
[38] Loiseau A, Tassin J-F. Model nanocomposites based on laponite [62] Dey D, Bhattacharjee D, Chakraborty S, Hussain SA. Development
and poly (ethylene oxide): preparation and rheology. Macromole- of hard water sensor using fluorescence resonance energy transfer.
cules 2006; 39(26): 9185-91. Sens Actuators B Chem 2013; 184: 268-73.
[39] Cocard S, Tassin JF, Nicolai T. Dynamical mechanical properties [63] Ramamurthy V, Schanze KS. Solid state and surface photochemis-
of gelling colloidal disks 2000. try 2000.
[40] Mourchid A, Lecolier E, Van Damme H, Levitz P. On viscoelastic, [64] Jung H, Kim HM, Choy YB, Hwang SJ, Choy JH. Laponite-based
birefringent, and swelling properties of Laponite clay suspensions: nanohybrid for enhanced solubility and controlled release of itra-
revisited phase diagram. Langmuir 1998; 14(17): 4718-23. conazole. Int J Pharm 2008; 349(1-2): 283-90.
[41] Ramsay J. Colloidal properties of synthetic hectorite clay disper- [65] Gaharwar AK, Kishore V, Rivera C, et al. Physically crosslinked
sions: I. Rheology. J Colloid Interface Sci 1986; 109(2): 441-7. nanocomposites from silicate-crosslinked PEO: mechanical proper-
[42] Lezhnina MM, Grewe T, Stoehr H, Kynast U. Laponite blue: dis- ties and osteogenic differentiation of human mesenchymal stem
solving the insoluble. Angew Chem Int Ed Engl 2012; 51(42): cells. Macromol Biosci 2012; 12(6): 779-93.
10652-5. [66] Calabrese I, Gelardi G, Merli M, Liveri ML, Sciascia L. Clay-
[43] Elzbieciak M, Wodka D, Zapotoczny S, Nowak P, Warszynski P. biosurfactant materials as functional drug delivery systems: Slow-
Characteristics of model polyelectrolyte multilayer films containing ing down effect in the in vitro release of cinnamic acid. Appl Clay
laponite clay nanoparticles. Langmuir 2010; 26(1): 277-83. Sci 2017; 135: 567-74.
[44] Wang S, Wu Y, Guo R, et al. Laponite nanodisks as an efficient [67] Liu Y, Meng H, Konst S, Sarmiento R, Rajachar R, Lee BP. In-
platform for Doxorubicin delivery to cancer cells. Langmuir 2013; jectable dopamine-modified poly(ethylene glycol) nanocomposite
29(16): 5030-6. hydrogel with enhanced adhesive property and bioactivity. ACS
[45] Li K, Wang S, Wen S, et al. Enhanced in vivo antitumor efficacy of Appl Mater Interfaces 2014; 6(19): 16982-92.
doxorubicin encapsulated within laponite nanodisks. ACS Appl [68] Stempfle B, Große A, Ferse B, Arndt KF, Wöll D. Anomalous
Mater Interfaces 2014; 6(15): 12328-34. diffusion in thermoresponsive polymer-clay composite hydrogels
[46] Wang G, Maciel D, Wu Y, et al. Amphiphilic polymer-mediated probed by wide-field fluorescence microscopy. Langmuir 2014;
formation of laponite-based nanohybrids with robust stability and 30(46): 14056-61.
pH sensitivity for anticancer drug delivery. ACS Appl Mater Inter- [69] Ghadiri M, Chrzanowski W, Rohanizadeh R. Antibiotic eluting
faces 2014; 6(19): 16687-95. clay mineral (Laponite®) for wound healing application: an in vitro
[47] Mustafa R, Luo Y, Wu Y, Guo R, Shi X. Dendrimer- study. J Mater Sci Mater Med 2014; 25(11): 2513-26.
