ChapterPDF Available

Bionanomaterials for cancer therapy

Authors:

Abstract

Cancer is the malignant and abnormal growth of cells, being the second reason leading to killing more than 9 million people annually across the world. This number stands for one in six deaths worldwide because of cancer approximately. The most common therapies for this disease are chemotherapy, radiotherapy surgery, and hormone therapy. Despite the side effects of radiation therapy, this strategy is essential for treating half of the patients with this disease. Although chemotherapy’s strong anticancer effects are due to cytotoxic drugs’ nonselective action and the targeting of cancerous and healthy cells, much toxicity is unfortunately distributed within the patient’s body treatment. Therefore, with this type of treatment and after, it is evident that getting rid of the long-term devastative effects of heart toxicity, neurological disorders, infertility, nephropathy, and chronic liver damage will be inevitable...
CHAPTER 13
Bionanomaterials for cancer therapy
Monireh Ganjali
1
, Mansoureh Ganjali
2
, Mohammad Mahdi Adib Sereshki
3
,
Navid Ahmadinasab
4
, Arash Ghalandarzadeh
5
, Alaa A.A. Aljabali
6
,
Ahmed Barhoum
7,8
1
Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials
and Energy Research Center (MERC), Tehran, Iran;
2
Nour Zoha Materials Engineering Research
Group (NMERG), Tehran, Iran;
3
Department of Hematology and Oncology, Iran University of
Medical Sciences, Tehran, Iran;
4
Department of Modern Technologies, Mangrove Research Center,
University of Hormozgan, Bandar Abbas, Iran;
5
School of Metallurgy and Materials Engineering,
Iran University of Science and Technology (IUST), Tehran, Iran;
6
Faculty of Pharmacy, Department
of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan;
7
NanoStruc
Research Group, Chemistry Department, Faculty of Science, Helwan University, Ain Helwan, Cairo,
Egypt;
8
School of Chemical Sciences, Fraunhofer Project Centre, Dublin City University, Dublin, Ireland
1. Introduction
Cancer is the malignant and abnormal growth of cells, being the second reason leading to
killing more than 9 million people annually across the world. This number stands for one
in six deaths worldwide because of cancer approximately. The most common therapies for
this disease are chemotherapy, radiotherapy surgery, and hormone therapy [1]. Despite the
side effects of radiation therapy, this strategy is essential for treating half of the patients
with this disease [2,3]. Although chemotherapy’s strong anticancer effects are due to
cytotoxic drugs’ nonselective action and the targeting of cancerous and healthy cells,
much toxicity is unfortunately distributed within the patient’s body treatment. Therefore,
with this type of treatment and after, it is evident that getting rid of the long-term
devastative effects of heart toxicity, neurological disorders, infertility, nephropathy, and
chronic liver damage will be inevitable [4e8].
A way to improve these devastative effects of cancer drugs is to include various advanced
drug delivery systems in encapsulation and nanoparticle loading application. It can be
performed by releasing and controlling the drug with minimal or even no toxicity to the
tumor site. Research has focused on killing cancer cells by targeting them and maintaining
healthy cells. New carriers for achieving this aim have been designed for anticancer drugs
that bring the drug to its intended location by more accurately identifying the target [9].
Bionanotechnology: Emerging Applications of Bionanomaterials. https://doi.org/10.1016/B978-0-12-823915-5.00015-0
Copyright ©2022 Elsevier Inc. All rights reserved. 443
Among these, nanoparticles have received more attention because they have a more
straightforward production method and can be prepared through biocompatible polymers.
Because of the more incredible permeability of the vessels around the growth tissue than
the ordinary tissue vessels and because of the quick development of the requirement for
more oxygen and supplements, malignancy cells will want to ingest better medications
because of the improved penetrability and maintenance (EPR) wonder [10,11].
Nanoparticles discussed in this chapter can be well used in drug delivery. In previous
years, drug-carrying nanostructures’ ability, control, and access to authorized drug
molecules have also been made possible. Over the last half-century, advances in related
sciences, such as polymer and chemistry, biology, as well as mechanics, and physics, have
all been able to influence the diversity of nanocarriers and the various categories of
carriers, and they could introduce to medical science with unique characteristics and other
performance [12]. Among the applications of nanostructured materials in drug delivery
systems (DDSs) include drug carriers in diseases, such as cancer, cardiovascular disease,
and Alzheimer’s disease [13].
The primary purpose of designing a helpful DDS using technology is to target the drug to
specific cells or tissues, increasing its therapeutic effects, reducing its therapeutic dose,
and modifying biocompatible nanomaterials for drug delivery [13].
2. Cancer disease: types and statistics
Development of healthy cells into cancer cells is a condition that usually proceeds from a
precancerous lesion into some malignant tumor cells. Cancer is the world’s second-highest
source of death, reported at 9.6 million fatalities in 2018. Approximately one in six cancer
deaths worldwide are due to cancer. In low- and middle-economy countries, approximately
70% of deaths from cancer arise. The International Cancers Research Organization (IARC)
reports that 1 out of 5 men and 1 out of 6 women in the world grow cancer over their lives
and that 1 out of 8 men and 1 out of 11 women are killed by their illness, as shown in
Fig. 13.1.
Cancerous cells vary in several respects from normal cells because they become
unregulated and aggressive. An essential distinction is the reduced specialization of
cancerous cells than usual. Although regular cells grow into many different types of cells
with distinct roles, cancerous cells should not. As mentioned earlier, it is another reason
why cancer cells start to split without stopping, unlike healthy cells. Cancer cells are often
capable of missed signaling that cells typically avoid separating or initiating a mechanism
programmed cell death or apoptosis, which the body uses to clear unwanted cells. More
than 100 tumor forms occur. Cancer forms are typically named by the tissues or organs in
which cancer occurs. For instance, lung cancer occurs in lung tissue, and brain cancer
444 Chapter 13
begins in brain cells, as depicted in Fig. 13.2. Cancer can be categorized into four main
categories: (1) the most prevalent form of cancer is carcinomas. They consist of epithelial
cells, which cover the body’s [14] inner and outer layers. Several different types of
epithelial cells also come underneath a microscope as columns (2) sarcomas are bones and
soft tissue tumors, comprising muscle, fat, arteries of the blood, lymph vessels, and fibrous
tissue (such as ligaments and tendons [15]). (3) Leukemias are classified as tumors starting
in bone marrow blood-forming tissue. Muscular tumors are not produced in these cancers.
Afterward, the blood and bone marrow got to be overflowed by an endless sum of unusual
white blood cells (leukemia and leukemia impact), padding solid blood cells. The low rate
of healthy blood cells will make it increasingly challenging for the body to obtain oxygen,
regulate inflammation, or combat pathogens [16] (4). Lymphoma would be a lymphocytic
cancerous tumor that begins with (T cells or B cells). These are white blood cells that
combat disease and contribute to the immune response. In lymphoma, the lymph node,
lymphatic vessels, and other bodies are damaged by irregular lymphocytes [17].
3. Anticancer nanocarriers
In general, drug carriers can be classified into two main groups: (1) organic carriers,
including ceramic NPs, metal NPs, and carbon NPs, and (2) inorganic carriers include
Figure 13.1
Estimated percentages of the reported number of cancer cases distributed worldwide as the
International Agency for Research on Cancer (IARC) reports in 2018. source @IARC GLOBOCAN
2018.
Bionanomaterials for cancer therapy 445
liposomes, solid lipid nanoparticles (SLNs), polymeric micelles (polymeric micelles),
dendrimers, polymersomes, hydrogel NPs, and biodegradable polymers [19e22]. The most
common materials in drug delivery in the form of nanoparticles are polymers. Polymer
nanoparticles (PNPs) are defined as colloidal particles with sizes between 10 and 1000 nm
[23]. Moreover, they are composed of natural (chitosan, gelatin, sodium alginate, and
albumin) [24e27] or artificial biodegradable polymers, such as polylactic acid (PLA),
polyglycolides (PGAs), poly(lactide co-glycolides) (PLGAs), polyanhydrides,
Figure 13.2
Common cancer types, such as lung, breast, colorectal, esophagus, and kidney cancers due to
distinct factors [18].
446 Chapter 13
polyorthoesters, polycyanoacrylates, polycaprolactone, polyglutamic acid, polymalic acid,
poly(N-vinyl pyrrolidone), poly(methyl methacrylate), poly(vinyl alcohol), poly(acrylic
acid), polyacrylamide, poly(ethylene glycol) (PEG), and poly(methacrylic acid) [28e39],
which is biocompatible and nontoxic; more examples are depicted in Fig. 13.3.
Biocompatible and nontoxic are common characteristics of natural polymer nanoparticles;
however, their low solubility has limited application, when offered in various biological
membranes [40]. Such delivery exposes the nanoparticles to various pHs. This change in
pH and some other issues sometimes restrict their use. Features such as biocompatibility
are required for potential applications in tissue engineering, drug and gene delivery, and
new vaccination strategies.
Figure 13.3
Nanoparticle drug delivery system to selectively target the desired cells. Therapeutic agents are
loaded into appropriately selected polymer nanoparticles that may enter the target cells through
the process of endocytosis and release therapeutic agents inside the endolysosomal effect of the
cell cytoplasm. Alternatively, the cationic nanocarriers can fuse with the negatively charged
cellular plasma member and release the therapeutics into the desired cells. One of the
disadvantages of using nontargeted therapeutics as free drugs is the membrane-active drug efflux
pumps that expel the therapeutic from the cytoplasm to the outside of the cell.
Bionanomaterials for cancer therapy 447
3.1 Polymeric nanocomposite drug delivery
PNCs are a combination of different polymers with nanoparticles established themselves
as a suitable type of nanocomposite (NC) biomaterials for regenerative medicine as well
as drug delivery applications and are widely investigated as multifunctional nanomaterials.
These PNCs can be classified into four types with different nanofiller, which are discussed
in this section.
3.1.1 Polymerepolymer nanocomposites
Most researchers are currently focusing on polymer-based bioNCs. Their NCs in various
forms, such as micelles, hydrogels, polymers, and liposomes, play an essential role in
treating cancer with unparalleled design capacity, environmentally friendly nature, and
comfortable and cost-effective production. PNCs are composed of polymers and
nanomaterials in various types, shapes, and sizes. The interaction of the polymer matrix
and nanoparticles is significant in determining the hybrid structure’s compositional
properties. They can provide higher payloads, extended circulation time of drugs, drug
targeting, and solubility [18].
PNPs comprise a large group of compounds, including vesicular systems (nanocapsules)
and matrix systems (nanospheres) (see Fig. 13.4). The drug is trapped inside a polymer
cavity in nanocapsules; however, the drug is dispersed in a polymer matrix [41].
Figure 13.4
Various examples of nanomaterial constructed from various metallic and nonmetallic
nanomaterials, mesoporous silica nanoparticles (MSNs), iron oxide nanomaterials, carbon
nanotubes, dendrimer, micelle, quantum dots as coreeshell nanomaterials and their applications
as nanocarriers with surface functionalities for the selective targeting to the target cell, which will
lead to the therapeutic release after specific stimulants within the target cell [42].
448 Chapter 13
Abraxane was the first polymer nanoparticle introduced to the world pharmaceutical
market in 2005 by American Pharmaceutical Partners and American Bioscience company,
which comprised of nanoparticles of the drug paclitaxel, bound to albumin [43,44]
(Fig. 13.5). This formulation is fabricated to release a compound called chromophore-EL,
present in previous paclitaxel formulations by increasing solubility. Chromophore causes
severe allergies in certain patients that can threaten their life. With this great success of
Abraxane, it has been proven that nanotechnology can introduce products to overcome the
challenges of formulation science.
