ArticlePDF AvailableLiterature Review

Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development

Authors:

Abstract and Figures

The spread of metastatic cancer cell is the main cause of death worldwide. Cellular and molecular basis of the action of phytochemicals in the modulation of metastatic cancer highlights the importance of fruits and vegetables. Quercetin is a natural bioflavonoid present in fruits, vegetables, seeds, berries, and tea. The cancer-preventive activity of quercetin is well documented due to its anti-inflammatory, anti-proliferative and anti-angiogenic activities. However, poor water solubility and delivery, chemical instability, short half-life, and low-bioavailability of quercetin limit its clinical application in cancer chemoprevention. A better understanding of the molecular mechanism of controlled and regulated drug delivery is essential for the development of novel and effective therapies. To overcome the limitations of accessibility by quercetin, it can be delivered as nanoconjugated quercetin. Nanoconjugated quercetin has attracted much attention due to its controlled drug release, long retention in tumor, enhanced anticancer potential, and promising clinical application. The pharmacological effect of quercetin conjugated nanoparticles typically depends on drug carriers used such as liposomes, silver nanoparticles, silica nanoparticles, PLGA (Poly lactic-co-glycolic acid), PLA (poly(D,L-lactic acid)) nanoparticles, polymeric micelles, chitosan nanoparticles, etc. In this review, we described various delivery systems of nanoconjugated quercetin like liposomes, silver nanoparticles, PLGA (Poly lactic-co-glycolic acid), and polymeric micelles including DOX conjugated micelles, metal conjugated micelles, nucleic acid conjugated micelles, and antibody-conjugated micelles on in vitro and in vivo tumor models; as well as validated their potential as promising onco-therapeutic agents in light of recent updates.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.net
Anti-Cancer Agents in Medicinal Chemistry, 2019, 19, 1-19 1
REVIEW ARTICLE
1871-5206/19 $58.00+.00 © 2019 Bentham Science Publishers
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development
Manjula Vinayak1,* and Akhilendra K. Maurya1,2
1Biochemistry & Molecular Biology Laboratory, Centre for Advanced Study in Zoology, Institute of Science, Banaras Hindu Univer-
sity, Va ranasi-221005, India; 2Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences,
University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
Abstract: The spread of metastatic cancer cell is the main cause of death worldwide. Cellular and molecular
basis of the action of phytochemicals in the modulation of metastatic cancer highlights the importance of fruits
and vegetables. Quercetin is a natural bioflavonoid present in fruits, vegetables, seeds, berries, and tea. The
cancer-preventive activity of quercetin is well documented due to its anti-inflammatory, anti-proliferative and
anti-angiogenic activities. However, poor water solubility and delivery, chemical instability, short half-life, and
low-bioavailability of quercetin lim it its clinic al applicatio n in cancer chemoprevention. A better understanding
of the molecular mechanism of controlled and regulated drug delivery is essen tial for the development of novel
and effective therapies. To overcome the limitations of accessibility by quercetin, it can be delivered as nano-
conjugated quercetin. Nanoconjugated quercetin has attracted much attention due to its controlled drug release,
long retention in tumor, enhanced anticancer potential, and promising clinical application. The pharmacological
effect of quercetin conjugated nanoparticles typically depends on drug carriers used such as liposomes, silver
nanoparticles, silica nanoparticles, PLGA (Poly lactic-co-glycolic acid), PLA (poly(D,L-lactic acid)) nanoparti-
cles, polymeric micelles, chitosan nanoparticles, etc.
In this review, we described various delivery systems of nanoconjugated quercetin like liposomes, silver
nanoparticles, PLGA (Poly lactic-co-glycolic acid), and polymeric micelles including DOX conjugated micelles,
metal conjugated micelles, nucleic acid conjugated micelles, and antibody-conju gated mic elles on in vitro and in
vivo tumor models; as well as validated their potential as promising onco-therapeutic agents in lig ht of recent
updates.
Keywords: Drug delivery, bioavailability, cancer th erapy, nanoparticles, quercetin, PLGA.
1. INTRODUCTION
Although there is tremendous development in chemotherapeutic
drugs along with other therapies of cancer, the side effects caused
by drugs and drug resistance, poor bioavailability, nonspecific tar-
geting, and low therapeutic indices remain a challenge [1, 2]. Natu-
ral phytochemicals offer an emerging strategy for chemoprevention
of various diseases with lesser side effects. Several herbal ingredi-
ents and antioxidants have been reported to have anticancer effects
[3-5]. Scientific evidence suggests that phytochemicals may inhibit
the process of carcinogenesis and reduce the burden of a few can-
cers with certain limitations [6 -9].
A recent study suggests that phytochemicals serve as dynamic
surface ligands to control nanoparticle and protein interactions; of
which nanoparticles as carriers of phytochemicals [10]. Pharmacol-
ogical significance of quercetin has a long history due to its poten-
tial health-promoting effects against numerous diseases such as
cardiovascular, diabetes, thrombosis, neurological disorder, and
cancer as well as for longevity [11-13]. The importance of quercetin
is realized because of its potential in an ti-carcinogenic [14-17],
anti-inflammatory [18], anti-obesity [19], antioxidan t [20-22], anti-
viral [23], and antibacterial activities [24, 25]. Owing to its poor
aqueous solubility, low bioavailability, difficulty in crossing th e
*Address correspondence to this author at the Biochemistry & Molecular
Biology Laboratory, Centre for Advanced Study in Zoology, Institute of
Science , Banaras Hind u Univ ersity, Varanasi-221005, India;
E-mail: manjulavinayak@rediffmail.com
blood-brain-barrier, and poor chemical stability, its clinical applica-
tion is limited [26]. In order to overcome these inadequacies of
quercetin, many new drug delivery systems have been studied and
developed to increase solubility and dissolution, elevating bioavail-
ability, raising drug stab ility, controlled drug release, indorsing
antioxidant activity, and improving therapeutic effect [27-32].
2. FOOD SAFETY AND SOURCES OF QUERCETIN
Quercetin is a ubiquitous bioactive flavonoid. It occurs in a
wide variety of fruits, vegetables, and beverages [14]. Onion, black
tea, red wine as well as various fruit juices are identified as rich
dietary sources of quercetin [12]. Quercetin is estimated to be con-
sumed approximately 25-50 mg in the daily diet [15]. Dietary in-
take at estimated levels of quercetin is reported to have no undesir-
able health effects [33]. Quercetin has been utilized as a component
in food and pharm aceutical industries because of its health promis-
ing benefits. It has been marketed primarily as a dietary supplement
in the USA. Quercetin is commercially used as a supplement for
dog foods [34]. Quercetin shows no mutagenicity/genotoxicity in
vivo [12]. LD50 for quercetin is 160 mg and 3000 mg per kg body
weight in oral and intraperitoneal dose of mice, respectively [12].
Quercetin and its prodrug have entered phase 1 clinical trial for
human cancers [35, 36]. Although oral or intravenous administra-
tion of prodrug QC12 to the patient does not show promising clini-
cal significan ce, quercetin explores the possibility to fight against
other cancers [36]. Cmax and Tmax for quercetin are reported as 2.3 ±
1.5 µg/mL and 0.7 ± 0.3h, respectively [37].!
A R T I C L E H I S T O R Y
Received: April 29, 2019
Revised: May 23, 2019
Accepted: May 23, 2019
DOI:
10.2174/1871520619666190705150214
2 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19 , No. 0 Vinayak and Maurya
3. STRUCTURE AND SOLUBILITY OF QUERCETIN
Quercetin (3, 3’, 4’, 5, 7-pentahydroxyflavone) is composed of
3, 5, 7-trihydroxy-4H-1-benzopyran-4-one (A and C) and a 3, 4-
dihydroxy-phenyl ring (B). The biochemical activity of quercetin is
due to the presence of hydroxyl groups which exert antioxidant
activity by scavenging free radicals. Quercetin possesses almost all
structural elem ents with characteristics of an antioxidant lik e (i) an
orthodihydroxy or catechol group in ring B (ii) a 2, 3-double bond
and (iii) 3- and 5-OH groups with 4-oxo group. Several factors
including pH, heat, and metal ions affect the chem ical stability of
quercetin [22, 38]. Quercetin is a lipophilic compound (log P = 1.82
+/- 0.32), and it is moderately soluble in ethanol and highly soluble
in DMSO. However, its solubility in water is only approximately
0.01 mg/mL at room temperature [39]. The half-life of quercetin is
approximately 11-24 h. It is th erefore difficult to directly incorpo-
rate high levels of quercetin into water-based food matrix and needs
to improve its solubility, bioaccumulation, and delivery into the
cellular system to target certain diseases like cancer [40].
Chemical structure of quercetin (C15H10O7).
4. ANTICANCER ACTIVITY OF QUERCETIN
Anticancer activity of quercetin has been widely studied in
several in vivo and in vitro tumor models including lymphoma [11,
41-44], hepatocellular carcinoma [15], breast cancer [17, 45-47],
and colorectal can cer [48, 49]. Nanocarriers such as polymeric
nanoparticles, liposome, lipid nanoparticles, and micelles have been
deliberate to reduce the side effects of drugs. Co-encapsulation of
quercetin and vincristine with lipid-polymeric nanocarriers has
potential as a novel therapeutic approach to overcome chemo-
resistant lymphoma [42]. Quercetin and its natural glycosides are
known to decrease cell proliferation and induce genetic instability
[50, 51]. Quercetin is a Multi-Drug Resistance (MDR) modulator
and thus a potential chemosensitizer [52]. It is involved in reversion
of MDR via transporter-mediated active efflux by modulating P-gp
protein (glycoprotein encoded by the human MDR1 gene) and
MRP1 (Multidrug Resistance Protein). Quercetin has been shown to
improve its therapeutic efficacy in combination with drugs against
different types of cancer. Combinatorial effect of quercetin and
resveratrol has been found to be antiproliferative against HT-29
colon cancer via induction of zinc finger protein ZBTB10 and sup-
pression of oncogenic microRNA [53]. A recent study demon-
strated that quercetin improves the synergistic anticancer efficacy in
combination with PI-103, rottlerin, and G0 6983 in MCF-7 and
RAW 264.7 cells [17]. The lower and higher dose of quercetin
(0.1% or 1% quercetin, respectively) enhanced the anti-tumor ef-
fects of Trichostatin A (TSA) in mice [54]. The combination of
quercetin with gemcitabine has been shown a synergistic effect in
the inhibition of pancreatic cancer cells [55].
Targeting metastatic expansion by cell survival and prolifera-
tion in murine T-cell lymphoma (Dalton’s lymphoma), a fast-
growing and highly metastasized cancer, is a cru cial target of can-
cer therapy. We highlighted a multi-factorial approach of quercetin
towards lymphoma prevention in mice via modulation of cell cycle
proteins, oncogenes, and tumor suppressor genes. Quercetin is re-
ported to modulate various signaling molecules in the regulation of
cell proliferation, apoptosis as w ell as angiogenesis towards the
suppression of murine T-cell lymphoma [3, 11, 41, 43]. We re-
ported the first morphological evidence to show regression in
Dalton’s lymphoma growth in mice by quercetin treatment along
with improvement in longevity [41]. Being nontoxic to Dalton’s
lymphoma bearing mice, quercetin exhibits cytotoxic potential to
ascites cells [11, 41]. Cancerous cells avoid apoptosis or pro-
grammed cell death and promote uncontrolled cell proliferation via
a series of cellular events. Apoptosis is induced by extrinsic and
intrinsic pathways. Cellular toxicity induces the intrinsic pathway
of apoptosis via mitochondrial activation leading to caspase 9 cas-
cade, and extrin sic pathway via receptor-mediated caspase 8 signal-
ing cascade as well as via activation of NF-kB which regulates a
large number of inflammatory genes [56, 57]. Mitochondrial induc-
tion of apoptosis by quercetin has been reported in various cell
lines, including nasopharyngeal carcinoma, hepatocellular carci-
noma, breast cancer, leukemia, and oral squamous carcinoma [11,
15, 58-62]. Quercetin enhances the expression of BAX and Cyt C
release as well as modulates mitochondrial membrane potential in
HaCaT keratinocytes, MDA-MB-231 breast cancer and epidermoid
carcinoma KBv200 [63-66]. Quercetin-induced apoptosis via
upregulation of p21WAF1/CIP1 and Cdc2-cyclin B1 complex has
been reported in MCF-7 breast cancer and KYSE-510 squamous
cell carcinoma [67, 68]. W e reported that quercetin suppresses cell
survival and promotes apoptosis via differential localization of
TNFR1 as well as improved activity of caspase 9 in ascite cells of
Dalton’s lymphoma bearing mice [11]. It is associated with an in-
crease in active caspase 3, a critical executioner of apoptosis by
partial or total cleavage of PARP [11]. PARP is one of the main
cleavage targets of caspase 3 in vivo; however, it is cleaved by dif-
ferent caspases in vitro. Quercetin also induces apoptosis via activa-
tion of the reactive oxygen species-dependent ASK1p38 pathway
[69]. Quercetin inhibits TRPM7 channel and MAPK signalin g
pathway in gastric cancer [70]. It increases the levels of JNK and
p53-dependent Bax in BEAS-2B cells [71]. The p53-dependent
apoptosis by quercetin is reported in lung cancer A549 cells and
cervical cancer Hela cells [72, 73]. Quercetin increases ERK-
mediated cell death in HL-60 cells, suppresses Twist via p38MAPK
signaling, and causes mitochondrial depolarization through down-
regulation of IL-6/STAT3 signaling [74, 75]. Signaling mechanism
of anticarcinogenic activity of quercetin is mainly confined to its
antioxidant and proapoptotic role via regulation of p53, caspases,
cell cycle arrest, angiogenesis via COX-2, MMPs. Quercetin at-
tenuates P I3K-AKT signaling pathway in T-cell lymphoma exposed
to hydrogen peroxide [3]. Quercetin attenuates prostate cancer by
inhibiting anti-apoptotic and survival signaling [76]. Warburg’s
effect demonstrates the necessity of aerobic glycolysis or glycolytic
metabolism to provide enough energy for maintenance of cancer
growth. Quercetin decreases glycolytic metabolism by lowering
LDH-A activity in ascites cells of Dalton’s lymphoma bearing
mice, causing their increased susceptibility to death [41]. Glycolytic
metabolism maintains acidic pH in the tumor microenvironment,
which facilitates cancer growth. Acidic pH stimulates disruption of
adherence junctions, metastatic potential, proteinase activity, and
drug resistance.
Quercetin attenuates the major survival signals, like PI3K,
AKT, and ERK in carcinoma HepG2 cells [77]. PI3K is implicated
in cell surviv al, proliferation, and angiogenesis via activation of
AKT. Quercetin treatment leads to inactivation of PI3K suggesting
its role in the inhibition of ascite cell survival of lymphoma bearing
mice [41, 78]. Declined phosphorylation of BAD by quercetin in
lymphoma suggests modulation of PI3K-PDK1-AKT-BAD signal-
ing pathway by quercetin in regression of cell survival [44, 79].
Apart from this pathway, BAD is also modulated via ERK activated
p90 ribosomal S6 kinase. GSK-3β is another downstream effector
of AKT which modulates a variety of functions including cell sur-
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 3
vival and growth metabolism. Inactivation of GSK-3β after phos-
phorylation by AKT promotes cell proliferation, whereas dephos-
phorylation of GSK-3β promotes apoptosis and suppresses tumor
growth [80-82]. We reported a declined phosphorylation of GSK-
3β by quercetin suggesting inhibition of cell proliferation as well as
NF-κB mediated cell survival in Dalton’s lymphoma ascite cells
[44]. AKT also activates mTOR and promotes cell survival by in-
creasing mTOR-dependant nutrient uptake [82, 83]. Hyperactivated
PI3K-AKT-mTOR signaling has been well documented in a wide
range of cancer [3, 84-86]. AKT has been identified as master regu-
lators of IKK activity which activates NF-κB [87]. Oxidative stress
is one of th e major causes of NF-κB activation. Antioxidant quer-
cetin inhibits cell survival by downregulating the phosphorylation
of IκBα and by declining survival factor NF-κB in DL ascite cells.
Quercetin has been shown to inhibit aflatoxin B1-induced hepatic
damage in mice [88]. It protects against liver injury induced by
alcohol in rats. Dietary supplement of quercetin inhibits the devel-
opment of the carcinogen-induced rat mammary cancer, colonic
neoplasia, and oral carcinogenesis. PI3K-AKT-BAD pathway is
deactivated by PTEN, a phosphatase which regulates the level of
PIP3 by its de-phosphorylation. Enhanced level of tumor suppressor
PTEN by quercetin supports the decreased activity of PI3K and
subsequent signaling pathway. PTEN is frequently lost or mutated
in cancer. Induced level of PTEN by quercetin is correlated with
upregulation of tumor suppressor p53, a principal mediator of
growth arrest, senescence, and apoptosis in response to a broad
array of cellular damage. AKT mediated oncogenic activation acts
as an anti-apoptotic signal via rapid destabilization of p53 [89-91].
We reported the anticancer effect of quercetin via induced mRNA
expression and protein level of p53 [41]. Cancer preventive effect
of quercetin is also validated against fatty acid synthase in liver
cancer [92]. Quercetin downregulates the Notch / AKT/mTOR sig-
naling pathway in U937 leukemia cell [73]. Quercetin attenuates
PI3K-AKT signaling pathway exposed to hydrogen peroxide in T-
cell lymphoma [3]. Quercetin attenuates AKT-mTOR pathway
mediated autophagy induction by inhibiting cell mobility and gly-
colysis in breast cancer [93].
Angiogenesis is an essential characteristic adaptation of tumor
growth for a continuous supply of glucose and oxygen as well as to
get rid of its waste products [94]. The angiogenic factor VEGF-A is
required for neovascularization and for angiogenic remodeling [95].
PI3K-AKT signaling is reported to increase VEGF-A secretion by
HIF-1 dependent, or by HIF-1 independent mechanism [95]. Down-
regulated level of VEGF-A by quercetin suggests its ability to re-
gress angiogenesis in Dalton’s lymphoma bearing mice via PDK1
[44]. PDK1 is vital to endothelial cell migration in response to
VEGF stimulation in a PI3K dependent manner. Further, overex-
pression of PTEN inhibits angiogenesis and tumor growth [44, 96,
97]. Loss of AKT1 is reported to reduce NO production and impair
tumor angiogenesis. The inflammatory enzymes iNOS and COX-2
are implicated in inflammato ry diseases and tumor progression
[98]. We have further reported that quercetin reduces inflammatory
response by decreasing the activity of iNOS and COX-2 in ascite
cells of Dalton’s lymphoma bearing mice as well as in HepG2 cells
[15, 44]. However, both promoting and deterring actions of iNOS
during tumor development have been reported, probably depending
upon the local concentration of NO within the tumor microenvi-
ronment [16, 99]. COX-2 and tumor suppressor p53 have been
reciprocally regulated by quercetin in the prevention of cancer [15].
Such reciprocal relation is reported earlier [100]. Hepatocellular
carcinoma is one of the major health threats and the third leading
cause among cancer-related deaths globally. Quercetin attenuates
cell survival and cell proliferation of HepG2 cells and elicits apop-
tosis by enhancing the expression of BAX and p53 through down-
regulation of ROS, PI3K, PKC, and COX-2 [15]. The independent
regulation of p53 by quercetin has been reported against hepatocel-
lular carcinoma [101].
5. BIOAVAILABILITY OF QUERCETIN
Nanoparticles have been serving as a tool to enhance the effec-
tiveness of phytochemicals by increasing bioavailability [102].
Digestion of quercetin in the human body occurs in the mouth,
small intestine, liver, and kidneys where it undergoes glucuronida-
tion, sulfation or methylation [8, 103-108]. Distribution of quercetin
after the intravenous and oral administration has been shown in rat.
It entails that the intake of quercetin from daily diet can lead to the
accumulation of quercetin throughout the body [109]. Bioavailabil-
ity of quercetin is related to its bio-accessibility. However, the pres-
ence of sugar moieties increases bioavailability and differences in
quercetin conjugated glycosides influence its bioavailability [110,
111]. Quercetin bioavailability increases when it is consumed as an
integral food component. The size and polarities of these com-
pounds can cause difficulty crossing membranes in the gut. How-
ever, the efficacy of qu ercetin is limited due to its hydrophobici ty,
poor gastrointestinal absorption, and instability in physiological
medium, extensive xenobiotic metabolism in liver and intestine via
glucuronidation or sulfation; which collectively contribute to low
oral bioavailability of quercetin [40, 112-114]. Bioavailability of
quercetin obtained from onion is more due to its greater absorption
[110]. Quercetin ingestion with brief chain fructooligosaccharide
progresses quercetin bioavailability [115]. Solid dispersions have a
greater surface area that promotes dissolution in the intestinal lu-
men, thereby promoting bioavailability of quercetin with cereal
ingredients [116]. Variations in gut microbiota metabolism, dietary
history, genetic polymorphisms, and interindividual variation play a
significant role in the bioavailability of quercetin [117]. Available
literature highlights that despite the tremendous anticarcinogenic
potential, quercetin could not show clinical utility. Efficient deliv-
ery of quercetin to target cells and its retention at the site is neces-
sary for its utilization as a therapeutical agent.
6. APPROACHES FOR QUERCETIN DELIVERY
Nanomedicines have broad research and application prospects.
There are more than 51 FDA approved nanomedicines and 77
products are under consideration of clinical trials [118]. A better
understanding of the molecular mechanism of controlled and regu-
lated drug delivery is essential for the development of novel and
effective therapies. Nanotechnology provides a method to create
novel formulations for numerous hydrophobic drugs. Quercetin
delivery through nanoparticles has pulled in much consideration for
its improved anticancer potential and promising clin ical application
[40]. Utilization of delivery systems including nanoparticles, lipid-
based carriers, micelles, inclusion complexes, and conjugates-based
encapsulation has the potential to improve both stability and
bioavailability and thus imposes health benefits of quercetin. These
delivery carriers are widely used for enhancing drug solubility in
water, absorption, circulation, retention time, and ultimately target
specificity.
