You are on page 1of 12

Artificial Cells, Nanomedicine, and Biotechnology

An International Journal

ISSN: 2169-1401 (Print) 2169-141X (Online) Journal homepage: https://www.tandfonline.com/loi/ianb20

Nanoparticles as carriers for drug delivery in


cancer

Ankita Dadwal, Ashish Baldi & Raj Kumar Narang

To cite this article: Ankita Dadwal, Ashish Baldi & Raj Kumar Narang (2018) Nanoparticles as
carriers for drug delivery in cancer, Artificial Cells, Nanomedicine, and Biotechnology, 46:sup2,
295-305, DOI: 10.1080/21691401.2018.1457039

To link to this article: https://doi.org/10.1080/21691401.2018.1457039

Published online: 25 Jul 2018.

Submit your article to this journal

Article views: 4868

View related articles

View Crossmark data

Citing articles: 5 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ianb20
ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY
2018, VOL. 46, NO. S2, S295–S305
https://doi.org/10.1080/21691401.2018.1457039

Nanoparticles as carriers for drug delivery in cancer


Ankita Dadwala,b, Ashish Baldib and Raj Kumar Naranga
a
Department of Pharmaceutics, I.S.F. College of Pharmacy, Moga, India; bDepartment of Pharmaceutical Science and Technology, Maharaja
Ranjit Singh Punjab Technical University, Bathinda, India

ABSTRACT ARTICLE HISTORY


Cancer nanotherapeutics are swiftly progressing and are being applied to solve several limitations of Received 15 December 2017
conventional drug delivery systems such as non-specific biodistribution and targeting, lack of water Revised 17 March 2018
solubility and poor oral bioavailability. Advances in protein engineering and materials science have con- Accepted 21 March 2018
tributed to novel nanoscale targeting approaches that may bring new hope to cancer patients. Several
KEYWORDS
therapeutic nanocarriers have been approved for clinical use. Nanoparticles have been designed for Metallic nanocapsules;
optimal size and surface characteristics to improve their biodistribution and to increase their circulation thermoresponsive systems;
time in the bloodstream. By selectively using the unique pathophysiology of tumours, such as their viral nanoparticles
enhanced permeability and retention effect nanotherapeutics are able to carry loaded active drug to
cancer cells. In addition to this passive targeting mechanism, active targeting strategies using ligands or
antibodies directed against selected tumour targets magnify the specificity of these therapeutic nano-
particles. Drug resistance, another obstacle can also be overcome or reduced by using nanoparticles.
Multifunctional and multiplex nanoparticles are now being actively investigated and are on the horizon
as the next generation of nanoparticles, facilitating personalized and tailored cancer treatment.

Introduction nanoparticles offer many advantages as drug carrier systems,


there are still many limitations to be solved such as poor oral
Cancer remains one of the world’s most devastating diseases,
bioavailability, instability in circulation, inadequate tissue dis-
with more than 10 million new cases every year [1]. However,
tribution and toxicity. In this reviews provide perspective on
mortality has decreased in the past 2 years [2] owing to bet-
the use of nanotechnology as a fundamental tool in cancer
ter understanding of tumour biology and improved diagnos-
research and nanomedicine [7,8]. Here we focus on the types
tic devices and treatments. Current cancer treatments include
and characteristics of nanoparticles, how nanoparticles are
surgical intervention, radiation and chemotherapeutic drugs,
being used as drug delivery systems to kill cancer cells more
which often also kill healthy cells and cause toxicity to the
effectively and also to reduce or overcome drug resistance
patient. Conventional chemotherapeutic agents also do not
and how nanoparticles will be developed to improve their
show targeted action and are distributed non-specifically in
therapeutic efficacy and functionality in future can-
the body where they affect both cancerous and normal cells,
cer treatments.
thereby limiting the dose achievable within the tumour cells
and also resulting in suboptimal treatment due to excessive
toxicities. Molecular targeting therapy has emerged as one
Targeted delivery of nanoparticles
approach to overcome the lack of specificity of conventional
chemotherapeutic agents [3]. However, the resistance devel- Ideally, for the effectiveness of anticancer drugs in cancer
opment in cancer cells can dodge the cytotoxicity not only of treatment, they should first, after administration, be able to
conventional chemotherapeutics but also of newer molecular penetrate through the barriers in the body and reach the
targeting therapeutics [4]. By using both passive and active desired tumour tissues with minimal loss of their volume or
targeting strategies, intracellular concentration of drugs in activity in the blood circulation. Second, after reaching the
cancer cells can be improved by nanoparticles while avoiding desired site, drugs should have the ability to selectively kill
toxicity in normal cells (Figure 1) [5,6]. Passive targeting feats tumour cells without affecting normal cells. These two basic
the characteristic features of tumour biology that allows approaches are also associated with improvements in patient
nanocarriers to accumulate in a tumour by the enhanced per- survival and quality of life by increasing the intracellular con-
meability and retention (EPR) [2]. Active approaches achieve centration of drugs and reducing dose-limiting toxicities sim-
this by conjugating nanocarriers containing chemotherapeu- ultaneously. Increasingly, nanoparticles seem to have the
tics with molecules that bind to overexpressed antigens potential to satisfy both of these requirements for effective
or receptors on the target cells. However, although drug carrier systems.

CONTACT Raj Kumar Narang drrknisf@gmail.com Department of Pharmaceutics, ISF College of Pharmacy, Moga 142001, Punjab, India
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
S296 A. DADWAL ET AL.

Figure 1. Improving cancer treatment.