The hydrophilic layer of polymeric micelles (PMs) with coreeshell structure has
coreeshell PMs due to a hydrophilic shell making them a good candidate as a nanocarrier
for loading hydrophobic drugs into the core. Blanco et al. [45] reported the effect of
encapsulation of b-lapachone in PEGePLA NPs on A549 cell lysates and orthotopic
Figure 13.5
The usual surface modifications of polymeric nanoparticles, PEGylation, carboxylate, amine,
antibodies, aptamer, peptides, glycans, and surface charge. Generated nanomaterials could be
classified as natural nanomaterials, such as proteins, liposomes, dendrimers, and liposomes, and
synthetic nanomaterials, including coreeshell nanomaterials, hydrogels, carbon nanotypes, and
metallic nanoparticles. The obtained nanomaterials vary in their shape from spherical, cuboidal,
start, plate, triangular, and rod-shaped with various desired sizes [51].
Bionanomaterials for cancer therapy 449
Lewis lung carcinoma (LLC). The results indicated a drastically reduced tumor growth and
metastatic tumor burden when compared with free b-lapachone [45]. In another study,
MPEGePLA and Pluronic were employed to deliver docetaxel [46]. The investigation
showed that the volume of tumor size significantly reduced more when treated with mixed
micelles. Jun et al. demonstrated that CP570 micelles could be used as a carrier to deliver
docetaxel [47]. Their study showed that CP570 micelles did not significantly affect the
improvement of the pharmacokinetics of docetaxel. Furthermore, Park et al. compared
conjugated and encapsulated PLGAePEG MPs containing doxorubicin (DOX) [48]. The
drug release of the conjugated DOXePLGAePEG MPs was more significant than
encapsulating PLGePEG MPs due to gradually hydrolyzing conjugated DOXePLGA and
controlled release water solution DOXePLGA in the incubation medium. The stable
release of DOX from DOXePLGAePEG micelles was similarly observed in compounds
containing microspheres and prepared PLGA nanoparticles with various hydrophilic drugs
to PLGA [49,50]. PNPs surface has been modified with different methods, such as charge
modification, bioactive peptides graft, amphipathic compound graft, and siRNA to obtain
the appropriate functions to fit the tumor environment or properties of drugs as shown in
Fig. 13.5.
3.1.2 Polymericemagnetic nanocomposites
Magnetic NPs are widely used in magnetic resonance imaging (MRI), biosensors,
theranostics, delivery, magnetic hyperthermia, photodynamic therapy, and photothermal
ablation therapy [52e58]. However, magnetic NPs, due to aggregation and low stability in
biological environments, are often coated with or embedded in a polymer [59,60]. This
surface coating has a sufficient role in improving some MNPs’ properties such as
pharmacokinetics, systemic toxicity and clearance rate, nonspecific protein adsorption or
cell interactions, and sustained drug release. Magnetic NPecoated polymers are proposed
as new materials for releasing the chemotherapeutic cancer drugs in the desired tissue or
organ. Many researchers have extensively studied polymericemagnetic NCs for innovative
triggered release in controlled drug delivery. An overview of polymericemagnetic NCs for
drug delivery applications is listed in Table 13.1 [61].
In addition to chemotherapy drugs, implantable magnetic (Fe
3
O
4
)-reinforced
polydimethylsiloxane NCs for the localized breast cancer treatment via hyperthermia were
explored by Kan-Dapaah et al. [76]. It has been identified in the study that the thermal
dose coverage influences NC shape and size. Recently, an alternating current (AC)
magnetic field has developed a novel fabrication technique for synthesizing spherical
microactuators of PLGA/Mg-gFe
2
O
3
NCs [77]. The release rate results showed an
increased diffusion rate in the excited PLGA/Mg-gFe
2
O
3
NCs by the AC magnetic field.
450 Chapter 13
The schematic structures of eight biological source-based MNPs as drugs has been
approved by the US Food and Drug Administration (FDA), European Medicines Agency
(EMEA), Japanese Ministry of Health, and Australia’s Therapeutic Goods Administration
for different clinical applications and derivatives as shown in Fig. 13.6 [78e81].
Moreover, some other magnetic NPebased drugs including Feraheme, Endorem,
Gastromark, Lumiren, Ferumoxytol, Combidex, Radiogardase, Feridex, and NanoTherm
have been recently approved by the FDA and European Medicines Agency (EMA) for
various applications in iron deficiency, iron replacement therapy, the lymph node
metastases imaging, MRI, pharmaceutical contrast agents or oral antidotes for heavy metal
contamination in humans, and the treatment of intermittent glioblastoma multiforme
[82e84].
3.1.3 Polymeremetallic nanocomposites
Metallic NPs are nanosized metals with dimensions (length, width, or thickness) within the
size range of 1e100 nm and can be synthesized in different forms through various
Table 13.1: Distinct polymericemagnetic nanaocomposites for specific drug delivery
applications.
Magnetic nanoparticles Polymer Drug loaded/application References
Carbonyl iron Poly(butylcyanoacrylate) 5-Fluorouracil
Ftorafur
[62]
Fe
3
O
4
Starch-g-
poly(methylmethacrylate-co-
PEG-acrylamide)
Doxorubicin [63]
Fe
3
O
4
Poly(lactic acid)/PEG Doxorubicin
hydrochloride
[64]
Fe
3
O
4
APSePEGeTFEE Doxorubicin [65]
Fe
3
O
4
PEG-b-poly(4-
vinylbenzylphosphonate)
Doxorubicin [66]
Fe
3
O
4
@LaF3:Ce3
þ
, Tb3
þ
nanoparticles
Chitosan Paclitaxel [67]
Fe
3
O
4
Poly[(N-isopropylacrylamide-r-
acrylamide)-b-L-lactic acid]
Paclitaxel [68]
Fe
3
O
4
Cisplatin Heparin [69]
Fe
3
O
4
Chitosan Gemcitabine [70]
Fe
3
O
4
PEGylated PLGA Sorafenib [71]
Fe
3
O
4
Chitosan Bortezomib [72]
Fe
3
O
4
Chitosan Gemcitabine [73]
Mesoporous silica PEG-co-poly(vinyl pyridine) Doxorubicin [74]
Fe
3
O
4
Chitosanepolymethacrylic
acid
Doxorubicin [75]
Bionanomaterials for cancer therapy 451
methods [85e88]. Metallic NPs, such as silver (Ag), gold (Au), titanium (Ti), platinum
(Pt), palladium (Pd), and copper (Cu), are generally used as drug carriers and contrast
agents in cancer treatment [89e91]. The applications of these substances due to their
antiproliferative and apoptotic induction properties for use in cancer therapy have been
studied by many researchers [92e95].
Tumor blood vessels are often large and have an insufficient lymphatic system [96,97];
therefore, metallic-based NPs can easily penetrate the cancerous cells and kill them. The
cytotoxic effects of silver nanoparticles (AgNPs) on the MCF-7 breast cancer cell line
were studied in a previous work. The MTT assay results demonstrated a significant
reduction of cancer cell proliferation after 24 h [98]. In another work, the effect of silver
Figure 13.6
Schematic diagram depicting various inorganic nanodrug delivery platforms (NDDPs) loaded with
anticancer chemotherapeutic agents employed for cancer treatment. iNPs, inorganic
nanoparticles; MWCNTs, multiwalled carbon nanotubes; SWCNTs, single-walled carbon
nanotubes [42].
452 Chapter 13
(AgNPs) and gold nanoparticles (AuNPs) as an antitumor treatment in vitro against human
breast cancer cells (MCF-7) was investigated [99]. The mechanism of cytotoxic effects of
silver and gold nanoparticles (AgNPs, AuNPs) on cancer cells is proposed to be due to the
increasing disruption of reactive oxygen species (ROS) released by the nanoparticle
toward mitochondria. This disruption leads to reduced cellular energy production (ATP),
which eventually causes DNA damage and cell death [100,101]. However, it has been used
to combine AgNPs with an anticancer drug to get over chemoresistance and undesired side
effects of chemotherapeutic drugs. Gopinath et al. investigated the combination of AgNPs
and 5-fluorouracil (5-FU) effects on the baby hamster kidney (BHK21) and human colon
adenocarcinoma (HT29) cell lines [102]. The study revealed that AgNPs can act as a
therapeutic drug and represent a new chemosensitization strategy for future gene therapy
applications. Furthermore, the results indicated that AgNPs apoptotic cells’ mechanism
associated with mitochondrial membranes’ damage is same as the mechanism induced by
other drugs or gene therapy treatments.
DOX is an anthracycline type of chemotherapy with broad-spectrum activity and is used to
treat various cancer types. Hekmat et al. demonstrated that the combination of AgNPs and
DOX could alter DNA structure [103]. Moreover, the study identified that the AgNPs and
DOX improved the cytotoxic effect of DOX on MCF7 cells and T47D cells compared
with stand-alone DOX or AgNPs.
Platinum complexes are another typical metallic NPs that are introduced as cancer
treatment agents in 1968, due to various oxidation states, chelating properties, redox
potentials, uneven ground, and excited electronic states [104]. Thus, several approved
platinum-based drugs by the FDA, Japan, China, and Korea, are used in different cancer
treatments as listed in Table 13.2.
Nevertheless, the metallic NPs are unstable in the blood and have low cellular uptake in
cancer, limiting their clinical application. Besides, severe chronic kidney disease,
neurological disorder, and ototoxicity are the main effects of these materials.
Consequently, to overcome these problems, the surface of metallic NPs has been modified
with biocompatible polymers and encapsulated of drugs in nanosized drug carries to avoid
aggregation of nanosized metal and preserve their properties and increase the duration of
drug action without the loss of their volume and activity. On the other hand, inhibition of
tumor cells without harmful effects on healthy tissue should be considered [115,116].
The cytotoxicity effect of the conjugated DOX onto coreeshell Ag/PNCs was investigated
[117]. The coreeshell Ag/PNCs consist of AgNPs as a core, and three FDA-approved
polymers (PVA, PEG, and PVP) were as a shell. The MTT results showed that DOX/Ag
NCs are more cytotoxic to MCF-7 cells than normal 1BR hTERT cells. The results also
showed that DOXeAg/polymeric (PVA, PEG, and PVP) NCs showed IC50 at 10-fold
reduced doses than free DOX and DOXeAgNCs. Based on the experimental results, it can
Bionanomaterials for cancer therapy 453
be concluded that the efficiency of DOXeAg/polymeric (PVA, PEG, and PVP) NCs is the
same as DOX alone, but with less dose of DOX about 95%. In another study,
Venkatpurwar et al. synthesized novel carriers for anticancer drug delivery with capped
gold nanoparticles [118]. The result showed a significant enhancement in cytotoxicity of
DOXeAuNCs on human glioma cell lines (LN-229) compared with free DOX that can be
attributed to the increase of cellular internalization via AuNP-mediated endocytosis. The
role of AuNP-mediated endocytosis in the growth inhibition of cancer cells through the
release of the conjunction of methotrexateegold nanoparticles (MTXeAuNPs) inside
cancer cells was also reported by Chen et al. [119].
3.1.4 Polymer ceramic nanocomposites
Besides, hyperthermia (HT) is a novel strategy through the local application of heat to
inhibit cancer cells. HT is categorized into three groups, such as whole body, regional, and
localized. The body temperature in whole-body hyperthermia (WBH) is raised to 39.5 and
40.5"C (103.1 and 104.9"F) [120,121]. Indeed, this temperature is defined between 41.8
and 42"C (107.2e107.6"F) in Europe and the United States, where it is approximately
Table 13.2: Applications of platinum-based drugs.