Quercetin-containing self-nanoemulsifying drug delivery sys-
tem has improved oral bio availability [112]. Curcio et al. showed
the suitability of molecularly imprinted nanospheres for con-
trolled/sustained release of quercetin using methacrylic acid and
ethylene glycoldymethacrylate as a functional monomer and cross-
linking agent, respectively. The antiproliferative activity of quer-
cetin has maintained without interfering with the cell viability
[119]. Semi-engineered water-soluble isoquercetin and maltooligo-
syl isoquercetin are produced by manufactured glycosylation to
defeat solvency challenges and restorative methodologies. Liposo-
mal quercetin has significantly improved the solubility and
bioavailability of quercetin in MCF-7 breast cancer cell [120].
Regulated metabolism for nanosponge conjugated quercetin has
been demonstrated in breast cancer cells [121]. These delivery sys-
tems are generally designed to efficiently encapsulate an apprecia-
ble number of functional components to protect them against the
4 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19 , No. 0 Vinayak and Maurya
Table 1. Molecular and bioche mical target of anticancer effects of nanoconjugated quercetin.
Sr.
No.
Delivery
System
Advantages
Effects/ Target s
Refs.
Quercetin shows resistance to acidic conditions and promoted
the release in alkaline pH mimicking the intestinal environment
pH-sensitive r elease beh avio r and promising anticancer activ ity
against HeLa cells
Contro lled release, pr eventing q uercetin degradation, strong
antioxidant activity
[125]
[126]
[127]
Inhibition of anti-inflammatory activities in terms of COX-2
and NF-κB in MCF-10A cells
[128, 129]
Improved bioavailability and cellular uptake of quercetin in
MCF-7
[120]
Enhanced cellular uptake and effective nanocarrier for drug
delivery to a brain tumor
[63]
Improved intestinal absorption of quercetin
[130]
1
Liposomes
Transporters for both
lipophilic and hydr o-
philic
particles, targetability
Declination of cyclin D1, NF-κB, histone deacetylase 1 and
induction of caspase 3 in esophageal sq uamous cell carcinomas
Efficient antitumoral agent against glioma cells
[131]
[30]
Inhibition of bacterial propagation
[24]
Reduction in cytotoxicity, oxidative stress as well as improved
metabolic activity of Caco-2 cells
[132]
2
Silver
nanoparticles
High reactivity,
powerful antimicro-
bial activity, and low
propensity to induce
bacterial resistance
Reduced cell viability of DL cells
[133]
Sustained rele ase of quercetin as well as decreased cell viability
of breast cancer cells MDA-MB231
[134]
Improved cytotoxicity, cellular uptake and delivery o f quercetin
to SKBR3 breast cancer cells
[135, 136]
Improved cytotoxicity of quercetin on HaCaT cells and A431
cells
[137]
Inhibits the activity and expression of P-glycoprotein in MCF-
7/ADR cells
[138]
Induced apoptosis in HepG2 cells
[139]
Growth inhibition of HepG2 cells
[140]
Improved cell cytotoxicity, apoptosis, and reduc ed hi stone
deacetylases, reduced cell proliferation of HepG2 cells and
modulation of p21, cdk1, and AKT
[141]
3
Poly lactic-co-
glycolic acid
(PLGA)
Good biocompatibil-
ity, biodegradable
polymer, better load-
ing, and encapsulat-
ing efficacy
Significant increase in cytoto xicity on human lung adenocarci-
noma epithelial cells A549
[142]
Improved apoptosis and cell growth inhibition in CT26 cells
[139]
Induced apoptosis as well as enhanced permeability and reten-
tion in PC-3 cells
[143]
Increased cellular uptake of quercetin and longer half-life in
MCF-7
[52]
Increased cytotoxicity of MCF-7 and growth inhibition on H22
tumor-bearing mice
[144]
Induced apoptosis, mitochondrial transmembrane potential
change, decreased phosphorylation of p44/42 and AKT in
A2780S cells
[39]
Improved drug accumulation in A549 cancer cell line and
murine xenograft model
[145]
Increased IC50 values in MCF-7 cells
[146]
4
Polymeric
micelles
Small size (com-
monly <10nm),
thermodynamic
dependability, colloi-
dal dependability
Enhanced delivery and inhibition of growth of human erythro-
myelogenous leukemia cells (K562) and small lung carcinoma
cells (GLC4))
[147]
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 5
chemical or thermal degradation. Qu ercetin can be released at a
controlled rate and at the site of action or within a region of the
gastrointestinal tract [122, 123]. Therefore, the development of
nano-conjugated quercetin via nano-based carriers has gained much
attention for the improvement of delivery and bioavailability of
quercetin.
7. NANO-CONJUGATED QUERCETIN
Nanoparticle drug delivery system has increased impressive
consideration throughout the decad es. Therapeutics can be captured
in the nanoparticles to enhan ce bioavailability, to improve watery
dissolvability, to build dependability, and to control delivery [1, 16,
124]. These delivery vehicles are broadly pondered in translational
application for the revolution of anticancer action of quercetin
(Table 1).
Nanomedicine delivery system is preferred in cerebrum tumor
treatment utilizing gliom a-particular nanoparticles [148] and fer-
rocenyl complex (ansa-FcdiOH) through stealth Lipid Nanocap-
sules (LNCs) [149] because of their tunable physicochemical prop-
erties. Quercetin stacked self-nanoemulsifying drug conveyance
framework (QT -SNEDDS) advances cell reinforcement movement
of quercetin against breast cancer [150]. Oral organization of quer-
cetin stacked polymeric nanocapsules (N1QC) has been accounted
for expanded cerebrum take up and mitochondrial restriction [151].
Additionally, the enhanced calming impact of Zein nanoparticles
implanted quercetin was shown in the mouse model of incited en-
dotoxemia [152]. Improved bioavailability of quercetin conjugated
from almond gum nanoparticles without any toxicity has been stud-
ied in Caco-2 cells [153]. Specifically, nanoparticles have been
broadly explored to deliver anticancer drugs by methods for passive
targeting. The higher amount of nano-sized particles passively per-
meates through the leaky tumor blood vessel than the tight capillary
in healthy tissues [154]. Together with the insufficiency of the lym-
phatic arrangement of tumors, the nanoparticles are accumulated
and held longer in the tumors. This phenomenon is the so-called
Enhanced Permeability and Retention (EPR) effect. In this manner,
the polymeric nanoparticles are utilized to specifically deliver anti-
cancer drugs to the tumors. Nanoconjugated quercetin prompts EPR
through the dual mood of delivery i.e., active or passive transport.
Subsequently, it enhances drug release, solubility & stability, pro-
tection against endosomal degradation, inhibition of MDR as well
as absorption of free quercetin in the blood vessel which leads to
DNA fragmentation and programmed cell death or apoptosis at
metastatic sites as depicted in Fig. (1). Encapsulation of anti-cancer
agent in the nanoparticle is recognized as a novel strategy to over-
come resistance through several mechanisms including increasing
drug penetration into cancer cells, modulation of drug release, and
high endocytosis phenomenon. Several approaches have been gen-
erated to increase th e effi cacy of quercetin for cancer treatment
with a unique advantage and drawback of each formulation. To
overcome the burden or limitations of accessibility, quercetin can
be delivered through polymeric micelles [114, 155], liposomes [63,
129, 156], chitosan nanoparticles [125], silver nanoparticles [24],
PLGA (Poly lactic-co-glycolic acid) [137], PLA (poly(D,L-lactic
acid)) nanoparticles, and silica nanoparticles. In addition, delivery
systems may likewise shield the bioactive compounds from being
enzymatically met abolized or therm al/ light degradation and expan d
their stability [157]. Quercetin stacked solid lipid nanoparticles
enhance osteoprotective activity in po stmenopausal osteoporosis
[158]. Quercetin based solid lipid nanoparticle treatment recovers
bone loss over quercetin treatment group in ovariectomized rats.
Co-delivery with nanoconjugated quercetin enhances doxorubicin
mediated cytotoxicity against MCF-7 breast cancer cells. Phyto-
some technology can improve the efficacy of chemotherapeutics
potential of anticancerous drugs by increasing the permeability of
tumor cells. Poly (β-amino esters) polymers have emerged as highly
promising candidates for biomedical and drug delivery applications
Fig. (1). Schematic diagram showing targeting of metastatic cancer by polymeric quercetin. Nanoconjugated quercetin leads to Enhanced Permeability and
Retention (EPR) via dual mood of delivery i.e., active or passive transport. Subsequently, it enhances drug release, solubility & stability, protection against
endosomal degradation, inhibition of MDR as well as absorption of free quercetin in the blood vessel which leads to DNA fragmentation and programmed cell
death or apoptosis at metastatic sites.
6 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19 , No. 0 Vinayak and Maurya
due to their pH sensitive, tuneable, and degradable properties.
These polymeric systems can serve as pro-drug carriers for the
delivery of bioactive compounds which are biologically unstable
and suffer from poor aqueous solubility, low bioavailability such as
the antioxidant quercetin [155].
7.1. Liposomes
Among different approaches to achieve intestinal drug delivery,
the use of natural polymers holds clinical promises. Oral admini-
stration of polymer-coated liposomes has been proposed for the
targeted delivery of drugs to the inflamed intestinal mucosa [159].
Liposomes have attractive biological properties including general
biocompatibility, biodegradability, and ability to encapsulate both
hydrophilic and hydrophobic therapeutic agents. Encapsulation of
hydrophobic drug into liposome can create aqueous intravenously
injectable formulations. In the meantime, liposomes can regulate
drug release and protect the encapsulated drugs from undesired
effects of extern al conditions. Doxil is the first lipo somal drug for-
mulation approved by the Food and Drug Authority for the treat-
ment of AIDS associated with Kaposi’s sarcoma. Encapsulation of
quercetin by liposome enhances quercetin solubility and improves
anticancer activity through longer exposure to cancer cells with a
high drug concentration. Cancer therapy with a combination of
drugs is an important strategy in clinical use. Liposomal quercetin
enhances the sensitivity of cancer to thermotherapy and thermo-
chemoth erapy [64]. Liposome conjugated quercetin inhibits th e
upregulation of hsp70 and enhances apoptosis induced by hyper-
thermia and thermochemotherapy. However, systemic administra-
tion of liposomal quercetin could sensitize CT26 cells to ther-
motherapy and chemothermotherapy. Liposomal quercetin demon-
strates the most effective tumor growth inhibition in the JIMT-1
human breast tumor xenograft as compared to monotherapy and
free vincristine/quercetin combinations [160]. The most widely
used strategy is to develop controlled drug released liposomes by
coating the surface with inert and biocompatible polymers such as
Polyethylene Glycol (PEG). PEG is an exceptionally hydrated
adaptable polymer chain that can enhance retention time by stabiliz-
ing and shielding liposome from opsonisation. Moreover, PEG is
non-toxic and non-immunogenic, making it su itable for clinical
applications. Liposomal quercetin is highly accumulated and has
long retention in liver, tumor, and spleen rather than kidney and
lung. In addition, liposomal quercetin demonstrated significant
tumor growth inhibition activity in vivo [64]. Many cancer active
targeting liposomes are developed to deliver quercetin and to fur-
ther improve the accumulation of quercetin in cancer cells. Liver-
specific liposomal quercetin is prepared by galacto sylation. Target-
ing the galactosyl receptors on th e surface of hepatic cells allowed
liver-specific delivery of quercetin, wh ereas intravenous admini-
stration of galactosylated liposomal quercetin demonstrated higher
inhibitory activ ity on the development of hepatocarcinoma and on
oxidative damage in rat liver as compared to free quercetin [156].
Customary liposomes can be barely utilized, due to their resis-
tance to gastric pH, enzymatic debasement, biocompatibility, bio-
degradability, biorenewability, and bioadhesion but they can be
effortlessly ensured by a polymeric coating [161]. Chitosan is util-
ized to coat polyethylene glycol containing vesicles for controlled
delivery of quercetin in colon cancer [159, 161]. Cross-linked chi-
tosan-lipo some combination sy stem speaks to a promising combi-
nation of nanovesicles and for delivery of quercetin into the diges-
tive system [125]. Quercetin loaded chitosan-quinoline nanoparti-
cles display pH-sensitive release behavior and promising anticancer
activity against HeLa cells [126]. The quercetin loaded fu-
coidan/chitosan nanoparticles has shown to exhibit controlled re-
lease, preventing quercetin degradation, strong antioxidant activity,
and increasing its oral bioavailability [127]. Quercetin conjugated
liposomes have been utilized to provide quercetin in rodent brain
[63, 129]. Piperine is among different ingredients joined with quer-
cetin liposomes to instigate the delivery of quercetin and increase
its retention in the small intestinal su rface. A lginate, an anionic
biopolymer can expand encapsulation proficiency of liposome.
Formulations of nano-sized quercetin liposomes inhibit the
expression of COX-2 and NF-kB in MCF-10A Cells [128]. The
nano-sized quercetin encap sulated by liposomes enhanced the cellu-
lar uptake in MCF-7 breast cancer cells [120]. These polymers
penetrate the polar medium inside the cells and to protect them
against the highly toxic effect induced by cum ene hydroperoxide.
Liposomes build up especially at tumor sites due to their ability to
extravasate through pores in the capillary endothel ium. These pores
seem to be important for rapid angiogenesis in tumors. Quercetin
liposomes and spherical nanoparticles consisting of a phospholipid
bilayer with an aqueous compartment, own the unique capability to
effectu ally deliv er both water-soluble chemical antioxidants in the
aqueous phase and lipid-soluble chemical antioxidants in the lipid
bilayer. Their surface can be conjugated with specific ligands that
target liposomes to their location of action. The size, composition,
surface charge, and other formulation possessions of liposomes is
well documented to encounter the necessities of specific conditions.
Additionally, it is believable that gene therapy medications might
be delivered by liposomes. Nanoparticles with entrapped siRNA
inhibit bacterial propagation [24, 162]. Quercetin loaded magnetol-
iposomes are designed to study as a stable and efficient antitumoral
agent against glioma cells [30].
7.2. Silver Nanoparticles
Silver nanoparticles (AgNPs) have numerous applications in
healthcare, food packaging and conservation, and household mate-
rials. With their increasing use, human exposures to silver nanopar-
ticles become the current concern with potential unexpected health
complications mainly due to their high reactiv ity. The toxic effects
of silver nanoparticles on human cells and the environment have
been extensively studied [163]. Combined treatment with gemcit-
abine and AgNPs caused increased cytotoxicity and apoptosis in
A2780 cells [163]. However, normally silver toxicity has been well
detoxified by small periplasmic silver-binding proteins, which bind
silver at the cell surface and by efflux pumps propels the incoming
metals and protects the cytoplasm from toxicity. The organic matrix
contains silver binding proteins that provide amino acid moieties,
which serve as nucleation sites for the formation of silver nanopar-
ticles. Silver nanoparticles are the mo st widely explored nano-
agents because of their broad antimicrobial spectrum with powerful
antimicrobial activity and low propensity to induce bacterial resis-
tance [25]. AgNPs provid e extremely attractive scaffolds for the
creation of transfection agents as they exhibit several advantages
including bioinertia, ready synthesis, and easy functionalization
[138, 164-166]. Quercetin conjugated with silver nanoparticles
shows a synergistic effect on antibacterial activity [24]. However,
silver nanoparticles conjugated quercetin applies negligible impact
on numerous sorts of drug-resistant bacteria including Pseudo-
monas aeruginosa and Bacillus subtilis. Quercetin being a charac-
teristic natural antioxidant compound can possibly conjugate with
silver nanopart icles pertinent in the treatment of cancer. Numerous
parameters like reducing agents, temperature, concentration of salt
and microenvironment affect the conjugation of quercetin and silver
nanoparticles. Quercetin loaded AgNPs reduces cytotoxicity, oxida-
tive stress as well as increases the metabolic act ivity of quercetin in
Caco-2 cells [132]. Silver nanoparticles in conjugation with sele-
nium improve drug delivery of quercetin and reduce cell viability of
Dalton’s lymphoma cells [133].
AgNPs not only induce apoptosis but also sensitize cancer cells.
The anticancer property of starch-coated silver nanoparticles was
studied in normal human lung fibroblast cells (IMR-90) and human
glioblastoma cells (U251). AgNPs induced alterations in metabolic
activity, and increased oxidative stress leading to mitochondrial
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 7
damage and increased production of ROS, ending with DNA dam-
age. Among these two cell types, U251 cells show more sensitivity
than IMR-90 [167]. The cellular uptake of AgNPs occurred mainly
through endocytosis. AgNPs treated cells exhibited various abnor-
malities, including upregulation of metallothionein, downregulation
of major actin-binding protein, filamin, and mitotic arrest [167].
The analysis of cancer cell morphology suggests that biologically
synthesized AgNPs could significantly induce cell death. Silver
nanoparticles were used to target breast-cancer cells (SKBR3) and
leukemia cells (SP2/O). Chitosan-based nanocarrier (NC) delivery
of AgNPs induces apoptosis at very low concentrations [168].
Lower concentrations of nanocarriers with AgNPs showed better
inhibitory results than AgNPs alone. Chitosan-coated silver
nanotriangles (Chit-AgNTs) show an increased cell mortality rate.
The anticancer property of bacterial -AgNPs and fungal extract-
conjugated AgNPs (F-AgNPs) has been demonstrated in human
breast cancer MDA-MB-231 cells. Both biologically produced
AgNPs exhibited significant cytotoxicity. Recent studies indicate
cell-specific toxicity by AgNPs. Plant extract-mediated synthesis of
AgNPs showed more pronounced toxic effect in human lung carci-
noma cells (A549) than non-cancer cells like human lung cells,
indicating that AgNPs could target cell-specific toxicity, probably
based on the lower pH in the cancer cells [169]. AgNPs showed
significant toxicity in MCF7 and T47D cancer cells by higher en-
docytic activity than MCF10-A normal breast cell line [170]. The
nanoconjugated quercetin through silver nanoparticles can be em-
ployed in the treatment of tumor models via influen cing its cytotox-
icity, delivery, and targeting site. However, further preclinical and
clinical study is needed to establish a chemoth erapeu tic agent
against a certain form of can cer.
Food-related applications of silver nanoparticles need to gain a
better understanding of the cellular and molecular mechanisms
involved in their interaction with the cells of the gastrointestinal
tract as well as with gastrointestinal fluid s, food matrix, microflora,
etc. Therefore, the safety of nanoparticles in the food matrix and
cancer cells remains to be explored and further understood.
7.3. PLGA (Poly Lactic-Co-Glycolic Acid)
Combination of arginine-glycineaspartic acid - sorafenib- quer-
cetin (RGD-SRF-QT) nanoparticles could provide a promising
platform for co-delivery of multiple anticancer drugs for the
achievement of combinational therapy and could offer the potential
for enhancing the therapeutic efficacy on hepatocellular carcinoma
[140]. Anticancer efficacy of poly (lactic acid)-quercetin nanoparti-
cles has been reported for sustained release kinetics revealing novel
vehicle for the treatment of breast cancer [134, 171]. Quercetin-
loaded poly (lactic-co-glycolic acid)-d-α-tocopheryl polyethylene
glycol succinate nanoparticles could be used as a potential intrave-
nous dosage for treatment of liver cancer owing to the enhanced
pharmacological effects of quercetin with increased liver targeting
[139]. Tamoxifen (Tmx) embedded PLGA nanoparticles (PLGA-
Tmx) is prepared to evaluate its better DNA cleavage potential,
cytotoxicity using Dalton’s lymphoma ascite cells and MDA-
MB231 breast cancer cells. PLGA-Tmx shows excellent DNA
cleavage potential as compared to pure Tamoxifen raising better
bioavailability. Sustained-release kinetics of PLGA-Tmx nanoparti-
cles shows mu ch better anticancer efficacy through enhanced DN A
cleavage potential and nuclear fragmentation and, thereby, reveals a
novel vehicle for the treatment of cancer [134]. Poly (e-
caprolactone) (P(CL)) is one of the biodegradab le and biocompati-
ble polyester polymers. The cellular uptake of P (CL)-TPGS
nanoparticles by SKBR3 cells is reported through cholesterol-
dependent endocytosis. The P (CL)-TPGS nanoparticles show im-
proved toxicity and uptake efficiency and could be potentially used
for the delivery of quercetin to breast cancer cells [136]. The retar-
dation of drug release from the nanoparticles depends on tempera-
ture and crystallin ity of the polymer. However, other factors must
be considered such as the compatibility and interaction of polymer
and drug [135]. A few studies hav e established the investigation of
P (CL)-TPGS copolymers with and without other co-monomers for
the delivery of genistein, paclitaxel, and TRAIL/endostatin [172-
176]. Quercetin-PLGA nanoparticles can be used as effective drug
delivery systems for skin cancer treatment encompassing natural
drugs [137].
Metastatic breast cancer is the fundamental driver of death from
breast cancer. We have reported the efficacy of trans-copper (II) β-
dithioester complexes and homoleptic zinc (II) complexes ag ainst
breast cancer [177, 178]. However, medicines are not focused on or
successful at this stage probab ly because of the presence of Breast
Cancer Stem Cells (BCSCs). The propinquity of BCSC is the criti-
cal explanation behind resistance and failure of therapy. As BCSC
originates from normal breast stem cells and having self-renewal,
high proliferation rate, ability to generate heterogeneity, etc. fur-
ther, Mesenchymal to Epith elial Transition (MET) is important for
invasion, intravasation, circulation and extravasation; and ulti-
mately leads to colonization of metastatic cells. Afterward, Lungs,
bone, liver, and brain are the main site of metastasis for breast can-
cer. Nanoconjugated quercetin in this regard has attracted much of
interest du e to effective therapeutic approaches against various
molecular targets during breast cancer metastasis in vitro as well as
in vivo system [120, 134, 157, 179-185] as depicted in Fig. (2). The
combination of a chemotherapeutic drug with a chemosensitizer has
emerged as a promising strategy for cancers showing multidrug
resistance. The improved synergistic efficacy of doxorubicin with
Boron Nitride Quantum Dots (BNQDs) has been reported in terms
of anticancer activity via DNA cleavage, ROS accumulation, and
interaction of doxorubicin-BNQDs with DNA in MCF-7 cells [186].