Size and surface characteristics of nanoparticles reach tumour tissues by passing through these two particular
vascular structures.
Nanoparticles must have the ability to remain in the blood-
stream for a considerable time without being eliminated for
effective delivery of drug to the targeted tumour tissue. Passive and active targeting
Conventional surface particles and non-modified nanopar-
Nanocarriers come across numerous barriers in their route to
ticles are usually caught in the circulation by the reticulo-
the target, such as mucosal barriers and non-specific uptake
endothelial system, such as the liver and the spleen,
depending on their size and surface characteristics [9]. The [14,15]. To report the challenges in targeting tumours with
fate of injected nanoparticles can be controlled by adjusting nanotechnology, it is essential to combine the rational design
their size and surface characteristics. of nanocarriers with the fundamental understanding of
Surface characteristics. Surface characteristics of nanopar- tumour biology. General features of tumours include Poor
ticles are an important factor for determining their lifespan lymphatic drainage and leaky blood vessels. Whereas free
and destiny during circulation relating to their capture by drugs may diffuse non-specifically, a nanocarrier can escape
macrophages. Ideally, nanoparticles should have hydrophilic into the tumour tissues via the leaky vessels by the enhanced
surface so that they can escape macrophage capture [10]. permeability and retention effect (EPR effect) [16] (Figure 2).
This can be achieved by two methods, first coating the sur- There is rapid and defective angiogenesis (formation of new
face of nanoparticles with a hydrophilic polymer, such as Peg, blood vessels from existing ones) because of which there is
second, protects them from opsonization by repelling plasma increased permeability of blood vessels in tumour cells.
proteins; alternatively, nanoparticles can be formed from Furthermore, the dysfunctional lymphatic drainage in
block copolymers with hydrophilic and hydrophobic tumours also helps in retaining the accumulated nanocarriers
domains [11]. and allows them to release drugs into the locality of the
Size. In addition to their surface characteristics, there is tumour cells. Experiments using liposomes of different mean
also one more advantage of nanoparticles is that their size size suggest that the threshold vesicle size for extravasation
can be adjusted. The size of nanoparticles used in a drug into tumours is 400 nm [13], but other studies have shown
delivery system should be small enough to escape capture by that particles having diameters <200 nm are more effective
fixed macrophages that are lodged in the reticuloendothelial [17,18]. Based on clinical therapy passive targeting
system, such as the liver and spleen but should be large approaches suffer from several limitations. Some drugs can-
enough to prevent their rapid leakage into blood capillaries. not diffuse efficiently so targeting cells within tumour is not
The size of the sinusoid in the spleen and fenestra of the always possible and the random nature of the approach
Kuffer cells in the liver varies from 150 to 200 nm [12] and makes it difficult to control the process because of this lack
the size of gap junction between endothelial cells of the of control multiple-drug resistance (MDR) may induce – a
leaky tumour vasculature may vary from 100 to 600 nm [13]. situation where chemotherapy treatments fail patients shows
Thus, the size of nanoparticles should be up to 100 nm to resistance of cancer cells towards one or more drugs.
ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY S297

Figure 2. Schematic representation of different mechanisms by which nanocarriers can deliver drugs to tumours. (A) Passive tissue targeting and (B) active cellu-
lar targeting.

MDR occurs because of the overexpression of transporter pro- cancer (B-cell lymphoma) were labelled with an internalizing
teins that expel drugs from cells on the surface of cancer anti-CD19 ligand a more significant therapeutic outcome was
cells [4,19,20]. Expelling drugs inevitably lowers the thera- achieved rather than a non-internalizing anti-CD20 ligand
peutic effect and cancer cells soon develop resistance to a [26]. In contrast, owing to the bystander effect targeting
variety of drugs. The passive strategy is further limited nanocarriers to non-internalizing receptors may sometimes be
because certain tumours do not exhibit the EPR effect, and advantageous in solid tumours, where cells lacking the target
the permeability of vessels may not be the same throughout receptor can be killed through drug release at the surface of
a single tumour [21]. One way to overcome these limitations the neighbouring cells, where carriers can bind [27]. It is gen-
is to modify the nanocarriers in such a way that they actively erally known that higher binding affinity increases targeting
bind to specific cells after extravasation. This can be achieved efficacy. However, for solid tumours due to a “binding-site
by attaching targeting agents such as ligands – molecules barrier” high binding affinity can decrease penetration of
that can bind to specific receptors on the cell surface – to nanocarriers, where the nanocarrier binds to its target so
the surface of the nanocarrier by a variety of conjugation
strongly that penetration into the tissue is prevented. In add-
chemistries. Nanocarriers will recognize the receptor and bind
ition to enhanced affinity, multivalent binding effects may
to target cells through ligand–receptor interactions, and drug
also be used to improve targeting. The collective binding in a
will be released inside the cell (Figure 2). In general, targeting
multivalent interaction is much stronger than monovalent
agent which is used to deliver nanocarriers to cancer cells, it
binding. For example, dendrimer nanocarriers conjugated to
is imperative that the agent should binds with high selectivity
3–15 folate molecules showed a 2500–170,000-fold enhance-
to molecules that are uniquely expressed on the cell surface.
Other important consideration is that to maximize specificity, ment in dissociation constants (KD) over free folate when
a surface marker which can be an antigen or receptor should attaching to folate-binding proteins immobilized on a surface.
be overexpressed on target cells than in the normal cells. For This was attributed to the avidity of the multiple folic acid
example, to efficiently deliver liposomes to B-cell receptors groups on the periphery of the dendrimers [28].
using the anti-CD19 monoclonal antibody (mAb), the density
of receptors should be in the range of 104–105 copies per
cell. Those with lower density are less effectively targeted
[22]. In a breast cancer model, a receptor density of 105 cop- Types of nanoparticles used as drug delivery systems
ies of ErbB2 receptors per cell was necessary to improve
the therapeutic efficacy of an anti-ErbB2-targeted liposomal Nanoparticles useful as drug delivery systems these are sub-
doxorubicin relative to its non-targeted counterpart [23]. The micron sized particles (3–200 nm), devices or systems that
binding of certain ligands to their receptors may cause recep- can be made by using a variety of materials including lipids
tor-mediated internalization, which is often necessary if nano- (liposomes), polymers (polymeric nanoparticles, micelles or
carriers are to release drugs inside the cells [6,24,25]. For dendrimers), viruses (viral nanoparticles) and even organo-
example, when immunoliposomes targeted to human blood metallic compound (nanotubes; Table 1).
S298

Table 1. Representative examples of nanocarrier-based drugs on the market.


