Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

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

net/publication/334358651

Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development

Article  in  Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry - Anti-Cancer Agents) · July 2019
DOI: 10.2174/1871520619666190705150214

CITATIONS READS
11 347

2 authors:

Manjula Vinayak Akhilendra Kumar Maurya


Banaras Hindu University Banaras Hindu University
69 PUBLICATIONS   1,214 CITATIONS    20 PUBLICATIONS   317 CITATIONS   

SEE PROFILE SEE PROFILE

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

Indian Scientific Education and Technology Foundation (ISET Foundation) View project

Gut Microbial Metabolism for Colon Carcinogenesis View project

All content following this page was uploaded by Akhilendra Kumar Maurya on 19 March 2021.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.net
Anti-Cancer Agents in Medicinal Chemistry, 2019, 19, 1-19 1

REVIEW ARTICLE

Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development

Manjula Vinayak1,* and Akhilendra K. Maurya1,2

1
Biochemistry & Molecular Biology Laboratory, Centre for Advanced Study in Zoology, Institute of Science, Banaras Hindu Univer-
sity, Varanasi-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 limit its clinical application in cancer chemoprevention. A better understanding
ARTICLE HISTORY 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 nano-
conjugated quercetin. Nanoconjugated quercetin has attracted much attention due to its controlled drug release,
Received: April 29, 2019 long retention in tumor, enhanced anticancer potential, and promising clinical application. The pharmacological
Revised: May 23, 2019
Accepted: May 23, 2019 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-
DOI: cles, polymeric micelles, chitosan nanoparticles, etc.
10.2174/1871520619666190705150214
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.

Keywords: Drug delivery, bioavailability, cancer therapy, nanoparticles, quercetin, PLGA.

1. INTRODUCTION blood-brain-barrier, and poor chemical stability, its clinical applica-


tion is limited [26]. In order to overcome these inadequacies of
Although there is tremendous development in chemotherapeutic
quercetin, many new drug delivery systems have been studied and
drugs along with other therapies of cancer, the side effects caused
developed to increase solubility and dissolution, elevating bioavail-
by drugs and drug resistance, poor bioavailability, nonspecific tar-
ability, raising drug stability, controlled drug release, indorsing
geting, and low therapeutic indices remain a challenge [1, 2]. Natu-
ral phytochemicals offer an emerging strategy for chemoprevention antioxidant activity, and improving therapeutic effect [27-32].
of various diseases with lesser side effects. Several herbal ingredi-
ents and antioxidants have been reported to have anticancer effects 2. FOOD SAFETY AND SOURCES OF QUERCETIN
[3-5]. Scientific evidence suggests that phytochemicals may inhibit Quercetin is a ubiquitous bioactive flavonoid. It occurs in a
the process of carcinogenesis and reduce the burden of a few can- wide variety of fruits, vegetables, and beverages [14]. Onion, black
cers with certain limitations [6-9]. tea, red wine as well as various fruit juices are identified as rich
A recent study suggests that phytochemicals serve as dynamic dietary sources of quercetin [12]. Quercetin is estimated to be con-
surface ligands to control nanoparticle and protein interactions; of sumed approximately 25-50 mg in the daily diet [15]. Dietary in-
which nanoparticles as carriers of phytochemicals [10]. Pharmacol- take at estimated levels of quercetin is reported to have no undesir-
ogical significance of quercetin has a long history due to its poten- able health effects [33]. Quercetin has been utilized as a component
tial health-promoting effects against numerous diseases such as in food and pharmaceutical industries because of its health promis-
cardiovascular, diabetes, thrombosis, neurological disorder, and ing benefits. It has been marketed primarily as a dietary supplement
cancer as well as for longevity [11-13]. The importance of quercetin in the USA. Quercetin is commercially used as a supplement for
is realized because of its potential in anti-carcinogenic [14-17], dog foods [34]. Quercetin shows no mutagenicity/genotoxicity in
anti-inflammatory [18], anti-obesity [19], antioxidant [20-22], anti- vivo [12]. LD50 for quercetin is 160 mg and 3000 mg per kg body
viral [23], and antibacterial activities [24, 25]. Owing to its poor weight in oral and intraperitoneal dose of mice, respectively [12].
aqueous solubility, low bioavailability, difficulty in crossing the 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-
*Address correspondence to this author at the Biochemistry & Molecular cal significance, quercetin explores the possibility to fight against
Biology Laboratory, Centre for Advanced Study in Zoology, Institute of
Science, Banaras Hindu University, Varanasi-221005, India;
other cancers [36]. Cmax and Tmax for quercetin are reported as 2.3 ±
E-mail: manjulavinayak@rediffmail.com 1.5 µg/mL and 0.7 ± 0.3h, respectively [37].  

