You are on page 1of 16

CHAPTER 1

Introduction to Active, Smart, and


Intelligent Nanomaterials for Biomedical
Application
JYOTI AHLAWAT, MS • MAHESH NARAYAN, PHD

1 INTRODUCTION in 2001, which supports research, development, and


Nanomaterials are materials that have the smallest commercialization of nanotechnology. Furthermore, in
dimension in the nanometer range. These nanosized 2003 “Forward look on nanomedicine” was launched
systems have a size range from a few nanometers such by the European Science Foundation that aimed at
as micelles to several hundreds of nanometers like lipo- biomedical applications of nanotechnology and nano-
somes [1]. Comparatively, the size of a water molecule science. Later, in 2004 the high-level group European
is w0.16 nm, benzene is w0.43 nm, the diameter of technology was established by the European Commis-
DNA is w2 nm, the size of hemoglobin is w6.4 nm, sion with the aim to improve the quality of life and
and the thickness of human hair is 80,000 nm [2]. Inter- healthcare for the European citizens. As a follow-up,
actions between these nanomaterials and biomolecules eight national nanomedicine development centers
can be considered for both in vitro as well as in vivo were established by NIH in 2005 and 2006 with the
diagnosis [1]. In vitro, these nanomaterials allow recog- aim to determine how the cellular machinery at nano-
nition and capture of the biological molecules while scale works and then develop devices based on these
in vivo, these particulate assemblies are used for imag- principles for the detection, treatment, and prevention
ing purposes [1]. The properties and structure of these of various diseases. There are active scientific community
nanomaterials differ significantly from the bulk mate- and medical industrial partners in countries such as
rials and the atoms and molecules from which they France, Germany, Spain, and the United Kingdom where
are synthesized [3]. Therefore, nanoparticulate systems development in biomedical nanotechnology is signifi-
that can release a drug at an appropriate rate in response cant although the transfer of technology to the industries
to a specific physiological stimulus and at the destined is still not as effective as in North America [1].
target site are termed as active, smart, and intelligent In summary, nanotechnology refers to the study of
nanomaterials [4] (Fig. 1.1). material at the ultrasmall scale. The nanomaterials
It is noteworthy that there is explosive scientific operate at the nanoscale at which the biomolecules in-
growth in the field of nanoscience and technology side the living body work. At this scale, the surface-to-
due to the introduction of new methods of nanomate- volume ratio of the particle is large, which results in
rial synthesis and advancement in the analytical tech- the huge surface for interactions with the biological
niques for the characterization and manipulation of molecules. Furthermore, this results in shorter reaction
material in the nanometer range [3]. For instance, time, and therefore the miniaturized devices are more
gene targeting/drug targeting, chips for chemical/ sensitive, faster, and portable.
biochemical assays, DNA chips and arrays, nanosen-
sors, and nanocomposites are some of the applications
based on nanomaterials. 2 TYPES OF NANOMATERIALS
Nanotechnology is expanding rapidly due to support In simple and medium terms, the nanomaterials are
from researchers from diverse backgrounds such as divided into three generations [1]:
academia, clinicians, industry, and federal sectors. As a • First generation: these include nanocapsules and
result, National Nanotechnology Initiative was launched nanospheres

Intelligent Nanomaterials for Drug Delivery Applications. https://doi.org/10.1016/B978-0-12-817830-0.00001-1


Copyright © 2020 Elsevier Inc. All rights reserved. 1
2 Intelligent Nanomaterials for Drug Delivery Applications

FIG. 1.1 Schematic illustration of various strategic designs for nanomaterials.

• Second generation: these include nanoparticles promote targeted drug delivery, reduce the therapeutic
coated with hydrophilic polymeric envelope such as dose, and are stable over a wide range of temperature
polyethylene glycol (PEG). and pH [6,7]. The inorganic nanoparticles can be syn-
• Third generation: it involves attachment of ligand on thesized using physical and chemical methods. The
the surface of a biodegradable core coated with a physical method involves the deposition of vapor and
polymer. relies on the principle of breaking down the bulk pre-
This section of the chapter sheds light on some of the cursor into smaller nanoparticles. The second method,
nanoparticulate systems (Tables 1.1 and 1.2) used for chemical synthesis, requires the breakdown of the metal
biomedical applications. The main advantages of the ion into atoms in the presence of a stabilizing agent fol-
nanoparticulate system include the protection of drugs lowed by controlled aggregation of these atoms [7]. The
from degradation inside the body before reaching the latter is generally preferred over the physical mode of
destined location, increased circulation time, enhanced synthesis of nanoparticles. It is necessary to design inor-
drug delivery, lowered dose, controlled and sustained ganic nanoparticles with enhanced stability, increased
release of drug, and reduced toxicity. circulation time without reticuloendothelial clearance,
and neutrophil activation without compromising the
2.1 Inorganic Nanoparticles therapeutic efficacies. As most of the inorganic nanopar-
The inorganic nanoparticles can be defined as particles ticles have to be chemically or biologically modified
of metallic composition exhibiting at least one dimen- before use for cellular delivery, concerns such as
sion in the nanometer range [5]. These nanoparticles toxicity, carcinogenesis, and immunogenicity arising
act as excellent drug-delivery vehicles, are biocompat- due to the use of these nanoparticles must be addressed
ible, provide enhanced bioavailability of drugs, [6,8,9].
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 3

TABLE 1.1
Properties and Various Applications of Selected Nanoparticles.
Sr.
No. Nanoparticle Properties Applications References
1 Gold Optical and photothermal properties Antibacterial and antiviral [7,21e41]
due to surface plasmon resonance, applications, biosensing, MRI,
thermal, electrical, antiangiogenic, cancer diagnosis, and photothermal
catalytic, magnetic properties cancer therapy
2 Silver Optical properties due to surface Antibacterial and antifungal [21,42e65]
plasmon resonance (SPR), applications in water purification
antiangiogenic, structural, thermal, systems, paints and household
electrical, and catalytic properties products, antiviral applications
against HIV-I and monkey pox virus,
biosensing
3 Magnetic Magnetic, caloric, physical, and Biomedicine, cancer treatment, MRI, [66e81]
hydrodynamic properties drug delivery, removal of toxic metal
ions and antimicrobial applications
4 Mesoporous Porosity, controlled particle size, Drug delivery, diagnostic catalysis, [5]
silica-based stable over a wide range of pH and biosensing, and separation
temperature, chemical stability

TABLE 1.2
Drug-Delivery Application of Biodegradable Polymers: Synthetic and Natural.
SYNTHETIC BIODEGRADABLE POLYMERS NATURAL BIODEGRADABLE POLYMERS
Polyesters Polyoxalates Starch Dextran
Polyorthoesters Polyiminocarbonates Hyaluronic acid Chitosan
Polyanhydrides Polyurethanes Heparin e
Polydioxanones Polyphosphazenes Gelatin
Poly(a-cyanoacrylates) e Albumin

Reprinted with permission from Coulembier O, Degée P, Hedrick JL, Dubois P. From controlled ring-opening polymerization to biodegradable
aliphatic polyester: especially poly (b-malic acid) derivatives. Progress in Polymer Science. 2006;31(8):723e747.

2.1.1 Metal nanoparticles Silver is known for its antimicrobial activity since
Biomedical applications like the visualization of ancient times. In a study, Mahapatra and Karak showed
cellular components using electron microscopy, tar- the use of silver particles, in the nanometer range, on
geted and nontargeted delivery of the drug-loaded protheses, catheters, vascular grafts, and skin grafts
nanoparticles, detection, and diagnosis of various dis- due to their broad spectrum antimicrobial activity but
eases involve the use of metal nanoparticles [10e12]. was also found to be little toxic to mammalian cells
In a study, Mirkin et al. showed that gold nanopar- [15].
ticles conjugated with a specific oligonucleotide can
detect (due to color change) complementary DNA 2.1.2 Magnetic nanoparticles
strands [13]. In another study, Loo et al. found out These nanoparticles have gained popularity in recent
that gold nanoparticles when tagged with antibodies, years due to their special magnetic properties and
enzymes, or nucleotides had applications in several ability to act as a contrast agent for magnetic resonance
biosensor assays, drug- and gene-delivery systems, and imaging [16] and as a drug carrier [17]. However, the
biomedical-based imaging systems [14]. safety of using these nanoparticles remains unexplored.
4 Intelligent Nanomaterials for Drug Delivery Applications