System Characteristics Structure Examples of compounds References
Polymeric nanoparticles (polymer- (a) Water-soluble, nontoxic, Drugs are conjugated to the side Albumin-Taxol (Abraxane) [29–32]
A. DADWAL ET AL.

drug conjugates) (b) Biodegradable Surface modifica- chain of a linear polymer with a PGA-Taxol (Xyotax)
tion (pegylation) linker (cleavable bond) PGA-Camptothecin (CT-2106)
(c) Selective accumulation and reten- HPMA-DOX (PK1)
tion in tumour tissue (EPR effect) HPMA-DOX-galactosamine (PK2)
(d) Specific targeting of cancer cells
while sparing normal cells – recep-
tor-mediated targeting with
a ligand
Liposomes (a) Amphiphilic, biocompatible Self-assembling closed colloidal Pegylated liposomal DOX (Doxil) [33–35]
(b) Ease of modification structures composed of Non-pegylated liposomal
(c) Targeting potential lipid bilayers DOX (Myocet)
Liposomal daunorubicin (DaunoXome)
Polymeric micelles (a) Suitable carrier for water-insoluble Amphiphilic block copolymers assem- PEG-pluronic-DOX [36–38]
drug ble and form a micelle with a PEG-PAA-DOX (NK911)
(b) Biocompatible, self-assembling, hydrophobic core and hydro- PEG-PLA-Taxol (Genexol-PM)
biodegradable philic shell
(c) Ease of functional modification
(d) Targeting potential
Viral nanoparticles (a) Surface modification by mutagen- Protein cages, which are multivalent, HSP-DOX
esis or bioconjugation – multiva- self-assembled structures CPMV-DOX
lency
(b) Specific tumour targeting, multi-
functionality
(c) Defined geometry and remarkable
uniformity
(d) Biological compatibility and
inert nature
Carbon nanotubes (a) Water-soluble and biocompatible Carbon cylinders composed of CNT-MTX
through chemical modification benzene ring CNT-amphotericin B
(organic functionalization)
(b) Multifunctionality
Dendrimers (a) Biodistribution and PK can be Radially emerging hyperbranched PAMAM-MTX [39]
tuned synthetic polymer with regular pat- PAMAM-platinate
(b) High structural and chemical tern and repeated units
homogeneity
(c) Ease of functionalization, high
ligand density
(d) Controlled degradation
(e) Multifunctionality
ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY S299

Figure 3. Types of nanocarriers for drug delivery. (A) polymeric nanoparticles: polymeric nanoparticles in which drugs are conjugated to or encapsulated in poly-
mers. (B) Polymeric micelles: amphiphilic block copolymers that form to nanosized core/shell structure in aqueous solution. The hydrophobic core region serves as a
reservoir for hydrophobic drugs, whereas hydrophilic shell region stabilizes the hydrophobic core and renders the polymer to be water-soluble. (C) Dendrimers: syn-
thetic polymeric macromolecule of nanometer dimensions, which is composed of multiple highly branched monomers that emerge radially from the central core.
(D) Liposomes: self-assembling structures composed of lipid bilayers in which an aqueous volume is entirely enclosed by a membranous lipid bilayer. (E) Viral-based
nanoparticles: in general structure are the protein cages, which are multivalent, self-assembles structures. (F) Carbon nanotubes: carbon cylinders composed of ben-
zene rings.

Polymer-based drug carriers circumvent the shortcomings of their free drug counterparts
Depending on the method of preparation of polymeric-based [43]. Among them, Xyotax (PGA paclitaxel; [30]) and CT-2106
drug carriers, the drug is either covalently bound to or phys- (PGA-camptothecin; [31]) are now in clinical trials. HPMA and
ically entrapped in polymer matrix [40]. The resulting com- PEG are the most widely used non-biodegradable synthetic
pounds may have the structure of capsules (polymeric polymers [44]. PK1, which is a conjugate of HPMA with doxo-
nanoparticles or polymer–drug conjugates), amphiphilic core/ rubicin, was the synthetic polymer-drug conjugate to be eval-
shell (polymeric micelles), or may be hyperbranched macro- uated in clinical trials as an anticancer agent. A phase I
molecules (dendrimers; Figure 3). Polymers used as drug con- clinical trial has been completed in patients with a variety of
jugates can be divided into two groups one is natural tumours that were refractory or resistant to prior therapy
polymers and other is synthetic polymers. such as chemotherapy and/or radiation [32]. PK1 should be
further evaluated in the next level of clinical trials.

Polymeric nanoparticles (polymer–drug conjugates)