Biotin conjugated poly (ethylene glycol)-b-poly(ε-caprolactone)
nanoparticles encapsulating the chemotherapeutic drug doxorubicin
and the chemosensitizer quercetin (BNDQ) has a potential role in
the treatm ent of drug-resistant breast cancer [138]. BNDQ is more
effectively taken up with less efflux by doxorubicin-resistant MCF-
7 breast cancer cells (MCF-7/ADR cells) than by the cells treated
with the free drugs or non-biotin conjugated nanoparticles. BNDQ
exhibit ed clear inhibition of the activity and expression of p-
glycoprotein, an MDR marker in MCF-7/ADR cells [138]. Quer-
cetin-chitosan conjugate has been studied for oral delivery of
doxorubicin to improve its oral bioavailability by increasing its
water solubility, opening tight junction and bypassing the P-
glycoprotein in Caco-2 cells [118]. Quercetin loaded within
poly(lacticcoglycolic acid) biodegradable nanoparticles, plays
a major role in drug t argeting to tumors due to its ability to interact
with CD44 receptor [55]. Chitosan is used to coat polyethylene
glycol containing vesicles for controlled delivery of quercetin in
colon cancer [159]. Cross-linked chitosan-liposom e fusion system
represents a promising combination of nanovesicles and for delivery
of quercetin into intestine [125]. Quercetin conjugated liposomes
have been used to deliver quercetin in rat brain [129].
7.4. Polymeric Micelles
Polymeric micelles have attracted enormous attention with suc-
cess in clinical studies of cancer targeting. Polymeric micelles are
constructed through self-assembly of block copolymers as nano-
scaled drug carriers. These micelles have a core-shell structure
where the drug-loaded core is encompassed by a bio compatible
PEG shell of 10-100nm [187]. Engineering the micelle-forming
block copolymers endowed polymeric micelles with smart on-
demand functionalities such as environment-sensitivity and target-
ability. Polymeric m icelles have been considered as the most prom-
ising nano-carriers because their critical size, drug incorporation
efficiency, stability, and release rate can be squeezed by setting up
the basic block copolymers. Utilization of polymeric micelles has
been reported in some tumor models and in some clinical studies
[188]. Colorectal cancer developing in the large intestine has been
8 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19 , No. 0 Vinayak and Maurya
the third leading cause of cancer-related death globally. The en-
hanced delivery by quercetin conjugated monomethoxy poly (ethyl-
ene glycol)poly(ε-caprolactone) MPEG-PCL nanomicellar system
in the CT26 tumor model, indicates a promising potential applica-
tion against colorectal tumor chemotherapy [114]. Further, quer-
cetin loaded nanomicelles significantly increase drug accumulation
at the tumor site and exhibit superior anticancer activity in prostate
cancer [143].
Biodegradable polyesters such as poly (D, L-lactide-co-
glycolide), poly (D, L-lactide), poly (ecaprolactone), and long-chain
alkyl derivatives appear to be widely used as a core-forming poly-
mer [189, 190]. The utilities of polymeric micelles have been ac-
counted for in experimental tumor models in mice as well as in
clinical examinations. In this manner, polymeric micelles can un-
derstand protected and successful treatment, and offer personalized
meds for individual patients. Various molecular interactions such as
hydrophobic interactions, hydrogen bonding, electrostatic interac-
tion and metal complex formation in the core-forming segments can
be a driving force of the formation of polymeric micelles [191-193].
A wide range of therapeutic molecules including hydrophobic sub-
stances, charged compounds, and metal complexes can be stably
and efficiently incorporated into the micellar core, and their release
can be controlled in a sustained or environment-sensitive manner.
Several micelles formulations of anticancer drugs are currently
under evaluation in preclinical and clinical studies [187]. Novel
nanoparticles containing polymeric microspheres loaded with pacli-
taxel and quercetin utilize as a promising pneumonic delivery sys-
tem for combined chemotherapy [194].
Because of characteristic core-shell structures and limited size
dispersions in the scope of 10-100nm, the drug-loaded center of
polymeric micelles can hinder interaction with plasma proteins and
cells, avoiding the recognition of the micelle by the reticulo-
endothelial system in the bloodstream [195]. Th erefore, polymeric
micelles can display prolonged circulation with a half-life longer
than 10h. The polymeric micelles separate into the constitu ent
square copolymers, the extent of which is beneath the edge of
glomerular discharge, in this way keeping away from long haul
amassing in the body. Long-circulating polymeric micelles success-
fully aggregate in large tumor because of the expanded brokenness
of tumor neo vasculature and impeded lymphatic seepage. Subse-
quently, polymeric micelles release incorporated drugs in a sus-
tained or microenvironment responsive manner [187, 191]. In this
manner, polymeric micelles can accomplish tumor-specific drug
activity while limiting side effects in normal tissues [187]. Contin-
gent upon the definitions, polymeric micelles are masked by malig-
nancy cells, and after that apply the impact of drug in an organelle-
specific way [191]. In such a w ay, polymeric micelles potentially
circumvent drug efflux or intracellular detoxification mechanisms,
overcoming drug resistance in cancer cells [196]. Encapsulated
polymeric micelle in conjugation with curcumin attenuates the pro-
gression of colon cancer. Polymeric micelles are formed from vari-
ous block copolymers. Poly (ethylene glycol) is generally utilized
as a shell-shaping polymer because of its hydration property and
large excluded volume impact averting cooperation with seru m
proteins. The micellar core is composed of a variety of synthetic
polymers, which critically affect the critical prop erties of polymeric
micelles as drug vehicles, including size, association number, criti-
cal micelle concentration, drug loading and release, and stability in
the bloodstream [187]. Dev elopment of α-helix of poly (L-
glutamate) may incredibly add to delayed blood distribution and
accumulation of cisplatin-stacked micelles in tumors under stage III
clinical assessm ent [197]. The technique for drug incorporation can
be characterized into 'non-covalent' and 'covalent' convention. In
Fig. (2). Targeting metastasis breast cancer through nanoconjugated quercetin. Metastasis breast cancer is the fundamental driver of death from breast cancer.
Medications are not fo cused on or successful at this stage might be because of the presence of Breast Cancer Stem Cells (BCSCs). The propinquity of BCSC is
the critical explanation behind resistance and failure of therapy. BCSC originates from normal breast stem cells and having self-renewal, high proliferation
rate, ability to generate heterogeneity, etc. EMT is impo rtant for invasion, intravasation, circu lation, and extravasation; and ultimately leads to colonization of
metastatic cells by Mesenchymal to Epithelial Transition (MET). Lungs, bone, liver, and brain are the main site of metastasis for breast cancer. Nanoconju-
gated quercetin in this regard has attracted much of interest due to effective therapeutic approaches against various molecular targets in metastasis breast can-
cer in vitro as well as in vivo sy stem.
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 9
the non-covalent drug stacking technique, water-insoluble com-
pounds are physically ensnared into the micelle center by dialysis,
ultrasound-helped scattering or as oil in water emulsion. A rela-
tively high drug loading capacity of approximately 20% can be
achieved without a chemical modification of drug molecules. For
successful drug incorporation, the similarity between a drug mole-
cule and th e core shaping fragments should be comp ared. Addition-
ally, the properties of the center shaping sections, for example,
hydrophobicity, the glass transition temperature, level of crystallin-
ity, and optional structure (α-helix development) are vital as they
fundamentally influence the productivity and limit of drug stacking
and its discharge conduct.
Drug molecules are chemically conjugated to the core-forming
segments in the covalent drug loading system. For drug conjuga-
tion, the environment-responsive cleavable linkage is mistreated to
guarantee the drug release at the target site [191]. Because the tu-
mor microenvironment is known to develop an acidic condition due
to the production of lactate by predominant anaerobic glycolysis in
cancer cells (Warburg effect), the corrosive cleavable linkage, for
example, hydrazone bond is valuable for tumor-particular drug
discharge [191]. The PEG-β-PAA copolymers are helpful for on-
request drug incorporation because of the opportunity of the choice
of amino acids and flexible side chain adjustment.
7.4.1. DOX Conjugated Micelles
The ‘non-covalent’ drug incorporation through chemical conju-
gation of doxorubicin (DOX) to the side chain of PAA by a stable
amide linkage resulted in loss of cytotoxic movement of conjugated
DOX yet stabilized the physical capture of free DOX through the p-
p bond between the anthracycline structures of conjugated and un-
conjugated drugs [188, 198]. In this way, improv ement of micellar
core-forming portions relying upon drug molecule is possible. DOX
was conjugated through the hydrazone bond between the carbonyl
gathering at C13 of DOX and the hydrazide bunch acquainted with
poly (D, L-aspartate). The DOX conjugated square copolymers
shaped polymeric micelles, which indicated acidic pH-responsive
DOX discharge. Currently, the micellar formulation of a less car-
diotoxic epimer, epirubicin (code name NC-6300/ K-912) is under
phase I clinical study [199]. Co-delivery with nanoconjugated quer-
cetin upgrades DOX mediated cytotoxicity against MCF-7 cells.
Monoclonal antibody (MAb604.107) builds the affectability of
doxorubicin and shows higher affection for Notch1 in T acute lym-
phoblastic leukemia (T-ALL) [157].
7.4.2. Metal Conjugated Micelles
Notwithstanding hydrophilic particles, metal complexes incor-
porated PEG-β-PAA micelles are complexed with PEG-β-poly(L-
glutamate) through Pt(II)-carboxylate complex formation, leading
to the formation of narrowly distributed micelles with the size of 30
nm [187, 200]. In these frameworks, the reversible ligand exchange
reaction of Pt (II) allows the ideal influx of dynamic platinum com-
plexes from the micelles, warranting their intense cytotoxic activity.
After systemic administration, cisplatin-loaded micelles were re-
vealed to show prolonged circulation and effective tumor accumu-
lation, achieving remarkable in vivo antitumor efficacies with re-
duced side-effects. Currently, CDDP and DACHPt-loaded micelles
(code names NC-6004 and NC-4016) are under phase III and phase
I clinical studies, respectively [197]. Intracellular drug release using
the vehicles may enhance the drug poten cy. For example, N-(2-
hydroxypropyl) methacrylamide copolymer-DOX conjugate an d
pH-responsiv e DOX stacked polymeric micelles were accounted to
conquest the DOX-hindrance in cancer cells because of intracellular
drug release. Micelles accelerate drug release in low pH conditions
emulating the late endosomal condition. Confocal microscopic
observation uncovered that DACHPt loaded micelles accomplished
intracellular drug release under both in vitro and in vivo conditions
[196]. Thusly, DACHPt-stacked micelles d emonstrated excellen t in
vitro and in vivo antitumor activities against oxaliplatin-safe tumor
cells [196].
7.4.3. Nucleic Acid Conjugated Micelles
Nucleic acids-based drugs such as plasmid, antisense DNA, and
siRNA can be incorporated into polymeric micelles through polyion
complex formation between negatively charged nucleic acids and
positively charged PEG-β-PAA copolymers. Polymeric micelles
immensely enhan ce the stability of nucleic acids-based drugs under
in vivo conditions, prompting delayed blood circulation. The clini-
cally approved polyethylene glycol-coated (PEGylated) liposomes
such as Doxil and albumin nanoparticles named Abraxane have the
measure of roughly 100nm. These polymeric micelles are accumu-
lated in the tumor due to the EPR impact. Transport of nanoparti-
cles depends on th e origin of tumor cells and the microenvironment.
Solid tumors have a pore cut-off size bigger than 200nm apart from
some malignancies, for ex ample, glioblastoma (GBM). In such
manner, pancreatic tumors and diffuse-type gastric cancers (scir-
rhous gastric diseases) have trademark histological highlights by
less porous vasculature with pericyte scope and thick fibrosis,
which may be a hindrance to extravasation and intrusion of
nanoparticles [200]. Co-administration of 30 and 70nm micelles in
mice bearing tumors uncovered that 30-nm micelles demonstrate a
uniform intratumoral microdistribution while 70nm micelles indi-
cate heterogeneous limitation at perivascular dist ricts [200]. The
gathering and entrance of polymeric micelles in pancreatic malig-
nancy models rely upon their size, as the 30nm micelles can avoid
the difficulties in transvascular transport and in filtrate tumor
stroma. The 30nm DACHPt-stacked micelles demonstrated strik-
ingly drawn out survival in malignancy bearing mice and the com-
parable impact was seen in gastric tumor. The extent of polymeric
micelles is additionally vital for focusing on tumor metastasis.
Lymph nodes are regular courses for metastasis in a few tumors. It
is realized that nearby organization of nanoparticles to primary
tumors prompts collection in the neighboring lymph node; however,
the lymphatic vessels are clinically associated in the sentinel lymph
node biopsy [201]. The sentinel lymph node is defined as the first
lymph node or group of nodes draining metastasizing cancer cells
from the tumor. Lymph node metastasis is one of the most impor-
tant prognostic signs as it determines if cancer has spread beyond a
primary tumor into the very first draining lymph node or not. Senti-
nel lymph node biopsy has been considered as a standard of care in
early breast cancers [201]. Polymeric micelles can accumulate se-
lectively in lymph node metastases through th e blood vascular
route, which is believed to be specific to the active recruitment of
lymphocytes to lymph nodes. Organization of COX-2 inhibitor
(celecoxib) brought about a huge reduction in collection of polym-
eric micelles in pre angiogenic metastases; therefore, the inflamma-
tory microenvironment seems to be a mechanism for the retention
of micelles in the metastatic niche.
7.4.4. Antibody Conjugated Micelles
Antibody and its fragments are likewise valuable as targetable
ligands to design effectively targetable micelles. Antibody conju-
gated micelles (immunomicelles) have been accounted against os-
teopontin, an epidermal development factor receptor for delivery of
antitumor drugs. Immunomicelles are conjugated with antagonistic
to Tissue Factor (TF) antibody parts, a known essential initiator of
blood coagulation, and assumes an imperative part in tumor multi-
plication, intrusion, and metastasis. Articulation level of TF on
cancer is related to the patient's prognosis. Anti-TF antibody frag-
ment conjugated micelles incorporating epirubicin and DACHPt
were efficiently internalized by TF-over expressing cancer cells and
showed superior in vitro and in vivo antitumor activity to non-
targeted micelles [202]. Immunomicelles can deliver several drug
particles per antibody which increase the choice of anticancer drugs
and practical outline. In this way, immunomicelles are more flexi-
10 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya
ble stages for drug delivery. Antibody-mediated treatments includ-
ing Antibody Drug Conjugates (ADCs) have indicated much poten-
tial in growth treatment by tumor-focused on delivery of cytotoxic
drugs. Targeted delivery of DOX through immune polymeric mi-
celles where antibody C225 against EGFR is coupled with poly (L-
glutamic acid)-co-PEG, shows improved activity in inhibiting the
growth of A431 cells than free DOX [203]. Coordination of anti-
bodies to drug stacked nanocarriers expands the appropriateness of
antibodies to an extensive variety of therapeutics. Antibody frag-
ment-installed polymeric micelle in conjugation with maleimide-
thiol has been studied for the treatment of pancreatic cancer [202].
However, there is a limitation of loads that can be delivered through
antibody drug conjugates. The measure of drugs that can be deliv-
ered by a solitary antibody is the real test being developed of anti-
body drug conjugates, since over-burdening may lessen the binding
affinity of the antibody or affect pharmacokin etics, solubility, and
stability of the drug. Generally, 2 to 4 drugs for each antibody can
be presented in a neutralizer drug conjugate for achieving a suc-
cessful therapeutic response. Certain cytotoxic drugs such as aur-
istatins, maytansines, and calicheamicins have been conjugated to
the antibody, which are thousand-fold high er cytotoxic than typical
anticancer drugs; however, there are some concerns of side effects
[196, 200, 204]. This antibody conjugated nanoparticles o ffer better
delivery system achieving superior therapeutic efficacy while re-
ducing side effects.
During the last two decades, polymeric micelles as drug vehi-
cles have made rapid p rogress and several formulations are already
being evaluated in late-stage clinical trials. The approval of these
micellar anticancer drugs and generation of innovative m icellar
nanomedicines with smart functionalities for a clinical trial is ex-
pected soon. Polymeric micelle such as Paclitaxel (PTX)-loaded
polymeric micelles, SP1049C, Genexol-PM, NK012, NK105,
NC6300, K-912, NK 6004 are now under clinical trial against cer-
tain form s of cancer like breast cancer, non-small-cell lung cancer
and ovarian can cer, adenocarcinoma of oesophagus, gastroesophag-
eal junction and stomach, NSCLC, renal, stomach and pancreatic
cancer, advance stomach cancer, liver cancer and solid tumors,
respectively [197-199, 202, 205-212] (Table 2).
7.5. Other Delivery System for Nanoconjugated Quercetin
Apart from several nano-sized platform s including polymeric
micelle, liposomes, PLGA (Poly lactic-co-glycolic acid), silver
nanoparticles; other carriers are also evolv ed for delivery of quer-
cetin. In this series, nanosponges, poly (β-amino esters) nanogels,
solid lipid, carbon nanotubes, and silicon quantum dots are under
evaluation nowadays for safe and efficient delivery of quercetin to
target various cancerous cells [121, 137, 158, 213-215] (Table 3).
In recent research, a certain new combination of quantum dots
is being explored for clinical imaging and drug targeting. Use of
quantum dots plays a central theme in nanotechnology for medical
imaging due to small size w ith special electrical and electronic
properties. Quantum dots have been connected in the biomedical
field because of subordinate optical properties, high fluorescence
quantum yields, amazing reliability against photobleaching, and
lower toxicity. Certain types of quantum dots such as boron nitride
quantum dots [165, 166, 186] and graphene oxide quantum dots
[213, 216, 217] are being studied for their fluorescence properties,
lower cytotoxicity, and increased accumulation in cancer cells;
which can provide sufficient information for drug delivery. Gra-
phene oxide has been implicated for controlled loading and targeted
delivery of doxorubicin in MCF-7 breast cancer cells [218]. Re-
cently, silicon quantum dots possess strong luminescence character-
istics as well as low inherent cytotoxicity compared to conventional
heavy metal quantum dots, thus making them a potential for bio-
logical applications. Quercetin encapsu lated silicon quantum dots
results in red fluorescent, inhibits hydrogen peroxide-induced DNA
damage, allows monitoring of delivery, improvement of aqueous
solubility and enhanced biocompatibility in HepG2 hepatocarci-
noma cells [213]. Quercetin encapsulated silicon quantum dots emit
red fluorescen ce allowing monitoring of delivery, inhibits hydrogen
peroxide-induced DNA damage, improves aqueous solubility,
and enhanced biocompatibility in HepG2 hepatocarcinoma cells
[213, 216].
Utilization of silica nanoparticles as controlled drug delivery
system has been studied because of their high stability, high surface
area and better ability to function alize by various ligands, high drug
loading efficiency and low in vivo toxicity. The increased oral
bioavailability of drug after loading with mesoporous silica
nanoparticles has been studied [219]. The US Food and Drug Ad-
ministration (FDA) permitted silica nanoparticles for the first hu-
man clinical trial against cancer. Quercetin incorporated with
aminopropyl functionalized mesoporous silica nanoparticles p ro-
vides better nanocarrier for quercetin delivery. It is reported as an
improved chemo-preventive agent for in vitro system [220]. Al-
though silica nanoparticles conjugated quercetin delivery sy stem
requires the evaluation of toxicity so that it may be utilized against
cancer in the near futu re. Further, there is a need for monitored
bioimaging to understand the function of the delivery system more
precisely. Phytosome innovation can enhance the adequacy of che-
motherapeutics capability of anticancerous tranquilizes by expand-
ing the permeability of tumor cells. Poly (β-amino esters) polymers
have emerged as highly promising candidates for drug delivery and
biomedical applications due to their pH sensitivity, tuneable and
degradable properties. These polymeric frameworks can fill in
as pro fessional drug transporters for the delivery of bioactive
compounds such as quercetin which experience the poor watery
solvency and low bioavailability. Treatment of quercetin based
solid lipid nanoparticles recovers bone loss in ovariectomized rats
[158].
Additionally, the discovery, characterization, and implication of
carbon nanotubes having unexpected el ectrical, mechanical, an d
Table 2. Polymeric micelles under clinical evaluation.
Sr. No.
Polymeric Micelle
Cancer
Clinical Evaluation
Refs.
1
Paclitaxel (PTX)-loaded polymeric micelles
Breast can cer, non-small-cell lung cancer, and ovarian cancer
Approved
[206, 207]
2
SP1049C
Adenocarcinoma of oesophagus, gastroesophageal junction and stomach
Clinical p hase III
[208]
3
Genexol-PM
NSCLC
Clinical p hase II
[202]
4
NK012
Renal, sto mach and Panc reatic c ancer
Clinical p hase I/II
[209]
5
NK105
Advance stomach cancer
Clinical p hase II
[210]
6
NC6300
Liver cancer
Clinical p hase I
[199, 211]
7
K-912
Solid tumors
Clinical p hase I
[198]
8
NK 6004
Solid tumors
Clinical p hase I/II
[197, 205, 212]
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 11
thermal properties further explore the possibility of drug delivery
and targeting of anticancerous drugs. The release of quercetin from
carbon nanotubes attributes the increased hydrophilicity and solu-
bility of quercetin at acid microenvironment of cancer [214]. Im-
proved efficiency and reduced toxicity of quercetin carbon nano-
tube composite implicated in HeLa cells, suggests the consid erable
potential advantages of quercetin-carbon nanotube composite in
cancer therapy [215]. Furth er, the suitability of the nanosponges has
been implicated not only as a dual release drug delivery system but
also with regulated metabolism through nano network [137]. The
variety in crosslinking allows a dual release with regulated release
kinetics and recommends enhanced bioavailability supported by a
reduced metabolism. Dual drug delivery of tamoxifen and quercetin
with nanosponges in regulated metabolism has been suggested for
anticancer treatment [121]. An embodiment of polyester nano-
sponge co-stacked with quercetin is examined for successful load-
ing, release, and in vitro digestion. Lockhart et al. reported that
improved bioavailability of the nanoparticles formulations of nano-
sponge is supported by drug release, cytotoxicity, and enhanced
anti-cancer effects in the recovery condition [121].