A study carried out by Lu et al. showed that the biocom- biosensors, and biomedical devices. However, CNTs
patibility of carbon-coated iron nanoparticles is depen- have solubility issues in most of the solvents and are
dent on the surface chemistry of nanoparticle and on cytotoxic without any functionalization [5]. Therefore,
the cell type studied [18]. functionalization could be achieved by (1) additional
reactions on the tips and the sidewalls or (2) oxidation
2.1.3 Mesoporous silica system followed by carboxyl-based coupling. Furthermore,
The mesoporous silica system has gained intensive atten- functionalization improves the biocompatibility of
tion around the world in the past decade because of its the CNTs and makes them soluble [5]. Such functional-
highly ordered structure, large pore size, worm-like inte- ization also helps in the penetration of the CNTs across
rior, and huge surface area [19]. These properties make it the cell membrane due to its hydrophilicity [5].
an ideal candidate for the hosting and delivery of a vari-
ety of molecules such as drugs and protein [9]. 2.4 Liposomes
As these microspheres have a size in the range of bac- Liposomes are defined as bilayered vesicles composed of
teria and can trigger immune response, to ensure effort- inner aqueous volume entirely surrounded by a lipid
less endocytosis by cells without any cell toxicity, the outer membrane [86]. These nanoparticulate systems
size of mesoporous silica nanoparticles can be modu- can vary in size from few nanometers to several micro-
lated between 50 and 300 nm [9]. meters [87,88]. Furthermore, properties of the liposome
The mesoporous silica-based nanoparticle MCM-41 have been found to depend on vesicle size, surface
is one of the important synthesized materials [20]. charge, lipid composition, and preparation method
Although biocompatibility is not so strong, it has employed [88]. For instance, liposomal vesicles with a
been employed for drug delivery because of their bioac- size greater than few nanometers have been investigated
tivity. Other mesoporous silica materials include SBA: to play a role in Mononuclear Phagocyte System (MPS)
SBA-15, SBA-16, SBA-1, SBA-3, HMS, and MSU [20]. clearance and complement activation and hence requires
additional methods for the prevention of opsonization
2.2 Micelles [89]. Therefore, surface modification using approaches
Micelles are coreeshell structure formed by the self- such as coating with linear dextran, PEG, and polyviny-
assembly of amphiphiles in the aqueous environment lalcohol can help in protection from MPS uptake,
when the concentration of these amphiphiles exceeds increasing their circulation time and hence sustained
the critical micelles concentration [82]. Most of the drug release [90e92]. These vesicular systems are consid-
micellar drug-delivery systems are composed of a hy- ered successful therapeutic drug carriers and have been
drophilic outer corona that consists of PEG and an in- used as carriers for many anticancer agents such as
ner low molecular weight hydrophobic core-forming cisplatin [93] and paclitaxel [94] and antibiotics such
block. Such nanoparticulate systems have a size below as ciprofloxacin [95] and amikacin [96]. Furthermore, li-
100 nm [83e85]. The hydrophobic interactions are posomes can be surface-modified with a ligand for
the major driving force for the formation of such ther- achieving targeted drug delivery [97].
modynamically stable structures [3]. Moreover, these
micellar systems possess reduced toxicity and have a po- 2.5 Polymer-Based Nanoparticles
larity gradient (from the outer hydrophilic corona to the These nanoparticles possess a coreeshell structure and
inner hydrophobic core) that helps in the solubilization are made up of biodegradable polymers and copolymers
of the poorly soluble hydrophobic compounds. This to increase circulation time in the body and reduce MPS
system is further classified into phospholipid, pluronic, uptake [5,98]. For example, some of the well-studied
poly(L-amino acid), and polyester micelles. Finally, the nanoparticles are poly(glycolic acid), poly(lactic acid),
pharmacokinetics, biodistribution, and circulation time poly(methylmethacrylate), and poly(lactic-co-glycolic
of drug, either in the core (nonpolar) or corona (polar acid) [98e100]. These are FDA approved and are biode-
molecule) or in between the core and shell (intermedi- gradable in nature. Furthermore, the corona consists of
ate polarity), are increased compared to the free drug. hydrophilic polymers, such as PEG and PVP and the
core is a polymeric matrix for the encapsulation of
2.3 Carbon Nanotubes hydrophobic drugs [5]. The drug molecule can be
Carbon nanotubes are self-assembling atoms of carbon either physically adsorbed or chemically linked to the
arranged in the form of a tube [86]. These tubes have surface or entrapped inside the nanoparticle. Moreover,
good electrical, mechanical, and surface properties mak- these polymeric nanoparticles have a size below
ing them an excellent candidate for drug delivery, 1000 nm [86].
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 5

3 CHARACTERISTICS OF NANOMATERIALS • desorption of surface-bound drug


3.1 Particle Size • diffusion of drugs from the nanoparticle core
The particle size can be determined using photon- • matrix erosion
correlation spectroscopy or dynamic light scattering. • Combination of erosion and diffusion processes.
Photon-correlation spectroscopy measures the diameter Rate of diffusion or erosion determines the release
by light scattering and using Brownian motion. Scan- of drugs in the case of nanospheres. For instance, if
ning electron microscopy and transmission electron mi- the rate of diffusion is faster than the erosion of the
croscopy are used to verify the results obtained using matrix, then the release mechanism is diffusion
photon-correlation microscopy. dependent. Moreover, the burst release of drug from
Particle size and size distribution determine the sta- nanoparticles is due to the weak interaction between
bility of nanoparticles, targeting ability, toxicity, in vivo the drug and the matrix [108]. For instance, if the sur-
fate, drug release profile, and loading of drugs. For face of nanoparticle is coated with polymers such as
example, smaller particles have a large surface area-to- PEO and PEG, then the release profile is diffusion
volume ratio, implying the presence of drugs near or dependent from the polymeric barrier due to the ionic
at the surface of nanoparticles allowing the faster release interaction between the drug and the auxiliary
of drugs following zero-order kinetics [102]. Further- ingredients.
more, the degradation of polymer also depends on
the particle size, for example, poly lactic-co-glycolic
acid (PLGA) [103]. 4 TYPES OF TARGETING
4.1 Passive Targeting
3.2 Surface Properties To begin with, passive targeting is the basic targeting
Nanomaterials, when administered through the intrave- strategy employed by the smart nanoparticulate system
nous route, can be recognized and cleared by the [109]. The enhanced permeability and retention (EPR)
phagocytes [104]. In addition, hydrophobicity of the effect or passive targeting refers to the preferential
nanoparticles determines opsonization and clearance localization of nanomaterials in the pathological tis-
by MPS. Therefore, to enhance the lifetime of these sues described by leaky vasculature and poor
nanomaterials in vivo and to minimize opsonization, lymphatic draining [110e113]. Nanoparticles such as
surface modification of nanoparticles with hydrophilic polymers, liposomes, and micelles take advantage of
polymers such as PEG, polyethylene oxide (PEO), and the leaky tumor vasculature allowing entrapment and
polysorbate 80 (Tween 80) is done. Furthermore, the accumulation inside the tumors [113,114]. Moreover,
zeta potential can be used to determine the charge on a direct relationship between passive targeting and
the surface of the nanoparticles [105]. For instance, a leaky vascularization has been documented in a
charge greater than 30 mV on nanoparticles provides different study [115]. Furthermore, prolonged
stability and prevents aggregation. circulation in the bloodstream can be achieved by
conjugating PEG onto the surface of drug-loaded
3.3 Drug Loading nanomaterials resulting in enhanced therapeutic
Drug loading can be done using two methods: efficiency of these nanomaterials compared to free
• Incorporation method: it involves the incorporation drug molecules [109]. For example, Doxil
of drugs during the time of nanoparticle development. (doxorubicin-loaded PEGylated liposome) has pro-
• Adsorption methods: it requires the adsorption of longed circulation half-life, avoids high plasma level,
drugs after nanoparticle development. and has minimized reticuloendothelial system uptake
Loading of the drug depends on the solubility of the and clearance [116].
drug in the solid polymer or liquid dispersion agents (ma-
trix) that in turn depends on the composition of matrix, 4.2 Active Targeting
drugepolymer interactions, and molecular weight [107]. Active targeting, also referred as receptor-mediated tar-
It has been previously reported that active substances geting or ligand-based targeting, involves the use of a
such as drugs and peptides show enhanced entrapment variety of targeting ligands such as peptides and anti-
efficiency at or near their pI (isoelectric point) [107]. bodies to allow targeted delivery and uptake by the
target cells expressing high levels of the specific bio-
3.4 Drug Release markers (e.g., cell surface receptors or cell adhesion
The drug release rate depends on the following: molecules). These targeting moieties can be either phys-
• solubility of the drug ically or chemically conjugated on the surface of the
6 Intelligent Nanomaterials for Drug Delivery Applications

TABLE 1.3
Various Examples of Ligands, Targets, and Nanocarrier Systems Used for Active Drug Targeting.
Examples of
Targeting Ligands Targets Nanomaterials References
Folate Folate receptor Liposomes [123e125]
Albumin nanoparticles
Transferrin Transferrin receptor Liposomes [126e129]
Nanogels
Insulin Insulin receptor Magnetic nanoparticles [130]
Antibodies and their fragments
Anti-HER2 and its fragments HER-2 receptor Liposomes [131]
OX26 (anti-transferrin) Transferrin receptor Liposomes [132]
323/A3 Epithelial glycoproteins Liposomes [133]
Anti-Flk-1 Vascular endothelial growth factor Lipid nanoparticles [134]
receptor 2 (Flk-1)
Anti-CD19 CD-19 epitope Liposomes [135]
Peptides
Luteinizing hormone-releasing Luteinizing hormone-releasing Polymeric nanoparticles [136]
hormone hormone receptor aminopeptidase
(CD13)
NGR peptide Aminopeptidase N (CD13) Liposomes [137]
Gelatinase inhibitory peptide MMP-2 and MMP-9 gelatinase Liposomes [138]
CTTHWGFTLC Albumin nanoparticles

Adapted with permission from O.M. Koo et al., 2005.