Albumin, chitosan and heparin are naturally occurring poly-
mers and have been used as a material of choice for the Polymeric micelles (amphiphilic block copolymers)
delivery of DNA, oligonucleotides and protein, as well as Amphiphilic block copolymers are responsible for the func-
drugs. Recently, serum albumin is included as a carrier for the tional properties of micelles, which accumulate to form a
formulation of paclitaxel nanoparticle [nanometer-sized albu- nanosized core/shell structure in aqueous media (Figure 3(B)).
min-bound paclitaxel (Abraxane); Figure 3(A)], has been used For hydrophobic drugs hydrophobic core region serves as a
in the clinic for the treatment of metastatic breast cancer reservoir, while the hydrophilic shell region helps in stabiliz-
[29]. Besides metastatic breast cancer, Abraxane has also ing the hydrophobic core and renders the polymers water-
been evaluated in clinical trials for many other cancers soluble, making the particle a suitable candidate for i.v.
including non-small-cell lung cancer (phase II trial) and administration [45]. There are two methods of drug loading
advanced non-haematologic malignancies (phase I and into a polymeric micelle: first is physical encapsulation [36]
pharmacokinetics trials; [41,42]). PGA was the first biodegrad- and second is chemical covalent attachment [37]. The first
able polymer to be used for conjugate synthesis among other polymeric micelle formulation of paclitaxel, Genexol-PM (PEG-
synthetic polymers such as N-(2-hydroxypropyl)-methacryla- poly(D,L-lactide)-paclitaxel), is a cremophor-free polymeric
mide copolymer (HPMA), polystyrene-maleic anhydride micelle-formulated paclitaxel. A phase I trial and pharmacoki-
copolymer, polyethylene glycol (PEG) and poly-L-glutamic netic study has been conducted in patients with advanced
acid (PGA). Several representative chemotherapeutics that are refractory malignancies [38]. Multifunctional polymeric
used widely in the clinic have been tested as conjugates with micelles containing targeting ligands, imaging and thera-
PGA in vitro and in vivo and showed encouraging abilities to peutic agents are being actively developed [46] and will
S300 A. DADWAL ET AL.

become conventional among several models of the micellar Viral nanoparticles


formulation in the near future.
A variety of viruses including cowpea mosaic virus, canine
parvovirus, cowpea chlorotic mottle virus and bacteriophages
have been developed for biomedical and nanotechnology
Dendrimers applications that include drug delivery and tissue targeting
Dendrimers are synthetic polymeric macromolecules of nano- (Figure 3(E)). By using chemical or genetic means on the cap-
meter dimensions, composed of multiple highly branched sid surface a number of targeting molecules and peptides
monomers that emerge outward from the central core can be displayed in a biologically functional. Therefore, for
(Figure 3(C)). Properties allied with these dendrimers such as specific tumour targeting in vivo several ligands or antibodies,
including transferrin, folic acid and single-chain antibodies,
their monodisperse size, multivalency, modifiable surface
have been conjugated to viruses [48]. Besides this artificial
functionality, water solubility and available internal cavity
targeting, a subset of viruses, such as canine parvovirus, have
make them attractive for drug delivery [4]. Dendrimer which
natural affinity for receptors such as transferrin receptors that
is most widely used as a scaffold is polyamidoamine dendri-
are up-regulated on a variety of tumour cells [49]. By target-
mer, which was conjugated with cisplatin [39]. The easily
ing heat shock protein, a dual-function protein cage with spe-
modifiable surface characteristic of dendrimers permits them
cific targeting and doxorubicin encapsulation has been
to be simultaneously conjugated with several molecules such
developed [50,51] the permeability of vessels and the EPR
as imaging contrast agents, targeting ligands or therapeutic effect may not be the same throughout a single tumour [10].
drugs, yielding a dendrimer-based multifunctional drug deliv- To overcome these limitations is to programme the nanocar-
ery system [4]. riers in such a way so they actively bind to specific cells after
extravasation. This binding may be achieved by attaching tar-
geting agents such as ligands molecules that bind to specific
receptors on the cell surface to the surface of the nanocarrier
Lipid-based drug carriers
by a variety of conjugation chemistries. Nanocarriers will rec-
Liposomes ognize and bind to target cells through ligand–receptor inter-
First described in 1965, liposomes are one of the first nano- actions, and bound carriers are internalized before the drug
particle platforms to be applied in medicine [47]. Today, there is released inside the cell. In general, when using a targeting
are more than 11 formulations which are approved for clinical agent to deliver nanocarriers to cancer cells, it is imperative
use, with many more in clinical and preclinical development. that the agent binds with high selectivity to molecules that
Liposomes are self-assembling closed colloidal structures are uniquely expressed on the cell surface. Other important
composed of lipid bilayers having a spherical shape in which considerations are outlined below. To maximize specificity, a
an outer lipid bilayer surrounds a central aqueous space surface marker (antigen or receptor) should be overexpressed
(Figure 3(D)) [17]. Their biocompatible and biodegradable on target cells relative to normal cells. For example, to effi-
compositions, as well as their unique ability to encapsulate ciently deliver liposomes to B-cell receptors using the anti-
hydrophilic agents in their aqueous core and hydrophobic CD19 monoclonal antibody (mAb), the density of receptors
agents within their lamellae, make liposomes excellent thera- should be in the range of 104–105 copies per cell. Those
with lower density are less effectively targeted [11]. In a
peutic carriers. To improve their stability and circulation half-
breast cancer model, a receptor density of 105 copies of
life, liposomes can also be coated with polymers such as
ErbB2 receptors per cell was necessary to improve the thera-
polyethylene glycol (PEG) [17]. Liposomal drug formulations
peutic efficacy of an anti-ErbB2-targeted liposomal doxorubi-
typically improve the pharmacokinetics and biodistribution of
cin relative to its non-targeted counterpart [14]. The binding
a drug. For example, pegylated liposomal doxorubicin
of certain ligands to their receptors may cause receptor-medi-
reduces the volume of distribution of doxorubicin from
ated internalization, which is often necessary if nanocarriers
1000 l/m2 in the free drug form to 2.8 l/m2 by restricting
are to release drugs inside the cell [13,15,16]. For example, a
the distribution within the plasma. Furthermore, it can
more significant therapeutic outcome was achieved when
achieve higher drug concentrations within tumour while immunoliposomes targeted to human blood cancer (B-cell
reducing drug concentration in normal tissues, such as heart lymphoma) were labelled with an internalizing anti-CD19 lig-
[48]. Currently, several kinds of cancer drugs have been and rather than a non-internalizing anti-CD20 ligand [17]. In
applied to this lipid-based system using a variety of prepar- contrast, targeting nanocarriers to non-internalizing receptors
ation methods. Among them, liposomal formulations of the may sometimes be advantageous in solid tumours owing to
anthracyclines doxorubicin (Doxil, Myocet) and daunorubicin the bystander effect, where cells lacking the target receptor
(DaunoXome) are approved for the treatment of metastatic can be killed through drug release at the surface of the
breast cancer and AIDS-related Kaposi’s sarcoma [33,39]. neighbouring cells, where carriers can bind [18]. It is generally
Besides these approved agents, many liposomal chemothera- known that higher binding affinity increases targeting effi-
peutics are currently being evaluated in clinical trials [34]. cacy. However, for solid tumours, there is evidence that high
The next generation of liposomal drugs may be immunolipo- binding affinity can decrease penetration of nanocarriers due
somes, which selectively deliver the drug to the desired sites to a “binding-site barrier”, where the nanocarrier binds to its
of action [35]. target so strongly that penetration into the tissue is
ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY S301