CONCLUSION AND FUTURE PERSPECTIVE
Nanoparticles have been extensively investigated to deliver
anticancer drugs due to their enhanced anticancer potential and
promising clinical application. Recent research in nanoconjugated
quercetin is proposed to overcome its limitations of low bioavail-
ability, chemical instability, and short biological half-life. The
pharmacological effect of quercetin conjugated nanoparticles
mostly depends on drug carriers used. Quercetin conjugated polym-
eric micelles, liposomes, silver nanoparticles, PLGA (Poly lactic-
co-glycolic acid) have shown better inhibition of tumor growth in
vivo. Controlled drug release, long retention in tumor, improved
efficiency in invagin ation and extravagation through pores in the
capillary endothelium of tumor by nanoconjugated drugs have built
the future hope of their clinical application in metastatic cancer.
Delivery of quercetin nanoparticles and their anticancer efficacy are
mostly confined to laboratory animals and in vitro system. How-
ever, certain other nanoconjugated anticancer drugs are under clini-
cal trials. Despite numerous in vitro and in vivo anti-cancer studies
of quercetin formulations, there exist certain limitations for clinical
translation such as cost, safety, and side effects. Locating key target
molecules, safe and stable delivery systems with superior strategy
to overcome resistance and reduce the side effects of chemothera-
peutic agents are the main goals of ideal cancer treatment. Addition
of cancer cell-specific targeting moieties on the nanoparticles has
been evolved to not only enhance target-specific delivery of nano-
formulations but also to reduce their interaction with the normal
cells, p reventing side effects of drugs.
AUTHOR CONTRIBUTIONS
All authors have participated in planning or drafting of the
manuscript and approved the final version.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This work was supported by Department of Science and Tech-
nology, India (Grant No. SR/S0/AS-97/2007); and University
Grants Commission, India (Grant Nos. F.31-217/2005; F.40-
209/2011 SR).
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or other-
wise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Senapati, S.; Mahanta, A.K.; Kumar, S.; Mait i, P. Controlled d rug
delivery vehicles for cancer treatment and their performance. Sig-
nal Transduct. Target. Ther., 2018, 3, 7.
[http://dx.doi.org/10.1038/s41392-017-0004-3] [PMID: 29560283]
[2] Maurya, A.K.; Vinayak, M. Breast cancer stem cell mediated aber-
rant signaling and epithelial-mesenchymal transition targets: hope
for breast cancer therapy. Int. J. Cancer Oncol., 2016, 3(3), 1-7.
[http://dx.doi.org/10.15436/2377-0902.16.1035]
[3] Maurya, A.K.; Vinayak, M. PI-103 and Quercetin Attenuate PI3K-
AKT Signaling Pathway in T- Cell Lymphoma Exposed to Hydro-
gen Peroxide. PLoS One, 2016, 11(8), e0160686.
[http://dx.doi.org/10.1371/journal.pone.0160686] [PMID: 27494022]
[4] Das, L.; Vinayak, M. Curcumin Modulates Glycolytic Metabolism
and Inflammatory Cytokines via Nrf 2 in Dalton’s Lymphoma As-
cites Cells In Vivo. Antican cer. Agents Med. Chem., 2018, 18(12),
1779-1791.
[http://dx.doi.org/10.2174/1871520618666180604093802] [PMID:
29866021]
[5] Vinayak, M. Molecular Action of Herbal Antioxidants in Regula-
tion of Cancer Growth: Scope for Novel Anticancer Drugs. Nutr.
Cancer, 2018, 70(8), 1199-1209.
[http://dx.doi.org/10.1080/01635581.2018.1539187] [PMID:
30463447]
[6] Neuhouser, M.L. Dietary flavonoids and cancer risk: evidence from
human population studies. Nutr. Cancer, 2004, 50(1), 1-7.
[http://dx.doi.org/10.1207/s15327914nc5001_1] [PMID: 15572291]
Table 3. Other delivery systems involved in anticancer effects of nanoconjugated quercetin.
Sr. No.
Chemical/Polymer
Advantages
Effects/ Target s
Refs.
1
Silicon quantum dots
Measure subordinate optical properties, high fluo-
rescence quantum yields, astounding soundness
against photob leachi ng and lowe r tox icity
Inhibition of hydrogen peroxide induced DNA damage, monitoring
of delivery, improvement of aqueous solubility and enhanced
biocompatibility in HepG2 cells
[213]
2
Carbon nanotub es
Surprising electrical, mechan ical and thermal prop-
erties, targetability
Increased hydrophilicity and solubility of quercetin at acidic cancer
microenvironment
Improved efficiency and reduced toxicity of quercetin-carbon
nanotube composite implicated in HeLa cells
[214]
[215]
3
Solid lipid
Great tolerability and biodegradability, high encap-
sulation effectiv eness, targetability
Improved osteoprotective activity in post-menopausal osteoporosis
[158]
4
Poly (β-amino esters)
nanogels
pH sensitivity, tuneable and degradable properties
Protection against induced cellu lar oxidative stress
[137]
5
Nanosponges
Regulated metabolism an d protec tion against in-
duced cellular oxidative stress
Regulated metabolism of quercet in in cancer therapy
[121]
12 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya
[7] Tang, N.P.; Zhou, B.; Wang, B.; Yu, R.B.; Ma, J. Flavonoids in-
take and risk of lung cancer: a meta-analysis. Jpn. J. Clin. Oncol.,
2009, 39(6), 352-359.
[http://dx.doi.org/10.1093/jjco/hyp028] [PMID: 19351659]
[8] Cui, Y.; Morgenstern, H.; Greenland, S.; Tashkin, D.P.; Mao, J.T.;
Cai, L.; Cozen, W.; Mack, T.M.; Lu, Q.Y.; Zhang, Z.F. Dietary
flavonoid intake and lung cancer--a population-based case-control
study. Cancer, 2008, 112(10), 2241-2248.
[http://dx.doi.org/10.1002/cncr.23398] [PMID: 18327817]
[9] Forcad os, G.E.; James, D.B.; Sallau, A.B.; Muhammad, A.; Ma-
beta, P. Oxidative Stress and Carcinogenesis: Potential of Phyto-
chemicals in Breast Cancer Therapy. Nutr. Cancer, 2017, 69(3),
365-374.
[http://dx.doi.org/10.1080/01635581.2017.1267777] [PMID:
28103111]
[10] Abraham, A.N.; Sharma, T.K.; Bansal, V.; Shukla, R. Phytochemi-
cals as Dynamic Surface Ligands To Control Nanoparticle-Protein
Interactions. ACS Omega, 2018, 3(2), 2220-2229.
[http://dx.doi.org/10.1021/acsomega.7b01878] [PMID: 30023827]
[11] Maurya, A.K.; Vinayak, M. Modulation of PKC signaling and
induction of apoptosis through suppression of reactive oxygen spe-
cies and tumor necrosis factor receptor 1 (TNFR1): key role of
quercetin in cancer prevention. Tumour Biol., 2015, 36(11), 8913-
8924.
[http://dx.doi.org/10.1007/s13277-015-3634-5] [PMID: 26076811]
[12] D’Andrea, G. Quercetin: A flavonol with multifaceted therapeutic
applications? Fitoterapia, 2015, 106, 256-271.
[http://dx.doi.org/10.1016/j.fitote.2015.09.018] [PMID: 26393898]
[13] Amanzadeh, E.; Esmaeili, A.; Rahgozar, S.; Nourbakhshnia, M. In.
Rev. Neurosci., 2019, 0
[14] Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.;
Mehmoo d, Z.; Mubarak, M.S. Anticancer potential of quercetin: A
comprehensive review. Phytother. Res., 2018, 32(11), 2109-2130.
[http://dx.doi.org/10.1002/ptr.6155] [PMID: 30039547]
[15] Maurya, A.K.; Vinayak, M. Anticarcinogenic action of quercetin
by downregulation of phosphatidylinositol 3-kinase (PI3K) and
protein kinase C (PKC) via induction of p53 in h epato cellular car-
cinoma (HepG2) cell line. Mol. Biol. Rep., 2015, 42(9), 1419-1429.
[http://dx.doi.org/10.1007/s11033-015-3921 -7] [PMID: 26311153]
[16] Kumari, A.; Yadav, S.K.; Pakade, Y.B.; Kumar, V.; Singh, B.;
Chaudhary, A.; Yadav, S.C. Nanoencapsulation and characteriza-
tion of Albizia chinensis isolated antioxidant quercitrin on PLA
nanoparticles. Colloids Surf. B Biointerfaces, 2011, 82(1), 224-232.
[http://dx.doi.org/10.1016/j.colsurfb.2010.08.046] [PMID: 20870396]
[17] Maurya, A.K.; Vinayak, M. Improved synergistic anticancer effi-
cacy of quercetin in combination with PI-103, rottlerin, and G0
6983 against MCF-7 and RAW 264.7 cells. In Vitro Cell. Dev.
Biol. Anim., 2019, 55(1), 36-44.
[http://dx.doi.org/10.1007/s11626-018-0309-8] [PMID: 30413935]
[18] Kleemann, R.; Verschuren, L.; Morrison, M.; Zadelaar, S.; van Erk,
M.J.; Wielinga, P.Y.; Kooistra, T. Anti-inflammatory, anti-
proliferative and anti-atherosclerotic effects of quercetin in human
in vitro and in vivo models. Atherosclerosis, 2011, 218(1), 44-52.
[http://dx.doi.org/10.1016/j.atherosclerosis.2011.04.023] [PMID:
21601209]
[19] Nabavi, S.F.; Russo, G.L.; Daglia, M.; Nabavi, S.M. Role of quer-
cetin as an alternative for obesity treatment: you are what you eat!
Food Chem., 2015, 179, 305-310.
[http://dx.doi.org/10.1016/j.foodchem.2015.02.006] [PMID:
25722169]
[20] Xu, D.; Hu, M-J.; Wang, Y-Q.; Cui, Y-L. Antioxidant Activities of
Quercetin and Its Complexes for Medicinal Application. Mole-
cules, 2019, 24(6), 1123.
[http://dx.doi.org/10.3390/molecules24061123] [PMID: 30901869]
[21] Dueñas, M.; González-Manzano, S.; González-Paramás, A.; San-
tos-Buelga, C. Antioxidant evaluation of O-methylated metabolites
of catechin, epicatechin and quercetin. J. Pharm. Biomed. Anal.,
2010, 51(2), 443-449.
[http://dx.doi.org/10.1016/j.jpba.2009.04.007] [PMID: 19442472]
[22] Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin:
from antioxidant to nutraceutical. Eur. J. Pharmacol., 2008, 585(2-3),
325-337.
[http://dx.doi.org/10.1016/j.ejphar.2008.03.008] [PMI D: 18417116]
[23] Ganesan, S.; Faris, A.N.; Comstock, A.T.; Wang, Q.; Nanua, S.;
Hershenson, M.B.; Saj jan, U.S. Quercetin inhibits rhinovirus repli-
cation in vitro and in vivo. Antiviral Res., 2012, 94(3), 258-271.
[http://dx.doi.org/10.1016/j.antiviral.2012.03.005] [PMID:
22465313]
[24] Sun, D.; Zhang, W.; Li, N.; Zhao, Z.; Mou, Z.; Yan g, E.; Wang, W.
Silver nanoparticles-quercetin conjugation to siRNA against drug-
resistant Bacillus subtilis for effective gene silencing: in vitro and
in vivo. Mater. Sci. En g. C, 2016, 63, 522-534.
[http://dx.doi.org/10.1016/j.msec.2016.03.024] [PMID: 27040247]
[25] Rai, M.; Yadav, A.; Gade, A . Silver nanoparticles as a new genera-
tion of antimicrobials. Biotechnol. Adv., 2009, 27(1), 76-83.
[http://dx.doi.org/10.1016/j.biotechadv.2008.09.002] [PMID:
18854209]
[26] Han, Q.; Wang, X.; Cai, S.; Liu, X.; Zhang, Y.; Yang, L.; Wang,
C.; Yang, R. Quercetin nanoparticles with enhanced bioavailability
as multifunctional agents toward amyloid induced neurotoxicity. J.
Mater. Chem. B Mater. Biol. Med., 2018, 6(9), 1387-1393.
[http://dx.doi.org/10.1039/C7TB03053C]
[27] George, D.; Maheswari, P.U.; Begum, K.M.M.S. Synergic formu-
lation of onion peel quercetin loaded chitosan-cellulose hydrogel
with green zinc oxide nanoparticles towards controlled release,
biocompatibility, antimicrobial and anticancer activity. Int. J. Biol.
Macromol., 2019, 132, 784-794.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.04.008] [PMID:
30951778]
[28] Alidadi, H.; Khorsandi, L.; Shirani, M. Effects of Quercetin on
Tubular Cell Apopto sis and Kidney Damage in Rats Induced by Ti-
tanium Dioxide Nanoparticles. Malays. J. Med. Sci., 2018, 25(2),
72-81.
[http://dx.doi.org/10.21315/mjms2018.25.2.8] [PMID: 30918457]
[29] Abdelhalim, M.A.K.; Qaid, H.A.; Al-Mohy, Y.; Al-Ayed, M.S.
Effects of quercetin and arginine on the nephrotoxicity and lipid
peroxidation induced by gold nanoparticles in vivo. Int. J.
Nanomedicine, 2018, 13, 7765-7770.
[http://dx.doi.org/10.2147/IJN.S183281] [PMID: 30538457]
[30] Cruz Dos Santos, S.; Osti Silva, N.; Dos Santos Espinelli, J.B.J.;
Germani Marinho, M.A.; Vieira Borges, Z.; Bruzamarello Caon
Branco, N.; Faita, F.L.; Meira Soares, B.; Horn, A.P.; Parize, A.L.;
Rodrigues de Lima, V. Molecular interactions and physico-
chemical characterization of quercetin-loaded magnetolipo somes.
Chem. Phys. Lipids, 2019, 218, 22-33.
[http://dx.doi.org/10.1016/j.chemphyslip.2018.11.010] [PMID:
30508514]
[31] Wen, P.; Hu, T.G.; Li, L.; Zong, M.H.; Wu, H. A colon-specific
delivery system for quercetin with enhanced cancer prevention
based on co-axial electrospinning. Food Funct., 2018, 9(11), 5999-
6009.
[http://dx.doi.org/10.1039/C8FO01216D] [PMID: 30382268]
[32] Ma, J.J.; Yu, Y.G.; Yin, S.W.; Tang, C.H.; Yang , X.Q. Cellular
Uptake and Intracellular Antioxidant Activity of Zein/Chitosan
Nanoparticles Incorporated with Quercetin. J. Agric. Food Chem.,
2018, 66(48) , 12783-12793.
[http://dx.doi.org/10.1021/acs.jafc.8b04571] [PMID: 30406660]
[33] Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.F.; Flamm,
G.W.; Williams, G.M.; Lines, T.C. A critical review of the data re-
lated to the safety of quercetin and lack of evidence of in vivo tox-
icity, including lack of genotoxic/carcinogenic properties. Food
Chem. Toxicol., 2007, 45(11), 2179-2205.
[http://dx.doi.org/10.1016/j.fct.2007.05.015] [PMID: 17698276]
[34] Reinboth, M.; Wolffram, S.; Abraham, G.; Ungemach, F.R.; Cer-
mak, R. Oral bioavailability of quercetin from different quercetin
glycosides in dogs. Br. J. Nutr., 2010, 104(2), 198-203.
[http://dx.doi.org/10.1017/S000711451000053X] [PMID: 20230651]
[35] Hirpara, K.V.; Aggarwal, P.; Mukherjee, A.J.; Joshi, N.; Burman,
A.C. Quercetin and its derivatives: synthesis, pharmacological uses
with special emphasis on anti-tumor properties and prodrug with
enhanced bio-availability. Anticancer. Agents Med. Chem., 2009,
9(2), 138-161.
[http://dx.doi.org/10.2174/187152009787313855] [PMID:
19199862]
[36] Mulholland, P.J.; Ferry, D.R.; Anderso n, D.; Hussain, S.A.; Young,
A.M.; Cook, J.E.; Hodgkin, E.; Seymour, L.W.; Kerr, D.J. Pre-
clinical and clinical study of QC12, a water-soluble, pro-drug of
quercetin. Ann. Oncol., 2001, 12(2), 245-248.
[http://dx.doi.org/10.1023/A:1008372017097] [PMID: 11300332]
[37] Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of
Biological Importance of Quercetin: A Bioactive Flavonoid. Phar-
macogn. Rev., 2016, 10(20), 84-89.
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 13
[http://dx.doi.org/10.4103/0973-7847.194044] [PMID: 28082789]
[38] Ezzati Nazhad Dolatabadi, J.; Mok htarzadeh, A.; Ghareghoran,
S.M.; Dehghan, G. Synthesis, Characterization and Antioxidant
Property of Quercetin-Tb(III) Complex. Adv. Pharm. Bull., 2014,
4(2), 101-104.
[PMID: 24511472]
[39] Gao, X.; Wang, B.; Wei, X.; Men, K.; Zheng, F.; Zhou, Y.; Zheng,
Y.; Gou, M.; Huang, M.; Guo, G.; Huang, N.; Qian, Z.; Wei, Y.
Anticancer effect and mechanism of polymer micelle-encapsulated
quercetin on ovarian cancer. Nanoscale, 2012, 4(22), 7021-7030.
[http://dx.doi.org/10.1039/c2nr32181e] [PMID: 23044718]
[40] Nam, J.S.; Sharma, A.R.; Nguyen, L.T.; Chakraborty, C.; Sharma,
G.; Lee, S.S. Application of Bioactive Quercetin in Oncotherapy:
From Nutrition to Nanomedicin e. Molecules, 2016, 21(1), E108.
[http://dx.doi.org/10.3390/molecules21010108] [PMID: 26797598]
[41] Maurya, A.K.; Vinayak, M. Qu ercetin regresses Dalton’s lym-
phoma growth via suppression of PI3K/AKT signaling leading to
upregulation of p53 and decrease in energy metabolism. Nutr. Can-
cer, 2015, 67(2), 354-363.
[http://dx.doi.org/10.1080/01635581.2015.990574] [PMID:
25658812]
[42] Zhu, B.; Yu, L.; Yue, Q. Co-delivery of vincristine and quercetin
by nanocarriers for lymphoma combination chemotherapy. Biomed.
Pharmacother., 2017, 91, 287-294.
[http://dx.doi.org/10.1016/j.biopha.2017.02.112] [PMID:
28463792]
[43] Maurya, A.K.; Vin ayak, M. PI-103 attenuates PI3K-AKT signaling
and induces apoptosis in murineT-cell lymphoma. Leuk. Lym-
phoma, 2017, 58(5), 1153-1161.
[http://dx.doi.org/10.1080/10428194.2016.1225207] [PMID:
27658642]
[44] Maurya, A.K.; Vinayak, M. Quercetin Attenuates Cell Survival,
Inflammation, and Angiogenesis via Modulation of AKT Signaling
in Murine T-Cell Lymphoma. Nutr. Cancer, 2017, 69(3), 470-480.
[http://dx.doi.org/10.1080/01635581.2017.1267775] [PMID:
28107044]
[45] Rivera Rivera, A.; Castillo-Pichardo, L.; Gerena, Y.; Dharmaward-
hane, S. Anti-Breast Cancer Potential of Quercetin via the
Akt/AMPK/Mammalian Target of Rapamycin (mTOR) Signaling
Cascade. PLoS One, 2016, 11(6), e01 57251.
[http://dx.doi.org/10.1371/journal.pone.0157251] [PMID: 27285995]
[46] Seo, H.S.; Ku, J.M.; Choi, H.S.; Choi, Y.K.; Woo, J.K.; Kim, M.;
Kim, I.; Na, C.H.; Hur, H.; Jang, B.H.; Shin, Y.C.; Ko, S.G. Quer-
cetin induces caspase-dependent extrinsic apoptosis through inhibi-
tion of sign al transducer and activator of transcription 3 signaling
in HER2-overexpressing BT-474 breast cancer cells. Oncol. Rep.,
2016, 36(1), 31-42.
[http://dx.doi.org/10.3892/or.2016.4786] [PMID: 27175602]
[47] Subramaniam, D.; Kaush ik, G.; Dandawate, P.; Anant, S. Targeting
cancer stem cells for chemoprevention of pancreatic cancer. Curr.
Med. Ch em., 2017.
[PMID: 28137215]
[48] Gavrilas, L.I.; Ionescu, C.; Tudoran, O.; Lisencu, C.; Balacescu,
O.; Miere, D. The Role of Bioactive Dietary Components in Modu-
lating miRNA Expression in Colorectal Cancer. Nutrients, 2016,
8(10), E590. [http://dx.doi.org/10.3390/nu8100590] [PMID:
27681738]
[49] Saleem, T.H.; A ttya, A.M.; Ahmed, E.A.; Ragab, S.M.; Ali Abdal-
lah, M.A.; Omar, H.M. Possible Protective Effects of Quercetin
and Sodium Gluconate Against Colon Cancer Induction by Di-
methylhydrazine in Mice. Asian Pac. J. Cancer Prev., 2015,
16(14), 5823-5828.
[http://dx.doi.org/10.7314/APJCP.2015.16.14.5823] [PMID:
26320457]
[50] Engen, A.; Maeda, J.; Wozniak, D.E.; Brents, C.A.; Bell, J.J.; Ue-
saka, M.; Aizawa, Y.; Kato, T.A. Induction of cytotoxic and geno-
toxic responses by natural and novel quercetin glycosides. Mutat.
Res. Genet. Toxicol. Environ. Mutagen., 2015, 784-785, 15-22.