nanomaterials [117,118]. Table 1.3 summarizes the clearance resulting in a lack of clinical application of
various targeting ligands used to functionalize nanoma- such ligand functionalized nanoparticulate systems
terials. However, there is still a lot of debate going on [122] (Fig. 1.2).
whether ligand functionalized nanomaterials have the
ability to provide enhanced nanomaterial accumulation 4.3 Stimuli-Responsive Targeting
at the targeted site (e.g., malignant cells) compared to A third type of targeting termed as stimuli-responsive
nontargeted nanomaterials [119]. For example, a targeting is currently receiving attention around the
similar level of accumulation in breast cancer xenograft globe as it allows triggered drug release [114]. In this,
models, overexpressing HER2 antigen, was observed for the nanoparticulate system can undergo physicochem-
both HER2 monoclonal antibody fragments conjugated ical changes in its structure resulting in release of drug
liposomes and nonfunctionalized liposomes [120]. at a particular location and time when subjected to
Pharmacodynamic differences were suggested as the endogenous stimuli such as low pH, presence of redox
reason for the improved antitumor effect and enhanced gradients or certain enzymes in tumor microenviron-
drug delivery of the targeted nanoparticulate nanome- ment, or exogenous stimuli such as light, heat, elec-
dicine formulation in vivo [121]. So far, denileukin dif- tric/magnetic fields, and ultrasound [114]. For
titox was the only FDA-approved nanoparticles in the example, Thermodox (thermosensitive liposomal
market, which too was clinically discontinued in doxorubicin) has temperature-sensitive lipids and poly-
2016, for the treatment of cancer using active targeting mers. Such combination allows the nanomaterial to
[121]. Reasons such as unspecificity of the targeting remain stable at physiologic temperature and later un-
moiety, dose loss due to lysosomal digestion, and im- dergo phase change increasing their permeability
mune clearance are considered as key factors in blood upon heating hence triggering the release of the drug
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 7

occurrence and are considered life threating. Nanopar-


ticles such as liposomes, dendrimers, lipid- and
polymer-based micelles, and polymeric nanoparticles
are a choice for the improvement of therapy of such
life-threatening diseases. For example, nanoparticulate
systems such as polyamidoamine (PAMAM) den-
drimers were evaluated for the delivery of poorly solu-
ble antiasthma drug beclomethasone dipropionate
[142]. In another study, inorganic nanoparticles,
organic nanoparticles, and metallic nanoparticles
were synthesized and tested for pulmonary immune
hemostasis [143]. Furthermore, polymeric drug-
delivery vehicles are of considerable interest to the
scientist, as these can be surface-modified and copoly-
merized for application in the treatment of lung dis-
eases. These include gelatin, chitosan, PLGA, PEG,
and alginate [145].

5.2 Ophthalmology
Eye is a tiny intricate organ, and drug delivery
following this route is of keen interest for the pharma-
FIG. 1.2 Different types of nanoparticles for the ceutical scientists around the globe [142]. Conditions
improvement of therapy for pulmonary diseases. such as corneal disorder, for example, glaucoma and
eye infection, for example, conjunctivitis requires
ocular administration of the drug. Nanoparticulate
[139]. Furthermore, nanomaterials that have pendant systems such as liposomes and pharmacosomes have
acidic or basic groups are termed “pH-sensitive” and been observed to improve residence time, corneal
can accept or lose protons depending on the surround- permeability, and bioavailability of drug molecules
ing pH and thus are believed to take advantage of the [146,147]. For instance, PLGA-loaded pranoprofen
low pH of the tumor microenvironment (6.4e6.7) nanoparticles, in the form of a hydrogel, for
[140,141]. Such pH sensitivity of the nanomaterials is ophthalmic administration were evaluated. As a result,
considered beneficial for the delivery of thermolabile improved pharmacokinetic in comparison to the free
drugs [114]. drug was observed [148]. In a different study,
curcumin-loaded polyethylene glycol-poly caprolac-
tone (MePEG-PCL) nanoparticles were evaluated for
5 NANOMATERIALS IN MEDICINE: THE ophthalmic administration of curcumin and were re-
IDEAL SCALE ported to show enhanced retention of drugs in the
In the past few years, nanomedicine has gained the cornea with improved efficiency [149]. Furthermore,
trust of scientists around the globe for the local deliv- DC-TMCNS (diclofenac-loaded N-trimethyl chitosan
ery of drugs while addressing pharmacokinetic and nanoparticles) were evaluated for improved bioavail-
pharmacodynamic drawbacks associated with the use ability of drugs [150].
of conventional free drug formulations [142,143].
Various nanoparticulate drug-delivery systems for 5.3 Cardiovascular System
enhancing therapeutic efficacy include liposomes, A huge number of people are affected by the cardiovas-
solid lipid nanoparticles, dendrimers, polymeric nano- cular disorder in developing countries, with reported
particles, micelles, and solid metal-containing nano- morbidity close to 80% and is reported to occur almost
particles. This section of the chapter describes the equally in both men and women [151]. Nanoparticu-
major fields of interest for drug-delivery applications late systems such as sirolimus-loaded chitosan-based
using nanomaterials. liposomes were evaluated for restenosis treatment
and were found to be effective [152]. In a different
5.1 Pulmonology study, rat myocardial ischemic model was tested with
Lung diseases such as lung carcinoma, asthma, and vascular endothelial growth factor-loaded polymeric
chronic obstructive pulmonary disease have a huge nanoparticles for the delivery of cytokines [153].
8 Intelligent Nanomaterials for Drug Delivery Applications

Moreover, CoQ10 (Coenzyme Q10) can be used in the to bloodebrain barrier integrity compared to cationic
treatment of myocardial ischemia owing to its role in nanoparticles and high concentration of anionic nano-
the mitochondrial electron transport chain. However, particles [163]. Related to this, Kreuter et al. in 2004
its poor pharmaceutical properties limit its use. There- showed that coating nanoparticle surfaces with a surfac-
fore, CoQ10-loaded polymeric nanoparticles were tant such as polysorbate (Tween) enhanced the penetra-
developed to improve the poor pharmacokinetic prop- tion of drug-loaded nanoparticles across the
erties of CoQ10 alone. bloodebrain barrier [164]. Furthermore, binding of
nanoparticle with lipoprotein receptors
5.4 Oncology (apolipoprotein-E and low-density lipoprotein) on
Cancer is of the leading cause of death around the brain capillary endothelial cells was suggested as the
globe. Cancer patients often face the cytotoxic effect reason for this uptake of drug by the brain [165]. In
of chemotherapeutic drugs and therefore nanoparticu- addition, PEG-PLA nanoparticles surface functionalized
late drug-delivery systems can help in increasing the with lectin were developed for translocation to the
life span and quality of patient’s life by minimizing brain via inhalation route [166]. In a different study,
systemic toxicity of the chemotherapeutics and by Gao et al. in 2006 showed twofold brain uptake of
increasing the tumor specificity [154]. For instance, the nanoparticle. Wheat germ agglutinin, which binds
Doxil (doxorubicin-loaded liposome) is an FDA- to N-acetyl-D-glucosamine and sialic acid present in
approved nanoparticle for the treatment of ovarian the nasal cavity, was used [167].
cancer. The half-life of this nanoparticulate system is In summary, nanoparticles can be surface-modified
100 times more than the free doxorubicin with overall with ligands to provide targeted drug delivery and
reduced cardiotoxicity [155,156]. Furthermore, nab- reduced systemic toxicity [142]. These drug-delivery
Ptx (Abraxane) is an albumin-bound Ptx used in the nanoforms can provide enhanced therapeutic efficacy
treatment of lung, breast, and pancreatic cancer of the drug. Advancement in the analytical techniques
[157]. Nab-Ptx in combination with gemcitabine has led to the manufacturing of nanomaterials for the
(Gem) was FDA-approved for pancreatic ductal adeno- delivery of therapeutics for the treatment of pulmonary,
carcinoma treatment in 2013 [158]. It acts by targeting ophthalmological, cardiovascular diseases, and cancer
cancer cells in the G2/M stage of the cell cycle. In a therapy and crossing bloodebrain barrier. Therefore,
study, Liu et al. developed a phospholipid-modified the future of the nanoparticulate nanomedicine is
cationic PAMAM-siMDR1 complex for the treatment propitious.
of human breast cancer cells [159]. This new complex
was reported to show increased cellular uptake of 6 TOXICOLOGICAL ISSUES OF
siRNA targeting the MDR1 gene and hence enhanced NANOMATERIALS
MDR1 gene silencing [157]. In a different study, Li
In the past few years, the special properties of nanoma-
et al. showed that HSPC (hR3-siMDR1-PAMAM com-
terials are being utilized for designing the toxicological
plex) could reverse MDR in adriamycin‑resistant Mich-
test for checking the hazardous or toxic effect of the
igan Cancer Foundation-7 cells (MCF/ADR) breast
engineered nanomaterials. Recent studies with cationic
cancer cell line [160]. This complex showed reduced
nanoparticles such as gold and polystyrene showed he-
toxicity and improved endosomal escape with
molysis and blood clotting, whereas anionic nanopar-
enhanced cellular uptake.
ticles were observed to be comparatively less
cytotoxic. Moreover, studies with carbon-based nano-
5.5 BraindThe Ultimate Target for Drug- particles such as SWCNT and MWCNT showed platelet
Delivery Application aggregation whereas no such aggregation was observed
Specific targeting to the brain is challenging for drug- in the case of C60-fullerenes (a building block for these
delivery applications, as free drugs including many CNTs) [168]. In addition, reactive oxygen species gener-
small molecules cannot cross the braineblood barrier. ation, mitochondrial dysfunction, and lipid peroxida-
In a study, the passage of nanoparticle (about tion were observed in keratinocytes and bronchial
200 nm) was reported [161]. In a different study carried epithelium when cells were incubated with a high
out by Kreuter et al. in 2003, the same effect was not dose of single-walled carbon nanotubes [169,170]. In
found for a different type of nanoparticle [162]. another study, neutral and low concentration of anionic
Furthermore, Lockman et al. in 2004 reported that nanoparticles was observed to cause less damage to the
nanoparticles with a neutral charge and low concentra- bloodebrain barrier integrity compared to the cationic
tion of anionic nanoparticles were considered less toxic and high concentration of anionic nanoparticles. For
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 9