Figure 4. Temperature-based mechanisms of drug delivery.

prevented [19]. In addition to enhanced affinity, multivalent 63 versus 70% in 48 h and 53 versus 62% in 72 h). The
binding effects (or avidity) may also be used to improve tar- increased drug efficacy could be driven by SWNT-mediated
geting. The collective binding in a multivalent interaction is cell internalization [53]. The multiple covalent functionaliza-
much stronger than monovalent binding. For example, den- tions on the sidewall or tips of carbon nanotubes allow them
drimer nanocarriers conjugated to 3–15 folate molecules to carry several molecules at once, and this strategy provides
showed a 2500–170,000-fold enhancement in dissociation a fundamental advantage in the treatment of cancer.
constants (KD) over free folate when attaching to folate-bind-
ing proteins immobilized on a surface. This was attributed to
Thermoresponsive systems
the avidity of the multiple folic acid groups on the periphery
of the dendrimers [20]. Thermoresponsive drug delivery is among the most explored
stimuli-responsive strategies and has been widely explored in
oncology. Thermoresponsiveness is usually governed by a
Carbon nanotubes
nonlinear sharp change with temperature in the properties of
Carbon nanotubes are carbon cylinders composed of ben- at least one component of the nanocarrier material. Such a
zene rings (Figure 3(G)) that have been applied in biology as sharp response triggers the release of the drug following a
sensors for detecting DNA and protein, diagnostic devices for variation in the surrounding temperature. Ideally, thermosen-
the discrimination of different proteins from serum samples, sitive nanocarriers should retain their load at body tempera-
and carriers to deliver vaccine or protein [47]. Carbon nano- ture (37  C), and rapidly deliver the drug within a locally
tubes are completely insoluble in all solvents, generating heated tumour (40–42  C) Figure 4 to nanochemotherapeu-
some health concerns and toxicity problems. However, the tics counteract rapid blood-passage time and washout from
introduction of chemical modification to carbon nanotubes the tumour.
can render them water-soluble and functionalized so that
they can be linked to a wide variety of active molecules such
Metallic nanocapsules
as peptides, proteins, nucleic acids and therapeutic agents
[17]. Antifungal agents (amphotericin B) or anticancer drugs The advantage of using a magnetic field relies on the differ-
(methotrexate) have been covalently linked to carbon nano- ent nature that the magnetic response can take, which can
tubes with a fluorescent agent (FITC). In an in vitro study, be a magnetic guidance under a permanent magnetic field, a
drugs bound to carbon nanotubes were shown to be more temperature increase when an alternating magnetic field is
effectively internalized into cells compared with free drug applied, or both when alternately used. Therefore, magnetic-
alone and to have potent antifungal activity [48,52]. Single- ally responsive systems allow for diversity in the drug delivery
walled carbon nanotube (SWNT)-based drug delivery system pathway. Furthermore, there is the possibility of performing
was developed by conjugation of human serum albumin magnetic resonance imaging, and hence to associate diag-
(HSA) nanoparticles for loading of antitumour agent PTX. The nostics and therapy within a single system (the so-called
SWNT-based drug carrier displayed high intracellular delivery theranostic approach) [54]. Magnetic guidance is typically
efficiency (cell uptake rate of 80%) in breast cancer MCF-7 obtained by focusing an extracorporeal magnetic field on the
cells, as examined by fluorescence-labelled drug carriers, sug- biological target during the injection of a magnetically
gesting the needle-shaped SWNT-HSA drug carrier was able responsive nanocarrier. This concept has demonstrated great
to transport drugs across cell membrane despite its macro- potential in experimental cancer therapy because of
molecular structure. The drug loading on SWNT-based drug improved drug accumulation inside solid-tumour models
carrier was through high binding affinity of PTX to HSA pro- Figure 5. Candidate nanosystems for such a therapeutic
teins. The PTX formulated with SWNT-HSA showed greater approach are core–shell nanoparticles (a magnetic core made
growth inhibition activity in MCF-7 breast cancer cells than of magnetite (Fe3O4) coated with silica or polymer) [55,56]
PTX formulated with HSA nanoparticle only (cell viability of magnetoliposomes (Fe3O4 or maghemite (Fe2O3) nanocrystals
S302 A. DADWAL ET AL.