[http://dx.doi.org/10.1016/j.mrgentox.2015.04.007] [PMID:
26046972]
[51] Maurya, A.K.; Vinayak, M. Abstract A07: Decline in the growth of
murine T-cell lymphoma via modulation of PI3K signaling path-
way: Key role of quercetin and PI-103. Mol. Cancer Ther., 2015,
14(7)(Suppl.), A07-A07.
[52] Chen, L.C.; Chen, Y.C.; Su, C.Y.; Hong, C.S.; Ho, H.O.; Sheu,
M.T. Development and characterization of self-assembling lecithin-
based mixed polymeric micelles containing quercetin in cancer
treatment and an in vivo pharmacokinetic study. Int. J. Nanomedi-
cine, 2016, 11, 1557-1566.
[PMID: 27143878]
[53] Del Follo-Martinez, A.; Banerjee, N.; Li, X.; Safe, S.; Mertens-
Talcott, S. Resveratrol and quercetin in combination have antican-
cer activity in colon cancer cells and repress oncogenic microRNA-
27a. Nutr. Cancer, 2013, 65(3), 494-504.
[http://dx.doi.org/10.1080/01635581.2012.725194] [PMID:
23530649]
[54] Chan, S-T.; Chuang, C-H.; Lin, Y-C.; Liao, J-W.; Lii, C-K.; Yeh,
S-L. Quercetin enhances the antitumor effect of trichostatin A and
suppresses muscle wasting in tumor-bearing mice. Food Funct.,
2018, 9(2), 871-879.
[http://dx.doi.org/10.1039/C7FO01444A] [PMID: 29292417]
[55] Serri, C.; Quagliariello, V.; Iaffaioli, R.V.; Fusco, S.; Botti, G.;
Mayol, L.; Biondi, M. Combination therapy for the treatment of
pancreatic cancer through hyaluronic acid-decorated nanoparticles
loaded with quercetin and gemcitabine: A preliminary in vitro
study. J. Cell. Physiol., 2019, 234(4), 4959-4969.
[http://dx.doi.org/10.1002/jcp.27297] [PMID: 30334571]
[56] Flusberg, D.A.; Sorger, P.K. Surviving apop tosis: life-death signal-
ing in single cells. Tren ds Cell Biol., 2015, 25(8), 446-458.
[http://dx.doi.org/10.1016/j.tcb.2015.03.003] [PMID: 25920803]
[57] Pobezinskaya, Y.L.; Liu, Z. The role of TRADD in death receptor
signaling. Cell Cycle, 2012, 11(5), 871-876.
[http://dx.doi.org/10.4161/cc.11.5.19300] [PMID: 22333735]
[58] Russo, M.; Spagnuolo, C.; Bilotto, S.; Tedesco, I.; Maiani, G.;
Russo, G.L.; Russo, M.; Spagnuolo, C.; Bilotto, S.; Tedesco, I.;
Maiani, G.; Russo, G.L. Inhibition of protein kinase CK2 by quer-
cetin enhances CD95-mediated apoptotis in a human thymus-
derived T cell line. Food Res. Int., 2014.
[http://dx.doi.org/10.1016/j.foodres.2014.05.022]
[59] Chou, C.C.; Yang, J.S.; Lu, H.F.; Ip, S.W.; Lo, C.; Wu, C.C.; Lin,
J.P.; Tang, N.Y.; Chung, J.G.; Chou, M.J.; Teng, Y.H.; Chen, D.R.
Quercetin-mediated cell cycle arrest and apoptosis involving acti-
vation of a caspase cascade through the mitochondrial pathway in
human breast cancer MCF-7 cells. Arch. Pharm. Res., 2010, 33(8),
1181-1191.
[http://dx.doi.org/10.1007/s12272-010-0808-y] [PMID: 20803121]
[60] Haghiac, M.; Walle, T. Quercetin induces necrosis and apoptosis in
SCC-9 oral cancer cells. Nutr. Cancer, 2005, 53(2), 220-231.
[http://dx.doi.org/10.1207/s15327914nc5302_11] [PMID:
16573383]
[61] Mozhgan Farzami Sepeh r, S.B.J.B.H. The Cuscuta kotschyan a
effects on breast cancer cells line MCF7. J. Med. Plants Res., 2011,
5(27), 6344-6351.
[http://dx.doi.org/10.5897/JMPR11.1048]
[62] Niu, G.; Yin, S.; Xie, S.; Li, Y.; Nie, D.; Ma, L.; Wang, X.; Wu, Y.
Quercetin induces apoptosis by activating caspase-3 and regulating
Bcl-2 and cyclooxygenase-2 pathways in human HL-60 cells. Acta
Biochim. Biophys. Sin. (Shanghai), 2011, 43(1), 30-37.
[http://dx.doi.org/10.1093/abbs/gmq107] [PMID: 21173056]
[63] Hu, J.; Wang, J.; Wang, G.; Yao, Z.; Dang, X. Pharmacokinetics
and antitumor efficacy of DSPE-PEG2000 poly meric liposomes
loaded with quercetin and temozolomide: Analysis of their effec-
tiveness in enhancing the chemosensitization of drug-resistant
glioma cells. Int. J. Mol. Med., 2016, 37(3), 690-702.
[http://dx.doi.org/10.3892/ijmm.2016.2458] [PMID: 26782731]
[64] Hu, J.; Yu, Q.; Zhao, F.; Ji, J.; Jiang, Z.; Chen, X.; Gao, P.; Ren,
Y.; Shao, S.; Zhang, L.; Yan, M. Protection of Quercetin against
Triptolide-induced apoptosis by suppressing oxidative stress in rat
Leydig cells. Chem. Biol. Interact., 2015, 240, 38-46.
[http://dx.doi.org/10.1016/j.cbi.2015.08.004] [PMID: 26277538]
[65] Shen, S.C.; Lee, W.R.; Yang, L.Y.; Tsai, H.H.; Yang, L.L.; Chen,
Y.C. Quercetin enhancement of arsenic-induced apoptosis via
stimulating ROS-dependent p53 protein ubiquitination in human
HaCaT keratinocytes. Exp. Dermatol., 2012, 21(5), 370-375.
[http://dx.doi.org/10.1111/j.1600-0625.2012.01479.x] [PMID:
22509835]
[66] Zhang, J.Y.; Yi, T.; Liu, J.; Zhao, Z.Z .; Chen, H.B. Quercetin in-
duces apoptosis via the mitocho ndrial pathway in KB and KBv200
cells. J. Agric. Food Chem., 2013, 61(9), 2188-2195.
[http://dx.doi.org/10.1021/jf305263r] [PMID: 23410218]
[67] Zhang, Q.; Zhao, X.H.; Wang, Z.J. Cytotoxicity of flavones and
flavonols to a human esophageal squamous cell carcinoma cell line
14 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya
(KYSE-510) by induction of G2/M arrest and apoptosis. Toxicol. In
Vitro, 2009, 23(5), 797-807.
[http://dx.doi.org/10.1016/j.tiv.2009.04.007] [PMID: 19397994]
[68] Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo,
Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest
and apoptosis in human breast cancer cells by quercetin. Int. J. On-
col., 2001, 19(4), 837-844.
[http://dx.doi.org/10.3892/ijo.19.4.837] [PMID: 11562764]
[69] Lee, Y.K.; Hwang, J.T.; Kwon, D.Y.; Surh, Y.J.; Park, O.J. Induc-
tion of apoptosis by quercetin is mediated through AMPKal-
pha1/ASK1/p38 pathway. Cancer Lett., 2010, 292(2), 228-236.
[http://dx.doi.org/10.1016/j.canlet.2009.12.005] [PMID: 20083342]
[70] Kim, M.C.; Lee, H .J.; Lim, B.; Ha, K.T.; Kim, S.Y.; So, I.; Kim,
B.J. Quercetin induces apoptosis by inhibiting MAPKs and TRPM7
channels in AGS cells. Int. J. Mol. Med., 2014, 33(6), 1657-1663.
[http://dx.doi.org/10.3892/ijmm.2014.1704] [PMID: 24647664]
[71] Lee, K.H.; Yoo, C.G. Simultaneous inactivation of GSK-3β sup-
presses quercetin-induced apoptosis by inhibiting the JNK path-
way. Am. J. Physiol. Lung Cell. Mol. Physiol., 2013, 304(11),
L782-L789.
[http://dx.doi.org/10.1152/ajplung.00348.2012] [PMID: 23564507]
[72] Vidya Priyadarsini, R.; Senthil Murugan, R.; Maitreyi, S.; Rama-
lingam, K.; Karunagaran, D.; Nagini, S. The flavonoid quercetin
induces cell cycle arrest and mitochondria-mediated apoptosis in
human cervical cancer (HeLa) cells through p53 induction and NF-
κB inhibition. Eur. J. Pharmacol., 2010, 649(1-3), 84-91.
[http://dx.doi.org/10.1016/j.ejphar.2010.09.020] [PMID:
20858478]
[73] Chen, X.; Dong, X.S.; Gao, H.Y.; Jiang, Y.F.; Jin, Y.L.; Chang,
Y.Y.; Chen, L.Y.; Wang, J.H. Suppression of HSP27 increases the
anti-tumor effects of quercetin in human leukemia U937 cells. Mol.
Med. Rep., 2016, 13(1), 689-696.
[http://dx.doi.org/10.3892/mmr.2015.4600] [PMID: 26648539]
[74] Mukherjee, A.; Kh uda-Bukhsh, A.R. Quercetin Down-regulates IL-
6/STAT-3 Signals to Induce Mitochondrial-mediated Apoptosis in
a Nonsmall- cell Lung-cancer Cell Line, A549. J. Pharmacopunc-
ture, 2015, 18(1), 19-26.
[http://dx.doi.org/10.3831/KPI.2015.18.002] [PMID: 25830055]
[75] Ranganathan, S.; Halagowder, D.; Sivasithambaram, N.D. Quer-
cetin Suppresses Twist to Induce Apoptosis in MCF-7 Breast Can-
cer Cells. PLoS One, 2015, 10(10), e0141370.
[http://dx.doi.org/10.1371/journal.pone.0141370] [PMID:
26491966]
[76] Ward, A.B.; Mir, H.; Kapur, N.; Gales, D.N.; Carriere, P.P.; Singh,
S. Quercetin inhibits prostate cancer by attenuating cell survival
and inhibiting anti-apoptotic pathways. World J. Surg. Oncol.,
2018, 16(1), 108.
[http://dx.doi.org/10.1186/s12957-018-1400-z] [PMID: 29898731]
[77] Granado-Serrano, A.B.; Mart ín, M.A .; Bravo, L.; Goya, L.; Ramos,
S. Quercetin induces apopto sis via caspase activation, regulation of
Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a
human hepatoma cell line (HepG2). J. Nutr., 2006, 136(11), 2715-
2721.
[http://dx.doi.org/10.1093/jn/136.11.2715] [PMID: 17056790]
[78] Walker, E.H.; Pacold, M.E.; Perisic, O.; Stephens, L.; Hawkins,
P.T.; Wy mann, M.P.; Williams, R.L. Structural determinants of
phosphoinositide 3-kinase inhibition by wortmannin, LY294002,
quercetin, myricetin, and staurosporine. Mol. Cell, 2000, 6(4), 909-
919.
[http://dx.doi.org/10.1016/S1097-2765(05)00089-4] [PMID:
11090628]
[79] Datta, S.R.; Dudek, H.; Tao, X.; Masters, S.; Fu, H.; Gotoh, Y.;
Greenberg, M.E. Akt phosphorylation of BAD couples survival
signals to the cell-intrinsic death machinery. Cell, 1997, 91(2), 231-
241.
[http://dx.doi.org/10.1016/S0092-8674(00)80405-5] [PMID:
9346240]
[80] Pap, M.; Cooper, G.M. Role of glycogen synthase kinase -3 in th e
phosphatidylinositol 3-Kinase/Akt cell survival pathway. J. Biol.
Chem., 1998, 273(32), 19929-19932.
[http://dx.doi.org/10.1074/jbc.273.32.19929] [PMID: 9685326]
[81] Dickey, A.; Schleicher, S.; Leahy, K.; Hu, R.; Hallahan, D.; Tho-
tala, D.K. GSK-3β inhibition promotes cell death, apoptosis, and in
vivo tumor growth delay in neuroblastoma Neuro-2A cell line. J.
Neurooncol., 2011, 104(1), 1 45-153.
[http://dx.doi.org/10.1007/s11060-010-0491-3] [PMID: 21161565]
[82] Chiang, G.G.; Abraham, R.T. Phosphorylation of mammalian
target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6
kinase. J. Biol. Chem., 2005, 280(27), 25485-25490.
[http://dx.doi.org/10.1074/jbc.M501707200] [PMID: 15899889]
[83] Hung, C.M.; Garcia-Haro, L.; Sparks, C.A.; Guertin, D.A. mTOR-
dependent cell survival mechanisms. Cold Spring Harb. Perspect.
Biol., 2012, 4(12), a008771.
[http://dx.doi.org/10.1101/cshperspect.a008771] [PMID:
23124837]
[84] Simpson, D.R.; Mell, L.K.; Cohen, E.E. Targeting the
PI3K/AKT/mTOR pathway in squamo us cell carcinoma of the
head and neck. Oral Oncol., 2015, 51(4), 291-298.
[http://dx.doi.org/10.1016/j.oraloncology.2014.11.012] [PMID:
25532816]
[85] Mabuchi, S.; Kuroda, H.; Takahashi, R.; Sasa no, T. The
PI3K/AKT/mTOR pathway as a the rapeutic target in ova rian can-
cer. Gynecol. Oncol., 2015, 137(1), 173-179.
[http://dx.doi.org/10.1016/j.ygyno.2015.02.003] [PMID:
25677064]
[86] Martelli, A.M.; Evangelisti, C.; Chiarini, F.; Grimaldi, C.; Cappel-
lini, A.; Ognibene, A.; McCubrey, J.A. The emerging role of the
phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin
signaling network in normal myelopoiesis and leukemogenesis.
Biochim. Biophys. Acta, 2010, 1803(9), 991-1002.
[http://dx.doi.org/10.1016/j.bbamcr.2010.04.005] [PMID:
20399811]
[87] Bai, D.; Ueno, L.; Vogt, P.K. Akt-mediated regulation of NFkap-
paB and the essentialness of NFkappaB for the oncogenicity of
PI3K and Akt. Int. J. Cancer, 2009, 125(12), 2863-2870.
[http://dx.doi.org/10.1002/ijc.24748] [PMID: 19609947]
[88] Choi, K.C.; Chung, W.T.; Kwon, J.K.; Yu, J.Y.; Jang, Y.S.; Park,
S.M.; Lee, S.Y.; Lee, J.C. Inhibitory effects of quercetin on afla-
toxin B1-induced hepatic damage in mice. Food Chem. Toxicol.,
2010, 48(10) , 2747-2753.
[http://dx.doi.org/10.1016/j.fct.2010.07.001] [PMID: 20621152]
[89] Ventura, A.; Kirsch, D.G.; McLaughlin, M.E.; Tuveson, D.A.;
Grimm, J.; Lintault, L.; Newman, J.; Reczek, E.E.; Weissleder, R.;
Jacks, T. Restoration of p53 function leads to tumour regression in
vivo. Nature, 2007, 445(71 28), 661-665.
[http://dx.doi.org/10.1038/nature05541] [PMID: 17251932]
[90] Moll, U.M.; Petrenko, O. The MDM2-p53 interaction. Mol. Cance r
Res., 2003, 1(14), 1001-1008.
[PMID: 14707283]
[91] Mayo, L.D.; Donner, D.B. The PTEN, Mdm2, p53 tumor suppres-
sor-oncoprotein network. Trends Biochem. Sci., 2002, 27(9), 462-
467.
[http://dx.doi.org/10.1016/S0968-0004(02)02166-7] [PMID:
12217521]
[92] Zhao, P.; Mao, J.M.; Zhang, S.Y.; Zhou, Z.Q.; Tan, Y.; Zhang, Y.
Quercetin induces HepG2 cell apoptosis by inhibiting fatty acid
biosynthesis. Oncol. Lett., 2014, 8(2), 765-769.
[http://dx.doi.org/10.3892/ol.2014.2159] [PMID: 25009654]
[93] Jia, L.; Huang, S.; Yin, X.; Zan, Y.; Guo, Y.; Han, L. Quercetin
suppresses the mobility of breast cancer by suppressing glycolysis
through Akt-mTOR pathway mediated autophagy induction. Life
Sci., 2018, 208, 123-130.
[http://dx.doi.org/10.1016/j.lfs.2018.07.027] [PMID: 30025823]
[94] Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and
lymphangiogenesis. Nat. Rev. Mol. Cell Biol., 2007, 8(6), 464-478.
[http://dx.doi.org/10.1038/nrm2183] [PMID: 17522591]
[95] Graupera, M.; Potente, M. Regulation of angio genesis by PI3K
signaling networks. Exp. Cell Res., 2013, 319(9), 1348-1355.
[http://dx.doi.org/10.1016/j.yexcr.2013.02.021] [PMID: 23500680]
[96] Ma, F.; Zhan g, L.; Westlu nd, K.N. Reactive oxygen species medi-
ate TNFR1 increase after TRPV1 activation in mouse DRG neu-
rons. Mol. Pain, 2009, 5, 31.
[http://dx.doi.org/10.1186/1744-8069-5-31] [PMID: 19531269]
[97] Fang, J.; Ding, M. ; Yang, L.; Liu, L.Z.; Jiang, B.H.
PI3K/PTEN/AKT signaling regulates prostate tumor a ngiogenesis.
Cell. Signal., 2007, 19(12), 2487-2497.
[http://dx.doi.org/10.1016/j.cellsig.2007.07.025] [PMID:
17826033]
[98] Sobolewski, C.; Cerella, C.; Dicato, M.; Ghibelli, L.; Diederich, M .
The role of cyclooxygenase-2 in cell proliferation and cell death in
human malignancies. Int. J. Cell Biol., 2010, 2010, 215158.
[http://dx.doi.org/10.1155/2010/215158] [PMID: 20339581]
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 15
[99] Das, L.; Vinayak, M. Long term effect of curcumin in restoration
of tumour suppressor p53 and phase-II antioxidant enzymes via ac-
tivation of Nrf2 signalling and modulation of inflammation in pre-
vention of cancer. PLoS One, 2015, 10(4), e0124000.
[http://dx.doi.org/10.1371/journal.pone.0124000] [PMID:
25860911]
[100] Subbaramaiah, K.; Altorki, N.; Chung, W.J.; Mestre, J.R.; Sampat,
A.; Dannenberg, A.J. Inhibition of cyclooxygenase-2 gene expres-
sion by p53. J. Biol. Chem., 1999, 274(16), 10911-10915.
[http://dx.doi.org/10.1074/jbc.274.16.10911] [PMID: 10196169]
[101] Jeon, J-S.; Kwon , S.; Ban, K.; Kwon Hong, Y.; Ahn, C.; Sung, J-
S.; Choi, I. Regulation of the Intracellular ROS Level Is Critical for
the Antiproliferative Effect of Quercetin in the Hepatocellular Car-
cinoma Cell Line HepG2. Nutr. Cancer, 2019, 71(5), 861-869.
[http://dx.doi.org/10.1080/01635581.2018.1559929] [PMID:
30661409]
[102] Jadhav, N.R.; Nadaf, S.J.; Lohar, D.A.; Ghagare, P.S.; Powar, T.A.
Phytochemicals Formulated As Nanoparticles: Inventions, Recent
Patents and Future Prospects. Recent Pat. Drug Deliv. Formul.,
2017, 11(3), 173-186.
[http://dx.doi.org/10.2174/1872211311666171120102531] [PMID:
29165100]
[103] Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L.
Polyphenols: food sources and bioavailability. Am. J. Clin. Nutr.,
2004, 79(5), 727-747.
[http://dx.doi.org/10.1093/ajcn/79.5.727] [PMID: 15113710]
[104] Weldin, J.; Jack, R.; Dugaw, K.; Kapur, R.P. Quercetin, an over-
the-counter supplement, causes neuroblastoma-like elevation of
plasma homovanillic acid. Pediatr. Dev. Pathol., 2003, 6(6), 547-
551.
[http://dx.doi.org/10.1007/s10024-003-5061-7] [PMID: 15018454]
[105] Konishi, Y.; Zhao, Z.; Shimizu, M. Phenolic acids are absorbed
from the rat stomach with different absorption rates. J. Agric. Food
Chem., 2006, 54(20), 7539-7543.
[http://dx.doi.org/10.1021/jf061554+] [PMID: 17002419]
[106] Németh, K.; Plumb, G. W.; Berrin, J.G.; Juge, N.; Jacob, R.; Naim,
H.Y.; Williamson, G.; Swallow, D.M.; Kroon, P.A. Deglycosyla-
tion by small intestinal epithelial cell beta-glucosidases is a critical
step in the absorption and metabolism of dietary flavonoid gly-
cosides in humans. Eur. J. Nutr., 2003, 42(1), 29-42.
[http://dx.doi.org/10.1007/s00394-003-0397-3] [PMID: 12594539]
[107] Murota, K.; Terao, J. Quercetin appe ars in the lymph of unanesthe-
tized rats as its ph ase II metabolites after administered into the
stomach. FEBS Lett., 2005, 579(24), 5343-5346.
[http://dx.doi.org/10.1016/j.febslet.2005.08.060] [PMID:
16194534]
[108] Spencer, J.P.; Kuhnle, G.G.; Wi lliams, R.J.; Rice-Evans, C. Intra-
cellular metabolism and bioactivity of quercetin and its in vivo me-
tabolites. Biochem. J., 2003, 372(Pt 1), 173-181.
[http://dx.doi.org/10.1042/bj20021972] [PMID: 12578560]
[109] de Boer, V.C.; Dihal, A.A.; van der Woude, H.; Arts, I.C.; Wolf-
fram, S.; Alink, G.M.; Rietjens, I.M.; Keijer, J .; Hollman, P.C. Tis-
sue distribution of quercetin in rats and pigs. J. Nutr., 2005, 135(7),
1718-1725.
[http://dx.doi.org/10.1093/jn/135.7.1718] [PMID: 15987855]
[110] Lee, J.; Mitchell, A.E. Pharmacokinetics of quercetin absorption
from apples and onions in healthy humans. J. Agric. Food Chem.,
2012, 60(15), 38 74-3881.