instance, Net et al. in a study showed that nanoparticles 7 CONCLUSION AND OUTLOOK
induce oxidative stress by the production of reactive ox- Recent advances in the nanoparticulate drug-delivery
ygen species in the brain that has been implicated in the system are set to spread rapidly. For decades, nanopar-
pathogenesis of Alzheimer’s and Parkinson’s disease ticles are used for drug-delivery applications to reduce
[171]. Furthermore, gold nanorods were found to be toxicity and side effects associated with the use of free
considerably cytotoxic, and this was attributed to the drugs. In addition, these can help in overcoming
stabilizer Cetyltrimethylammonium Bromide (CTAB) various physiological barriers such as bloodebrain bar-
used during its synthesis. For silica nanoparticles, rier. Smart-drug delivery involves enhancing the thera-
increased dose and exposure time caused increased peutic index while minimizing the cytotoxic effect
toxicity in vitro. It was observed to be due to the gener- using multifunctional nanomaterials. Moreover, the
ation of reactive oxygen species and reduced gluta- physicochemical properties of the nanoparticles such
thione levels resulting in the onset of oxidative stress as size and structure can be modified according to the
inside the cell [172]. severity of disease for attaining maximal therapeutic
For the application of nanotechnology in medicine, benefits. For instance, FA-DABA-SMA polymers employ
knowledge of the safety and toxicological issues of the three levels of targeting different from older formula-
free drug, different nanoparticulate delivery systems, ef- tions that reply to the EPR effect [114]. Therefore, opti-
fect of rate of drug release at target and nontarget sites, mizing the drug release profile and employing different
and interaction of these nanomaterials with the targeting strategies are of keen interest and importance
biological components inside the body needs full atten- in this drug-delivery field but the cost of production,
tion [173]. For instance, increased complexity of manufacturing, regulatory standards, and technical dif-
nanomaterials such as surface coating and ligand ficulties must be addressed simultaneously. In addition,
attachment can affect biodistribution, bioavailability, a key point that was overlooked for so many years was
and biocompatibility of nanomaterials following the risks of these carrier platforms to patients. Overall, a
in vivo administration [174,175]. Moreover, compre- scientist should consider minimizing the complexity of
hensive bioassays to assess absorption, distribution, the nanoparticulate system for generating clinically
metabolism, excretion, cytotoxicity, immunotoxicity, translatable nanosized therapeutics. So far, we have lit-
and genotoxicity before preclinical evaluations for toxi- tle knowledge about the basics of the interaction of
cological responses to nanomaterials are essential these nanomaterials with the biological components
[176]. For example, rats administered with Doxil such as living cells and organs inside the organism.
showed decreased bacterial blood clearance due to Therefore, the collaboration between those working in
macrophage suppression [177]. particle toxicology and the development of various
Some of the testing approaches (under develop- drug-delivery nanomaterials are necessary; then, we
ment) for comparative and simultaneous risk potential will see how nanotechnology will bring real added
analysis of many nanomaterials include computational values in a cost-effective way.
modeling, high-throughput screening method, and
high content screening [178]. In addition, special
emphasis on the discrimination in toxicity of drug- REFERENCES
loaded and “empty” nanoparticles should be taken [1] Boisseau P, Loubaton B. Nanomedicine, nanotechnology
into account, as the slow or nondegrading nanomateri- in medicine. Physical Rendering 2011;12:620e36.
als may persist and accumulate at the site causing [2] Faraji AH, Wipf P. Nanoparticles in cellular drug
delivery. Bioorganic and Medicinal Chemistry 2009;
chronic inflammatory reactions.
17(8):2950e62.
In summary, there is a tendency of nanosized [3] Rao CN, et al. Nanomaterials-An introduction. In: The
(small) particles to be more toxic than the larger parti- chemistry of nanomaterials: synthesis, properties and
cles having the same chemical composition. Various applications. Wiley; 2006. p. 1e11.
studies focus on the reduction in the toxicity of the [4] James HP, John R, Alex A, Anoop KR. Smart polymers for
loaded drug but do not describe anything regarding the controlled delivery of drugsea concise overview. Acta
the toxic effect of the carrier molecule itself. Costigan Pharmaceutica Sinica B 2014;4(2):120e7.
et al. in 2006 reviewed the various evidence of the [5] Safari J, Zarnegar Z. Advanced drug delivery systems:
potentially hazardous effect of nanomaterials in health- nanotechnology of health design A review. Journal of
care products. However, it is believed that not all the Saudi Chemical Society 2014;18(2):85e99.
[6] Xu ZP, Zeng QH, Lu GQ, Yu AB. Inorganic nanoparticles
toxic effect for the use of nanomaterials is known at
as carriers for efficient cellular delivery. Chemical Engi-
this moment [179e188]. neering Science 2006;61(3):1027e40.
10 Intelligent Nanomaterials for Drug Delivery Applications

[7] Kango S, Kalia S, Celli A, Njuguna J, Habibi Y, Kumar R. [22] Frens G. Controlled nucleation for the regulation of the
Surface modification of inorganic nanoparticles for particle size in monodisperse gold suspensions. Nature
development of organiceinorganic nanocompositesda Physical Science 1973;241(105):20.
review. Progress in Polymer Science 2013;38(8): [23] Sun Y, Mayers B, Xia Y. Metal nanostructures with
1232e61. hollow interiors. Advanced Materials 2003;15(7-8):
[8] Cho K, Wang XU, Nie S, Shin DM. Therapeutic nanopar- 641e6.
ticles for drug delivery in cancer. Clinical Cancer [24] Kundu S, Panigrahi S, Praharaj S, Basu S, Ghosh SK,
Research 2008;14(5):1310e6. Pal A, Pal T. Anisotropic growth of gold clusters to
[9] Ensign LM, Cone R, Hanes J. Oral drug delivery with gold nanocubes under UV irradiation. Nanotechnology
polymeric nanoparticles: the gastrointestinal mucus 2007;18(7):075712.
barriers. Advanced Drug Delivery Reviews 2012;64(6): [25] Kundu S, Peng L, Liang H. A new route to obtain high-
557e70. yield multiple-shaped gold nanoparticles in aqueous so-
[10] Ranjan AP, Mukerjee A, Helson L, Vishwanatha JK. Scale lution using microwave irradiation. Inorganic Chemistry
up, optimization and stability analysis of Curcumin C3 2008;47(14):6344e52.
complex-loaded nanoparticles for cancer therapy. Jour- [26] Elghanian R, Storhoff JJ, Mucic RC, Letsinger RL,
nal of Nanobiotechnology 2012;10(1):38. Mirkin CA. Selective colorimetric detection of polynu-
[11] Sankari Z, Adeli H, Adeli A. Intrahemispheric, interhemi- cleotides based on the distance-dependent optical prop-
spheric, and distal EEG coherence in Alzheimer’s disease. erties of gold nanoparticles. Science 1997;277(5329):
Clinical Neurophysiology 2011;122(5):897e906. 1078e81.
[12] Alivisatos AP, Gu W, Larabell C. Quantum dots as [27] Jain PK, Huang X, El-Sayed IH, El-Sayed MA. Noble
cellular probes. Annual Review of Biomedical Engineer- metals on the nanoscale: optical and photothermal
ing 2005;7:55e76. properties and some applications in imaging, sensing,
[13] Rosi NL, Giljohann DA, Thaxton CS, Lytton-Jean AK, biology, and medicine. Accounts of Chemical Research
Han MS, Mirkin CA. Oligonucleotide-modified gold 2008;41(12):1578e86.
nanoparticles for intracellular gene regulation. Science [28] Zhang Y, Schwartzberg AM, Xu K, Gu C, Zhang JZ. Elec-
2006;312(5776):1027e30. trical and thermal conductivities of gold and silver nano-
[14] Loo C, Lin A, Hirsch L, Lee MH, Barton J, Halas N, et al. particles in solutions and films and electrical field
Nanoshell-enabled photonics-based imaging and ther- enhanced Surface-Enhanced Raman Scattering (SERS).
apy of cancer. Technology in Cancer Research & Treat- In: Physical chemistry of interfaces and nanomaterials
ment 2004;3(1):33e40. IV, vol. 5929. International Society for Optics and Pho-
[15] Mahapatra SS, Karak N. Hyperbranched aromatic poly- tonics; August 2005. p. 592912.
amines with s-triazine rings. Journal of Applied Polymer [29] Arvizo RR, Rana S, Miranda OR, Bhattacharya R,
Science 2007;106(1):95e102. Rotello VM, Mukherjee P. Mechanism of anti-angiogenic
[16] Idée JM, Port M, Raynal I, Schaefer M, Le Greneur S, property of gold nanoparticles: role of nanoparticle size
Corot C. Clinical and biological consequences of trans- and surface charge. Nanomedicine: Nanotechnology,
metallation induced by contrast agents for magnetic Biology and Medicine 2011;7(5):580e7.
resonance imaging: a review. Fundamental and Clinical [30] Chen X, Zhao D, Zhao L, An Y, Ma R, Shi L, et al. Optic
Pharmacology 2006;20(6):563e76. and catalytic properties of gold nanoparticles tuned by
[17] Chen H, Khemtong C, Yang X, Chang X, Gao J. Nanoni- homopolymers. Science in China, Series B: Chemistry
zation strategies for poorly water-soluble drugs. Drug 2009;52(9):1372e81.
Discovery Today 2011;16(7e8):354e60. [31] Gréget R, Nealon GL, Vileno B, Turek P, Mény C, Ott F,
[18] Lu AH, Salabas EE, Schüth F. Magnetic nanoparticles: et al. Magnetic properties of gold nanoparticles: a room-
synthesis, protection, functionalization, and temperature quantum effect. ChemPhysChem 2012;
application. Angewandte Chemie International Edition 13(13):3092e7.
2007;46(8):1222e44. [32] Baffou G, Quidant R. Thermo-plasmonics: using
[19] Meek ST, Greathouse JA, Allendorf MD. Metal-organic metallic nanostructures as nano-sources of heat. Laser
frameworks: a rapidly growing class of versatile and Photonics Reviews 2013;7(2):171e87.
nanoporous materials. Advanced Materials 2011; [33] Ortega MA, Rodriguez L, Castillo J, Piscitelli V,
23(2):249e67. Fernandez A, Echevarria L. Thermo-optical properties
[20] Kresge CT, Leonowicz ME, Roth WJ, Vartuli JC, Beck JS. of gold nanoparticles in colloidal systems. Journal of
Ordered mesoporous molecular sieves synthesized by a Optics A: Pure and Applied Optics 2008;10(10):
liquid-crystal template mechanism. Nature 1992; 104024.
359(6397):710. [34] Gil-Tomás J, Tubby S, Parkin IP, Narband N, Dekker L,
[21] Frederix F, Friedt JM, Choi KH, Laureyn W, Campitelli A, Nair SP, Street C. Lethal photosensitisation of Staphylo-
Mondelaers D, et al. Biosensing based on light absorp- coccus aureus using a toluidine blue Oetioproninegold
tion of nanoscaled gold and silver particles. Analytical nanoparticle conjugate. Journal of Materials Chemistry
Chemistry 2003;75(24):6894e900. 2007;17(35):3739e46.
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 11