Drugs Applications References
Cisplatin Testicular cancer, ovarian
cancer, cervical cancer, breast
cancer, bladder cancer, head
and neck cancer, esophageal
cancer, lung cancer,
mesothelioma, brain tumors,
and neuroblastoma
[105,108e111]
Carboplatin Ovarian cancer, lung, head and
neck, endometrial, esophageal,
bladder, breast, and cervical;
central nervous system or germ
cell tumors; osteogenic
sarcoma; and as preparation
for a stem cell or bone marrow
transplant
[105,108e110]
Oxaliplatin Advanced cancer of the colon
and rectum
[105,106,109]
Nedaplatin Advanced or recurrent
esophageal cancer
[107,108]
Lobaplatin Breast cancer, esophageal
squamous cell carcinoma,
gastric carcinoma, lung cancer,
melanoma, and ovarian cancer
[107,113,114]
Heptaplatin Advanced gastric cancer [108,112]
454 Chapter 13
43e44"C (109e111"F) in Japan and Russia. This method raises the patient’s body
temperature by immersing in a hot water bath, heating blankets, or ultraviolet radiation.
Nevertheless, the body temperature in the case of local/regional hyperthermia remains
unchanged. All of these methods have certain side effects, such as pain at the site,
infection, bleeding, blood clots, swelling, burns, blistering, and damage to the skin,
muscles, and nerves near the treated area, nausea, vomiting, diarrhea, and problems with
heart, blood vessels, and other major organs [122].
Hyperthermia based on the combination of polymeric matrix embedding magnetic
bioceramic fillers is proposed to solve the challenges in conventional HT. By the
hyperthermia method, ferromagnetic glasseceramics (FGCs) have shown promising
outcomes for the treatment of deep tumors, such as osteosarcoma cells [123]. Kokubo
et al. synthesized the FGCs granule and showed that when it was located around the
tumors, strongly bonding it and bone, a bone-like apatite complex around the tumors is
formed [124]. Hence, tumor cells were inhibited due to efficient heat after exposure
toward an alternating magnetic field.
As a typical carrier material, ceramic nanoparticles have been extensively exploited for
imaging-guided therapy, including silica [125], NIR spectra of 5% Er
3þ
-doped
glasseceramics [126], and lanthanide-doped upconversion nanoparticles (UCNPs) [8e12].
Previous reports indicated that biomacromolecules were closely related to the bioinspired
mineralization process. For instance, the mineralization of CaCO
3
[127] can be induced
and regulated by regenerated silk fibroin (SF)-based nanostructures through an oriented
crystallization process [26,27]. A series of magnetic glasseceramics containing different
additions of P
2
O
5
or MnO
2
in the system Fe
2
O
3
eCaOeZnOeSiO
2
eB
2
O
3
for localized
treatment of cancer was studied by Abdel-Hameed et al. [128,129]. Increasing P
2
O
5
or
MnO
2
in the composite influences on thermal effect and decreases it. Tumor hypoxia is
caused by the rapidly proliferating cancer cells enclosed in the distorted tumor
vasculatures, which severely hinders the treatment efficacy of photodynamic therapy
(PDT) owing to the insufficient oxygen supply in the tumor microenvironment [28e30].
Significantly, MnO
2
nanoparticles can induce enzymatic decomposition of H
2
O
2
into water
and oxygen and thus may work as a crucial mediator to diminish tumor hypoxia [34,35].
Furthermore, MnO
2
can decompose in response to other tumor-specific environmental
cues, such as glutathione (GSH) or local acidity, and the produced Mn
2þ
is an excellent
contrast agent for T1-magnetic resonance (MR) imaging of tumoral tissues [130].
In a recent study, Yang and coworkers clinically confirmed that photosensitizer
indocyanine green (ICG) and chemotherapeutic drug DOX were coimmobilized into silk
fibroin (SF) nanoparticles, where MnO
2
mineralized the particle surface through a
bioinspired crystallization process (Fig. 13.7)[130].
Bionanomaterials for cancer therapy 455
Among the different ceramic nanoparticles developed so far, mesoporous SiO
2
has
attracted the scientific community’s attention for being applied as a drug delivery system
capable of controlling the drug release in space and time. Modifications of SiO
2
NPs with
different types of polymers were performed so far to imprint them a stimulus-responsive
behavior (namely, pH, redox potential, adenosine triphosphate, enzyme, or temperature) to
allow their application in cancer therapy, which is schematically represented in Fig. 13.8.
Several efforts have been executed via MSNs to target specific tissues through passive or
active targeting procedures [131].
Figure 13.7
Schematic of the synthetic procedure of SF@MnO
2
/ICG/DOX (SMID) nanoparticles as a
multifunctional drug delivery platform for in vivo MR/fluorescence imaging-assisted trimodal
therapy of cancer [130].
456 Chapter 13
In the beginning, MSNs were developed as an anticancer drug delivery system, mainly
based on their efficacy to store a high amount of chemotherapeutics into pores and exploit
the enhanced permeability and retention (EPR) effect for passive targeting to tumor
tissues. This part of the chapter will discuss the EPR effect and passive targeted cancer
therapy using MSNs. Later, MSN surface modifications by conjugating targeting ligands
were introduced to enhance MSN uptake in targeted cells. Different targeting moieties
have been employed to the surface of MSNs, e.g., small molecules, aptamers, short
peptides, antibodies, and antibody fragments [131].
Nasab et al. fabricated chitosan nanoparticles that are coated with curcumin [132]. The
results showed that mesoporous silica nanocarriers, refined by chitosan as a pH-responsive
polymer, improved curcumin’s solubility in the aqueous media and could be acted as a
nanocarrier against the U87MG glioblastoma cancer cell line. In another study, a
coreeshell system containing chitosanepoly(methacrylic acid) (CSePMAA) shells and
MSN cores was studied [133]. According to this study, the drug-releasing and release
behavior of loaded DOX hydrochlorideeMSN/CSPMAA composite showed high drug-
loading capacity encapsulation efficiency 22.3 #0.3% and 95.7 #2.0%, respectively.
Osteosarcoma (OS) or osteogenic sarcoma (OGS) (or only bone cancer) is a type of
cancerous tumor in a bone that is usually found in the long bones around the knee.
This type of cancer is found in children, adolescents, and young adult men than women [135].
Figure 13.8
Schematic representation shows different stimuli-responsive behaviors of mesoporous silica nano-
particles for cancer therapy [134].
Bionanomaterials for cancer therapy 457
This type of cancer is found in children, adolescents, and young adult men than women
[135]. Among the most common treatments for osteosarcoma are surgery, chemotherapy,
and radiation therapy.
Moreover, patients with higher-grade tumors are treated with a new method known as
neoadjuvant therapy (chemotherapy given before surgery) in recent times [136]. Apart
from the potential clinical benefits of chemotherapy drugs, due to localization of OS in a
wide area and cutting off from the blood vascular system resulting from crushing healthy
vessels [137], the defective drug reaches the tumor.
Additionally, since OS is spread to other tissues, a sensitive platform to recognize healthy
and tumor cells is required. Therefore, polymericeceramic NCs have received much
attention as a potential basis for one such advanced platform. The novel
hydroxyapatiteepolyvinyl alcoholedoxorubicin anticancer drug (HApePVAeDOX) NC
was developed for obstructive sleep apnea (OSA) treatment [138]. It was shown that the
DOXeHApePVA NC is highly toxic toward osteosarcoma cells (MG 63) with better
biological properties. Son et al. developed drug-loaded calcium phosphate (CaP) NC
system containing an anticancer drug, such as caffeic acid (CA-NP), chlorogenic acid
(CG-NP), or cisplatin (CP-NP) in the presence of alginate as a polymer template to control
the release rate of drugs for overall survival (OS) treatment [139]. The anticancer activity
of cisplatin-loaded CP-NP NC was higher than another NC on OS and dependent on drugs
and time concentration overall survival.
Furthermore, drug-loaded NCs were released at lower pH. Mondal et al. [140] utilized an
easy synthetic method with a two-step hydrothermal and wet chemical deposition
technique for the synthesis of iron oxide nanoparticles (Fe
3
O
4
), hydroxyapatite (HAp), and
coated hydroxyapatite (Fe
3
O
4
eHAp) for the treatment of cancer via magnetic
hyperthermia approach. First, a controlled hydrothermal synthesis procedure was
employed to synthesize Fe
3
O
4
nanoparticles followed by coating with HAp. The
cytotoxicity study of synthesized iron oxide, HAp, and Fe
3
O
4
eHAp nanoparticles was
performed with MG-63 osteosarcoma cell lines. The Fe
3
O
4
eHAp nanoparticles were
further studied for in vitro hyperthermia assessment. The promising results encourage the
future application of Fe
3
O
4
eHAp nanomaterials as a nanoheater for magnetic
hyperthermiaemediated cancer therapy (Fig. 13.9). Along this line, DOX-loaded
degradable MSNs with hyaluronic acid (HA) decoration (DOX-loaded HA-PMSNs) for
cancer treatment were synthesized by Palanikumar et al. [141]. The release profile of
DOX@MSN/CS-PMAA depicted that the DOX release rate was more dependent on pH in
DOX@MSN/CS-PMAA than DOX@MSN, whereas a faster release behavior was
observed with decreasing pH from 7.4 to 6.8 and 5.5 in DOX@MSN/CS-PMAA system
(Fig. 13.9). This issue is attributed to the effect of electrostatic interaction between DOX
and polymer nanoshells.
458 Chapter 13
Nanoselenium-poly-L-lactic acid (nSe-PLLA) NCs have been prepared to reduce bone
cancer cell functions by Stolzoff and Webster [142]. The SeNPePLLA samples had a
greater alkaline phosphatase (ALP) activity than healthy osteoblast tissue. Antioxidant-
loaded phosphorylated chitosan nanohydroxyapatite (HAp/PCS) and phosphorylated
chitosanenanohydroxyapatiteegingerol (HAP/PCS/G) composite scaffold for bone tissue
engineering was developed by Sumathra et al. to accelerate cell proliferation [143]. The
investigation showed that HAP/PCS/G composite scaffold had good properties as large
pore size, biocompatibility, and cytotoxicity and could be used as an osteoconductive
scaffold. In another study, Wu et al. prepared JQ1 (a bromodomain inhibitor)-loaded
n-HAp with and without Medronate (BP) as an amino acid protein [144]. According to
this study, the OS cell was selected by loaded n-HA with BP. The positive effects of this
synergy are evidenced by the consistently favorable effect achieved by HAp/BP/JQ1, not
Figure 13.9
Magnetic hydroxyapatite (IO-HAp)emediated hyperthermia study to treat cancer [140].
Bionanomaterials for cancer therapy 459
only reduced OS cell migration and viability of differentiated OS cells more than any
other HAp composition, but also selectively necrotized OS cells, similar to HAp/JQ1 and
HAp/BP.
Silica nanostructures (n-SiO
2
) as nanocarriers, due to advantages, such as biocompatible,
highly porous, easy-to-control fabrication, cost-effective, and easy to modify for
functionalization nature, have been widely used in drug delivery [145e148]. MSNs were
first synthesized by Kresge et al. [149]. The study reported that the fabrication of
interaction of silanol group of mesoporous n-SiO
2
(MSNs) with cellular membranes could
cause toxicity [149]. Therefore, surface modification of the MSNs with a polymer shell is
highly required to utilize them for potential anticancer as well as other biomedical
applications [150].