Functionalized Laponite Nanodisks as a Platform for Anticancer [70] Fraile JM, Garcia-Martin E, Gil C, et al. Laponite as carrier for
Drug Delivery. Nanomaterials (Basel) 2015; 5(4): 1716-31. controlled in vitro delivery of dexamethasone in vitreous humor
[48] Wu Y, Guo R, Wen S. Folic acid-modified laponite nanodisks for models. Eur J Pharm Biopharm 2016; 108: 83-90.
targeted anticancer drug delivery. J Mater Chem B Mater Biol Med [71] Gibbs DM, Black CR, Hulsart-Billstrom G, et al. Bone induction at
2014; 2(42): 7410-8. physiological doses of BMP through localization by clay nanopar-
[49] Chen G, Li D, Li J. Targeted doxorubicin delivery to hepatocarci- ticle gels. Biomaterials 2016; 99: 16-23.
noma cells by lactobionic acid-modified laponite nanodisks. New J [72] Golafshan N, Rezahasani R, Tarkesh Esfahani M, Kharaziha M,
Chem 2015; 39(4): 2847-55. Khorasani SN. Nanohybrid hydrogels of laponite: PVA-Alginate as
[50] de Melo JV, Cosnier S, Mousty C, Martelet C, Jaffrezic-Renault N. a potential wound healing material. Carbohydr Polym 2017; 176:
Urea biosensors based on immobilization of urease into two oppo- 392-401.
sitely charged clays (laponite and Zn-Al layered double hydrox- [73] Koshy ST, Zhang DKY, Grolman JM, Stafford AG, Mooney DJ.
ides). Anal Chem 2002; 74(16): 4037-43. Injectable nanocomposite cryogels for versatile protein drug deliv-
[51] Mousty C. Biosensing applications of clay-modified electrodes: a ery. Acta Biomater 2018; 65: 36-43.
review. Anal Bioanal Chem 2010; 396(1): 315-25. [74] Boyer C, Figueiredo L, Pace R, et al. Laponite nanoparticle-
[52] Poyard S, Jaffrezic-Renault N, Martelet C, Cosnier S, Labbé P, associated silated hydroxypropylmethyl cellulose as an injectable
Besombes JL. A new method for the controlled immobilization of reinforced interpenetrating network hydrogel for cartilage tissue
enzyme in inorganic gels (laponite) for amperometric glucose bio- engineering. Acta Biomater 2018; 65: 112-22.
sensing. Sens Actuators B Chem 1996; 33(1): 44-9. [75] Mahdavinia GR, Soleymani M, Etemadi H, Sabzi M, Atlasi Z.
[53] Coche-Guerente L, Labbé P, Mengeaud V. Amplification of am- Model protein BSA adsorption onto novel magnetic chito-
perometric biosensor responses by electrochemical substrate recy-
Laponite-based Nanomaterials for Biomedical Applications Current Pharmaceutical Design, 2019, Vol. 25, No. 00 19

san/PVA/laponite RD hydrogel nanocomposite beads. Int J Biol [100] Ambre AH, Katti KS, Katti DR. Nanoclay based composite scaf-
Macromol 2018; 107(Pt A): 719-29. folds for bone tissue engineering applications. J Nanotechnol Eng
[76] Park JK, Choy YB, Oh JM, Kim JY, Hwang SJ, Choy JH. Con- Med 2010; 1(3): 031013.
trolled release of donepezil intercalated in smectite clays. Int J [101] Frydrych M, Wan C, Stengler R, O'Kelly KU, Chen B. Structure
Pharm 2008; 359(1-2): 198-204. and mechanical properties of gelatin/sepiolite nanocomposite
[77] Ruiz-Hitzky E, Aranda P, Darder M, Rytwo G. Hybrid materials foams. J Mater Chem 2011; 21(25): 9103-11.
based on clays for environmental and biomedical applications. J [102] Baker KC, Manitiu M, Bellair R, Gratopp CA, Herkowitz HN,
Mater Chem 2010; 20(42): 9306-21. Kannan RM. Supercritical carbon dioxide processed resorbable
[78] Aguzzi C, Cerezo P, Viseras C, Caramella C. Use of clays as drug polymer nanocomposite bone graft substitutes. Acta Biomater
delivery systems: possibilities and limitations. Appl Clay Sci 2007; 2011; 7(9): 3382-9.
36(1): 22-36. [103] Haraguchi K. Nanocomposite hydrogels. Curr Opin Solid State
[79] Viseras C, Aguzzi C, Cerezo P, Bedmar MC. Biopolymer–clay Mater Sci 2007; 11(3): 47-54.
nanocomposites for controlled drug delivery. Mater Sci Technol [104] Haraguchi K, Takehisa T. Nanocomposite hydrogels: a unique
2008; 24(9): 1020-6. organic-inorganic network structure with extraordinary mechanical,
[80] Wheeler PA, Wang J, Baker J, Mathias LJ. Synthesis and charac- optical, and swelling/de-swelling properties. Adv Mater 2002;
terization of covalently functionalized laponite clay. Chem Mater 14(16): 1120.