[35] Perni S, Piccirillo C, Pratten J, Prokopovich P, [49] Rodriguez-Sanchez L, Blanco MC, Lopez-Quintela MA.
Chrzanowski W, Parkin IP, Wilson M. The antimicrobial Electrochemical synthesis of silver nanoparticles. The
properties of light-activated polymers containing meth- Journal of Physical Chemistry B 2000;104(41):9683e8.
ylene blue and gold nanoparticles. Biomaterials 2009; [50] Haes AJ, Van Duyne RP. A nanoscale optical biosensor:
30(1):89e93. sensitivity and selectivity of an approach based on the
[36] Gu H, Ho PL, Tong E, Wang L, Xu B. Presenting vanco- localized surface plasmon resonance spectroscopy of
mycin on nanoparticles to enhance antimicrobial triangular silver nanoparticles. Journal of the American
activities. Nano Letters 2003;3(9):1261e3. Chemical Society 2002;124(35):10596e604.
[37] Di Gianvincenzo P, Marradi M, Martínez-Ávila OM, [51] Gurunathan S, Lee KJ, Kalishwaralal K,
Bedoya LM, Alcamí J, Penadés S. Gold nanoparticles Sheikpranbabu S, Vaidyanathan R, Eom SH. Antiangio-
capped with sulfate-ended ligands as anti-HIV agents. genic properties of silver nanoparticles. Biomaterials
Bioorganic and Medicinal Chemistry Letters 2010; 2009;30(31):6341e50.
20(9):2718e21. [52] Khan MAM, Kumar S, Ahamed M, Alrokayan SA,
[38] Debouttière PJ, Roux S, Vocanson F, Billotey C, Beuf O, AlSalhi MS. Structural and thermal studies of silver
Favre-Réguillon A, et al. Design of gold nanoparticles for nanoparticles and electrical transport study of their
magnetic resonance imaging. Advanced Functional Ma- thin films. Nanoscale Research Letters 2011;6(1):434.
terials 2006;16(18):2330e9. [53] Chen D, Qiao X, Qiu X, Chen J. Synthesis and electrical
[39] Huang X, Qian W, El-Sayed IH, El-Sayed MA. The poten- properties of uniform silver nanoparticles for electronic
tial use of the enhanced nonlinear properties of gold applications. Journal of Materials Science 2009;44(4):
nanospheres in photothermal cancer therapy. Lasers in 1076e81.
Surgery and Medicine: The Official Journal of the Amer- [54] Xu R, Wang D, Zhang J, Li Y. Shape-dependent catalytic ac-
ican Society for Laser Medicine and Surgery 2007;39(9): tivity of silver nanoparticles for the oxidation of styrene.
747e53. ChemistryeAn Asian Journal 2006;1(6):888e93.
[40] Huang X, El-Sayed IH, Qian W, El-Sayed MA. Cancer cell [55] Naderi S, Ghaderi A, Solaymani S, Golzan MM. Struc-
imaging and photothermal therapy in the near-infrared tural, optical and thermal properties of silver colloidal
region by using gold nanorods. Journal of the American nanoparticles. The European Physical Journal-Applied
Chemical Society 2006;128(6):2115e20. Physics 2012;58(2).
[41] Agarwal A, Huang SW, O’donnell M, Day KC, Day M, [56] Feng QL, Wu J, Chen GQ, Cui FZ, Kim TN, Kim JO.
Kotov N, Ashkenazi S. Targeted gold nanorod contrast A mechanistic study of the antibacterial effect of silver
agent for prostate cancer detection by photoacoustic ions on Escherichia coli and Staphylococcus aureus. Journal
imaging. Journal of Applied Physics 2007;102(6): of Biomedical Materials Research 2000;52(4):662e8.
064701. [57] Smetana AB, Klabunde KJ, Marchin GR, Sorensen CM.
[42] Iravani S, Korbekandi H, Mirmohammadi SV, Biocidal activity of nanocrystalline silver powders and
Zolfaghari B. Synthesis of silver nanoparticles: chemical, particles. Langmuir 2008;24(14):7457e64.
physical and biological methods. Research in Pharma- [58] Nanda A, Saravanan M. Biosynthesis of silver nanopar-
ceutical Sciences 2014;9(6):385. ticles from Staphylococcus aureus and its antimicrobial ac-
[43] Pingali KC, Rockstraw DA, Deng S. Silver nanoparticles tivity against MRSA and MRSE. Nanomedicine:
from ultrasonic spray pyrolysis of aqueous silver Nanotechnology, Biology and Medicine 2009;5(4):
nitrate. Aerosol Science and Technology 2005;39(10): 452e6.
1010e4. [59] Kim KJ, Sung WS, Suh BK, Moon SK, Choi JS, Kim JG,
[44] Lee I, Han SW, Kim K. Simultaneous preparation of Lee DG. Antifungal activity and mode of action of silver
SERS-active metal colloids and plates by laser nano-particles on Candida albicans. Biometals 2009;
ablation. Journal of Raman Spectroscopy 2001; 22(2):235e42.
32(11):947e52. [60] Baram-Pinto D, Shukla S, Perkas N, Gedanken A, Sarid R.
[45] Long D, Wu G, Chen S. Preparation of oligochitosan sta- Inhibition of herpes simplex virus type 1 infection by sil-
bilized silver nanoparticles by gamma irradiation. Radi- ver nanoparticles capped with mercaptoethane sulfonate.
ation Physics and Chemistry 2007;76(7):1126e31. Bioconjugate Chemistry 2009;20(8):1497e502.
[46] Bönnemann H, Richards RM. Nanoscopic metal parti- [61] Lara HH, Ayala-Nuñez NV, Ixtepan-Turrent L,
cles synthetic methods and potential applications. Eu- Rodriguez-Padilla C. Mode of antiviral action of silver
ropean Journal of Inorganic Chemistry 2001;2001(10): nanoparticles against HIV-1. Journal of Nanobiotech-
2455e80. nology 2010;8(1):1.
[47] Mallick K, Witcomb MJ, Scurrell MS. Polymer stabilized [62] Lu L, Sun RW, Chen R, Hui CK, Ho CM, Luk JM,
silver nanoparticles: a photochemical synthesis route. Che CM. Silver nanoparticles inhibit hepatitis B virus
Journal of Materials Science 2004;39(14):4459e63. replication. Antiviral Therapy 2008;13(2):253.
[48] Navaladian S, Viswanathan B, Viswanath RP, [63] Sun L, Singh AK, Vig K, Pillai SR, Singh SR. Silver nano-
Varadarajan TK. Thermal decomposition as route for particles inhibit replication of respiratory syncytial virus.
silver nanoparticles. Nanoscale Research Letters 2007; Journal of Biomedical Nanotechnology 2008;4(2):
2(1):44. 149e58.
12 Intelligent Nanomaterials for Drug Delivery Applications