4. Conclusion
This chapter explains NCs with different nanofillers and their great potential as a single
platform with multifunctional capabilities for slow and controlled drug delivery emerging
as an attractive approach as compared with the traditional treatment containing
chemotherapy drugs, radiation, and surgical excision; NCs functionalized with a targeting
agent present a novel therapeutic approach. The entrapment procedure of chemotherapy
drugs into the NCs has resulted in more significant drug loading and slower drug release
properties. However, despite many remarkable studies, some critical questions need to be
answered toward clinical applications. Therefore, more data are required about the
appropriate shape and size nanofiller currently doped into the polymers. Subsequently,
more attempts should be made to improve nanofillers’ synthesis and properties for
successfully transporting chemo drugs to purposed targets. Besides, extra in vitro and
in vivo experiment for enhancing drug delivery from these nanocarriers is demanded.
Furthermore, several types of PNCs cannot be scaled up due to the high-cost process. In
this regard, nanocarriers based on PNCs should be fabricated at low cost and fast. The
next major challenge encountered in PNC for drug delivery is their biocompatibility and
acceptability since synthetic materials with body cells in vivo experiments are different
from in vitro tests and in biological media. Besides, fundamental challenges in PNC
systems are stability, solubility, permeability, and sustained release.
References
[1] V. Schirrmacher, From chemotherapy to biological therapy: a review of novel concepts to reduce the
side effects of systemic cancer treatment, Int. J. Oncol. 54 (2019) 407e419, https://doi.org/10.3892/
ijo.2018.4661.
[2] R. Baskar, K.A. Lee, R. Yeo, K.W. Yeoh, Cancer and radiation therapy: current advances and future
directions, Int. J. Med. Sci. 9 (2012) 193e199, https://doi.org/10.7150/ijms.3635.
460 Chapter 13
[3] S. Siva, M.P. MacManus, R.F. Martin, O.A. Martin, Abscopal effects of radiation therapy: a clinical
review for the radiobiologist, Cancer Lett. 356 (2015) 82e90, https://doi.org/10.1016/
j.canlet.2013.09.018.
[4] C.S. Lee, E.J. Ryan, G.A. Doherty, Gastro-intestinal toxicity of chemotherapeutics in colorectal cancer:
the role of inflammation, World J. Gastroenterol. 20 (2014) 3751e3761, https://doi.org/10.3748/
wjg.v20.i14.3751.
[5] R.M. Tru
¨eb, Chemotherapy-induced alopecia, Curr. Opin. Support. Palliat. Care 4 (2010) 281e284,
https://doi.org/10.1097/SPC.0b013e3283409280.
[6] D. Liu, A. Ahmet, L. Ward, P. Krishnamoorthy, E.D. Mandelcorn, R. Leigh, J.P. Brown, A. Cohen,
H. Kim, A practical guide to the monitoring and management of the complications of systemic
corticosteroid therapy, Allergy Asthma Clin. Immunol. 9 (2013), https://doi.org/10.1186/1710-1492-9-30.
[7] S.H. Chu, Y.J. Lee, E.S. Lee, Y. Geng, X.S. Wang, C.S. Cleeland, Current use of drugs affecting the
central nervous system for chemotherapy-induced peripheral neuropathy in cancer patients: a systematic
review, Support. Care Cancer 23 (2015) 513e524, https://doi.org/10.1007/s00520-014-2408-8.
[8] M. Ewertz, C. Qvortrup, L. Eckhoff, Chemotherapy-induced Peripheral Neuropathy in Patients Treated
with Taxanes and Platinum Derivatives, 2015.
[9] J.P. Gillet, M.M. Gottesman, Mechanisms of multidrug resistance in cancer, Methods Mol. Biol. 596
(2010) 47e76, https://doi.org/10.1007/978-1-60761-416-6_4.
[10] D. Poncelet, Microencapsulation: fundamentals, methods and applications, in: Surface Chemistry in
Biomedical and Environmental Science, 2006, pp. 23e34.
[11] B. Haley, E. Frenkel, Nanoparticles for Drug Delivery in Cancer Treatment, 2008.
[12] J. Shi, A.R. Votruba, O.C. Farokhzad, R. Langer, Nanotechnology in drug delivery and tissue
engineering: from discovery to applications, Nano Lett. 10 (2010) 3223e3230, https://doi.org/10.1021/
nl102184c.
[13] M.M. Yallapu, M. Jaggi, S.C. Chauhan, Scope of Nanotechnology in Ovarian Cancer Therapeutics, 2010.
[14] B.A. Nacev, K.B. Jones, A.M. Intlekofer, S.E. Jamie, C.D. Allis, W.D. Tap, M. Ladanyi, T.O. Nielsen,
The epigenomics of sarcoma, Nat. Rev. Cancer 20 (2020) 608e623.
[15] H. Lu, R.P. Graham, R. Seethala, D. Chute, Intraductal carcinoma of salivary glands harboring TRIM27-
RET fusion with mixed low grade and apocrine types, Head Neck Pathol. 14 (2020) 239e245, https://
doi.org/10.1007/s12105-018-0996-1.
[16] M. Hallek, Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment,
Am. J. Hematol. 94 (2019) 1266e1287.
[17] K. Tsukasaki, A. Marc¸ais, R. Nasr, K. Kato, T. Fukuda, O. Hermine, A. Bazarbachi, Diagnostic
approaches and established treatments for adult t cell leukemia lymphoma, Front. Microbiol. 11 (2020)
1207.
[18] Study examines whether blood test can identify early cancers. National Cancer Institute. https://www.
cancer.gov/news-events/cancer-currents-blog/2020/cancerseek-blood-test-detect-early-cancer.
[19] Y. Li, K. Xiao, K.S. Lam, Reversible Cross-Linked Polymeric Nanoplatform in Drug Delivery, 2014.
[20] A.K. Mishra, Nanomedicine for Drug Delivery and Therapeutics, 2013.
[21] R.M. Donlan, P. Wu, D.W. Grainger, B.D. Ratner, An Introduction to Materials in Medicine, 2004.
[22] D. Chenthamara, S. Subramaniam, S.G. Ramakrishnan, S. Krishnaswamy, M.M. Essa, F.H. Lin,
M.W. Qoronfleh, Therapeutic efficacy of nanoparticles and routes of administration, Biomater. Res. 23
(2019), https://doi.org/10.1186/s40824-019-0166-x.
[23] M. Sharma, Chapter 18. Transdermal and intravenous nano drug delivery systems: present and future, in:
Applications of Targeted Nano Drugs and Delivery Systems, 2019, pp. 499e550.
[24] C.A. Farrugia, M.J. Groves, Gelatin behaviour in dilute aqueous solution: designing a nanoparticulate
formulation, J. Pharm. Pharmacol. 51 (1999) 643e649, https://doi.org/10.1211/0022357991772925.
[25] T. Irimia, C.E. Dinu-Pı
ˆrvu, M.V. Ghica, D. Lupuleasa, D.L. Muntean, D.I. Udeanu, L. Popa, Chitosan-
based in situ gels for ocular delivery of therapeutics: a state-of-the-art review, Mar. Drugs 16 (2018),
https://doi.org/10.3390/md16100373.
Bionanomaterials for cancer therapy 461
[26] C. Schwinger, S. Koch, U. Jahnz, P. Wittlich, N.G. Rainov, J. Kressler, High throughput encapsulation of
murine fibroblasts in alginate using the JetCutter technology, J. Microencapsul. 19 (2002) 273e280,
https://doi.org/10.1080/02652040110105328.
[27] B. Luppi, F. Bigucci, G. Corace, A. Delucca, T. Cerchiara, M. Sorrenti, L. Catenacci, A.M. Di Pietra,
V. Zecchi, Albumin nanoparticles carrying cyclodextrins for nasal delivery of the anti-Alzheimer drug
tacrine, Eur. J. Pharmaceut. Sci. 44 (2011) 559e565, https://doi.org/10.1016/j.ejps.2011.10.002.
[28] N. Shokri, H. Akbari Javar, F. Sh, A. Khalaj, R. Khoshayand, Dinarvand, F. Atyabi, A. Nomani,
E. Azizi, Preparation and evaluation of poly (caprolactone fumarate) nanoparticles containing
doxorubicin HC1, DARU J. Pharm. Sci. 19 (2011) 12e22.
[29] P.K. Ghosh, Hydrophilic polymeric nanoparticles as drug carriers, Indian J. Biochem. Biophys. 37 (2000)
273e282.
[30] B. Kateb, K. Chiu, K.L. Black, V. Yamamoto, B. Khalsa, J.Y. Ljubimova, H. Ding, R. Patil,
J.A. Portilla-Arias, M. Modo, D.F. Moore, K. Farahani, M.S. Okun, N. Prakash, J. Neman, D. Ahdoot,
W. Grundfest, S. Nikzad, J.D. Heiss, Nanoplatforms for constructing new approaches to cancer
treatment, imaging, and drug delivery: what should be the policy? Neuroimage 54 (2011) https://doi.org/
10.1016/j.neuroimage.2010.01.105.
[31] H.M. Mansour, M.J. Sohn, A. Al-Ghananeem, P.P. DeLuca, Materials for pharmaceutical dosage forms:
molecular pharmaceutics and controlled release drug delivery aspects, Int. J. Mol. Sci. 11 (2010)
3298e3322, https://doi.org/10.3390/ijms11093298.
[32] S. Sundar, J. Kundu, S.C. Kundu, Biopolymeric nanoparticles, Sci. Technol. Adv. Mater. 11 (2010)
014104, https://doi.org/10.1088/1468-6996/11/1/014104.
[33] H. Tian, Z. Tang, X. Zhuang, X. Chen, X. Jing, Biodegradable synthetic polymers: preparation,
functionalization and biomedical application, Prog. Polym. Sci. 37 (2012) 237e280, https://doi.org/
10.1016/j.progpolymsci.2011.06.004.
[34] B. Guo, P.X. Ma, Synthetic biodegradable functional polymers for tissue engineering: a brief review, Sci.
China Chem. 57 (2014) 490e500, https://doi.org/10.1007/s11426-014-5086-y.
[35] L.S. Nair, C.T. Laurencin, Biodegradable polymers as biomaterials, Prog. Polym. Sci. 32 (2007)
762e798, https://doi.org/10.1016/j.progpolymsci.2007.05.017.
[36] J.C. Leroux, E. Alle
´mann, F. De Jaeghere, E. Doelker, R. Gurny, Biodegradable nanoparticlesdfrom
sustained release formulations to improved site specific drug delivery, J. Contr. Release (1996) 339e350.
[37] E.W. Neuse, Synthetic polymers as drug-delivery vehicles in medicine, Met. Based Drugs 2008 (2008),
https://doi.org/10.1155/2008/469531.
[38] N. Saito, N. Murakami, J. Takahashi, H. Horiuchi, H. Ota, H. Kato, T. Okada, K. Nozaki, K. Takaoka,
Synthetic biodegradable polymers as drug delivery systems for bone morphogenetic proteins, Adv. Drug
Deliv. Rev. 57 (2005) 1037e1048, https://doi.org/10.1016/j.addr.2004.12.016.
[39] A. Ludwig, The use of mucoadhesive polymers in ocular drug delivery, Adv. Drug Deliv. Rev. 57 (2005)
1595e1639, https://doi.org/10.1016/j.addr.2005.07.005.
[40] R. Solaro, F. Chiellini, A. Battisti, Targeted delivery of protein drugs by nanocarriers, Materials 3 (2010)
1928e1980, https://doi.org/10.3390/ma3031928.
[41] A.M. Ribeiro, F. Veiga, A. Figueiras, Biodegradable polymeric nanostructures: design and advances in
oral drug delivery for neurodegenerative disorders, in: Nanostructures for Oral Medicine, 2017,
pp. 61e86.