2005; 17(11): 3012-8. [105] Okay O, Oppermann W. Polyacrylamide-clay nanocomposite hy-
[81] Bujdák J, Danko M, Chorvát Jr D, Czímerová A, Sýkora J, Lang K. drogels: Rheological and light scattering characterization. Macro-
Selective modification of layered silicate nanoparticle edges with molecules 2007; 40(9): 3378-87.
fluorophores. Appl Clay Sci 2012; 65: 152-7. [106] Xu K, Wang J, Xiang S, Chen Q, Zhang W, Wang P. Study on the
[82] de Paiva LB, Morales AR, Díaz FRV. Organoclays: properties, synthesis and performance of hydrogels with ionic monomers and
preparation and applications. Appl Clay Sci 2008; 42(1): 8-24. montmorillonite. Appl Clay Sci 2007; 38(1): 139-45.
[83] Takahashi T, Yamada Y, Kataoka K, Nagasaki Y. Preparation of a [107] Chang CW, van Spreeuwel A, Zhang C, Varghese S. PEG/clay
novel PEG-clay hybrid as a DDS material: dispersion stability and nanocomposite hydrogel: a mechanically robust tissue engineering
sustained release profiles. J Control Release 2005; 107(3): 408-16. scaffold. Soft Matter 2010; 6(20): 5157-64.
[84] Mongondry P, Nicolai T, Tassin J-F. Influence of pyrophosphate or [108] Wang J, Lin L, Cheng Q, Jiang L. A strong bio-inspired layered
polyethylene oxide on the aggregation and gelation of aqueous PNIPAM-clay nanocomposite hydrogel. Angew Chem Int Ed Engl
laponite dispersions. J Colloid Interface Sci 2004; 275(1): 191-6. 2012; 51(19): 4676-80.
[85] Gonçalves M, Figueira P, Maciel D, et al. Antitumor efficacy of [109] Wang C, Wang S, Li K, et al. Preparation of laponite bioceramics
doxorubicin-loaded laponite/alginate hybrid hydrogels. Macromol for potential bone tissue engineering applications. PLoS One 2014;
Biosci 2014; 14(1): 110-20. 9(6): e99585.
[86] Jung H, Kim HM, Choy YB, Hwang SJ, Choy JH. Itraconazole– [110] Nair BP, Sindhu M, Nair PD. Polycaprolactone-laponite composite
Laponite: Kinetics and mechanism of drug release. Appl Clay Sci scaffold releasing strontium ranelate for bone tissue engineering
2008; 40(1–4): 99-107. applications. Colloids Surf B Biointerfaces 2016; 143: 423-30.
[87] De Beule K, Van Gestel J. Pharmacology of itraconazole. Drugs [111] Gao T, Wang W, Wang A. A pH-sensitive composite hydrogel
2001; 61(Suppl. 1): 27-37. based on sodium alginate and medical stone: synthesis, swelling,
[88] Jain S, Sehgal VN. Itraconazole: an effective oral antifungal for and heavy metal ions adsorption properties. Macromol Res 2011;
onychomycosis. Int J Dermatol 2001; 40(1): 1-5. 19(7): 739-48.
[89] Xiao S, Castro R, Maciel D, et al. Fine tuning of the pH-sensitivity [112] Wang W, Wang J, Wang A. pH-Responsive nanocomposites from
of laponite-doxorubicin nanohybrids by polyelectrolyte multilayer methylcellulose and attapulgite nanorods: Synthesis, swelling and
coating. Mater Sci Eng C 2016; 60: 348-56. absorption performance on heavy metal ions. Journal of Macromo-
[90] Nair BP, Sharma CP. Poly(lactide-co-glycolide)-laponite-F68 lecular Science. Part A 2012; 49(4): 306-15.
nanocomposite vesicles through a single-step double-emulsion [113] Pacelli S, Paolicelli P, Moretti G, et al. Gellan gum methacrylate
method for the controlled release of doxorubicin. Langmuir 2012; and laponite as an innovative nanocomposite hydrogel for biomedi-
28(9): 4559-64. cal applications. Eur Polym J 2016; 77: 114-23.
[91] Li K, Wang S, Wen S, et al. Enhanced in vivo antitumor efficacy of [114] Osmałek T, Froelich A, Tasarek S. Application of gellan gum in
doxorubicin encapsulated within laponite nanodisks. ACS Appl pharmacy and medicine. Int J Pharm 2014; 466(1-2): 328-40.