[64] Elechiguerra JL, Burt JL, Morones JR, Camacho- [78] Babes L, Denizot B, Tanguy G, Le Jeune JJ, Jallet P. Syn-
Bragado A, Gao X, Lara HH, Yacaman MJ. Interaction thesis of iron oxide nanoparticles used as MRI contrast
of silver nanoparticles with HIV-1. Journal of Nanobio- agents: a parametric study. Journal of Colloid and Inter-
technology 2005;3(1):6. face Science 1999;212(2):474e82.
[65] Rogers JV, Parkinson CV, Choi YW, Speshock JL, [79] Chertok B, Moffat BA, David AE, Yu F, Bergemann C,
Hussain SM. A preliminary assessment of silver nano- Ross BD, Yang VC. Iron oxide nanoparticles as a drug de-
particle inhibition of monkeypox virus plaque livery vehicle for MRI monitored magnetic targeting of
formation. Nanoscale Research Letters 2008;3(4):129. brain tumors. Biomaterials 2008;29(4):487e96.
[66] Jolivet JP, Chanéac C, Tronc E. Iron oxide chemistry. [80] Khodabakhshi A, Amin MM, Mozaffari M. Synthesis of
From molecular clusters to extended solid networks. magnetite nanoparticles and evaluation of its efficiency
Chemical Communications 2004;(5):481e3. for arsenic removal from simulated industrial
[67] Wan S, Huang J, Yan H, Liu K. Size-controlled prepara- wastewater. Iranian Journal of Environmental Health
tion of magnetite nanoparticles in the presence of graft Science and Engineering 2011;8(3):189e200.
copolymers. Journal of Materials Chemistry 2006; [81] Tran N, Mir A, Mallik D, Sinha A, Nayar S, Webster TJ.
16(3):298e303. Bactericidal effect of iron oxide nanoparticles on Staphy-
[68] Albornoz C, Jacobo SE. Preparation of a biocompatible lococcus aureus. International Journal of Nanomedicine
magnetic film from an aqueous ferrofluid. Journal of 2010;5:277.
Magnetism and Magnetic Materials 2006;305(1): [82] Ball V, Maechling C. Isothermal microcalorimetry to
12e5. investigate non specific interactions in biophysical
[69] Hou Y, Yu J, Gao S. Solvothermal reduction synthesis chemistry. International Journal of Molecular Sciences
and characterization of superparamagnetic magnetite 2009;10(8):3283e315.
nanoparticles. Journal of Materials Chemistry 2003; [83] Kwon GS. Polymeric micelles for delivery of poorly
13(8):1983e7. water-soluble compounds. Critical Reviews in Therapeu-
[70] Pascal C, Pascal JL, Favier F, Elidrissi Moubtassim ML, tic Drug Carrier Systems 2003;20(5).
Payen C. Electrochemical synthesis for the control of [84] Torchilin VP. PEG-based micelles as carriers of contrast
g-Fe2O3 nanoparticle size. Morphology, microstructure, agents for different imaging modalities. Advanced
and magnetic behavior. Chemistry of Materials 1999; Drug Delivery Reviews 2002;54(2):235e52.
11(1):141e7. [85] Ashok B, Arleth L, Hjelm RP, Rubinstein I, Önyüksel H.
[71] Franzel L, Bertino MF, Huba ZJ, Carpenter EE. Synthesis In vitro characterization of PEGylated phospholipid mi-
of magnetic nanoparticles by pulsed laser ablation. celles for improved drug solubilization: effects of PEG
Applied Surface Science 2012;261:332e6. chain length and PC incorporation. Journal of Pharma-
[72] Vijayakumar R, Koltypin Y, Felner I, Gedanken A. Sono- ceutical Sciences 2004;93(10):2476e87.
chemical synthesis and characterization of pure [86] Ahlawat J, Henriquez G, Narayan M. Enhancing the de-
nanometer-sized Fe3O4 particles. Materials Science and livery of chemotherapeutics: role of biodegradable poly-
Engineering: A 2000;286(1):101e5. meric nanoparticles. Molecules 2018;23(9):2157.
[73] Bandyopadhyay M, Bhattacharya J. Magnetic and caloric [87] New RR, editor. Liposomes. IRL at Oxford University
properties of magnetic nanoparticles: an equilibrium Press; 1990.
study. Journal of Physics: Condensed Matter 2006; [88] Sethi V, Önyüksel H, Rubinstein I. Liposomal vasoactive
18(49):11309. intestinal peptide. In: Methods in enzymology, vol. 391.
[74] Akbarzadeh A, Samiei M, Davaran S. Magnetic nanopar- Academic Press; 2005. p. 377e95.
ticles: preparation, physical properties, and applications [89] Allen TM, Chonn A. Large unilamellar liposomes with
in biomedicine. Nanoscale Research Letters 2012;7(1): low uptake into the reticuloendothelial system. FEBS
144. Letters 1987;223(1):42e6.
[75] Martchenko I, Dietsch H, Moitzi C, Schurtenberger P. [90] Papahadjopoulos D, Allen TM, Gabizon A, Mayhew E,
Hydrodynamic properties of magnetic nanoparticles Matthay K, Huang SK, et al. Sterically stabilized lipo-
with tunable shape anisotropy: prediction and experi- somes: improvements in pharmacokinetics and anti-
mental verification. The Journal of Physical Chemistry tumor therapeutic efficacy. Proceedings of the National
B 2011;115(49):14838e45. Academy of Sciences 1991;88(24):11460e4.
[76] Roca AG, Costo R, Rebolledo AF, Veintemillas- [91] Lasic DD, Martin FJ, Gabizon A, Huang SK,
Verdaguer S, Tartaj P, Gonzalez-Carreno T, et al. Progress Papahadjopoulos D. Sterically stabilized liposomes: a
in the preparation of magnetic nanoparticles for applica- hypothesis on the molecular origin of the extended cir-
tions in biomedicine. Journal of Physics D: Applied culation times. Biochimica et Biophysica Acta (BBA)-
Physics 2009;42(22):224002. Biomembranes 1991;1070(1):187e92.
[77] Chan DC, Kirpotin DB, Bunn Jr PA. Synthesis and eval- [92] Takeuchi H, Kojima H, Yamamoto H, Kawashima Y.
uation of colloidal magnetic iron oxides for the site- Evaluation of circulation profiles of liposomes coated
specific radiofrequency-induced hyperthermia of with hydrophilic polymers having different molecular
cancer. Journal of Magnetism and Magnetic Materials weights in rats. Journal of Controlled Release 2001;
1993;122(1e3):374e8. 75(1e2):83e91.
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 13

[93] Zamboni WC, Gervais AC, Egorin MJ, Schellens JH, [107] Calvo P, Remunan-Lopez C, Vila-Jato JL, Alonso MJ.
Zuhowski EG, Pluim D, et al. Systemic and tumor dispo- Novel hydrophilic chitosan-polyethylene oxide nano-
sition of platinum after administration of cisplatin or particles as protein carriers. Journal of Applied Polymer
STEALTH liposomal-cisplatin formulations (SPI-077 Science 1997;63(1):125e32.
and SPI-077 B103) in a preclinical tumor model of [108] Magenheim B, Levy MY, Benita S. A new in vitro tech-
melanoma. Cancer Chemotherapy and Pharmacology nique for the evaluation of drug release profile from
2004;53(4):329e36. colloidal carriers-ultrafiltration technique at low
[94] Zhang JA, Anyarambhatla G, Ma L, Ugwu S, Xuan T, pressure. International Journal of Pharmaceutics 1993;
Sardone T, Ahmad I. Development and characterization 94(1e3):115e23.
of a novel Cremophor® EL free liposome-based [109] Matsumura Y, Maeda H. A new concept for macromo-
paclitaxel (LEP-ETU) formulation. European Journal of lecular therapeutics in cancer chemotherapy: mecha-
Pharmaceutics and Biopharmaceutics 2005;59(1): nism of tumoritropic accumulation of proteins and
177e87. the antitumor agent smancs. Cancer Research 1986;
[95] Kadry AA, Al-Suwayeh SA, Abd-Allah AR, Bayomi MA. 46(12 Part 1):6387e92.
Treatment of experimental osteomyelitis by liposomal [110] Bae YH. Drug targeting and tumor heterogeneity. Jour-
antibiotics. Journal of Antimicrobial Chemotherapy nal of Controlled Release: Official Journal of the
2004;54(6):1103e8. Controlled Release Society 2009;133(1):2.
[96] Donald PR, Sirgel FA, Venter A, Smit E, Parkin DP, Van [111] Bae YH, Park K. Targeted drug delivery to tumors: myths,
de Wal BW, Mitchison DA. The early bactericidal activity reality and possibility. Journal of Controlled Release
of a low-clearance liposomal amikacin in pulmonary 2011;153(3):198.
tuberculosis. Journal of Antimicrobial Chemotherapy [112] Jain RK, Stylianopoulos T. Delivering nanomedicine to
2001;48(6):877e80. solid tumors. Nature Reviews Clinical Oncology 2010;
[97] Dagar S, Krishnadas A, Rubinstein I, Blend MJ, 7(11):653.
Önyüksel H. VIP grafted sterically stabilized liposomes [113] Wang M, Thanou M. Targeting nanoparticles to cancer.
for targeted imaging of breast cancer: in vivo studies. Pharmacological Research 2010;62(2):90e9.
Journal of Controlled Release 2003;91(1e2):123e33. [114] Kalaydina RV, Bajwa K, Qorri B, Decarlo A,
[98] Gessner A, Waicz R, Lieske A, Paulke BR, Mäder K, Szewczuk MR. Recent advances in “smart” delivery sys-
Müller RH. Nanoparticles with decreasing surface hydro- tems for extended drug release in cancer therapy. Inter-
phobicities: influence on plasma protein adsorption. In- national Journal of Nanomedicine 2018;13:4727.
ternational Journal of Pharmaceutics 2000;196(2): [115] Theek B, Gremse F, Kunjachan S, Fokong S, Pola R,
245e9. Pechar M, et al. Characterizing EPR-mediated passive
[99] Zweers ML, Engbers GH, Grijpma DW, Feijen J. In vitro drug targeting using contrast-enhanced functional ultra-
degradation of nanoparticles prepared from polymers sound imaging. Journal of Controlled Release 2014;182:
based on DL-lactide, glycolide and poly (ethylene 83e9.
oxide). Journal of Controlled Release 2004;100(3): [116] Nichols JW, Bae YH. EPR: evidence and fallacy. Journal
347e56. of Controlled Release 2014;190:451e64.
[100] Radwan MA, Zaghloul IY, Aly ZH. In vivo performance [117] Li X, Szewczuk MR, Malardier-Jugroot C. Folic
of parenteral theophylline-loaded polyisobutylcyanoa- acid-conjugated amphiphilic alternating copolymer
crylate nanoparticles in rats. European Journal of Phar- as a new active tumor targeting drug delivery
maceutical Sciences 1999;8(2):95e8. platform. Drug Design, Development and Therapy
[101] Deleted in review 2016;10:4101.
[102] Redhead HM, Davis SS, Illum L. Drug delivery in poly [118] Byrne JD, Betancourt T, Brannon-Peppas L. Active target-
(lactide-co-glycolide) nanoparticles surface modified ing schemes for nanoparticle systems in cancer
with poloxamer 407 and poloxamine 908: in vitro char- therapeutics. Advanced Drug Delivery Reviews 2008;
acterisation and in vivo evaluation. Journal of 60(15):1615e26.
Controlled Release 2001;70(3):353e63. [119] Ferrari M. Nanovector therapeutics. Current Opinion in
[103] Dunne M, Corrigan OI, Ramtoola Z. Influence of parti- Chemical Biology 2005;9(4):343e6.
cle size and dissolution conditions on the degradation [120] Kirpotin D, Park JW, Hong K, Zalipsky S, Li WL, Carter P,
properties of polylactide-co-glycolide particles. Bioma- et al. Sterically stabilized anti-HER2 immunoliposomes:
terials 2000;21(16):1659e68. design and targeting to human breast cancer cells
[104] Müller R, Maaben S, Weyhers H, Mehnert W. Phagocytic in vitro. Biochemistry 1997;36(1):66e75.
uptake and cytotoxicity of solid lipid nanoparticles (SLN) [121] Kirpotin DB, Drummond DC, Shao Y, Shalaby MR,
sterically stabilized with poloxamine 908 and poloxamer Hong K, Nielsen UB, et al. Antibody targeting of long-
407. Journal of Drug Targeting 1996;4(3):161e70. circulating lipidic nanoparticles does not increase tumor
[105] Couvreur P, Barratt G, Fattal E, Vauthier C. Nanocapsule localization but does increase internalization in animal
technology: a review. Critical Reviews in Therapeutic models. Cancer Research 2006;66(13):6732e40.
Drug Carrier Systems 2002;19(2). [122] Chen WC, Zhang AX, Li SD. Limitations and niches of
[106] Deleted in review the active targeting approach for nanoparticle drug
14 Intelligent Nanomaterials for Drug Delivery Applications