Figure 5. Magnetically responsive systems.

encapsulated in liposomes) [57] and porous metallic nanocap- Enhanced permeability and retention effect
sules [58]. Most core–shell nanoparticles have shown promis-
Tumour vasculatures are generally abnormal, with aberrant
ing results in vitro, yet only some of them have demonstrated
branching and leaky walls [21]. This leakiness is due to the
improved tumour accumulation and anticancer pharmaco-
decreased number of pericytes and rapid proliferation of
logical efficacy in various in vivo models. To avoid limitations
endothelial cells. These characteristics result in large pores in
related to physical drug entrapment (for instance, uncon-
the tumour vasculatures, ranging from 100 nm to several hun-
trolled burst release or poor drug loading), the drugs and the
dred nanometers in diameter, as compared to normal vessel
nanocarriers can be covalently linked [56,59]. For example,
junctions of 5–10 nm [22]. These large pores allow higher vas-
Fe3O4 nanocrystals loaded into squalene–gemcitabine conju-
cular permeability and hydraulic conductivity in tumours, ena-
gate nanoassemblies exhibiting high drug payloads have
bling macromolecules such as nanoparticles to pass into
demonstrated the absence of burst release, enhancement of
tumours [21,23]. In normal tissue, lymphatic system clears the
the magnetic resonance imaging contrast in the targeted
macromolecules. However, solid tumours are generally char-
L1210 solid-tumour nodule and significant thera-
acterized by impaired lymphatics [6]. Proliferating tumour
peutic efficacy [59]. cells compress lymphatic vessels and collapse most of the
vessels, especially at the centre of tumours. The impaired
Nanoshells lymphatic system coupled with increased permeability of
tumour vasculature results in the EPR effect. Nanoparticles,
Owing to their non-invasiveness and the possibility of remote like other macromolecules, have extended retention times in
spatial and temporal control, in the past few years a large tumour, which results in higher concentrations than in
variety of photoresponsive systems has been engineered to plasma or in other tissues. Thus, nanoparticles can achieve
achieve on-demand drug release in response to illumination passive targeting to tumours through the EPR effect.
of a specific wavelength (in the ultraviolet, visible or near-
infrared (NIR) regions) Figure 6. The different strategies
available rely on either repeatable on–off drug-release or Nanoparticle clearance by the mononuclear
one-time event triggered by photosensitiveness-induced phagocyte system
structural modifications of the nanocarriers. For instance, To fully take advantage of the EPR effect, nanoparticles must
doxorubicin-loaded hollow gold nanospheres showed acceler- remain in circulation long enough for tumour accumulation.
ated drug release when irradiated at 808 nm, allowing However, nanoparticles are prone to clearance by the mono-
enhanced anticancer activity and reduced systemic toxicity nuclear phagocyte system, previously called the reticuloendo-
compared with the free-drug treatment [60] thelial system. The MPS is part of the immune system that is
mainly responsible for clearing macromolecules from circula-
Important concepts in nanoparticle drug delivery tion. Immunotoxin therapy of cancer [24]. The MPS comprises
bone marrow progenitors, blood monocytes and tissue mac-
for cancer
rophages. It also includes the Kupffer cells of the liver and
There are several general concepts that are important in macrophages of the spleen, which are responsible for clear-
nanoparticle drug delivery. These include the enhanced per- ance of macromolecules from circulation. Nanoparticles can
meability and retention (EPR) effect, nanoparticle clearance interact with MPS cells and lead to their opsonization. Since
by the mononuclear phagocyte system (MPS) and desirable premature elimination from circulation will prevent nanopar-
nanoparticle characteristics for cancer applications. ticles from accumulating in tumours, much effort has been
ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY S303

Figure 6. Light-triggered drug delivery systems.

devoted to creating “stealth” nanoparticles. The most com- well-designed clinical trials to further the development of
mon strategy has been grafting PEG or other macromolecules these drugs. Clinical investigators should fully understand
such as polysaccharides onto the nanoparticle surface [25]. the particular nanoparticles they are investigating and design
The presence of PEG or other molecules enables steric stabil- trials that take advantage of nanoparticle properties. More
ization, preventing protein adsorption, interactions among complex targeted systems, which can release nanochemo-
particles and interactions with immune cells. therapeutics at a target site when exposed to external stimuli
such as light and temperature, are also under development.
Another potential is to develop more nanoparticles capable
Future directions
of delivering combination chemotherapeutics. CPX-351, a lip-
Although nanomedicine is a relatively new branch of science, osomal formulation of cytarabine and daunorubicin, showed
its translation into clinical care has been rapid. The unique promising results in its first human study [18]. Together with
properties of nanoparticle drug carriers make them well the progression of nanoscale drug delivery systems, advances
suited for oncology applications. Nanoparticle chemothera- in nanoscale imaging suggest the potential for the develop-
peutics are composed to influence the treatment of most ment of multifunctional “smart” nanoparticles that may facili-
cancers. However, there are still limited clinical data and a tate the realization of individualized cancer therapy. Almost
limited number of nanotherapeutics approved for clinical use. all types of nanoparticles including polymeric nanoparticles
To fully understand the advantages and disadvantages of [61], nanocrystals [62], polymeric micelles [17], dendrimers
nanoparticle therapeutics, more clinical data are needed it [63] and carbon nanotubes [64] have been evaluated for their
can also help in identifying the best applications for nanoche- suitability as multifunctional nanoparticles that can be
motherapeutics. Thus, it is crucial to develop and carry out applied for simultaneous in vivo imaging and treatment of
S304 A. DADWAL ET AL.