Mater Interfaces 2014; 6(15): 12328-34. [115] Gaharwar AK, Schexnailder P, Kaul V, et al. Highly Extensible
[92] Roozbahani M, Kharaziha M, Emadi R. pH sensitive dexametha- Bio-­‐Nanocomposite Films with Direction-­‐Dependent Properties.
sone encapsulated laponite nanoplatelets: Release mechanism and Adv Funct Mater 2010; 20(3): 429-36.
cytotoxicity. Int J Pharm 2017; 518(1-2): 312-9. [116] Nair BP, Sindhu M, Nair PD. Polycaprolactone-laponite composite
[93] Gonçalves M, Figueira P, Maciel D, et al. pH-sensitive scaffold releasing strontium ranelate for bone tissue engineering
Laponite(®)/doxorubicin/alginate nanohybrids with improved anti- applications. Colloids Surf B Biointerfaces 2016; 143: 423-30.
cancer efficacy. Acta Biomater 2014; 10(1): 300-7. [117] Wu CJ, Wilker JJ, Schmidt G. Robust and adhesive hydrogels from
[94] Qi R, Guo R, Shen M, et al. Electrospun poly (lactic-co-glycolic cross-linked poly(ethylene glycol) and silicate for biomedical use.
acid)/halloysite nanotube composite nanofibers for drug encapsula- Macromol Biosci 2013; 13(1): 59-66.
tion and sustained release. J Mater Chem 2010; 20(47): 10622-9. [118] Ordikhani F, Dehghani M, Simchi A. Antibiotic-loaded chitosan-
[95] Sowmya S, Bumgardener JD, Chennazhi KP, Nair SV, Jayakumar Laponite films for local drug delivery by titanium implants: cell
R. Role of nanostructured biopolymers and bioceramics in enamel, proliferation and drug release studies. J Mater Sci Mater Med 2015;
dentin and periodontal tissue regeneration. Prog Polym Sci 2013; 26(12): 269.
38(10): 1748-72. [119] Min J, Braatz RD, Hammond PT. Tunable staged release of thera-
[96] Varghese S, Theprungsirikul P, Ferran A, Hwang N, Canver A, peutics from layer-by-layer coatings with clay interlayer barrier.
Elisseeff J. Chondrogenic differentiation of human embryonic germ Biomaterials 2014; 35(8): 2507-17.
cell derived cells in hydrogels. In 2006 International Conference of [120] Wang C, Wang S, Li K, et al. Preparation of laponite bioceramics
the IEEE Engineering in Medicine and Biology Society 2006 Aug for potential bone tissue engineering applications. PLoS One 2014;
30 (pp. 2643-2646). IEEE. 9(6): e99585.
[97] Nuttelman CR, Tripodi MC, Anseth KS. Synthetic hydrogel niches [121] Li C. A targeted approach to cancer imaging and therapy. Nat
that promote hMSC viability. Matrix Biol 2005; 24(3): 208-18. Mater 2014; 13(2): 110-5.
[98] Ahearne M, Yang Y, Liu K. Mechanical characterisation of hydro- [122] Lee N, Hyeon T. Designed synthesis of uniformly sized iron oxide
gels for tissue engineering applications nanoparticles for efficient magnetic resonance imaging contrast
[99] Parlato M, Reichert S, Barney N, Murphy WL. Poly(ethylene gly- agents. Chem Soc Rev 2012; 41(7): 2575-89.
col) hydrogels with adaptable mechanical and degradation proper- [123] Estelrich J, Escribano E, Queralt J, Busquets MA. Iron oxide
ties for use in biomedical applications. Macromol Biosci 2014; nanoparticles for magnetically-guided and magnetically-responsive
14(5): 687-98. drug delivery. Int J Mol Sci 2015; 16(4): 8070-101.
20 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Das et al.

[124] Degen CL, Poggio M, Mamin HJ, Rettner CT, Rugar D. Nanoscale [136] Winey KI, Vaia RA. Nanocomposites. MRS BULLETIN 2007; 32
magnetic resonance imaging. Proc Natl Acad Sci USA 2009; [137] LeBaron PC, Wang Z, Pinnavaia TJ. Polymer-layered silicate
106(5): 1313-7. nanocomposites: an overview. Appl Clay Sci 1999; 15(1): 11-29.
[125] Kuehn S, Hickman SA, Marohn JA. Advances in mechanical detec- [138] Eric TL, Richard AM. Integrated genetic analysis microsystems. J
tion of magnetic resonance. J Chem Phys 2008; 128(5): 052208. Phys D Appl Phys 2004; 37(23): R245.