delivery. European Journal of Nanomedicine 2012; [136] Dharap SS, Qiu B, Williams GC, Sinko P, Stein S,
4(2e4):89e93. Minko T. Molecular targeting of drug delivery systems
[123] Ni S, Stephenson SM, Lee RJ. Folate receptor targeted de- to ovarian cancer by BH3 and LHRH peptides. Journal
livery of liposomal daunorubicin into tumor cells. Anti- of Controlled Release 2003;91(1e2):61e73.
cancer Research 2002;22(4):2131e5. [137] Pastorino F, Brignole C, Marimpietri D, Cilli M,
[124] Koo OM, Rubinstein I, Onyuksel H. Role of nanotech- Gambini C, Ribatti D, et al. Vascular damage and
nology in targeted drug delivery and imaging: a concise anti-angiogenic effects of tumor vessel-targeted lipo-
review. Nanomedicine: Nanotechnology, Biology and somal chemotherapy. Cancer Research 2003;63(21):
Medicine 2005;1(3):193e212. 7400e9.
[125] Zhang L, Hou S, Mao S, Wei D, Song X, Lu Y. Uptake of [138] Medina OP, Kairemo K, Valtanen H, Kangasniemi A,
folate-conjugated albumin nanoparticles to the SKOV3 Kaukinen S, Ahonen I, et al. Radionuclide imaging of tu-
cells. International Journal of Pharmaceutics 2004; mor xenografts in mice using a gelatinase-targeting
287(1e2):155e62. peptide. Anticancer Research 2005;25(1A):33e42.
[126] Maruyama K, Ishida O, Kasaoka S, Takizawa T, [139] Kono K, Ozawa T, Yoshida T, Ozaki F, Ishizaka Y,
Utoguchi N, Shinohara A, et al. Intracellular targeting Maruyama K, et al. Highly temperature-sensitive lipo-
of sodium mercaptoundecahydrododecaborate (BSH) somes based on a thermosensitive block copolymer
to solid tumors by transferrin-PEG liposomes, for boron for tumor-specific chemotherapy. Biomaterials 2010;
neutron-capture therapy (BNCT). Journal of Controlled 31(27):7096e105.
Release 2004;98(2):195e207. [140] Adochite RC, Moshnikova A, Golijanin J, Andreev OA,
[127] Omori N, Maruyama K, Jin G, Wang SJ, Hamakawa Y, Katenka NV, Reshetnyak YK. Comparative study of tu-
Sato K, et al. Targeting of post-ischemic cerebral endo- mor targeting and biodistribution of pH (Low) Inser-
thelium in rat by liposomes bearing polyethylene tion Peptides (pHLIP® peptides) conjugated with
glycol-coupled transferrin. Neurological Research different fluorescent dyes. Molecular Imaging and
2003;25(3):275e9. Biology 2016;18(5):686e96.
[128] Ishida O, Maruyama K, Tanahashi H, Iwatsuru M, [141] Weerakkody D, Moshnikova A, El-Sayed NS,
Sasaki K, Eriguchi M, Yanagie H. Liposomes bearing Adochite RC, Slaybaugh G, Golijanin J, et al. Novel
polyethyleneglycol-coupled transferrin with intracel- pH-sensitive cyclic peptides. Scientific Reports 2016;6:
lular targeting property to the solid tumors in vivo. Phar- 31322.
maceutical Research 2001;18(7):1042e8. [142] Mir M, Ishtiaq S, Rabia S, Khatoon M, Zeb A, Khan GM,
[129] Vinogradov SV, Batrakova EV, Kabanov AV. Nanogels et al. Nanotechnology: from in vivo imaging system to
for oligonucleotide delivery to the brain. Bioconjugate controlled drug delivery. Nanoscale Research Letters
Chemistry 2004;15(1):50e60. 2017;12(1):500.
[130] Gupta AK, Berry C, Gupta M, Curtis A. Receptor- [143] Blanco E, Shen H, Ferrari M. Principles of nanoparticle
mediated targeting of magnetic nanoparticles using in- design for overcoming biological barriers to drug
sulin as a surface ligand to prevent endocytosis. IEEE delivery. Nature Biotechnology 2015;33(9):941.
Transactions on Nanobioscience 2003;2(4):255e61. [144] Deleted in review
[131] Park JW, Hong K, Kirpotin DB, Colbern G, Shalaby R, [145] Sung JC, Pulliam BL, Edwards DA. Nanoparticles for
Baselga J, et al. Anti-HER2 immunoliposomes: drug delivery to the lungs. Trends in Biotechnology
enhanced efficacy attributable to targeted delivery. Clin- 2007;25(12):563e70.
ical Cancer Research 2002;8(4):1172e81. [146] Wadhwa S, Paliwal R, Paliwal SR, Vyas SP. Nanocarriers
[132] Gosk S, Vermehren C, Storm G, Moos T. Targeting antid in ocular drug delivery: an update review. Current Phar-
transferrin receptor antibody (OX26) and OX26- maceutical Design 2009;15(23):2724e50.
conjugated liposomes to brain capillary endothelial [147] Puglia C, Offerta A, Carbone C, Bonina F, Pignatello R,
cells using in situ perfusion. Journal of Cerebral Blood Puglisi G. Lipid nanocarriers (LNC) and their applica-
Flow and Metabolism 2004;24(11):1193e204. tions in ocular drug delivery. Current Medicinal Chem-
[133] Fonseca MJ, Jagtenberg JC, Haisma HJ, Storm G. istry 2015;22(13):1589e602.
Liposome-mediated targeting of enzymes to cancer cells [148] Andrei G, Peptu CA, Popa M, Desbrieres J, Peptu C,
for site-specific activation of prodrugs: comparison with Gardikiotis F, et al. Formulation and evaluation of cefur-
the corresponding antibody-enzyme conjugate. Phar- oxim loaded submicron particles for ophthalmic
maceutical Research 2003;20(3):423e8. delivery. International Journal of Pharmaceutics 2015;
[134] Li L, Wartchow CA, Danthi SN, Shen Z, Dechene N, 493(1e2):16e29.
Pease J, et al. A novel antiangiogenesis therapy using an [149] Pradhan N, Guha R, Chowdhury S, Nandi S, Konar A,
integrin antagonist or antieFlk-1 antibody coated 90Y- Hazra S. Curcumin nanoparticles inhibit corneal
labeled nanoparticles. International Journal of Radiation neovascularization. Journal of Molecular Medicine
Oncology Biology Physics 2004;58(4):1215e27. 2015;93(10):1095e106.
[135] Sapra P, Allen TM. Ligand-targeted liposomal anti- [150] Asasutjarit R, Theerachayanan T, Kewsuwan P,
cancer drugs. Progress in Lipid Research 2003;42(5): Veeranodha S, Fuongfuchat A, Ritthidej GC. Develop-
439e62. ment and evaluation of diclofenac sodium loaded-N-
CHAPTER 1 Introduction to Active, Smart, and Intelligent Nanomaterials 15