[http://dx.doi.org/10.1021/jf3001857] [PMID: 22439822]
[111] Kaşıkcı, M.B. B.N., Bioavailability of Quercetin. Curr. Res. Nutr.
Food Sci., 2016, 4, 146-151 .
[http://dx.doi.org/10.12944/CRNFSJ.4.Special-Issue-October.20]
[112] Tran, T.H.; Guo, Y.; Song, D.; Bruno, R.S.; Lu, X. Quercetin -
containing self-nanoemulsifying drug delivery system for improv-
ing oral bioavailability. J. Pharm. Sci., 2014, 103(3), 840-852.
[http://dx.doi.org/10.1002/jps.23858] [PMID: 24464737]
[113] Dian, L.; Yu, E.; Chen, X.; Wen, X.; Zhang, Z.; Qin, L.; Wang, Q.;
Li, G.; Wu, C. Enhancing oral bioavailability of quercetin using
novel soluplus polymeric micelles. Nanoscale Res. Lett., 2014,
9(1), 2406.
[http://dx.doi.org/10.1186/1556-276X-9-684] [PMID: 26088982]
[114] Xu, G.; Shi, H.; Ren, L.; Gou, H.; Gong, D.; Gao, X.; Huang, N.
Enhancing the anti-colon cancer activity of quercetin by self-
assembled micelles. Int. J. Nanomedicine, 2015, 10, 2051-2063.
[PMID: 25844036]
[115] Matsumura, Y.; Kataoka, K. Preclinical and clinical studies of
anticancer agent-incorporating polymer micelles. Cancer Sci.,
2009, 100(4), 572-579.
[http://dx.doi.org/10.1111/j.1349-7006.2009.01103.x] [PMID:
19462526]
[116] Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of
quercetin bioavailability. J. Nutr. Biochem., 2015, 26(3), 201-210.
[http://dx.doi.org/10.1016/j.jnutbio.2014.10.008] [PMID:
25468612]
[117] Santos, A.F.A.G .I.A.B. M.P.A.T.L.T.K.V.G.W. C.N. Bioavailability
of Quercetin in Humans with a Focus on Interindividual Variation.
Compr. Rev. Food Sci. Food Saf., 2018, 17, 714-731.
[http://dx.doi.org/10.1111/1541-4337.12342]
[118] Mu, Y.; Fu, Y.; Li, J.; Yu, X.; Li, Y.; Wang, Y.; Wu, X.; Zhang,
K.; Kong, M.; Feng, C.; Chen, X. Multifunctional quercetin conju-
gated chitosan nano-micelles with P-gp inhibition and permeation
enhancement of anticancer drug. Carbohydr. Polym., 2019, 203,
10-18.
[http://dx.doi.org/10.1016/j.carbpol.2018.09.020] [PMID:
30318192]
[119] Ahmad, N.; Ahmad, R.; Naqvi, A.A.; Alam, M.A.; Abdur Rub, R.;
Ahmad, F.J. Enhancement of Quercetin Oral Bioavailability by
Self-Nanoemulsifying Drug Delivery System and their Quantifica-
tion Through Ultra High Performance Liquid Chromatography and
Mass Spectrometry in Cerebral Ischemia. Drug Res. (Stuttg.), 2017,
67(10), 564-575.
[http://dx.doi.org/10.1055/s-0043-109564] [PMID: 28561230]
[120] Rezaei-Sadabady, R.; Eidi, A.; Zarghami, N.; Barzegar, A. Intracel-
lular ROS protection efficiency and free radical-scavenging activity
of quercetin and quercetin-encapsulated liposomes. Artif. Cells
Nanomed. Biotechnol., 2016, 44(1), 128-134.
[http://dx.doi.org/10.3109/21691401.2014.926456] [PMID:
24959911]
[121] Lockhart, J.N.; Stevens, D.M.; Beezer, D.B.; Kravitz, A.; Harth, E.
Dual drug delivery of tamoxifen and quercetin: Regulated metabo-
lism for anticancer treatment with nanosponges. J Control Rele ase.,
2015, 220(Pt B), 751-757.
[122] des Rieux, A.; Fievez, V.; Garinot, M.; Schneider, Y.J.; Préat, V.
Nanoparticles as potential oral delivery systems of proteins and
vaccines: a mechanistic approach. J. Control. Release, 2006,
116(1), 1-27.
[http://dx.doi.org/10.1016/j.jconrel.2006.08.013] [PMID:
17050027]
[123] McClements, D.J.; Decker, E.A.; Park, Y.; Weiss, J. Structural
design principles for delivery of bioactive components in nutraceu-
ticals and functional foods. Crit. Rev. Food Sci. Nutr., 2009, 49 (6),
577-606.
[http://dx.doi.org/10.1080/10408390902841529] [PMID:
19484636]
[124] Rodriguez-Nogales, C.; Noguera, R.; Patrick, C.; Blanco-Prieto,
M.J.J. Therapeutic opportunities in neuroblastoma using nanotech-
nology. Journal of Pharmacology and Experimental Therapeutics,
2019, jpet.118.255067.
[http://dx.doi.org/10.1124/jpet.118.255067]
[125] Caddeo, C.; Díez-Sales, O.; Pons, R.; Carb one, C.; Ennas, G.;
Puglisi, G.; Fadda, A.M.; Manconi, M. Cross-linked chito-
san/liposome hybrid system for the intestinal delivery of quercetin.
J. Colloid Interface Sci., 2016, 461, 69-78.
[http://dx.doi.org/10.1016/j.jcis.2015.09.013] [PMID: 26397912]
[126] Rahimi, S.; Khoee, S.; Ghandi, M. Preparation and characterization
of rod-like chitosan-quinoline nanoparticles as pH-responsive
nanocarriers for quercetin delivery. Int. J. Biol. Macromol., 2019,
128, 279-289.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.01.137] [PMID:
30695722]
[127] Barbosa, A.I.; Costa Lima, S.A.; Reis, S. Application of pH-
Responsive Fucoidan/Chitosan Nanoparticles to Improve Oral
Quercetin Delivery. Molecules, 2019, 24(2), 346.
[http://dx.doi.org/10.3390/molecules24020346] [PMID: 30669398]
[128] Aroonsri Priprem, S.S. Hye-Kyung Na, Young-Joon Surh, Malyn
Chulasiri, Effect of Formulations of Nanosized Quercetin
Liposomes on COX-2 and NF-kB in MCF-10A Cells. Pharm.
Nanotechnol., 2013, 1(1), 26-34.
[http://dx.doi.org/10.2174/2211738511301010026]
16 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya
[129] Priprem, A.; Watanatorn, J.; Sutthiparinyanont, S.; Phachonpai,
W.; Muchimapura, S. Anxiety and cognitive effects of quercetin
liposomes in rats. Nanomedicine (Lond.), 2008, 4(1), 70-78.
[http://dx.doi.org/10.1016/j.nano.2007.12.001] [PMID: 18249157]
[130] Ren, J.; Fang, Z.; Jiang, L.; Du, Q. Quercetin-containing self-
assemble proliposome preparation and evaluation. J. Liposome
Res., 2017, 27(4), 335-342.
[http://dx.doi.org/10.1080/08982104.2016.1239635] [PMID:
27667265]
[131] Zheng, N.G.; Mo, S.J.; Li, J.P.; Wu, J.L. Anti-CSC effects in hu-
man esophageal squamous cell carcinomas and Eca109/9706 cells
induced by nano liposomal quercetin alone or combine d with CD
133 antiserum. Asian Pac. J. Cancer Prev., 2014, 15(20), 8679-
8684.
[http://dx.doi.org/10.7314/APJCP.2014.15.20.8679] [PMID:
25374189]
[132] Martirosyan, A.; Grintzalis, K.; Polet, M.; Laloux, L.; Schneider,
Y.J. Tuning the inflammatory response to silver nanoparticles via
quercetin in Caco-2 (co-)cultures as model of the human intestinal
mucosa. Toxicol. Lett., 2016, 253, 36-45.
[http://dx.doi.org/10.1016/j.toxlet.2016.04.018] [PMID: 27113704]
[133] Mittal, A.K.; Kumar, S.; Banerjee, U.C. Quercetin and gallic acid
mediated synthesis of bimetallic (silver and selenium) nanoparti-
cles and their antitumor and antimicrobial potential. J. Colloid In-
terface Sci., 2014, 431, 194-199.
[http://dx.doi.org/10.1016/j.jcis.2014 .06.0 30] [PMID: 25000181]
[134] Pandey, S.K.; Patel, D.K.; Maurya, A.K.; Thakur, R.; Mishra, D.P.;
Vinayak, M.; Haldar, C.; Maiti, P. Controlled release of drug and
better bioavailability using poly(lactic acid-co-glycolic acid)
nanoparticles. Int. J. Biol. Macromol., 2016, 89, 99-110.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.04.065] [PMID:
27112980]
[135] Suksiriworapong, J.; Sripha, K.; Kreuter, J.; Junyaprasert, V.B.
Functionalized (poly(ɛ-caprolactone))-poly(ethylene glycol)
nanoparticles with grafting nicotinic acid as drug carriers. Int. J.
Pharm., 2012, 423(2), 562-570.
[http://dx.doi.org/10.1016/j.ijpharm.2011.11.033] [PMID:
22155410]
[136] Suksiriworapong, J.; Phoca, K.; Ngamsom, S.; Sripha, K.; Moong-
karndi, P.; Junyaprasert, V.B. Comparison of poly(ε-caprolactone)
chain lengths of poly(ε-capro lactone)-co-d-α-tocopheryl-
poly(ethylene glycol) 1000 succinate nanoparticles for enhance-
ment of quercetin delivery to SKBR3 breast cancer cells. Eur. J.
Pharm. Bioph arm., 2016, 101, 15-24.
[http://dx.doi.org/10.1016/j.ejpb.2016.01.008] [PMID: 26802701]
[137] Gupta, A.; Kaur, C.D.; Saraf, S.; Saraf, S. Formulation, characteri-
zation, and evaluation of ligand-conjugated biodegradable quer-
cetin nanoparticles for active targeting. Artif. Cells Nanomed. Bio-
technol., 2016, 44(3), 960-970.
[PMID: 25813566]
[138] Lv, L.; Liu, C.; Chen, C.; Yu, X.; Chen, G.; Shi, Y.; Qin, F.; Ou, J.;
Qiu, K.; Li, G. Quercetin and doxorubicin co-encapsulated biotin
receptor-targeting nanoparticles for minimizing drug resistance in
breast cancer. Oncotarget, 2016, 7(22), 32184-32199.
[http://dx.doi.org/10.18632/oncotarget.8607] [PMID: 27058756]
[139] Guan, X.; Gao, M.; Xu, H.; Zhang, C.; Liu, H.; Lv, L.; Deng, S.;
Gao, D.; Tian, Y. Quercetin-loaded poly (lactic-co-glycolic acid)-
d-α-tocopheryl polyethylene glycol 1000 succinate nan oparticles
for the targeted treatment of liver cancer. Drug Deliv., 2016, 23(9),
3307-3318.
[http://dx.doi.org/10.1080/10717544.2016.1176087] [PMID:
27067032]
[140] Wang, C.; Su, L.; Wu, C.; Wu, J.; Zhu, C.; Yuan, G. RGD peptide
targeted lipid-coated nanoparticles for combinatorial delivery of
sorafenib and quercetin against hepatocellular carcinoma. Drug
Dev. Ind. Pharm., 2016, 42(12), 1938-1944.
[http://dx.doi.org/10.1080/03639045.2016.1185435] [PMID:
27142812]
[141] Bishayee, K.; Khuda-Bukhsh, A.R.; Huh, S.O. PLGA-Loaded
Gold-Nanoparticles Precipitated with Quercetin Downregulate
HDAC-Akt Activities Controlling Proliferation and Activate p53-
ROS Crosstalk to Induce Apoptosis in Hepatocarcinoma Cells.
Mol. Cells, 2015, 38(6), 518-527.
[http://dx.doi.org/10.14348/molcells.2015.2339] [PMID:
25947292]
[142] Pimple, S.; Manjappa, A.S.; Ukawala, M.; Mu rthy, R .S. PLGA
nanoparticles loaded with etoposide and quercetin dihydrate indi-
vidually: in vitro cell line study to ensure advantage of combination
therapy. Cancer Nanotechnol., 2012, 3(1-6), 25-36.
[http://dx.doi.org/10.1007/s12645-012-0027-y] [PMID: 26069494]
[143] Zhao, J.; Liu, J.; Wei, T.; Ma, X.; Cheng, Q.; Huo, S.; Zhang, C.;
Zhang, Y.; Duan, X.; Liang, X.J. Quercetin-loaded nanomicelles to
circumvent human castration-resistant prostate cancer in vitro and
in vivo. Nanoscale, 2016, 8(9), 5126-5138.
[http://dx.doi.org/10.1039/C5NR08966B] [PMID: 26875690]
[144] Pang, X.; Lu, Z.; Du, H.; Yang, X.; Zhai, G. Hyaluronic acid-
quercetin conjugate micelles: synthesis, characterization, in vitro
and in vivo evaluation. Colloids Surf. B Biointerfaces, 2014, 123,
778-786.
[http://dx.doi.org/10.1016/j.colsurfb.2014.10.025] [PMID: 25454664]
[145] Tan, B.J.; Liu, Y.; Chang, K.L.; Lim, B.K.; Chiu, G.N. Perorally
active nanomicellar formulation of quercetin in the treatment of
lung can cer. Int. J. Nanomedicine, 2012, 7, 651-661.
[PMID: 22334787]
[146] Zhao, L.; Shi, Y.; Zou, S.; Sun, M.; Lil, L.; Zhail, G. Formulation
and in vitro evaluation of quercetin loaded polymeric micelles
composed of pluronic P123 and D-a-tocopheryl polyethylene gly-
col succinate. J. Biomed. Nanotechnol., 2011, 7(3), 358-365.
[http://dx.doi.org/10.1166/jbn.2011.1298] [PMID: 21830476]
[147] Khonkarn, R.; Mankhetkorn, S.; Hennink, W.E.; Okonogi, S. PEG-
OCL micelles for quercetin solubilization and inhibition of cancer
cell growth. Eur. J. Pharm. Biopharm., 2011, 79(2), 268-275.
[http://dx.doi.org/10.1016/j.ejpb.2011.04.011] [PMID: 21596135]
[148] Sun, C.; Ding, Y.; Zhou, L.; Shi, D.; Sun, L.; Webster, T.J.; Shen,
Y. Noninvasive nanoparticle strategies for brain tumor targeting.
Nanomedicine., 2017, 13(8), 2605-2621.
[http://dx.doi.org/10.1016/j.nano.2017.07.009] [PMID: 28756093]
[149] Lainé, A.L.; Clavreul, A.; Rousseau, A.; Tétaud, C.; Vessieres, A.;
Garcion, E.; Jaouen, G.; Aubert, L.; Guilbert, M.; Benoit, J.P.;
Toillon, R.A.; Passirani, C. Inhibition of ectopic glioma tumor
growth by a potent ferrocenyl drug loaded into stealth lipid nano-
capsules. Nanomedicine., 2014, 10(8), 16 67-1677.
[http://dx.doi.org/10.1016/j.nano.2014.05.002] [PMID: 24842766]
[150] Jain, A.K.; Thanki, K.; Jain, S. Novel self-nanoemulsifying formu-
lation of quercetin: Implications of pro-oxidant activity on the anti-
cancer efficacy. Nanomedicine., 2014, 10(5), 959-969.
[http://dx.doi.org/10.1016/j.nano.2013.12.010] [PMID: 24407148]
[151] Ghosh, S.; Sarkar, S.; Choudhury, S.T.; Ghosh, T.; Das, N.
Triphenyl phosphonium coated nano-quercetin for oral de livery:
Neuroprotective effects in attenuating age related global moderate
cerebral ischemia reperfusion injury in rats. Nanomedicine., 2017,
13(8), 2439-2450.
[http://dx.doi.org/10.1016/j.nano.2017.08.002] [PMID: 28822845]
[152] Penalva, R.; González-Navarro, C.J.; Gamazo, C.; Esparza, I.;
Irache, J.M. Zein nanoparticles for oral delivery of quercetin:
Pharmacokinetic studies and preventive anti-inflammatory effects
in a mouse model of endotoxemia. Nanomedicine (Lond.), 2017,
13(1), 103-110.
[http://dx.doi.org/10.1016/j.nano.2016.08.033] [PMID: 27615118]
[153] Sedaghat Doost, A.; Kassozi, V.; Grootaert, C.; Claeys, M.; Dewet-
tinck, K.; Van Camp, J.; Van der Meeren, P. Se lf-assembly, func-
tionality, and in-vitro properties of quercetin loaded nanoparticles
based on shellac-almond gum biological macromolecules. Int. J.
Biol. Macromol., 2019, 129, 1024 -1033.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.02.071] [PMID:
30794898]
[154] Fang, J.; Nakamura, H.; Maeda, H. The EPR effect: Unique fea-
tures of tumor blood vessels for drug delivery, factors involved,
and limitations and augmentation of the effect. Adv. Drug Deliv.
Rev., 2011, 63(3), 136-151.
[http://dx.doi.org/10.1016/j.addr.2010.04.009] [PMID: 20441782]
[155] Patra, A.; Satpathy, S.; Shenoy, A.K.; Bush, J.A.; Kazi, M.; Hus-
sain, M.D. Formulation and evaluation of mixed polymeric mi-
celles of quercetin for treatment of breast, ovarian, and multidrug
resistant cancers. Int. J. Nanomedicine, 2018, 13, 2869-2881.
[http://dx.doi.org/10.2147/IJN.S153094] [PMID: 29844670]
[156] Men, K.; Duan, X.; Wei, X.W.; Gou, M.L.; Huang, M.J.; Chen,
L.J.; Qian, Z.Y.; Wei, Y.Q. Nanoparticle-delivered quercetin for
cancer therapy. Anticancer. Agents Med. Chem., 2014, 14(6), 826-
832.
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 17
[http://dx.doi.org/10.2174/1871520614666140521122932] [PMID:
24851877]
[157] Sharma, G.; Park, J.; Sharma, A.R.; Jung, J.S.; Kim, H.; Chak-
raborty, C.; Song, D.K.; Lee, S.S.; Nam, J.S. Methoxy
poly(ethylene glycol)-poly(lactide) nanoparticles encapsulating
quercetin act as an effective anticancer agent by inducing apoptosis
in breast cancer. Pharm. Res., 2015, 32(2), 723-735.
[http://dx.doi.org/10.1007/s11095-014-1504-2] [PMID: 25186442]
[158] Ahmad, N.; Banala, V.T.; Kushwaha, P.; Karvande, A.; Sharma, S.;
Tripathi, A.K.; Verma, A.; Trivedi, R.; Mishra, P.R. Quercetin-
loaded solid lipid nan oparticles improve osteoprotective activity in
an ovariectomized rat model: a preventive strategy for post-
menopausal osteoporosis. RSC Advances, 2016, 6(100), 97613-
97628.
[http://dx.doi.org/10.1039/C6RA17141A]
[159] Castangia, I.; Nácher, A.; Caddeo, C.; Merino, V.; Díez-Sales, O.;
Catalán-Latorre, A.; Fernàndez-Busquets, X.; Fadda, A.M.; Man-
coni, M. Therapeutic efficacy of quercetin enzyme-responsive
nanovesicles for the treatment of experimental colitis in rats. Acta
Biomater., 2015, 13, 216-227.
[http://dx.doi.org/10.1016/j.actbio.2014.11.017] [PMID: 25463498]
[160] Singh, S.K.; Singh, S.; Lillard, J.W., Jr; Singh, R. Drug delivery
approaches for breast cancer. Int. J. Nanomedicine, 2017, 12, 6205-
6218.
[http://dx.doi.org/10.2147/IJN.S140325] [PMID: 28883730]
[161] Gulbake, A.; Jain, S.K. Chitosan: a potential polymer for colon-
specific drug delivery system. Expert Opin. Drug Deliv., 2012,
9(6), 713-729.
[http://dx.doi.org/10.1517/17425247.2012.682148] [PMID:
22530707]
[162] Han, L.; Zhao, J.; Zhang, X.; Cao, W.; Hu, X.; Zou, G.; Duan, X.;
Liang, X.J. Enhanced siRNA delivery and silencing gold-chitosan
nanosystem with surface charge-reversal polymer assembly and
good biocompatibility. ACS Nano, 2012, 6(8), 7340-7351.
[http://dx.doi.org/10.1021/nn3024688] [PMID: 22838646]
[163] Yuan, Y.G.; Peng, Q.L.; Gurunathan, S. Silver nanoparticles en-
hance the apoptotic potential of gemcitabine in human ovarian can-
cer cells: combination therapy for effective cancer treatment. Int. J.
Nanomedicine, 2017, 12, 6487-6502.
[http://dx.doi.org/10.2147/IJN.S135482] [PMID: 28919750]
[164] Yoo, G.; Jeong, S.H.; Ryu, W.I.; Lee, H.; Kim, J.H.; Bae, H.C.;
Son, S.W. Gene expression analysis reveals a functional role for
the Ag-NPs-induced Egr-1 transcriptional factor in human kerati-
nocytes. Mol. Cell. Toxicol., 2014, 10(2), 149-156.
[http://dx.doi.org/10.1007/s13273-014-0016-9]
[165] Lin, J.; Huang, Z.; Wu, H.; Zhou, W.; Jin, P.; Wei, P.; Zhang, Y.;
Zheng, F.; Zhang, J.; Xu, J.; Hu, Y.; Wang, Y.; Li, Y.; Gu, N.;
Wen, L. Inhibition of autophagy enhances the anticancer activity of
silver nanoparticles. Autophagy, 2014, 10(11), 2006-2020.
[http://dx.doi.org/10.4161/auto.36293] [PMID: 25484080]
[166] Lin, L.; Xu, Y.; Zhang, S.; Ross, I.M.; Ong, A. C.; Allwood, D.A.