[126] Weissleder R, Nahrendorf M, Pittet MJ. Imaging macrophages with [139] Blyth DJ, Poynter SJ, Russell DA. Calcium biosensing with a sol-
nanoparticles. Nat Mater 2014; 13(2): 125-38. gel immobilized photoprotein. Analyst (Lond) 1996; 121(12):
[127] Mulder WJ, Jaffer FA, Fayad ZA, Nahrendorf M. Imaging and 1975-8.
nanomedicine in inflammatory atherosclerosis. Science transla- [140] Mahmoudi M, Lynch I, Ejtehadi MR, Monopoli MP, Bombelli FB,
tional medicine 2014; 6(239): 239-9. Laurent S. Protein-nanoparticle interactions: opportunities and
[128] Felbeck T, Hoffmann K, Lezhnina MM, Kynast UH, Resch-Genger challenges. Chem Rev 2011; 111(9): 5610-37.
U. Fluorescent Nanoclays: Covalent Functionalization with Amine [141] Walkey CD, Chan WCW. Understanding and controlling the inter-
Reactive Dyes from Different Fluorophore Classes and Surface action of nanomaterials with proteins in a physiological environ-
Group Quantification. J Phys Chem C 2015; 119(23): 12978-87. ment. Chem Soc Rev 2012; 41(7): 2780-99.
[129] Tzitzios V, Basina G, Bakandritsos A, et al. Immobilization of [142] Gunawan C, Lim M, Marquis CP, Amal R. Nanoparticle–protein
magnetic iron oxide nanoparticles on laponite discs - an easy way corona complexes govern the biological fates and functions of
to biocompatible ferrofluids and ferrogels. J Mater Chem 2010; nanoparticles. J Mater Chem B Mater Biol Med 2014; 2(15): 2060-
20(26): 5418-28. 83.
[130] Haraguchi K, Takehisa T, Ebato M. Control of cell cultivation and [143] Felbeck T, Lezhnina MM, Resch-Genger U, Kynast UH. Red emis-
cell sheet detachment on the surface of polymer/clay nanocompo- sive nanoclay hybrids in transparent aqueous dispersion—towards
site hydrogels. Biomacromolecules 2006; 7(11): 3267-75. optical applications in biophotonics. J Luminescence 2016;
[131] Takezawa T, Mori Y, Yoshizato K. Cell culture on a thermo- 169(Part B): 728-32.
responsive polymer surface. Biotechnology (N Y) 1990; 8(9): 854- [144] Sackett DL, Wolff J. Nile red as a polarity-sensitive fluorescent
6. probe of hydrophobic protein surfaces. Anal Biochem 1987;
[132] Akiyama Y, Kikuchi A, Yamato M, Okano T. Ultrathin poly(N- 167(2): 228-34.
isopropylacrylamide) grafted layer on polystyrene surfaces for cell [145] Murugan NA, Kongsted J, Rinkevicius Z, Ågren H. Color model-
adhesion/detachment control. Langmuir 2004; 20(13): 5506-11. ing of protein optical probes. Phys Chem Chem Phys 2012; 14(3):
[133] Gaharwar AK, Schexnailder PJ, Jin Q, Wu CJ, Schmidt G. Addi- 1107-12.
tion of chitosan to silicate cross-linked PEO for tuning osteoblast [146] Anand U, Jash C, Mukherjee S. Protein unfolding and subsequent
cell adhesion and mineralization. ACS Appl Mater Interfaces 2010; refolding: a spectroscopic investigation. Phys Chem Chem Phys
2(11): 3119-27. 2011; 13(45): 20418-26.
[134] Schexnailder PJ, Gaharwar AK, Bartlett RL II, Seal BL, Schmidt [147] Lourenço AVS, Kodaira CA, Ramos-Sanchez EM, et al. Lumines-
G. Tuning cell adhesion by incorporation of charged silicate cent material based on the [Eu(TTA)3(H2O)2] complex incorpo-
nanoparticles as cross-linkers to polyethylene oxide. Macromol Bi- rated into modified silica particles for biological applications. J
osci 2010; 10(12): 1416-23. Inorg Biochem 2013; 123: 11-7.
[135] Giannelis EP. Polymer layered silicate nanocomposites. Adv Mater
1996; 8(1): 29-35.

View publication stats

You might also like