[42] S. Hossen, M.K. Hossain, M.K. Basher, M.N.H. Mia, M.T. Rahman, M.J. Uddin, Smart nanocarrier-
based drug delivery systems for cancer therapy and toxicity studies: a review, J. Adv. Res. 15 (2019)
1e18.
[43] N. Desai, Nanoparticle albumin-bound paclitaxel (Abraxane®), in: Albumin in Medicine: Pathological
and Clinical Applications, 2016, pp. 101e119.
462 Chapter 13
[44] R. Palumbo, F. Sottotetti, G. Trifiro
`, E. Piazza, A. Ferzi, A. Gambaro, E.G. Spinapolice, E. Pozzi,
B. Tagliaferri, C. Teragni, A. Bernardo, Nanoparticle albumin-bound paclitaxel (Nab-paclitaxel) as
second-line chemotherapy in HER2-negative, taxane-pretreated metastatic breast cancer patients:
prospective evaluation of activity, safety, and quality of life, Drug Des. Dev. Ther. 9 (2015) 2189e2199,
https://doi.org/10.2147/DDDT.S79563.
[45] E. Blanco, E.A. Bey, C. Khemtong, S.G. Yang, J. Setti-Guthi, H. Chen, C.W. Kessinger, K.A. Carnevale,
W.G. Bornmann, D.A. Boothman, J. Gao, B-lapachone micellar nanotherapeutics for non-small cell lung
cancer therapy, Cancer Res. 70 (2010) 3896e3904, https://doi.org/10.1158/0008-5472.CAN-09-3995.
[46] C.F. Mu, P. Balakrishnan, F.D. Cui, Y.M. Yin, Y.B. Lee, H.G. Choi, C.S. Yong, S.J. Chung, C.K. Shim,
D.D. Kim, The effects of mixed MPEG-PLA/Pluronic®copolymer micelles on the bioavailability and
multidrug resistance of docetaxel, Biomaterials 31 (2010) 2371e2379, https://doi.org/10.1016/
j.biomaterials.2009.11.102.
[47] Y.J. Jun, V.B. Jadhav, J.H. Min, J.X. Cui, S.W. Chae, J.M. Choi, I.S. Kim, S.J. Choi, H.J. Lee,
Y.S. Sohn, Stable and efficient delivery of docetaxel by micelle-encapsulation using a tripodal
cyclotriphosphazene amphiphile, Int. J. Pharm. 422 (2012) 374e380, https://doi.org/10.1016/
j.ijpharm.2011.10.052.
[48] T.G. Park, Degradation of poly(d,l-lactic acid) microspheres: effect of molecular weight, J. Contr.
Release 30 (1994) 161e173, https://doi.org/10.1016/0168-3659(94)90263-1.
[49] H.S. Yoo, J.E. Oh, K.H. Lee, T.G. Park, Biodegradable nanoparticles containing doxorubicin-PLGA
conjugate for sustained release, Pharm. Res. 16 (1999) 1114e1118, https://doi.org/10.1023/
A:1018908421434.
[50] H.S. Yoo, K.H. Lee, J.E. Oh, T.G. Park, In vitro and in vivo anti-tumor activities of nanoparticles based
on doxorubicin-PLGA conjugates, J. Contr. Release 68 (2000) 419e431, https://doi.org/10.1016/S0168-
3659(00)00280-7.
[51] B. Li, Q. Li, J. Mo, H. Dai, Drug-loaded polymeric nanoparticles for cancer stem cell targeting, Front.
Pharmacol. 8 (2017) 51, https://doi.org/10.3389/fphar.2017.00051.
[52] C. Corot, P. Robert, J.M. Ide
´e, M. Port, Recent Advances in Iron Oxide Nanocrystal Technology for
Medical Imaging, 2006.
[53] K. Ulbrich, K. Hola
´,V.!
Subr, A. Bakandritsos, J. Tu!
cek, R. Zbo!
ril, Targeted drug delivery with polymers
and magnetic nanoparticles: covalent and noncovalent approaches, release control, and clinical studies,
Chem. Rev. 116 (2016) 5338e5431, https://doi.org/10.1021/acs.chemrev.5b00589.
[54] O.L. Gobbo, K. Sjaastad, M.W. Radomski, Y. Volkov, A. Prina-Mello, Magnetic nanoparticles in cancer
theranostics, Theranostics 5 (2015) 1249e1263, https://doi.org/10.7150/thno.11544.
[55] Y. Yang, L. Jing, X. Li, L. Lin, X. Yue, Z. Dai, Hyaluronic acid conjugated magnetic prussian
blue@quantum dot nanoparticles for cancer theranostics, Theranostics 7 (2017), https://doi.org/10.7150/
thno.17411.
[56] R.A. Revia, M. Zhang, Magnetite nanoparticles for cancer diagnosis, treatment, and treatment
monitoring: recent advances, Mater. Today 19 (2016) 157e168, https://doi.org/10.1016/
j.mattod.2015.08.022.
[57] A. Singh, S.K. Sahoo, Magnetic nanoparticles: a novel platform for cancer theranostics, Drug Discov.
Today 19 (2014) 474e481, https://doi.org/10.1016/j.drudis.2013.10.005.
[58] A.J. Cole, V.C. Yang, A.E. David, Cancer theranostics: the rise of targeted magnetic nanoparticles,
Trends Biotechnol. 29 (7) (2011) 323e332.
[59] M. Kawashita, K. Kawamura, Z. Li, PMMA-based bone cements containing magnetite particles for the
hyperthermia of cancer, Acta Biomater. 6 (2010) 3187e3192, https://doi.org/10.1016/
j.actbio.2010.02.047.
Bionanomaterials for cancer therapy 463
[60] C. Wang, J. Yan, X. Cui, D. Cong, H. Wang, Preparation and characterization of magnetic hollow
PMMA nanospheres via in situ emulsion polymerization, Colloids Surf. A Physicochem. Eng. Asp. 363
(2010) 71e77, https://doi.org/10.1016/j.colsurfa.2010.04.016.
[61] N.L. Shimanovskii, Targeted transport of drugs by iron oxide nanoparticles, Russ. J. Gen. Chem. 84
(2014) 391e406, https://doi.org/10.1134/S1070363214020418.
[62] J.L. Arias, F. Linares-Molinero, V. Gallardo, A
´.V. Delgado, Study of carbonyl iron/
poly(butylcyanoacrylate) (core/shell) particles as anticancer drug delivery systems. Loading and release
properties, Eur. J. Pharmaceut. Sci. 33 (2008) 252e261, https://doi.org/10.1016/j.ejps.2007.12.005.
[63] N. Zohreh, S.H. Hosseini, A. Pourjavadi, Hydrazine-modified starch coated magnetic nanoparticles as an
effective pH-responsive nanocarrier for doxorubicin delivery, J. Ind. Eng. Chem. 39 (2016) 203e209,
https://doi.org/10.1016/j.jiec.2016.05.029.
[64] M. Frounchi, S. Shamshiri, Magnetic nanoparticles-loaded PLA/PEG microspheres as drug carriers,
J. Biomed. Mater. Res. 103 (2015) 1893e1898.
[65] J. Wu, Y. Wang, W. Jiang, S. Xu, R. Tian, Synthesis and characterization of recyclable clusters of
magnetic nanoparticles as doxorubicin carriers for cancer therapy, Appl. Surf. Sci. 321 (2014) 43e49,
https://doi.org/10.1016/j.apsusc.2014.09.184.
[66] M. Hałupka-Bryl, M. Bednarowicz, B. Dobosz, R. Krzyminiewski, T. Zalewski, B. Wereszczy"
nska,
G. Nowaczyk, M. Jarek, Y. Nagasaki, Doxorubicin loaded PEG-b-poly(4-vinylbenzylphosphonate) coated
magnetic iron oxide nanoparticles for targeted drug delivery, J. Magn. Magn Mater. 384 (2015)
320e327, https://doi.org/10.1016/j.jmmm.2015.02.078.
[67] R. Mangaiyarkarasi, S. Chinnathambi, S. Karthikeyan, P. Aruna, S. Ganesan, Paclitaxel conjugated
Fe3O4@LaF3: Ce3þ, Tb3þnanoparticles as bifunctional targeting carriers for Cancer theranostics
application, J. Magn. Magn Mater. 399 (2016) 207e215, https://doi.org/10.1016/j.jmmm.2015.09.084.
[68] M.M. Lin, Y.J. Kang, Y. Sohn, D.K. Kim, Dual targeting strategy of magnetic nanoparticle-loaded and
RGD peptide-activated stimuli-sensitive polymeric micelles for delivery of paclitaxel, J. Nanoparticle
Res. 17 (2015), https://doi.org/10.1007/s11051-015-3033-2.
[69] M. Fazilati, Anti-neoplastic applications of heparin coated magnetic nanoparticles against human ovarian
cancer, J. Inorg. Organomet. Polym. Mater. 24 (2014) 551e559, https://doi.org/10.1007/s10904-013-
0005-9.
[70] M. Parsian, G. Unsoy, P. Mutlu, S. Yalcin, A. Tezcaner, U. Gunduz, Loading of Gemcitabine on chitosan
magnetic nanoparticles increases the anti-cancer efficacy of the drug, Eur. J. Pharmacol. 784 (2016)
121e128, https://doi.org/10.1016/j.ejphar.2016.05.016.
[71] Y.J. Li, M. Dong, F.M. Kong, J.P. Zhou, Folate-decorated anticancer drug and magnetic nanoparticles
encapsulated polymeric carrier for liver cancer therapeutics, Int. J. Pharm. 489 (2015) 83e90, https://
doi.org/10.1016/j.ijpharm.2015.04.028.
[72] G. Unsoy, S. Yalcin, R. Khodadust, P. Mutlu, O. Onguru, U. Gunduz, Chitosan magnetic nanoparticles
for pH responsive Bortezomib release in cancer therapy, Biomed. Pharmacother. 68 (2014) 641e648,
https://doi.org/10.1016/j.biopha.2014.04.003.
[73] J.L. Arias, L.H. Reddy, P. Couvreur, Fe
3
O
4
/chitosan nanocomposite for magnetic drug targeting to
cancer, J. Mater. Chem. 22 (2012) 7622e7632, https://doi.org/10.1039/c2jm15339d.
[74] A. Pourjavadi, Z.M. Tehrani, C. Bennett, PEG-co-polyvinyl pyridine coated magnetic mesoporous silica
nanoparticles for pH-responsive controlled release of doxorubicin, Int. J. Polym. Mater. Polym. Biomater.
64 (2015) 570e577, https://doi.org/10.1080/00914037.2014.996706.
[75] M. Zarouni, R. Salehi, A. Akbarzadeh, N. Samadi, S. Davaran, F. Ramezani, H. Dariushnejad,
Biocompatible polymer coated paramagnetic nanoparticles for doxorubicin delivery: synthesis and
anticancer effects against human breast cancer cells, Int. J. Polym. Mater. Polym. Biomater. 64 (2015)
718e726, https://doi.org/10.1080/00914037.2014.1002129.
[76] K. Kan-Dapaah, N. Rahbar, W. Soboyejo, Implantable magnetic nanocomposites for the localized
treatment of breast cancer, J. Appl. Phys. 116 (2014), https://doi.org/10.1063/1.4903736.
464 Chapter 13
[77] S.M. Mirvakili, Q.P. Ngo, R. Langer, Polymer nanocomposite microactuators for on-demand chemical
release via high-frequency magnetic field excitation, Nano Lett. 20 (2020) 4816e4822, https://doi.org/
10.1021/acs.nanolett.0c00648.