Fabrication and luminescence of monolayered boron nitride quan-
tum dots. Small, 2014, 10(1), 60-65.
[http://dx.doi.org/10.1002/smll.201301001] [PMID: 23839969]
[167] AshaRani, P.V.; Low Kah Mun, G.; Hande, M.P.; Valiyaveettil, S.
Cytotoxicity and genotoxicity of silver nanoparticles in human
cells. ACS Nano, 2009, 3(2), 279-290.
[http://dx.doi.org/10.1021/nn800596w] [PMID: 19236062]
[168] Sanpui, P.; Chattopadhyay, A.; Ghosh, S.S. Induction of apoptosis
in cancer cells at low silver nanoparticle concentrations using chi-
tosan nanocarrier. ACS Appl. Mater. Interfaces, 2011, 3(2), 218-
228.
[http://dx.doi.org/10.1021/am100840c] [PMID: 21280584]
[169] Gurunathan, S.; Jeong, J.K.; Han, J.W.; Zhang, X.F.; Park, J.H.;
Kim, J.H. Multidimensional effects of biologically synthesized sil-
ver nanoparticles in Helicobacter pylori, Helicobacter felis, and
human lung (L132) and lung carcinoma A549 cells. Nanoscale Res.
Lett., 2015, 10, 35.
[http://dx.doi.org/10.1186/s11671-015-0747-0] [PMID: 25852332]
[170] Ortega, F.G.; Fernández-Baldo, M.A.; Fernández, J.G.; Serrano,
M.J.; Sanz, M.I.; Diaz-Mochón, J.J.; Lorente, J.A.; Raba, J. Study
of antitumor activity in breast cell lines using silver nanoparticles
produced by yeast. Int. J. Nanomedicine, 2015, 10, 2021-2031.
[PMID: 25844035]
[171] Pandey, S.K.; Patel, D.K.; Maurya, A.K.; Thakur, R.; Mishra, D.P.;
Vinayak, M.; Haldar, C.; Maiti, P. Corrigendum to "Controlled re-
lease of drug and better bioavailability using poly(lactic acid-co-
glycolic acid) nanoparticles" [Int. J. Biol. Macromol. 89 (2016) 99-
110]. Int J Biol Macromol., 2018, 114, 1361.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.03.038] [PMID:
29674009]
[172] Zhang, H.; Liu, G.; Zeng, X.; Wu, Y.; Yang, C.; Mei, L.; Wang, Z.;
Huang, L. Fabrication of genistein-loaded biodegradable TPGS-b-
PCL nanoparticles for improved therapeutic effects in cervical can-
cer cells. Int. J. Nanomedicine, 2015, 10, 2461-2473.
[PMID: 25848264]
[173] Bernabeu, E.; Gonzalez, L.; Legaspi, M.J.; Moretton, M.A.; Chiap-
petta, D.A. Paclitaxel-Loaded TPGS-b-PCL Nanoparticles: In Vitro
Cytotoxicity and Cellular Uptake in MCF-7 and MDA-MB-231
Cells versus mPEG-b-PCL Nanoparticles and Abraxane®. J. Nano-
sci. Nanotechnol., 2016, 16(1), 160-170.
[http://dx.doi.org/10.1166/jnn.2016.10739] [PMID: 27398441]
[174] Bernabeu, E.; Cagel, M.; Lagomarsino, E.; Moretton, M.; Chiap-
petta, D.A. Paclitaxel: What has been done and the challenges re-
main ahead. Int. J. Pharm., 2017, 526(1-2), 474-495.
[http://dx.doi.org/10.1016/j.ijpharm.2017.05.016] [PMID:
28501439]
[175] Zheng, Y.; Chen, H.; Zeng, X.; Liu, Z.; Xiao , X.; Zhu, Y.; Gu, D.;
Mei, L. Surface modification of TPGS-b-(PCL-ran-PGA) n anop ar-
ticles with polyethyleneimine as a co-delivery system of TRAIL
and endostatin for cervical cancer gene therapy. Nanoscale Res.
Lett., 2013, 8(1), 161.
[http://dx.doi.org/10.1186/1556-276X-8-161] [PMID: 23570619]
[176] Jiang, L.; Li, X.; Liu, L.; Zhang, Q. Thiolated chitosan-modified
PLA-PCL-TPGS nanoparticles for oral chemotherapy of lung can-
cer. Nanoscale Res. Lett., 2013, 8(1), 66.
[http://dx.doi.org/10.1186/1556-276X-8-66] [PMID: 23394588]
[177] Yadav, M.K.; Maurya, A.K.; Rajput, G.; Manar, K.K.; Vinayak,
M.; Drew, M.G .B.; Singh, N. Synthesis, characterization, DNA
binding and cleavage activity of homoleptic zinc(II) β-
oxodithioester chelate complexes. J. Coord. Chem., 2017, 70(18),
3171-3185.
[http://dx.doi.org/10.1080/00958972.2017.1377835]
[178] Yadav, M.K.; Maurya, A.K.; Rajput, G.; Manar, K.K.; Vinayak,
M.; Drew, M.G.B.; Singh, N. New planar trans-copper(II) β-
dithioester chelate complexes: synthesis, characterization, antican-
cer activity and DNA-binding/cleavage studies. J. Coord. Chem.,
2017, 70(4), 565-583.
[http://dx.doi.org/10.1080/00958972.2016.1275589]
[179] Pool, H.; Quintan ar, D.; Figueroa, J.D.; Marin ho Mano , C.;
Bechara, J.E.H. God; #xed; nez, L.A.; Mendoza, S., Antioxidant
Effects of Quercetin and Catechin Encapsulated into PLGA
Nanoparticles. J. Nanomater., 2012, 2012, 12.
[http://dx.doi.org/10.1155/2012/145380]
[180] Jain, A.K.; Thanki, K.; Jain, S. Co-encapsulation of tamoxifen and
quercetin in polymeric nanoparticles: implications on oral bioavail-
ability, antitumor efficacy, and drug-induced toxicity. Mol. Pha rm.,
2013, 10(9), 3459-3474.
[http://dx.doi.org/10.1021/mp400311j] [PMID: 23927416]
[181] Balakrishnan, S.; Mukherjee, S.; Das, S.; Bhat, F.A.; Raja Singh,
P.; Patra, C.R.; Arunakaran, J. Go ld nanoparticles-conjugated quer-
cetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated
pathway in breast cancer cell lines (MCF-7 and MDA-MB-231).
Cell Biochem. Funct., 2017, 35(4), 217-231.
[http://dx.doi.org/10.1002/cbf.3266] [PMID: 28498520]
[182] Sun, M.; Nie, S.; Pan, X.; Zhang, R.; Fan, Z. ; Wang, S. Quercetin-
nanostructured lipid carriers: characteristics and anti-breast cancer
activities in vitro. Colloids Surf. B Biointerfaces, 2014, 113, 15-24.
[http://dx.doi.org/10.1016/j.colsurfb.2013.08.032] [PMID:
24060926]
[183] Sarkar, A.; Ghosh, S.; Chowdhury , S.; Pandey, B.; Sil, P.C. Tar-
geted delivery of quercetin loaded mesoporous silica nanoparticles
to the breast cancer cells. Biochim. Biophys. Acta, 2016, 1860(10),
2065-2075.
[http://dx.doi.org/10.1016/j.bbagen.2016.07.001] [PMID:
27392941]
[184] Aghapour, F.; Moghadamnia, A.A.; Nicolini, A.; Kani, S.N.M.;
Barari, L.; Morakabati, P.; Rezazadeh, L.; Kazemi, S. Quercetin
conjugated with silica nanoparticles inhibits tumor growth in MCF-
7 breast cancer cell lines. Biochem. Biophys. Res. Commun., 2018,
500(4), 860-865.
[http://dx.doi.org/10.1016/j.bbrc.2018.04.174] [PMID: 29698680]
18 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya
[185] Li, J.; Zhang, J.; Wang, Y.; Liang, X.; Wusiman, Z.; Yin, Y.; Shen,
Q. Synergistic inhibition of migration and invasion of breast cancer
cells by dual docetaxel/quercetin-loaded nanoparticles via
Akt/MMP-9 pathway. Int. J. Pharm., 2017, 523(1), 300-309.
[http://dx.doi.org/10.1016/j.ijpharm.2017.03.040] [PMID:
28336457]
[186] Umrao, S.; Maurya, A.K.; Shukla, V.; Grigoriev, A.; Ahuja, R.;
Vinayak, M.; Srivastava, R.R.; Saxena, P.S.; Oh, I.K.; Srivastava,
A. Anti-carcinogenic Activity of Blue Fluorescent h-BN Quantum
Dots: As an Effective Enhancer for DNA Cleavage Activity of
Anti-cancer Drug Doxorubicin In: Materials Today Bio; , 2019.
[187] Cabral, H.; Kataoka, K. Progress of drug-loaded polymeric mi-
celles into clinical studies. J. Control. Release, 2014, 190, 465-476.
[http://dx.doi.org/10.1016/j.jconrel.2014.06.042] [PMID:
24993430]
[188] Nishiyama, N.; Matsumura, Y.; Kataoka, K. Development of po-
lymeric micelles for targeting intractable cancers. Cancer Sci.,
2016, 107(7), 867-874.
[http://dx.doi.org/10.1111/cas.12960] [PMID: 27116635]
[189] Soleymani Abyaneh, H.; Vakili, M.R.; Zhang , F.; Choi, P.; La-
vasanifar, A. Rational design of block copolymer micelles to con-
trol burst drug release at a nanoscale dimension. Acta Biomater.,
2015, 24, 127-139.
[http://dx.doi.org/10.1016/j.actbio.2015.06.017] [PMID: 26093068]
[190] Fonge, H.; Hu ang, H.; Scollard, D.; Reilly, R.M.; Allen, C. Influ-
ence of formulation variables on the biodistribution of multifunc-
tional block copolymer micelles. J. Control. Release, 2012, 157(3),
366-374.
[http://dx.doi.org/10.1016/j.jconrel.2011.09.088] [PMID:
21982897]
[191] Bae, Y.; Kataoka, K. Intelligent polymeric micelles from functional
poly(ethylene glycol)-poly(amino acid) block copolymers. Adv.
Drug Deliv. Rev., 2009, 61(10), 768-784.
[http://dx.doi.org/10.1016/j.addr.2009.04.016] [PMID: 19422866]
[192] Matsukawa, N.; Matsumoto , M.; Shinoki, A.; Hagio, M.; Inoue, R.;
Hara, H. Nondigestible saccharides suppress the bacterial degrada-
tion of quercetin aglycone in the large intestine and enhance the
bioavailability of quercetin glucoside in rats. J. Agric. Food Chem.,
2009, 57(20) , 9462-9468.
[http://dx.doi.org/10.1021/jf9024079] [PMID: 19807098]
[193] Miyata, K.; Nishiyama, N.; Kataoka, K. Rational design of smart
supramolecular assemblies for gene delivery: chemical challenges
in the creation of artificial viruses. Chem. Soc. Rev., 2012, 41(7),
2562-2574.
[http://dx.doi.org/10.1039/C1CS15258K] [PMID: 22105545]
[194] Liu, K.; Chen, W.; Yang, T.; Wen, B.; Ding, D. ; Keidar, M.; Tang,
J.; Zhang, W. Paclitaxel and quercetin nanoparticles co-loaded in
microspheres to prolong retention time for pulmonary drug deliv-
ery. Int. J. Nanomedicine, 2017, 12, 8239-8255.
[http://dx.doi.org/10.2147/IJN.S147028] [PMID: 29180863]
[195] Mosqueira, V.C.; Legrand, P.; Gulik, A.; Bourdon, O.; Gref, R.;
Labarre, D.; Barratt, G. Relationship between complement activa-
tion, cellular uptake and surface physicochemical aspects of novel
PEG-modified nanocapsules. Biomaterials, 2001, 22(22), 2967-
2979.
[http://dx.doi.org/10.1016/S0142-9612(01)00043-6] [PMID:
11575471]
[196] Murakami, M.; Cabral, H.; Matsumoto, Y.; Wu, S.; Kano, M.R.;
Yamori, T.; Nishiyama, N.; Kataoka, K. Improving drug potency
and efficacy by nanocarrier-mediated subcellular targeting. Sci.
Transl. Med., 2011, 3(64), 64ra2.
[http://dx.doi.org/10.1126/scitranslmed.3001385] [PMID:
21209412]
[197] Mochida, Y.; Cabral, H.; Kataok a, K. Polymeric micelles for tar-
geted tumor therapy of platinum anticancer drugs. Expert Opin.
Drug Deliv., 2017, 14(12), 1423-1438.
[http://dx.doi.org/10.1080/17425247.2017.1307338] [PMID:
28290714]
[198] Takahashi, A.; Yamamoto, Y.; Yasunaga, M.; Koga, Y.; Kuroda,
J.; Takigahira, M.; Harada, M.; Saito, H.; Hayashi, T.; Kato, Y.;
Kinoshita, T.; Ohkohchi, N.; Hyodo, I.; Matsumura, Y. NC-6300,
an epirubicin-incorporatin g micelle, extends the antitumor effect
and reduces the cardiotoxicity of epirubicin. Cancer Sci., 2013,
104(7), 920-925.
[http://dx.doi.org/10.1111/cas.12153] [PMID: 23495762]
[199] Mukai, H.; Kogawa, T.; Matsubara, N.; Naito, Y.; Sasaki, M.;
Hosono, A. A first-in-human Phase 1 study of epirubicin-
conjugated polymer micelles (K-912/NC-6300) in patients with ad-
vanced or recurrent solid tumors. Invest. New Drugs, 2017, 35(3),
307-314.
[http://dx.doi.org/10.1007/s10637-016-0422-z] [PMID: 28054329]
[200] Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.;
Kimura, M.; Terada, Y.; Kano, M.R.; Miyazono, K.; Uesaka, M.;
Nishiyama, N.; Kataoka, K. Accumulation of sub-100 nm polym-
eric micelles in poorly permeable tumours depends on size. Nat.
Nanotechnol., 2011, 6(12), 815-823.
[http://dx.doi.org/10.1038/nnano.2011.166] [PMID: 22020122]
[201] Chintamani, ; Tandon, M.; Mishra, A.; Agarwal, U.; Saxena, S.
Sentinel lymp h node biopsy using dye alone method is reliable and
accurate even after neo-adjuvant chemotherapy in locally advanced
breast cancer--a prospective study. World J. Surg. Oncol., 2011, 9,
19.
[http://dx.doi.org/10.1186/1477-7819-9-19] [PMID: 21396137]
[202] Ahn, H.K.; Jung, M.; Sym, S.J.; Shin, D.B.; Kang, S.M.; Kyung,
S.Y.; Park, J.W.; Jeong, S.H.; Cho, E.K. A phase II trial of Cre-
morphor EL-free paclitaxel (Genexol-PM) and gemcitabin e in pa-
tients with advanced non-small cell lung cancer. Cancer Che-
mother. Pharmacol., 2014, 74(2), 277-282.
[http://dx.doi.org/10.1007/s00280-014-2498-5] [PMID: 24906423]
[203] Vega, J.; Ke, S.; Fan, Z.; Wallace, S.; Charsangavej, C.; Li, C.
Targeting doxorubicin to epidermal growth factor receptors by site-
specific conjugation of C225 to poly(L-glutamic acid) through a
polyethylene glycol spacer. Pharm. Res., 2003, 20(5), 826-832.
[http://dx.doi.org/10.1023/A:1023454107190] [PMID: 12751641]
[204] Reddy, S.T.; van der Vlies, A.J.; Simeon i, E.; Angeli, V.;
Randolph, G.J.; O’Neil, C.P.; Lee, L.K.; Swartz, M.A.; Hubbell,
J.A. Exploiting lymphatic transport and complement activation in
nanoparticle vaccines. Nat. Biotechnol., 2007, 25(10), 1159-1164.
[http://dx.doi.org/10.1038/nbt1332] [PMID: 17873867]
[205] Houdaihed, L.; Evans, J.C.; Allen, C. Overcoming the Road
Blocks: Advancement of Block Copolymer Micelles for Cancer
Therapy in the Clinic. Mol. Pharm., 2017, 14(8), 2503-2517.
[http://dx.doi.org/10.1021/acs.molpharmaceut.7b00188] [PMID:
28481116]
[206] Lee, K.S.; Chung, H.C.; Im, S.A.; Park, Y.H.; Kim, C.S.; Kim,
S.B.; Rha, S.Y.; Lee, M.Y .; Ro, J. Multicenter phase II trial of
Genexol-PM, a Cremophor-free, polymeric micelle formulation of
paclitaxel, in patients with metastatic breast cancer. Breast Cancer
Res. Treat., 2008, 108(2), 241-250.
[http://dx.doi.org/10.1007/s10549-007-9591-y] [PMID: 17476588]
[207] Kim, J.Y.; Do, Y.R.; Song, H.S.; Cho, Y.Y.; Ryoo, H.M.; Bae,
S.H.; Kim, J.G.; Chae, Y.S.; Kang, B.W.; Baek, J.H.; Kim, M.K.;
Lee, K.H.; Park, K. Mul ticenter Phase II Clinical Trial of Genexol-
PM® with Gemcitab ine in Advanced Biliary Tract Cancer. Anti-
cancer Res., 2017, 37(3), 1467-1473.
[http://dx.doi.org/10.21873/anticanres.11471] [PMID: 28314319]
[208] Sutton, D.; Nasongk la, N.; Blanco, E.; Gao, J. Functionalized mi-
cellar systems for cancer targeted drug delivery. Pharm. Res., 2007,
24(6), 1029-1046.
[http://dx.doi.org/10.1007/s11095-006-9223-y] [PMID: 17385025]
[209] Matsumura, Y. Preclinical and clinical studies of NK012, an SN-
38-incorporating polymeric micelles, which is designed based on
EPR effect. Adv. Drug Deliv. Rev., 2011, 63(3), 184-192.
[http://dx.doi.org/10.1016/j.addr.2010.05.008] [PMID: 20561951]
[210] Hamaguchi, T.; Kato, K.; Yasui, H.; Morizane, C.; Ikeda, M.;
Ueno, H.; Muro, K.; Yamada, Y.; Okusaka, T.; Shirao, K .; Shi-
mada, Y.; Nakahama, H.; Matsumura, Y. A phase I and pharma-
cokinetic study of NK105, a paclitaxel-incorporating micellar
nanoparticle formulation. Br. J. Cancer, 2007, 97(2), 170-176.
[http://dx.doi.org/10.1038/sj.bjc.6603855] [PMID: 17595665]
[211] Matsumura, Y. The drug discovery by nanomedicine and its clini-
cal experience. Jpn. J. Clin. Oncol., 2014, 44(6), 515-525.
[http://dx.doi.org/10.1093/jjco/hyu046] [PMID: 24755547]
[212] Wilson, R.H.; Plummer, R.; Adam, J.; Eatock, M.M.; Boddy, A.V.;
Griffin, M.; Miller, R.; Matsumura, Y.; Shimizu, T.; Calvert, H.
Phase I and pharmacokinetic study of NC-6004, a new platinum
entity of cisplatin-conjugated polymer forming micelles. Journal of
Clinical Oncology., 2008, 26(15_suppl), 2573-2573.
[213] Wang, C.; Wu, C.; Zhou, X.; Han, T.; Xin, X.; Wu, J.; Zhang, J.;
Guo, S. Enhancing cell nucleus accumulation and DNA cleavage
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 19
activity of anti-cancer drug via graphene quantum dots. Sci. Rep.,
2013, 3, 2852.
[http://dx.doi.org/10.1038/srep02852] [PMID: 24092333]
[214] Dolatabadi, J.E.N.; Jamali, A. A.; Hasanzadeh, M.; Omidi, Y
Quercetin Delivery into Cancer Cells with Single Walled Carbon
Nanotubes. Intern ati ona l Journal of Bioscience, Biochemistry and
Bioinformatics., 2011, 21(a), 25.
[http://dx.doi.org/10.7763/IJBBB.2011.V1.4]
[215] Cirillo, G.; Vittorio, O.; Hampel, S.; Iemma, F.; Parchi, P.; Cec-
chini, M.; Puoci, F.; Picci, N. Quercetin nanocomposite as novel
anticancer therapeutic: improved efficiency and reduced toxicity.
Eur. J. Pharm. Sci., 2013, 49(3), 359-365.
[http://dx.doi.org/10.1016/j.ejps.2013.04.008] [PMID: 23602995]
[216] Wang, Q.; Bao, Y.; Ahire, J.; Chao, Y. Co-encapsulation of biode-
gradable nanoparticles with silicon quantum dots and quercetin for
monitored delivery. Adv. Healthc. Mater., 2013, 2(3), 459-466.
[http://dx.doi.org/10.1002/adhm.201200178] [PMID: 23184534]
[217] Zhou, X.; Zhan g, Y.; Wang, C.; Wu, X.; Yang, Y .; Zheng, B.; Wu,
H.; Guo, S.; Zhang, J. Photo-Fenton reaction of graphene oxide: a
new strategy to prepare graphene quantum dots for DNA cleavage.
ACS Nano, 2012, 6(8), 6592-6599.
[http://dx.doi.org/10.1021/nn301629v] [PMID: 22813062]
[218] Zhang, L.; Xia, J.; Zhao, Q.; Liu, L.; Zhang, Z. Functional gra-
phene oxide as a nanocarrier for controlled loading and targeted de-
livery of mixed anticancer drugs. Small, 2010, 6(4), 537-544.
[http://dx.doi.org/10.1002/smll.200901680] [PMID: 20033930]
[219] Zhang, Y.; Wang, J.; Bai, X.; Jiang, T.; Zhang, Q.; Wan g, S.
Mesoporous silica nanoparticles for increasing the oral bioavail-
ability and permeation of poorly water soluble drugs. Mol. Ph arm.,
2012, 9(3), 505-513.
[http://dx.doi.org/10.1021/mp200287c] [PMID: 22217205]
[220] Sapino, S.; Ugazio, E.; Gastaldi , L.; Miletto, I.; Berlier, G.; Zonari,
D.; Oliaro-Bosso, S. Mesoporous silica as topical nanocarriers for
quercetin: characterization and in vitro studies. Eur. J. Pharm. Bio-
pharm., 2015, 89, 116-125.