cancers. Eventually, multiplex nanoparticles may be capable [23] Park JW, Hong K, Kirpotin DB, et al. Anti-HER2 immunoliposomes.
of detecting malignant cells (active targeting moiety), visualiz- Clin Cancer Res. 2002;8:1172–1181.
[24] Pastan I, Hassan R, Fitzgerald DJ, et al. Immunotoxin therapy of
ing their location in the body (real-time in vivo imaging), kill-
cancer. Nat Rev Cancer. 2006;6:559–565.
ing the cancer cells with minimal side effects by sparing [25] Peer D, Zhu P, Carman CV, et al. Selective gene silencing in acti-
normal cells (active targeting and controlled drug release or vated leukocytes by targeting siRNAs to the integrin lymphocyte
photothermal ablation) and monitoring treatment effects in function-associated antigen-1. Proc Natl Acad Sci USA.
real time. 2007;104:4095–4100.
[26] Sapra P, Allen TM. Internalizing antibodies are necessary for
improved therapeutic efficacy of antibody-targeted liposomal
Disclosure statement drugs. Cancer Res. 2002;62:7190–7194.
[27] Allen TM. Long-circulating (sterically stabilized) liposomes for tar-
The authors have declared no conflict of interest. The authors alone are geted drug delivery. Trends Pharmacol Sci. 1994;15:215–220.
responsible for the content and writing of the paper. [28] Hong S, Leroueil PR, Majoros IJ, et al. The binding avidity of a
nanoparticle-based multivalent targeted drug delivery platform.
Chem Biol. 2007;14:107–115.
References [29] Gradishar WJ, Tjulandin S, Davidson N, et al. Phase III trial of
nanoparticle albumin-bound paclitaxel compared with polyethy-
[1] Stewart BW, Kleihues P. 2003. World Cancer Report. Lyon: IARC lated castor oil-based paclitaxel in women with breast cancer. J
press. Clin Oncol. 2005;23:7794–7803.
[2] Society AC. 2007. Breast cancer facts & figures. Atlanta (GA): [30] Sabbatini P, Aghajanian C, Dizon D, et al. Phase II study of CT-
American Cancer Society. 2103 in patients with recurrent epithelial ovarian, fallopian tube,
[3] Ross JS, Schenkein DP, Pietrusko R, et al. Targeted therapies for
or primary peritoneal carcinoma. J Clin Oncol. 2004;22:4523–4531.
cancer 2004. Am J Clin Pathol. 2004;122:598–609. [31] Bhatt R, DE Vries P, Tulinsky J, et al. Synthesis and in vivo antitu-
[4] Cho K, Wang X, Nie S, et al. Therapeutic nanoparticles for drug
mor activity of poly (L-glutamic acid) conjugates of 20 (S)-campto-
delivery in cancer. Clin Cancer Res. 2008;14:1310–1316.
thecin. J Med Chem. 2003;46:190–193.
[5] Maeda H. The enhanced permeability and retention (EPR) effect in
[32] Vasey PA, Kaye SB, Morrison R, et al. Phase I clinical and pharma-
tumor vasculature: the key role of tumor-selective macromolecular
cokinetic study of PK1 [N-(2-hydroxypropyl) methacrylamide
drug targeting. Adv Enzyme Regul. 2001;41:189–207.
copolymer doxorubicin]: first member of a new class of chemo-
[6] Allen TM. Ligand-targeted therapeutics in anticancer therapy. Nat
therapeutic agents – drug-polymer conjugates. Clin Cancer Res.
Rev Cancer. 2002;2:750–763.
1999;5:83–94.
[7] Duncan R. Polymer conjugates as anticancer nanomedicines. Nat
[33] Markman M. Pegylated liposomal doxorubicin in the treatment of
Rev Cancer. 2006;6:688–701.
cancers of the breast and ovary. Expert Opin Pharmacother.
[8] Ferrari M. Cancer nanotechnology: opportunities and challenges.
2006;7:1469–1474.
Nat Rev Cancer. 2005;5:161–171.
[34] Rivera E. Current status of liposomal anthracycline therapy in
[9] Moghimi SM, Hunter AC, Murray JC. Long-circulating and target-
metastatic breast cancer. Clin Breast Cancer. 2003;4:S76–S83.
specific nanoparticles: theory to practice. Pharmacol Rev. 2001;53:
[35] Rosenthal E, Poizot-Martin I, Saint-Marc T, et al. Phase IV study of
283–318.
[10] Moghimi SM, Szebeni J. Stealth liposomes and long circulating liposomal daunorubicin (DaunoXome) in AIDS-related Kaposi sar-
nanoparticles: critical issues in pharmacokinetics, opsonization coma. Am J Clin Oncol. 2002;25:57–59.
[36] Batrakova E, Dorodnych TY, Klinskii EY, et al. Anthracycline antibi-
and protein-binding properties. Prog Lipid Res. 2003;42:463–478.
[11] Harris JM, Martin NE, Modi M. Pegylation. Clin Pharmacokinet. otics non-covalently incorporated into the block copolymer
2001;40:539–551. micelles: in vivo evaluation of anti-cancer activity. Br J Cancer.
[12] Wisse E, Braet F, Luo D, et al. Structure and function of sinusoidal 1996;74:1545–1552.
lining cells in the liver. Toxicol Pathol. 1996;24:100–111. [37] Nakanishi T, Fukushima S, Okamoto K, et al. Development of the
[13] Yuan F, Dellian M, Fukumura D, et al. Vascular permeability in a polymer micelle carrier system for doxorubicin. J Control Release.
human tumor xenograft: molecular size dependence and cutoff 2001;74:295–302.
size. Cancer Res. 1995;55:3752–3756. [38] Kim T-Y, Kim D-W, Chung J-Y, et al. Phase I and pharmacokinetic
[14] Couvreur P, Vauthier C. Nanotechnology: intelligent design to study of Genexol-Pm, a cremophor-free, polymeric micelle-formu-
treat complex disease. Pharm Res. 2006;23:1417–1450. lated paclitaxel, in patients with advanced malignancies. Clin
[15] Alonso MJ. Nanomedicines for overcoming biological barriers. Cancer Res. 2004;10:3708–3716.
Biomed Pharmacother. 2004;58:168–172. [39] Malik N, Evagorou EG, Duncan R. Dendrimer-platinate: a novel
[16] Matsumura Y, Maeda H. A new concept for macromolecular thera- approach to cancer chemotherapy. Anti-Cancer Drugs. 1999;10:
peutics in cancer chemotherapy: mechanism of tumoritropic accu- 767–776.
mulation of proteins and the antitumor agent smancs. Cancer [40] Rawat M, Singh D, Saraf S, et al. Nanocarriers: promising vehicle
Res. 1986;46:6387–6392. for bioactive drugs. Biol Pharm Bull. 2006;29:1790–1798.
Green M, Manikhas G, Orlov S, et al. AbraxaneV, a novel
R
[17] Torchilin VP. Recent advances with liposomes as pharmaceutical [41]
CremophorV-free, albumin-bound particle form of paclitaxel for
R
carriers. Nat Rev Drug Discov. 2005;4:145–160.
[18] Hobbs SK, Monsky WL, Yuan F, et al. Regulation of transport path- the treatment of advanced non-small-cell lung cancer. Ann Oncol.
ways in tumor vessels: role of tumor type and microenvironment. 2006;17:1263–1268.
Proc Natl Acad Sci USA. 1998;95:4607–4612. [42] Nyman DW, Campbell KJ, Hersh E, et al. Phase I and pharmacokin-
[19] Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: etics trial of ABI-007, a novel nanoparticle formulation of pacli-
role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58. taxel in patients with advanced nonhematologic malignancies. J
[20] Peer D, Margalit R. Fluoxetine and reversal of multidrug resistance. Clin Oncol. 2005;23:7785–7793.
Cancer Lett. 2006;237:180–187. [43] Li C. Poly (L-glutamic acid)–anticancer drug conjugates. Adv Drug
[21] Jain RK. Barriers to drug delivery in solid tumors. Sci Am. Deliv Rev. 2002;54:695–713.
1994;271:58–65. [44] Duncan R. The dawning era of polymer therapeutics. Nat Rev
[22] de Menezes DEL, Pilarski LM, Allen TM. In vitro and in vivo target- Drug Discov. 2003;2:347–360.
ing of immunoliposomal doxorubicin to human B-cell lymphoma. [45] Adams ML, Lavasanifar A, Kwon GS. Amphiphilic block copolymers
Cancer Res. 1998;58:3320–3330. for drug delivery. J Pharm Sci. 2003;92:1343–1355.
ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY S305