[78] F. Pereira, J. Aires-de-Sousa, Computational methodologies in the exploration of marine natural product
leads, Mar. Drugs 16 (2018), https://doi.org/10.3390/md16070236.
[79] J. Wiese, J.F. Imhoff, Marine bacteria and fungi as promising source for new antibiotics, Drug Dev. Res.
80 (2019) 24e27, https://doi.org/10.1002/ddr.21482.
[80] D.J. Newman, G.M. Cragg, Drugs and drug candidates from marine sources: an assessment of the
current “state of play”, Planta Med. 82 (9/10) (2016) 775e789.
[81] W.H. Gerwick, B.S. Moore, Lessons from the past and charting the future of marine natural products
drug discovery and chemical biology, Chem. Biol. 19 (2012) 85e98, https://doi.org/10.1016/
j.chembiol.2011.12.014.
[82] K.M. Krishnan, Biomedical nanomagnetics: a spin through possibilities in imaging, diagnostics, and
therapy, IEEE Trans. Magn. 46 (2010) 2523e2558, https://doi.org/10.1109/TMAG.2010.2046907.
[83] M.G. Harisinghani, J. Barentsz, P.F. Hahn, W.M. Deserno, S. Tabatabaei, C.H. van de Kaa, J. de la
Rosette, R. Weissleder, Noninvasive detection of clinically occult lymph-node metastases in prostate
cancer, N. Engl. J. Med. 348 (2003) 2491e2499, https://doi.org/10.1056/nejmoa022749.
[84] R. Bawa, G.F. Audette, I. Rubinstein, Handbook of Clinical Nanomedicine: Nanoparticles, Imaging,
Therapy and Clinical Applications, 2016, pp. 1e1662.
[85] C. Louis, O. Pluchery, Gold Nanoparticles for Physics, Chemistry and Biology, 2012.
[86] B. Wiley, Y. Sun, Y. Xia, Synthesis of silver nanostructures with controlled shapes and properties, Acc.
Chem. Res. 40 (2007) 1067e1076, https://doi.org/10.1021/ar7000974.
[87] P.G. Jamkhande, N.W. Ghule, A.H. Bamer, M.G. Kalaskar, Metal nanoparticles synthesis: an overview
on methods of preparation, advantages and disadvantages, and applications, J. Drug Deliv. Sci. Technol.
53 (2019), https://doi.org/10.1016/j.jddst.2019.101174.
[88] G. Mansoureh, V. Parisa, Synthesis of metal nanoparticles using laser ablation technique, in: Emerging
Applications of Nanoparticles and Architectural Nanostructures: Current Prospects and Future Trends,
2018, pp. 575e596.
[89] M.Z. Ahmad, S. Akhter, G.K. Jain, M. Rahman, S.A. Pathan, F.J. Ahmad, R.K. Khar, Metallic
nanoparticles: technology overview and drug delivery applications in oncology, Expert Opin. Drug Deliv.
7 (2010) 927e942, https://doi.org/10.1517/17425247.2010.498473.
[90] C. Krishnaraj, P. Muthukumaran, R. Ramachandran, M.D. Balakumaran, P.T. Kalaichelvan, Acalypha
indica Linn: biogenic synthesis of silver and gold nanoparticles and their cytotoxic effects against
MDA-MB-231, human breast cancer cells, Biotechnol. Rep. 4 (2014) 42e49, https://doi.org/10.1016/
j.btre.2014.08.002.
[91] R. Vijayan, S. Joseph, B. Mathew, Indigofera tinctoria leaf extract mediated green synthesis of silver and
gold nanoparticles and assessment of their anticancer, antimicrobial, antioxidant and catalytic properties,
Artif. Cells Nanomed. Biotechnol. 46 (2018) 861e871, https://doi.org/10.1080/21691401.2017.1345930.
[92] R. Foldbjerg, D.A. Dang, H. Autrup, Cytotoxicity and genotoxicity of silver nanoparticles in the human
lung cancer cell line, A549, Arch. Toxicol. 85 (2011) 743e750, https://doi.org/10.1007/s00204-010-
0545-5.
[93] P.V. AshaRani, G.L.K. Mun, M.P. Hande, S. Valiyaveettil, Cytotoxicity and genotoxicity of silver
nanoparticles in human cells, ACS Nano 3 (2009) 279e290, https://doi.org/10.1021/nn800596w.
[94] M.R. Kamala Priya, P.R. Iyer, Anticancer studies of the synthesized gold nanoparticles against MCF 7
breast cancer cell lines, Appl. Nanosci. 5 (2015) 443e448, https://doi.org/10.1007/s13204-014-0336-z.
[95] S.A. Loutfy, M.B. Mohamed, N.T. Abdel-Ghani, N. Al-Ansary, W.A. Abdulla, O.M. El-Borady,
Y. Hussein, M.H. Eldin, Metallic nanomaterials as drug carriers to decrease side effects of chemotherapy
(in vitro: cytotoxicity study), J. Nanopharm. Drug Deliv. 1 (2013) 138e149, https://doi.org/10.1166/
jnd.2013.1010.
Bionanomaterials for cancer therapy 465
[96] V. Torchilin, Tumor delivery of macromolecular drugs based on the EPR effect, Adv. Drug Deliv. Rev.
63 (2011) 131e135, https://doi.org/10.1016/j.addr.2010.03.011.
[97] M. Upreti, A. Jyoti, P. Sethi, Tumor microenvironment and nanotherapeutics, Transl. Cancer Res. 2
(2013) 309e319, https://doi.org/10.3978/j.issn.2218-676X.2013.08.11.
[98] M. Ganjali, N. Ahmadi Nasab, M. Ganjali, Cytotoxic effects of laser-synthesized silver nanoparticles on
human breast adenocarcinoma cells, in: Materials Today: Proceedings, 2018, pp. 15567e15572.
[99] S.A. Loutfy, N.A. Al-Ansary, N.T. Abdel-Ghani, A.R. Hamed, M.B. Mohamed, J.D. Craik, T.A. Salah
Eldin, A.M. Abdellah, Y. Hussein, M.T.M. Hasanin, S.E.I. Elbehairi, Anti-proliferative activities of
metallic nanoparticles in an in vitro breast cancer model, Asian Pac. J. Cancer Prev. 16 (2015)
6039e6046, https://doi.org/10.7314/APJCP.2015.16.14.6039.
[100] C. Buzea, I.I. Pacheco, K. Robbie, Nanomaterials and nanoparticles: sources and toxicity, Biointerphases
2 (2007) MR17eMR71, https://doi.org/10.1116/1.2815690.
[101] M.E. Selim, A.A. Hendi, Gold nanoparticles induce apoptosis in MCF-7 human breast cancer cells,
Asian Pac. J. Cancer Prev. 13 (2012) 1617e1620, https://doi.org/10.7314/APJCP.2012.13.4.1617.
[102] P. Gopinath, S.K. Gogoi, A. Chattopadhyay, S.S. Ghosh, Implications of silver nanoparticle induced cell
apoptosis for in vitro gene therapy, Nanotechnology 19 (2008), https://doi.org/10.1088/0957-4484/19/7/
075104.
[103] A. Hekmat, A.A. Saboury, A. Divsalar, The effects of silver nanoparticles and doxorubicin combination
on DNA structure and its antiproliferative effect against T47D and MCF7 cell lines, J. Biomed.
Nanotechnol. 8 (2012) 968e982, https://doi.org/10.1166/jbn.2012.1451.
[104] B. Rosenberg, L. VanCamp, J.E. Trosko, V.H. Mansour, Platinum compounds: a new class of potent
antitumour agents [24], Nature 222 (1969) 385e386, https://doi.org/10.1038/222385a0.
[105] L. Cai, C. Yu, L. Ba, Q. Liu, Y. Qian, B. Yang, C. Gao, Anticancer platinum-based complexes with non-
classical structures, Appl. Organomet. Chem. 32 (4) (2018) e4228.
[106] C. Moncharmont, P. Auberdiac, A. Me
´lis, S. Afqir, C. Pacaut, C. Chargari, Y. Merrouche, N. Magne
´,
Cisplatine ou carboplatine, telle est la question, Bull. Cancer 98 (2) (2011) 164e175.
[107] V. Pinzani, F. Bressolle, I. Johanne Haug, M. Galtier, J.P. Blayac, P. Balme
`s, Cisplatin-induced renal
toxicity and toxicity-modulating strategies: a review, Cancer Chemother. Pharmacol. 35 (1) (1994) 1e9.
[108] D. Gibson, Platinum(IV) anticancer prodrugs-hypotheses and facts, Dalton Trans. 45 (2016)
12983e12991, https://doi.org/10.1039/c6dt01414c.
[109] P.J. O’Dwyer, J.P. Stevenson, S.W. Johnson, Clinical pharmacokinetics and administration of established
platinum drugs, Drugs 59 (2000) 19e27, https://doi.org/10.2165/00003495-200059004-00003.
[110] S. Dilruba, G.V. Kalayda, Platinum-based drugs: past, present and future, Cancer Chemother. Pharmacol.
77 (2016) 1103e1124, https://doi.org/10.1007/s00280-016-2976-z.
[111] N.J. Wheate, S. Walker, G.E. Craig, R. Oun, The status of platinum anticancer drugs in the clinic and in
clinical trials, Dalton Trans. 39 (2010) 8113e8127, https://doi.org/10.1039/c0dt00292e.
[112] M.J. McKeage, Lobaplatin: a new antitumour platinum drug, Expert Opin. Investig. Drugs 10 (2001)
119e128, https://doi.org/10.1517/13543784.10.1.119.
[113] H. Cao, Y. Feng, L. Chen, C. Yu, Lobaplatin inhibits prostate cancer proliferation and migration through
regulation of BCL2 and BAX, Dose-Response 17 (2019), https://doi.org/10.1177/1559325819850981.
[114] K.H. Lee, M.S. Hyun, H.-K. Kim, H.M. Jin, J. Yang, H.S. Song, Y.R. Do, H.M. Ryoo, J.S. Chung,
D.Y. Zang, H.-Y. Lim, J.Y. Jin, C.Y. Yim, H.S. Park, J.S. Kim, C.H. Sohn, S.N. Lee, Randomized,
multicenter, phase III trial of heptaplatin 1-hour infusion and 5-fluorouracil combination chemotherapy
comparing with cisplatin and 5-fluorouracil combination chemotherapy in patients with advanced gastric
cancer, Cancer Res. Treat. 41 (2009) 12, https://doi.org/10.4143/crt.2009.41.1.12.
[115] T.K. Sau, A.L. Rogach, F. Ja
¨ckel, T.A. Klar, J. Feldmann, Properties and applications of colloidal
nonspherical noble metal nanoparticles, Adv. Mater. 22 (16) (2010) 1805e1825.
[116] R.A. Sperling, P.R. Gil, F. Zhang, M. Zanella, W.J. Parak, Biological applications of gold nanoparticles,
Chem. Soc. Rev. 37 (2008) 1896e1908, https://doi.org/10.1039/b712170a.
466 Chapter 13
[117] N.M. Elbaz, L. Ziko, R. Siam, W. Mamdouh, Core-shell silver/polymeric nanoparticles-based
combinatorial therapy against breast cancer in-vitro, Sci. Rep. 6 (2016) 30729e30738, https://doi.org/
10.1038/srep30729.
[118] V. Venkatpurwar, A. Shiras, V. Pokharkar, Porphyran capped gold nanoparticles as a novel carrier for
delivery of anticancer drug: in vitro cytotoxicity study, Int. J. Pharm. 409 (2011) 314e320, https://
doi.org/10.1016/j.ijpharm.2011.02.054.