[http://dx.doi.org/10.1016/j.ejpb.2014.11.022] [PMID: 25478737]
DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The Edito-
rial Department reserves the right to make minor modifications for further improvement of the manuscript.
... Additionally, the highly reactive and pH-sensitive nature of quercetin prone it to chemical alterations when passing through the acidic environment of the gastrointestinal tract, undergoing deprotonation, which furthers the flavonoid's lack of solubility and bioavailability [ZZM21]. In attempts to ameliorate this issue, recent studies and developments in quercetin applications encompass a wide range of drug delivery systems for the flavonoid, including lipid-based nanoparticles, polymerbased nanoparticles, and metallic nanoparticles [VM19]. In this review, it will be discussed how the numerous pharmacological properties of quercetin can be harnessed through the use of novel drug delivery systems for the potential treatment of neurodegenerative disorders. ...
... As quercetin's primary struggles are aqueous insolubility and a lack of chemical stability, research has aimed to find carriers that remedy these issues while simultaneously preserving and enhancing the flavonoid's health benefits [KTMC22]. The most successful delivery systems concerning these efforts include polymer-based, lipid-based, and metallic nanoparticles [VM19]. ...
... Utilizing delivery systems like NPs can improve plant extracts' stability and bioavailability, improving their health benefits. These NPs are widely used for enhancing drug solubility in water, absorption, circulation, retention time, and target specificity [76]. ...
Article
Background: Plants are used in medicine because they are low-cost, widely available, and have few side effects (compared to pharmacological treatment). Plants have phytocompounds with antidiabetic properties that can be delivered using nanoparticles (NPs). Objective: To describe the antidiabetic properties of green synthesized NPs (GSNPs) and their characterization methods. Methods: Three databases were searched using the terms “type 2 diabetes mellitus,” “antidiabetic effects,” “phytochemicals,” “plants,” and “nanoparticles.” Studies describing the antidiabetic effects (in vitro or animal models) of NPs synthesized by plant extracts and characterizing them through UV-Vis spectroscopy, FTIR, XRD, SEM, TEM, and DLS were included. Results: 16 studies were included. In vitro studies reported enzyme inhibition values between 11% (H. polyrhizus) and 100% (A. concinna) for alfa-amylase and between 41.1% (M. zapota) and 100% (A. concinna) for alfa-glucosidase. Animal studies with Wistar Albino rats having diabetes (induced by alloxan or streptozotocin) reported improved blood glucose, triglycerides, total cholesterol, LDL, and HDL after treatment with GSNPs. Regarding characterization, NP sizes were measured with DLS (25-181.5 nm), SEM (52.1-91 nm), and TEM (8.7-40.6 nm). The surface charge was analyzed with zeta potential (-30.7 to -2.9 mV). UV-Vis spectroscopy was employed to confirm the formations of AgNPs (360-460 nm), AuNPs (524-540 nm), and ZnONPs (300-400 nm), and FTIR was used to identify plant extract functional groups. Conclusions: GSNP characterization (shape, size, zeta potential, and others) is essential to know the viability and stability, which are important to achieve health benefits for biomedical applications. Studies reported good enzyme inhibition percentages in in vitro studies, decreasing blood glucose levels and improving lipid profiles in animal models with diabetes. However, these studies had limitations in the methodology and potential risk of bias, so results need careful interpretation.
... Similarly, experiments using aged obese mice fed a high-fat diet demonstrated that treatment with D + Q reduces SA-β-gal + adipocytes and p16 mRNA expression in the adipose tissue (Sierra-Ramirez et al. 2020). Dasatinib is a tyrosine kinase inhibitor that primarily targets senescent adipocyte progenitors, while quercetin eliminates senescent cells by inhibiting the anti-apoptotic protein BCL-xL, hypoxia-inducible factor-1α, and other aging-related anti-apoptotic proteins (Marunaka et al. 2017;Vinayak and Maurya 2019). Quercetin can interact with protein kinase C δ and Janus kinase 2 to inhibit ultraviolet (UV)-induced cyclooxygenase-2 and MMP-1 expression, as well as collagen degradation in human skin and skin fibroblasts (Shin et al. 2019b). ...
Article
Full-text available
The skin’s protective functions are compromised over time by both endogenous and exogenous aging. Senescence is well-documented in skin phenotypes, such as wrinkling and sagging, a consequence of the senescence-associated secretory phenotype (SASP) that involves the accumulation of senescent fibroblasts, chronic inflammation, and collagen remodeling. Although therapeutic approaches for eliminating senescent cells from the skin are available, their efficacy remains unclear. Accordingly, we aimed to examine the effects of dasatinib in combination with quercetin (D + Q) on senescent human skin fibroblasts and aging human skin. Senescence was induced in human dermal fibroblasts (HDFs) using approaches such as long-term passaging, ionizing radiation, and doxorubicin treatment. The generated senescent cells were treated with D + Q or vehicle. Additionally, a mouse-human chimera model was generated by subcutaneously transplanting whole-skin grafts of aged individuals onto nude mice. Mouse models were administered D + Q or vehicle by oral gavage for 30 days. Subsequently, skin samples were harvested and stained for senescence-associated beta-galactosidase. Senescence-associated markers were assessed by western blotting, reverse transcription-quantitative PCR and histological analyses. Herein, D + Q selectively eliminated senescent HDFs in all cellular models of induced senescence. Additionally, D + Q-treated aged human skin grafts exhibited increased collagen density and suppression of the SASP compared with control grafts. No adverse events were observed during the study period. Collectively, D + Q could ameliorate skin aging through selective elimination of senescent dermal fibroblasts and suppression of the SASP. Our findings suggest that D + Q could be developed as an effective therapeutic approach for combating skin aging.
... The variance in quercetin absorption may be due to factors such as quercetin derivative solubility, the type of sugar groups linked to quercetin, and the sugar coupling sites [40]. Various technical methods, including enzyme modification, nanotechnology, and target carriers, have been identified in relevant research as effective approaches for enhancing the bioavailability of quercetin [1,41,42]. ...
Article
Full-text available
Fat synthesis and lipolysis are natural processes in growth and have a close association with health. Fat provides energy, maintains physiological function, and so on, and thus plays a significant role in the body. However, excessive/abnormal fat accumulation leads to obesity and lipid metabolism disorder, which can have a detrimental impact on growth and even harm one’s health. Aside from genetic effects, there are a range of factors related to obesity, such as excessive nutrient intake, inflammation, glycometabolism disease, and so on. These factors could serve as potential targets for anti-obesity therapy. Quercetin is a flavonol that has received a lot of attention recently because of its role in anti-obesity. It was thought to have the ability to regulate lipid metabolism and have a positive effect on anti-obesity, but the processes are still unknown. Recent studies have shown the role of quercetin in lipid metabolism might be related to its effects on inflammatory responses and glycometabolism. The references were chosen for this review with no date restrictions applied based on the topics they addressed, and the databases PubMed and Web of Sicence was used to conduct the references research, using the following search terms: “quercetin”, “obesity”, “inflammation”, “glycometabolism”, “insulin sensitivity”, etc. This review summarizes the potential mechanisms of quercetin in alleviating lipid metabolism through anti-inflammatory and hypoglycemic signaling pathways, and describes the possible signaling pathways in the interaction of inflammation and glycometabolism, with the goal of providing references for future research and application of quercetin in the regulation of lipid metabolism.
... In addition, quercetin has been shown to activate Jun N-terminal kinases (JNKs) and increase JNK-dependent Bax and p53 expression in normal bronchial epithelial cells. Thus, evidence suggests that the apoptotic effects of quercetin may result from the inhibition of HSP kinases, followed by the downregulation of HSP-70 and HSP-90 protein expression [101]. ...
Article
Full-text available
Quercetin is a flavonoid with a low molecular weight that belongs to the human diet’s phenolic phytochemicals and nonenergy constituents. Quercetin has a potent antioxidant capacity, being able to capture reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive chlorine species (ROC), which act as reducing agents by chelating transition-metal ions. Its structure has five functional hydroxyl groups, which work as electron donors and are responsible for capturing free radicals. In addition to its antioxidant capacity, different pharmacological properties of quercetin have been described, such as carcinostatic properties; antiviral, antihypertensive, and anti-inflammatory properties; the ability to protect low-density lipoprotein (LDL) oxidation, and the ability to inhibit angiogenesis; these are developed in this review.
... Unformulated quercetin absorption was only 11 ng/mL. Barras et al. [137] used a liposome loaded with quercetin that increased its solubility 100-fold. However, there is no information about the precise absorption levels. ...
Article
Full-text available
Cells are separated from the environment by a lipid bilayer membrane that is relatively impermeable to solutes. The transport of ions and small molecules across this membrane is an essential process in cell biology and metabolism. Monocarboxylate transporters (MCTs) belong to a vast family of solute carriers (SLCs) that facilitate the transport of certain hydrophylic small compounds through the bilipid cell membrane. The existence of 446 genes that code for SLCs is the best evidence of their importance. In-depth research on MCTs is quite recent and probably promoted by their role in cancer development and progression. Importantly, it has recently been realized that these transporters represent an interesting target for cancer treatment. The search for clinically useful monocarboxylate inhibitors is an even more recent field. There is limited pre-clinical and clinical experience with new inhibitors and their precise mechanism of action is still under investigation. What is common to all of them is the inhibition of lactate transport. This review discusses the structure and function of MCTs, their participation in cancer, and old and newly developed inhibitors. Some suggestions on how to improve their anticancer effects are also discussed.
Article
Quercetin is a unique bioactive flavonoid, and is an excellent antioxidant and has anti-tumor effects by regulating different tumor-related processes like proliferation, apoptosis, invasion, and spread. The latest investigations reveal that quercetin may have the capability to influence DNA methylation modification, one of the primary factors in the development of tumors. Despite the fact that quercetin has significant therapeutic properties, its use as an anti-tumor medicine is constrained by its poor solubility, short half-life, and ineffective tumor targeting. Here, we review the structure and properties of quercetin, its capacity for DNA methylation modification in tumors, and the possibility of nanoscale delivery of quercetin for future tumor treatment.
Article
Full-text available
Blue fluorescent hexagonal boron nitride quantum dots (h-BNQDs) of ∼10 nm size as an effective enhancer for DNA cleavage activity of anticancer drug doxorubicin (DOX) were synthesized using simple one-step hydrothermal disintegration of exfoliated hexagonal boron nitride at very low temperature ∼ 120 °C. Boron nitride quantum dots (BNQDs) at a concentration of 25 μg/ml enhanced DNA cleavage activity of DOX up to 70% as checked by converting supercoiled fragment into nicked circular PBR322 DNA. The interaction of BNQDs with DOX is proportional to the concentration of BNQDs, with binding constant K b ∼0.07338 μg/ml. In addition, ab initio theoretical results indicate that DOX is absorbed on BNQDs at the N-terminated edge with binding energy -1.075 eV and prevented the normal replication mechanisms in DNA. BNQDs have been shown to kill the breast cancer cell MCF-7 extensively as compared with the normal human keratinocyte cell HaCaT. The cytotoxicity of BNQDs may be correlated with reduced reactive oxygen species level and increased apoptosis in MCF-7 cells, which may be liable to enhance the anticancerous activity of DOX. The results provide a base to develop BNQD-DOX as a more effective anticancer drug.
Article
Full-text available
Quercetin is a bioactive compound that is widely used in botanical medicine and traditional Chinese medicine due to its potent antioxidant activity. In recent years, antioxidant activities of quercetin have been studied extensively, including its effects on glutathione (GSH), enzymatic activity, signal transduction pathways, and reactive oxygen species (ROS) caused by environmental and toxicological factors. Chemical studies on quercetin have mainly focused on the antioxidant activity of its metal ion complexes and complex ions. In this review, we highlight the recent advances in the antioxidant activities, chemical research, and medicinal application of quercetin.
Article
Full-text available
Quercetin, an antioxidant flavonoid, has been known that it can induce the cell cycle arrest and apoptosis of hepatocellular carcinoma (HCC) cells by the stabilization or induction of p53. Here, we found that quercetin reduced the proliferation of HepG2 cells significantly, but not Huh7 cells. Interestingly, quercetin down-regulated the intracellular ROS level in HepG2 cells, but not Huh7 cells. Functional study using siRNA showed that the proliferation of HepG2 cells was still regulated by quercetin in the absence of p53. Furthermore, we confirmed the effect of quercetin on HepG2 cells by H2O2 supplementation. This study demonstrates that the antiproliferative effect of quercetin on HCC cells can be mediated by reducing intracellular ROS, which is independent of p53 expression.
Article
Full-text available
Polymeric nanoparticles based on fucoidan and chitosan were developed to deliver quercetin as a novel functional food. Through the polyelectrolyte self-assembly method, fucoidan/chitosan (F/C) nanoparticles were obtained with three different weight ratios (1/1, 3/1, and 5/1). The content of quercetin in the fucoidan/chitosan nanoparticles was in the range 110 ± 3 to 335 ± 4 mg·mL−1, with the increase of weight ratio of fucoidan to chitosan in the nanoparticle. Physicochemically stable nanoparticles were obtained with a particle size within the 300–400 nm range and surface potential higher than +30 mV for the 1F/1C ratio nanoparticle and around −30 mV for the 3F/1C and 5F/1C ratios nanoparticles. The 1F/1C ratio nanoparticle became larger and more unstable as the pH increased from 2.5 to 7.4, while the 3F/1C and 5F/1C nanoparticles retained their initial characteristics. This result indicates that the latter nanoparticles were stable along the gastrointestinal tract. The quercetin-loaded fucoidan/chitosan nanoparticles showed strong antioxidant activity and controlled release under simulated gastrointestinal environments (in particular for the 3F/1C and 5F/1C ratios), preventing quercetin degradation and increasing its oral bioavailability.
Article
The therapeutic prospective of a novel carrier based delivery of phyto-derived quercetin (QE) extracted from onion peel waste was studied in the present work. Dialdehyde cellulose (DAC) developed from sugarcane bagasse (SCB) cellulose effectively crosslinked chitosan hydrogel. The hydrogel matrices were embedded with green zinc oxide nanoparticles (ZnO NPs), phyto-synthesized using musk melon seeds. The hybrid carrier formulation was characterised using analytical techniques such as XRD, FTIR and SEM analysis. The onion peel drug (OPD) analysed for its bioactive components using HPLC was reported as a potential source of QE. Taguchi optimization for the QE drug loading in the nanohybrid hydrogel indicated a remarkable improvement by 39% in comparison to drug loading in hydrogel without ZnO NPs. The maximum QE release was obtained at an optimal drug loading condition with pH 5.0, which favoured the anticancer applications. The drug release revealed a Fickian diffusion mechanism by adopting various kinetic models. The commercial QE and QE in OPD (QE/OPD) loaded nanohybrid hydrogels were found to inhibit the growth of Staphylococcus aureus and Trichophyton rubrum strains. The biocompatibility and anticancer properties of the QE/OPD loaded nanohybrid hydrogels were established against normal L929 murine fibroblast cells and A431 human skin carcinoma cell lines respectively.
Article
Quercetin-fortified nanoparticles were prepared from almond gum (AG), a novel biological macromolecule, and Tween 80 (T80) as stabilizers and shellac (SH) as core material using an antisolvent precipitation method. The final nanoparticles were prepared by 0.67% SH, 0.02% Q, 0.5% AG and 0.1% w/v T80 using the stirring speed of 750 rpm at a dosing rate of 0.5 ml/min. The morphology of the particles was characterized using Cryo-SEM and TEM microscopy. The average particle size was 135 ± 8 nm with a polydispersity index of 0.252 ± 0.01 and an encapsulation efficiency of 97.7 ± 1.2%. At pH 7.4 (intestinal pH), quercetin-loaded nanoparticles showed significantly (p < 0.05) higher antioxidant activity compared to free quercetin while the degradation of quercetin was lower in the nanoparticles compared to free quercetin at the similar pH. Quercetin loaded in nanoparticles was successfully found to be 2 times more available for uptake than free quercetin at pH 7.4. MTT and SRB assays revealed that no significant (p > 0.05) toxicity was observed for Caco-2 cells treated with quercetin-loaded nanoparticles with a dilution factor of 100. This study provides information about the formulation of promising nanocarriers using biological macromolecules for oral delivery of bioactive compounds.
Article
Quercetin is a polyphenolic flavonoid, which is frequently found in fruits and vegetables. The antioxidant potential of quercetin has been studied from subcellular compartments, that is, mitochondria to tissue levels in the brain. The neurodegeneration process initiates alongside aging of the neurons. It appears in different parts of the brain as Aβ plaques, neurofibrillary tangles, Lewy bodies, Pick bodies, and others, which leads to Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and other diseases. So far, no specific treatment has been identified for these diseases. Despite common treatments that help to prevent the development of disease, the condition of patients with progressive neurodegenerative diseases usually do not completely improve. Currently, the use of flavonoids, especially quercetin for the treatment of neurodegenerative diseases, has been expanded in animal models. It has also been used to treat animal models of neurodegenerative diseases. In addition, improvements in behavioral levels, as well as in cellular and molecular levels, decreased activity of antioxidant and apoptotic proteins, and increased levels of antiapoptotic proteins have been observed. Low bioavailability of quercetin has also led researchers to construct various quercetin-involved nanoparticles. The treatment of animal models of neurodegeneration using quercetin-involved nanoparticles has shown that improvements are observed in shorter periods and with use of lower concentrations. Indeed, intranasal administration of quercetin-involved nanoparticles, constructing superparamagnetic nanoparticles, and combinational treatment using nanoparticles such as quercetin and other drugs are suggested for future studies.
Article
Novel chitosan–quinoline nanoparticles as anticancer drug nanocarriers were prepared using 2-chloro-3-formylquinoline and 3-formylquinolin-2(1H)-one as non-toxic modifying agents via oil–in–water nanoemulsion technique. Chitosan–quinoline nanoparticles were characterized by FT–IR, UV–vis spectrophotometry, XRD, SEM, AFM and DLS techniques. The morphological and particle size studies demonstrated that drug–loaded chitosan–quinoline nanoparticles have a regular nanorod shape and monolithic structure with the desired particle size of 141 to 174.8 nm and a negative zeta potential of −2.4 to −14.1 mV. Drug loading capacity (LC) and encapsulation efficiency (EE) were achieved using quercetin as a hydrophobic anticancer drug and were about 4.8–9.6% and 65.8–77%, respectively. The in vitro release studies displayed great pH-sensitive release behavior. Evaluation of the anticancer efficacy of quercetin loaded chitosan–quinoline nanoparticles using the in vitro cytotoxicity studies against HeLa cells indicated that the chitosan nanoparticles are a promising candidate for the anticancer drugs delivery.
Article
Neuroblastoma (NB) is the most common extracranial solid tumor preferentially occurring in preschoolers. Its characteristic aggressiveness and heterogeneous clinical behavior are especially visible in relapsed or refractory cases and hamper therapeutic success. Although the introduction of novel antitumor agents, such as dinutuximab, isotretinoin, irinotecan, or I-131- metaiodobenzylguanidine, has increased survival rates, the situation in high-risk NB remains dismal. Moreover, treatment is particularly aggressive in these patients, leading to short- and long-term toxicities. The extensive research performed using nanotechnology in recent decades has prompted its application as a therapeutic alternative to overcome some of the common limitations of conventional chemotherapy. Nevertheless, the therapeutic role of nanomedicine in pediatric tumors like NB is not fully elucidated, and to date, only albumin-bound paclitaxel nanoparticles have reached clinic stages. In this review, we summarize the current therapeutic strategies for NB with special attention to the use of nanomedicine. We also highlight the preclinical studies on passive and active targeting nanodelivery of therapeutics in experimental NB models.
Article
The bioflavonoid quercetin may prevent magnetoliposomes oxidation, preserving their stability. In this work, the interaction between quercetin and asolectin-based magnetoliposomes was investigated by monitoring the hydration degree, vibrational, rotational and translational mobility parameters of the system as well as its thermodynamic properties. The efficiency of the encapsulation of maghemite magnetic nanoparticles was detected by high resolution-continuum source flame atomic absorption spectrometry (HR-CS FAAS). The magnetic behavior of the system was studied by vibrating sample magnetometry (VSM) technique. The size and surface charge of magnetoliposomes were detected by dynamic light scattering (DLS) and zeta potential (ζ-potential) measurements. The influence of quercetin on the physico-chemical parameters of the magnetoliposomes was evaluated by Fourier transform infrared spectroscopy (FTIR), ³¹P and ¹H nuclear magnetic resonance (NMR) and differential scanning calorimetry (DSC) techniques. In vitro antioxidant and antitumoral assays were also performed for the magnetoliposomes. An insertion of quercetin into magnetoliposomes reduced the efficiency of the encapsulation of maghemite nanoparticles by 11%, suggesting a significant interaction between flavonoid and nanoparticles in a specific region of the system. Quercetin discreetly decreased the saturation magnetization of magnetoliposomes, but did not affect the superparamagnetic behavior of the system. ³¹P and ¹H NMR results showed that quercetin did not alter the inverted hexagonal system phase state but decreased lipid polar head mobility. The flavonoid also seems to reorient the choline group above the bilayer phosphate membrane plane, as indicated by ζ-potential system values. FTIR, NMR and DSC responses showed that quercetin disordered the carbonyl and the methylene regions of the magnetoliposomes. Quercetin, as the nanoparticles, seems to be located in the polar head regions of magnetoliposomes, ordering it and diminishing the lipid intermolecular communication in the membrane carbonyl and non-polar regions. The lipid peroxidation of the magnetoliposomes was prevented 8-fold by the presence of quercetin in the system. Also, the flavonoid was responsible for a 45% reduction in the viability of glioma cells. Location and interactions between quercetin and magnetoliposomes components were discussed in order to be correlated with the results of biological activity, contributing to the design of more stable and efficient magnetoliposomes to be applied as contrast and antitumoral agents.