Trimethyl chitosan nanoparticles for ophthalmic use. [164] Kreuter J. Influence of the surface properties on
AAPS PharmSciTech 2015;16(5):1013e24. nanoparticle-mediated transport of drugs to the brain.
[151] Singh B, Garg T, Goyal AK, Rath G. Recent advance- Journal of Nanoscience and Nanotechnology 2004;
ments in the cardiovascular drug carriers. Artificial Cells, 4(5):484e8.
Nanomedicine, and Biotechnology 2016;44(1): [165] Michaelis K, Hoffmann MM, Dreis S, Herbert E,
216e25. Alyautdin RN, Michaelis M, et al. Covalent linkage of
[152] Haeri A, Sadeghian S, Rabbani S, Anvari MS, apolipoprotein e to albumin nanoparticles strongly en-
Ghassemi S, Radfar F, Dadashzadeh S. Effective attenua- hances drug transport into the brain. Journal of Pharma-
tion of vascular restenosis following local delivery of cology and Experimental Therapeutics 2006;317(3):
chitosan decorated sirolimus liposomes. Carbohydrate 1246e53.
Polymers 2017;157:1461e9. [166] De Jong WH, Borm PJ. Drug delivery and nanoparticles:
[153] Formiga FR, Pelacho B, Garbayo E, Abizanda G, applications and hazards. International Journal of
Gavira JJ, Simon-Yarza T, et al. Sustained release of Nanomedicine 2008;3(2):133.
VEGF through PLGA microparticles improves vasculo- [167] Gao X, Tao W, Lu W, Zhang Q, Zhang Y, Jiang X, Fu S.
genesis and tissue remodeling in an acute myocardial Lectin-conjugated PEGePLA nanoparticles: preparation
ischemiaereperfusion model. Journal of Controlled and brain delivery after intranasal administration. Bio-
Release 2010;147(1):30e7. materials 2006;27(18):3482e90.
[154] Danhier F, Feron O, Préat V. To exploit the tumor micro- [168] Radomski A, Jurasz P, Alonso-Escolano D, Drews M,
environment: passive and active tumor targeting of Morandi M, Malinski T, Radomski MW. Nanoparticle-
nanocarriers for anti-cancer drug delivery. Journal of induced platelet aggregation and vascular thrombosis.
Controlled Release 2010;148(2):135e46. British Journal of Pharmacology 2005;146(6):882e93.
[155] Rahman AM, Yusuf SW, Ewer MS. Anthracycline- [169] Sayes CM, Liang F, Hudson JL, Mendez J, Guo W,
induced cardiotoxicity and the cardiac-sparing effect of Beach JM, et al. Functionalization density dependence
liposomal formulation. International Journal of Nano- of single-walled carbon nanotubes cytotoxicity
medicine 2007;2(4):567. in vitro. Toxicology Letters 2006;161(2):135e42.
[156] Batist G. Cardiac safety of liposomal anthracyclines. Car- [170] Shvedova A, Castranova V, Kisin E, Schwegler-Berry D,
diovascular Toxicology 2007;7(2):72e4. Murray A, Gandelsman V, et al. Exposure to carbon
[157] Kundranda MN, Niu J. Albumin-bound paclitaxel in nanotube material: assessment of nanotube cytotoxicity
solid tumors: clinical development and future using human keratinocyte cells. Journal of Toxicology
directions. Drug Design, Development and Therapy and Environmental Health Part A 2003;66(20):
2015;9:3767. 1909e26.
[158] Bhaw-Luximon A, Jhurry D. New avenues for improving [171] Nel A, Xia T, Mädler L, Li N. Toxic potential of materials
pancreatic ductal adenocarcinoma (PDAC) treatment: at the nanolevel. Science 2006;311(5761):622e7.
selective stroma depletion combined with nano drug [172] Lin W, Huang YW, Zhou XD, Ma Y. In vitro toxicity of
delivery. Cancer Letters 2015;369(2):266e73. silica nanoparticles in human lung cancer cells. Toxi-
[159] Zhou L, Wang H, Li Y. Stimuli-responsive nanomedi- cology and Applied Pharmacology 2006;217(3):252e9.
cines for overcoming cancer multidrug resistance. Thera- [173] Hare JI, Lammers T, Ashford MB, Puri S, Storm G,
nostics 2018;8(4):1059. Barry ST. Challenges and strategies in anti-cancer nano-
[160] Li J, Liu J, Guo N, Zhang X. Reversal of multidrug resis- medicine development: an industry perspective.
tance in breast cancer MCF-7/ADR cells by h-R3- Advanced Drug Delivery Reviews 2017;108:25e38.
siMDR1-PAMAM complexes. International Journal of [174] Allen TM, Cullis PR. Drug delivery systems: entering the
Pharmaceutics 2016;511(1):436e45. mainstream. Science 2004;303(5665):1818e22.
[161] Olivier JC, Fenart L, Chauvet R, Pariat C, Cecchelli R, [175] Tinkle S, McNeil SE, Mühlebach S, Bawa R, Borchard G,
Couet W. Indirect evidence that drug brain targeting us- Barenholz Y, et al. Nanomedicines: addressing the scien-
ing polysorbate 80-coated polybutylcyanoacrylate tific and regulatory gap. Annals of the New York Acad-
nanoparticles is related to toxicity. Pharmaceutical emy of Sciences 2014;1313(1):35e56.
Research 1999;16(12):1836e42. [176] Gaspar R. Regulatory issues surrounding nanomedicines:
[162] Kreuter J, Ramge P, Petrov V, Hamm S, Gelperina SE, setting the scene for the next generation of
Engelhardt B, et al. Direct evidence that polysorbate- nanopharmaceuticals. Nanomedicine 2007;2(2):143e7.
80-coated poly (butylcyanoacrylate) nanoparticles [177] Storm G, Ten Kate MT, Working PK, Bakker-
deliver drugs to the CNS via specific mechanisms Woudenberg IA. Doxorubicin entrapped in sterically sta-
requiring prior binding of drug to the nanoparticles. bilized liposomes: effects on bacterial blood clearance
Pharmaceutical Research 2003;20(3):409e16. capacity of the mononuclear phagocyte system. Clinical
[163] Lockman PR, Koziara JM, Mumper RJ, Allen DD. Nano- Cancer Research 1998;4(1):111e5.
particle surface charges alter bloodebrain barrier integ- [178] Oomen AG, Bos PM, Fernandes TF, Hund-Rinke K,
rity and permeability. Journal of Drug Targeting 2004; Boraschi D, Byrne HJ, et al. Concern-driven integrated
12(9e10):635e41. approaches to nanomaterial testing and assessmente
16 Intelligent Nanomaterials for Drug Delivery Applications

report of the NanoSafety Cluster Working Group 10. extract and their anticancer activity. International Jour-
Nanotoxicology 2014;8(3):334e48. nal of Nanotechnology 2016;2(2):1e4.
[179] Costigan S. The toxicology of nanoparticles used in [184] Ahmad N, Bhatnagar S, Ali SS, Dutta R. Phytofabrication
health care products. Available at: the website of the of bioinduced silver nanoparticles for biomedical
Medicines and Healthcare products Regulatory Agency, applications. International Journal of Nanomedicine
Department of Health, UK. Accessed, 20. 2006. 2015;10:7019.
[180] Sahai N, Ahmad N, Gogoi M. Nanoparticles based drug [185] Ahmad N, Shree K, Srivastava M, Dutta R. Novel rapid
delivery for tissue regeneration using biodegradable biological approach for synthesis of silver nanoparticles
scaffolds: a review. Current Pathobiology Reports and its characterization. International Journal of Phar-
2018;6(4):219e24. macology 2014;1(1):28e31.
[181] Khatoon A, Khan F, Ahmad N, Shaikh S, Rizvi SMD, [186] Ahmad N, Bhatnagar S, Dubey SD, Saxena R, Sharma S,
Shakil S, et al. Silver nanoparticles from leaf extract of Dutta R. Nanopackaging in food and electronics. In:
Mentha piperita: eco-friendly synthesis and effect on Nanoscience in food and agriculture, vol. 4. Cham:
acetylcholinesterase activity. Life Sciences 2018;209: Springer; 2017. p. 45e97.
430e4. [187] Ahlawat J, Kumar V, Gopinath P. Carica papaya loaded
[182] Ahmad N, Bhatnagar S, Saxena R, Iqbal D, Ghosh AK, poly (vinyl alcohol)-gelatin nanofibrous scaffold for po-
Dutta R. Biosynthesis and characterization of gold nano- tential application in wound dressing. Materials Science
particles: kinetics, in vitro and in vivo study. Materials and Engineering: C 2019:109834.
Science and Engineering: C 2017;78:553e64. [188] Ahlawat J, Deemer EM, Narayan M. Chitosan nanopar-
[183] Ahmad N, Rizvi S, Sahai N, Dutta R. Biosynthesis, char- ticles rescue rotenone-mediated cell death. Materials
acterization of gold nanoparticles using M. indica leaf 2019;12(7):1176.

You might also like