[119] Y.H. Chen, C.Y. Tsai, P.Y. Huang, M.Y. Chang, P.C. Cheng, C.H. Chou, D.H. Chen, C.R. Wang,
A.L. Shiau, C.L. Wu, Methotrexate conjugated to gold nanoparticles inhibits tumor growth in a
syngeneic lung tumor model, Mol. Pharm. 4 (2007) 713e722, https://doi.org/10.1021/mp060132k.
[120] R.W.Y. Habash, R. Bansal, D. Krewski, H.T. Alhafid, Thermal therapy, part 2: hyperthermia techniques,
Crit. Rev. Biomed. Eng. 34 (2006) 491e542, https://doi.org/10.1615/CritRevBiomedEng.v34.i6.30.
[121] P. Wust, B. Hildebrandt, G. Sreenivasa, B. Rau, J. Gellermann, H. Riess, R. Felix, P. Schlag,
Hyperthermia in combined treatment of cancer, Lancet Oncol. 3 (2002) 487e497, https://doi.org/
10.1016/S1470-2045(02)00818-5.
[122] G.F. Baronzio, E.D. Hager, Hyperthermia in Cancer Treatment: A Primer, 2006.
[123] H. Tripathi, G.C. Pandey, A. Dubey, S.K. Shaw, N.K. Prasad, S.P. Singh, C. Rath, Superparamagnetic
manganese ferrite and strontium bioactive glass nanocomposites: enhanced biocompatibility and
antimicrobial properties for hyperthermia application, Adv. Eng. Mater. 23 (2021) 1e11, https://doi.org/
10.1002/adem.202000275.
[124] T. Kokubo, T. Hayashi, S. Sakka, T. Kitsugi, T. Yamamuro, Bonding between bioactive glasses, glass-
ceramics or ceramics in a simulated body fluid, Yogyo Kyokai Shi (J. Ceram. Soc. Jpn.) 95 (1987)
785e791, https://doi.org/10.2109/jcersj1950.95.1104_785.
[125] Q. Mu, H. Wang, M. Zhang, Nanoparticles for imaging and treatment of metastatic breast cancer, Expert
Opin. Drug Deliv. 14 (2017) 123e136.
[126] X. Shang, P. Chen, T. Jia, D. Feng, S. Zhang, J.Q. Zhenrong Sun, Upconversion luminescence
mechanisms of Er(3þ) ions under excitation of an 800 nm laser, Phys. Chem. Chem. Phys. 17 (2015)
11481e11489.
[127] A.N. Koo, K. Hyun Min, H. Jae Lee, J. Ho Jegal, S.C.L. Jae Won Lee, Calcium carbonate mineralized
nanoparticles as an intracellular transporter of cytochrome c for cancer therapy, Chem. Asian J. 10
(2015) 2380e2387.
[128] S.A.M. Abdel-Hameed, M.A. Marzouk, M.M. Farag, Effect of P
2
O
5
and MnO
2
on crystallization of
magnetic glass ceramics, J. Adv. Res. 5 (2014) 543e550, https://doi.org/10.1016/j.jare.2013.07.001.
[129] S.A.M. Abdel-Hameed, M.M. Hessien, M.A. Azooz, Preparation and characterization of some
ferromagnetic glass-ceramics contains high quantity of magnetite, Ceram. Int. 35 (2009) 1539e1544,
https://doi.org/10.1016/j.ceramint.2008.08.021.
[130] R. Yang, M. Hou, Y. Gao, S. Lu, L. Zhang, Z. Xu, M.L. Chang, P.X. Yuejun Kang, Biomineralization-
inspired crystallization of manganese oxide on silk fibroin nanoparticles for in vivo MR/fluorescence
imaging-assisted tri-modal therapy of cancer, Theranostics 9 (2019) 6314e6333.
[131] S. Barui, V. Cauda, Multimodal decorations of mesoporous silica nanoparticles for improved cancer
therapy, Pharmaceutics 12 (2020) 1e33.
[132] N.A. Nasab, H.H. Kumleh, M. Beygzadeh, S. Teimourian, M. Kazemzad, Delivery of curcumin by a pH-
responsive chitosan mesoporous silica nanoparticles for cancer treatment, Artif. Cells Nanomed.
Biotechnol. 46 (2018), https://doi.org/10.1080/21691401.2017.1290648.
[133] H. Tang, J. Guo, Y. Sun, B. Chang, Q. Ren, W. Yang, Facile synthesis of pH sensitive polymer-coated
mesoporous silica nanoparticles and their application in drug delivery, Int. J. Pharm. 421 (2011)
388e396, https://doi.org/10.1016/j.ijpharm.2011.10.013.
[134] J. Correia, Stimuli-responsive mesoporous silica nanoparticles for cancer therapy: a review, Microporous
Mesoporous Mater. 236 (2016) 141e157.
Bionanomaterials for cancer therapy 467
[135] A. Luetke, P.A. Meyers, I. Lewis, H. Juergens, Osteosarcoma treatmentewhere do we stand? a state of
the art review, Cancer Treat. Rev. 40 (4) (2014) 523e532.
[136] M.E. Anderson, Update on survival in osteosarcoma, Orthop. Clin. North Am. 47 (2016) 283e292,
https://doi.org/10.1016/j.ocl.2015.08.022.
[137] T. Roose, P.A. Netti, L.L. Munn, Y. Boucher, R.K. Jain, Solid stress generated by spheroid growth
estimated using a linear poroelasticity model, Microvasc. Res. 66 (2003) 204e212, https://doi.org/
10.1016/S0026-2862(03)00057-8.
[138] S. Ghosh, R.S.K. Raju, N. Ghosh, K. Chaudhury, S. Ghosh, I. Banerjee, N. Pramanik, Development and
physicochemical characterization of doxorubicin-encapsulated hydroxyapatiteepolyvinyl alcohol
nanocomposite for repair of osteosarcoma-affected bone tissues, C. R. Chem. 22 (2019) 46e57, https://
doi.org/10.1016/j.crci.2018.10.005.
[139] K. Dan Son, Y.J. Kim, Anticancer activity of drug-loaded calcium phosphate nanocomposites against
human osteosarcoma, Biomater. Res. 21 (2017), https://doi.org/10.1186/s40824-017-0099-1.
[140] S. Mondal, P. Manivasagan, S. Bharathiraja, M. Santha Moorthy, Van T. Nguyen, H.H. Kim, S.Y. Nam,
D.L. Kang, J.O. and, Hydroxyapatite coated iron oxide nanoparticles: a promising nanomaterial for
magnetic hyperthermia cancer treatment, Nanomaterials 7 (2017) 1e15.
[141] L. Palanikumar, J. Kim, J.Y. Oh, H. Choi, M.H. Park, C. Kim, J.H. Ryu, Hyaluronic acid-modified
polymeric gatekeepers on biodegradable mesoporous silica nanoparticles for targeted cancer therapy,
ACS Biomater. Sci. Eng. 4 (2018) 1716e1722, https://doi.org/10.1021/acsbiomaterials.8b00218.
[142] M. Stolzoff, T.J. Webster, Reducing bone cancer cell functions using selenium nanocomposites,
J. Biomed. Mater. Res. 104 (2016) 476e482, https://doi.org/10.1002/jbm.a.35583.
[143] M. Sumathra, M. Rajan, M.A. Munusamy, A phosphorylated chitosan armed hydroxyapatite
nanocomposite for advancing activity on: osteoblast and osteosarcoma cells, New J. Chem. 42 (2018)
12457e12466, https://doi.org/10.1039/c8nj01316k.
[144] V.M. Wu, J. Mickens, V. Uskokovi"
c, Bisphosphonate-functionalized hydroxyapatite nanoparticles for the
delivery of the bromodomain inhibitor JQ1 in the treatment of osteosarcoma, ACS Appl. Mater.
Interfaces 9 (2017) 25887e25904, https://doi.org/10.1021/acsami.7b08108.
[145] E. Vyskocilova
´, I. Lu!
sticka
´, I. Paterova
´, L. MacHova
´, L. Cerveny
´, Modified MCM-41 as a drug delivery
system for acetylsalicylic acid, Solid State Sci. 38 (2014) 85e89, https://doi.org/10.1016/
j.solidstatesciences.2014.10.004.
[146] F. Tang, L. Li, D. Chen, Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery,
Adv. Mater. 24 (2012) 1504e1534, https://doi.org/10.1002/adma.201104763.
[147] W. Cheng, C. Liang, L. Xu, G. Liu, N. Gao, W. Tao, L. Luo, Y. Zuo, X. Wang, X. Zhang, X. Zeng,
L. Mei, TPGS-functionalized polydopamine-modified mesoporous silica as drug nanocarriers for
enhanced lung cancer chemotherapy against multidrug resistance, Small 13 (2017), https://doi.org/
10.1002/smll.201700623.
[148] N. Ahmadi Nasab, H. Hassani Kumleh, M. Kazemzad, F. Ghavipanjeh, Application of spherical
mesoporous silica MCM-41 for adsorption of dibenzothiophene (A sulfur containing compound) from
model oil. Iran, J. Chem. Chem. Eng. 33 (2014) 37e42.
[149] C.T. Kresge, M.E. Leonowicz, W.J. Roth, J.C. Vartuli, J.S. Beck, Ordered mesoporous molecular sieves
synthesized by a liquid-crystal template mechanism, Nature 359 (1992) 710e712, https://doi.org/
10.1038/359710a0.
[150] A. Pourjavadi, Z.M. Tehrani, Mesoporous silica nanoparticles (MCM-41) coated PEGylated chitosan as a
pH-responsive nanocarrier for triggered release of erythromycin, Int. J. Polym. Mater. Polym. Biomater.
63 (2014) 692e697, https://doi.org/10.1080/00914037.2013.862534.
468 Chapter 13
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background: Toward designing new class of desired physicochemical properties of metallic nanomaterials such as spherical gold and silver nanoparticles capped with citrate to be used as a drug delivery system for anticancerous drug like 6-Mercapopurine (6-MP) against human breast cancer. This could increase curative effect of potent drug and lower side effects of high dose of anticancerous drug. Methods: Full charcterization for the prepared nanoparticles and drugs loaded nanoparticles was performed by TEM. Human breast cancer cell line (MCF-7) was maintained in the laboratory. Colourimetric cytotoxicity assay was established and standardized for testing all engineered nanomaterials. Results: Alteration of cell morphology was observed after treatment of MCF-7 with concentrated solution of silver nanoparticles (AgNPs) and 6-MP@AgNPs (10–3 M) capped with citrate after 24 hrs of cells exposure. Also, such concentration of AgNPs exhibited more aggressive cytotoxic effect than that exerted by AuNPs after 24 hrs of cell exposure (viability was 30% vs. 83 for both AgNPs and AuNPs, respectively). The concentrated solution of gold composite (10–3 M of 6-MP@AuNPs) exhibited double cytotoxic effect than that exerted by each of free 6-MP or AuNPs, after 24 hrs of cell exposure (43% viability for gold composite vs. 92% and 83% for both 6-MP and AuNPs, respectively). The effect of capping materials has been tested via investigating the cytotoxcity of AgNPs capped with lactose compared to that which capped with citrate. Our results indicate that Ag NPs capped with lactose is much more safer on cells than that capped with citrate. The antipro- liferative activity of tested nanomaterials against breast cancer was notable at different concentrations, for example, at 25 μM, gold composite showed aggressive cytotoxic effect than that exerted by silver composite capped with lactose (viability 59% vs. 80%), but smaller molar ratio of 6-MP used for synthesis silver composite may be useful as a safer drug carrier against human breast cancer. Conclusion: Our engineered metallic nanoparticles provided a flexible platform for enhancing anticancerous activity of 6-MP against human breast cancer. Silver nanocomposite capped with lactose may be useful as a drug carrier for 6-MP minimizing its side effect more than gold nanopaticles capped with citrate, but this require further studies.