[46] Nasongkla N, Bey E, Ren J, et al. Multifunctional polymeric [56] Hua M-Y, Liu H-L, Yang H-W, et al. The effectiveness of a magnetic
micelles as cancer-targeted, MRI-ultrasensitive drug delivery sys- nanoparticle-based delivery system for BCNU in the treatment of
tems. Nano Lett. 2006;6:2427–2430. gliomas. Biomaterials. 2011;32:516–527.
[47] Bangham A. Liposomes: the Babraham connection. Chem Phys [57] Plassat V, Wilhelm C, Marsaud V, et al. Anti-estrogen-loaded super-
Lipids. 1993;64:275–285. paramagnetic liposomes for intracellular magnetic targeting and
[48] Hofheinz R-D, Gnad-Vogt SU, Beyer U, et al. Liposomal encapsu- treatment of breast cancer tumors. Adv Funct Mater.
lated anti-cancer drugs. Anticancer Drugs. 2005;16:691–707. 2011;21:83–92.
[49] Wu J, Liu Q, Lee RJ. A folate receptor-targeted liposomal formula- [58] Zhang F, Braun GB, Pallaoro A, et al. Mesoporous multifunctional
tion for paclitaxel. Int J Pharm. 2006;316:148–153. upconversion luminescent and magnetic “nanorattle” materials for
[50] Manchester M, Singh P. Virus-based nanoparticles (VNPs): platform targeted chemotherapy. Nano Lett. 2011;12:61–67.
technologies for diagnostic imaging. Adv Drug Deliv Rev. [59] Arias JL, Reddy LH, Othman M, et al. Squalene based nanocompo-
2006;58:1505–1522. sites: a new platform for the design of multifunctional pharma-
[51] Singh P, Destito G, Schneemann A, et al. Canine parvovirus-like ceutical theragnostics. ACS Nano. 2011;5:1513–1521.
particles, a novel nanomaterial for tumor targeting. J [60] You J, Zhang R, Xiong C, et al. Effective photothermal chemother-
Nanobiotechnol. 2006;4:2. apy using doxorubicin-loaded gold nanospheres that target
[52] Minelli C, Lowe SB, Stevens MM. Engineering nanocomposite EphB4 receptors in tumors. Cancer Res. 2012;72:4777–4786.
materials for cancer therapy. Small. 2010;6:2336–2357. [61] Hurwitz HI, Fehrenbacher L, Hainsworth JD, et al. Bevacizumab in
[53] Shao W, Paul A, Rodes L, et al. A new carbon nanotube- combination with fluorouracil and leucovorin: an active regimen
based breast cancer drug delivery system: preparation and in vitro for first-line metastatic colorectal cancer. J Clin Oncol. 2005;
analysis using paclitaxel. Cell Biochem Biophys. 2015; 23:3502–3508.
71:1405–1414. [62] Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus ritux-
[54] Yang H-W, Hua M-Y, Liu H-L, et al. Self-protecting core-shell imab compared with CHOP alone in elderly patients with diffuse
magnetic nanoparticles for targeted, traceable, long half- large-B-cell lymphoma. N Engl J Med. 2002;346:235–242.
life delivery of BCNU to gliomas. Biomaterials. 2011; [63] Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a spe-
32:6523–6532. cific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid
[55] Zhang L, Wang T, Yang L, et al. General route to multifunctional leukemia. N Engl J Med. 2001;2001:1031–1037.
uniform yolk/mesoporous silica shell nanocapsules: a platform for [64] Wang AZ, Gu F, Zhang L, et al. Biofunctionalized targeted nano-
simultaneous cancer-targeted imaging and magnetically guided particles for therapeutic applications. Expert Opin Biol Ther. 2008;
drug delivery. Chem Eur J. 2012;18:12512–12521. 8:1063–1070.

You might also like