Article
Full-text available
Herein, the strontium bioactive glass with ferrite composites is studied for the application of biocompatibility and magnetic hyperthermia. The bioactive glass SrBG (46.1SiO2–21.9CaO–24.4Na2O–2.6P2O5–5SrO) prepared by the melt quench technique at 1400 °C and manganese ferrite (MnFe2O4) prepared by the modified microwave reflux oven are mixed together and sintered at 400 °C for 6 h for making composites. The in vitro bioactivity of these composites is observed by immersing in simulated body fluid (SBF). The growth of the precipitated hydroxyapatite phase observed after SBF treatment is confirmed by the X‐ray diffractometer (XRD), Fourier transform infrared (FTIR) spectrometer, and scanning electron microscope (SEM) results. The in vitro cell viability assessment of these composites with MG‐63 cell lines indicates that the composites are biocompatible in nature. Furthermore, these composites show the antibacterial effect on the E. coli and S. Aureus bacteria cells. The room temperature superparamagnetic behavior accompanied by the heating ability in these composites proves their potential role for hyperthermia applications.
Article
Full-text available
Adult T-cell leukemia-lymphoma (ATL) is a distinct mature T-cell malignancy caused by human T-cell leukemia/lymphotropic virus type I (HTLV-1) endemic in some areas in the world. HTLV-1 transmits through mother-to-child infection via breastfeeding, sexual intercourses, and blood transfusions. Early HTLV-1 infection, presumably through mother’s milk, is crucial in developing ATL. The estimated cumulative risk of the development of ATL in HTLV-1 carriers is a few percent after transmission from their mothers. The median age of ATL onset is about 70 in Japan and is now rising, whereas an overall mean age in the mid-forties is reported in other parts of the world. ATL is classified into four clinical subtypes (acute, lymphoma, chronic, and smoldering) defined by organ lesions and LDH/calcium values. In aggressive ATL (acute, lymphoma or unfavorable chronic types) and indolent ATL (favorable chronic or smoldering types), intensive chemotherapy followed by allogeneic hematopoietic stem cell transplantation and watchful waiting until disease progression has been recommended, respectively, in Japan. Based on a worldwide meta-analysis and multiple other retrospective studies, the antiviral combination of interferon alpha (IFN) and zidovudine (AZT) is recommended in many parts of the world in acute, chronic, and smoldering ATL whereas patients with the lymphoma subtype are treated with chemotherapy, either alone or combined with AZT/IFN. Several new agents have been approved for ATL by the Pharmaceutical and Medical Devices Agency (PMDA) after clinical trials, including an anti-CC chemokine receptor 4 monoclonal antibody, mogamulizumab; an immunomodulatory agent, lenalidomide; and an anti-CD30 antibody/drug conjugate, brentuximab vedotin.
Article
Full-text available
The presence of leaky vasculature and the lack of lymphatic drainage of small structures by the solid tumors formulate nanoparticles as promising delivery vehicles in cancer therapy. In particular, among various nanoparticles, the mesoporous silica nanoparticles (MSN) exhibit numerous outstanding features, including mechanical thermal and chemical stability, huge surface area and ordered porous interior to store different anti-cancer therapeutics with high loading capacity and tunable release mechanisms. Furthermore, one can easily decorate the surface of MSN by attaching ligands for active targeting specifically to the cancer region exploiting overexpressed receptors. The controlled release of drugs to the disease site without any leakage to healthy tissues can be achieved by employing environment responsive gatekeepers for the end-capping of MSN. To achieve precise cancer chemotherapy, the most desired delivery system should possess high loading efficiency, site-specificity and capacity of controlled release. In this review we will focus on multimodal decorations of MSN, which is the most demanding ongoing approach related to MSN application in cancer therapy. Herein, we will report about the recently tried efforts for multimodal modifications of MSN, exploiting both the active targeting and stimuli responsive behavior simultaneously, along with individual targeted delivery and stimuli responsive cancer therapy using MSN.
Article
Full-text available
On-demand delivery of substances has been demonstrated for various applications in the fields of chemistry and biomedical engineering. Single-pulse release profile has been shown previously for micro/nanoparticles in different form factors. However, to obtain a sustained release, a pulsatile release profile is needed. Here, we demonstrate such a release profile from magnetic nanocomposite polymer microspheres loaded with chemicals. By exciting the microparticles with AC magnetic fields, we could achieve up to 61% cumulative release over a five-day period. One of the main advantages of using a magnetic stimulus is that the properties of the environment (e.g., transparency, density, and depth) in which the particles are located do not affect the performance. The operating magnitude of the magnetic field used in this work is safe and does not interact with any non-metallic materials. The proposed approach can potentially be used in micro-chemistry, drug delivery, lab-on-chip, and micro-robots for drug delivery.
Article
Full-text available
In modern-day medicine, nanotechnology and nanoparticles are some of the indispensable tools in disease monitoring and therapy. The term "nanomaterials" describes materials with nanoscale dimensions (< 100 nm) and are broadly classified into natural and synthetic nanomaterials. However, "engineered" nanomaterials have received significant attention due to their versatility. Although enormous strides have been made in research and development in the field of nanotechnology, it is often confusing for beginners to make an informed choice regarding the nanocarrier system and its potential applications. Hence, in this review, we have endeavored to briefly explain the most commonly used nanomaterials, their core properties and how surface functionalization would facilitate competent delivery of drugs or therapeutic molecules. Similarly, the suitability of carbon-based nanomaterials like CNT and QD has been discussed for targeted drug delivery and siRNA therapy. One of the biggest challenges in the formulation of drug delivery systems is fulfilling targeted/specific drug delivery, controlling drug release and preventing opsonization. Thus, a different mechanism of drug targeting, the role of suitable drug-laden nanocarrier fabrication and methods to augment drug solubility and bioavailability are discussed. Additionally, different routes of nanocarrier administration are discussed to provide greater understanding of the biological and other barriers and their impact on drug transport. The overall aim of this article is to facilitate straightforward perception of nanocarrier design, routes of various nanoparticle administration and the challenges associated with each drug delivery method.
Article
Full-text available
Regenerated silk fibroin (SF) is a type of natural biomacromolecules with outstanding biocompatibility and biodegradability. However, stimulus-responsive SF-based nanocomplex has seldom been reported for application in tumor diagnosis and therapy. Methods: As a proof-of-concept study, a multifunctional SF@MnO2 nanoparticle-based platform was strategically synthesized using SF as a reductant and a template via a biomineralization-inspired crystallization process in an extremely facile way. Because of their mesoporous structure and abundant amino and carboxyl terminal residues, SF@MnO2 nanoparticles were co-loaded with a photodynamic agent indocyanine green (ICG) and a chemotherapeutic drug doxorubicin (DOX) to form a SF@MnO2/ICG/DOX (SMID) nanocomplex. Results: The obtained product was highly reactive with endogenous hydrogen peroxide (H2O2) in tumor microenvironment, which was decomposed into O2 to enhance tumor-specific photodynamic therapy (PDT). Moreover, SMID nanocomplex produced a strong and stable photothermal effect upon near-infrared (NIR) irradiation for photothermal therapy (PTT) owing to the distinct photothermal response of SF@MnO2 and stably conjugated ICG. The concurrent NIR fluorescence and magnetic resonance (MR) imaging in vivo both indicated effective tumor-specific enrichment of SMID nanoparticles via enhanced permeability and retention (EPR) effect. Animal studies further verified that SMID nanoparticles remarkably improved tumor inhibitive efficacy through combination PTT/PDT/chemotherapy with minimal systemic toxicity or adverse effect. Conclusion: This study demonstrated the promising potential of SF-based nanomaterial to address some of the key challenges in cancer therapy due to unfavorable tumor microenvironment for drug delivery.
Article
Full-text available
Lobaplatin is a diastereometric mixture of platinum (II) complexes, which contain a 1,2-bis (aminomethyl) cyclobutane stable ligand and lactic acid. Previous studies have showed that lobaplatin plays inhibiting roles in various types of tumors. However, the role of lobaplatin in prostate cancer remains unknown. Cell viability was detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Cell proliferation was detected by cell colony formation assay. Cell migration and invasion were determined by transwell migration and invasion assay. Cell apoptosis was detected by flow cytometry. The messenger RNA and protein expression levels were detected by quantitative real-time polymerase chain reaction and Western blot. Lobaplatin treatment inhibits cell viability, cell proliferation, cell migration, and invasion, while promotes cell apoptosis of prostate cancer cell lines DU145 and PC3. Meanwhile, lobaplatin treatment regulates apoptosis by downregulation of BCL2 expression and upregulation of BAX expression levels. Our study suggests lobaplatin inhibits prostate cancer proliferation and migration through regulation of BCL2 and BAX expression.
Article
Epigenetic regulation is critical to physiological control of development, cell fate, cell proliferation, genomic integrity and, fundamentally, transcriptional regulation. This epigenetic control occurs at multiple levels including through DNA methylation, histone modification, nucleosome remodelling and modulation of the 3D chromatin structure. Alterations in genes that encode chromatin regulators are common among mesenchymal neoplasms, a collection of more than 160 tumour types including over 60 malignant variants (sarcomas) that have unique and varied genetic, biological and clinical characteristics. Herein, we review those sarcomas in which chromatin pathway alterations drive disease biology. Specifically, we emphasize examples of dysregulation of each level of epigenetic control though mechanisms that include alterations in metabolic enzymes that regulate DNA methylation and histone post-translational modifications, mutations in histone genes, subunit loss or fusions in chromatin remodelling and modifying complexes, and disruption of higher-order chromatin structure. Epigenetic mechanisms of tumorigenesis have been implicated in mesenchymal tumours ranging from chondroblastoma and giant cell tumour of bone to chondrosarcoma, malignant peripheral nerve sheath tumour, synovial sarcoma, epithelioid sarcoma and Ewing sarcoma — all diseases that present in a younger patient population than most cancers. Finally, we review current and potential future approaches for the development of sarcoma therapies based on this emerging understanding of chromatin dysregulation. This Review discusses how the disease biology of many sarcomas is driven by chromatin pathway alterations ranging from dysregulation of DNA methylation, histone modifications and nucleosome remodelling to disruption of higher-order, 3D chromatin structure, with a view to use this knowledge to better develop targeted therapies for patients with sarcoma.
Article
Unforeseen changes in surface properties because of particle size have made nanoparticles very popular in the field of material science. Decrease in particle size to nano-size demonstrates peculiar and improved properties such as particle size distribution and morphology. This distinctive change in specific surface area is responsible for its high value and affects imperative parameters like surface reactivity. Application of nanoparticles in various fields like energy, medicines and nutrition has tremendously increased nowadays. In pharmaceutical and medicinal sciences, nanoparticles are playing imperative role. Several approaches are employed for the metallic nanoparticles preparation, which is categorized into two main types on as bottom up methods and top down methods depending on starting material of nanoparticle preparation. Certain metals have distinctive properties like antimicrobial property of gold and silver. Metal particles such as gold are widely used from ancient time for the medicines and Ayurvedic preparations in India and China. The use of metal nanoparticles is continuously increasing worldwide in biomedicine and allied disciplines. Nowadays researchers are focusing on metal nanoparticles, nanostructures and nanomaterial synthesis because of their conspicuous properties. Present review selectively focuses on different methods of nanoparticle preparations and its advantages, disadvantages, and applications.