1871-5206/19 $58.00+.00 © 2019 Bentham Science Publishers


2 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya

3. STRUCTURE AND SOLUBILITY OF QUERCETIN ported to modulate various signaling molecules in the regulation of
cell proliferation, apoptosis as well as angiogenesis towards the
Quercetin (3, 3’, 4’, 5, 7-pentahydroxyflavone) is composed of suppression of murine T-cell lymphoma [3, 11, 41, 43]. We re-
3, 5, 7-trihydroxy-4H-1-benzopyran-4-one (A and C) and a 3, 4- ported the first morphological evidence to show regression in
dihydroxy-phenyl ring (B). The biochemical activity of quercetin is Dalton’s lymphoma growth in mice by quercetin treatment along
due to the presence of hydroxyl groups which exert antioxidant with improvement in longevity [41]. Being nontoxic to Dalton’s
activity by scavenging free radicals. Quercetin possesses almost all lymphoma bearing mice, quercetin exhibits cytotoxic potential to
structural elements with characteristics of an antioxidant like (i) an ascites cells [11, 41]. Cancerous cells avoid apoptosis or pro-
orthodihydroxy or catechol group in ring B (ii) a 2, 3-double bond grammed cell death and promote uncontrolled cell proliferation via
and (iii) 3- and 5-OH groups with 4-oxo group. Several factors a series of cellular events. Apoptosis is induced by extrinsic and
including pH, heat, and metal ions affect the chemical stability of intrinsic pathways. Cellular toxicity induces the intrinsic pathway
quercetin [22, 38]. Quercetin is a lipophilic compound (log P = 1.82 of apoptosis via mitochondrial activation leading to caspase 9 cas-
+/- 0.32), and it is moderately soluble in ethanol and highly soluble cade, and extrinsic pathway via receptor-mediated caspase 8 signal-
in DMSO. However, its solubility in water is only approximately ing cascade as well as via activation of NF-kB which regulates a
0.01 mg/mL at room temperature [39]. The half-life of quercetin is large number of inflammatory genes [56, 57]. Mitochondrial induc-
approximately 11-24 h. It is therefore difficult to directly incorpo- tion of apoptosis by quercetin has been reported in various cell
rate high levels of quercetin into water-based food matrix and needs lines, including nasopharyngeal carcinoma, hepatocellular carci-
to improve its solubility, bioaccumulation, and delivery into the noma, breast cancer, leukemia, and oral squamous carcinoma [11,
cellular system to target certain diseases like cancer [40]. 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]. We 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
Chemical structure of quercetin (C15H10O7). cleavage targets of caspase 3 in vivo; however, it is cleaved by dif-
ferent caspases in vitro. Quercetin also induces apoptosis via activa-
4. ANTICANCER ACTIVITY OF QUERCETIN tion of the reactive oxygen species-dependent ASK1⁄ p38 pathway
[69]. Quercetin inhibits TRPM7 channel and MAPK signaling
Anticancer activity of quercetin has been widely studied in pathway in gastric cancer [70]. It increases the levels of JNK and
several in vivo and in vitro tumor models including lymphoma [11, p53-dependent Bax in BEAS-2B cells [71]. The p53-dependent
41-44], hepatocellular carcinoma [15], breast cancer [17, 45-47], apoptosis by quercetin is reported in lung cancer A549 cells and
and colorectal cancer [48, 49]. Nanocarriers such as polymeric cervical cancer Hela cells [72, 73]. Quercetin increases ERK-
nanoparticles, liposome, lipid nanoparticles, and micelles have been mediated cell death in HL-60 cells, suppresses Twist via p38MAPK
deliberate to reduce the side effects of drugs. Co-encapsulation of signaling, and causes mitochondrial depolarization through down-
quercetin and vincristine with lipid-polymeric nanocarriers has regulation of IL-6/STAT3 signaling [74, 75]. Signaling mechanism
potential as a novel therapeutic approach to overcome chemo- of anticarcinogenic activity of quercetin is mainly confined to its
resistant lymphoma [42]. Quercetin and its natural glycosides are antioxidant and proapoptotic role via regulation of p53, caspases,
known to decrease cell proliferation and induce genetic instability cell cycle arrest, angiogenesis via COX-2, MMPs. Quercetin at-
[50, 51]. Quercetin is a Multi-Drug Resistance (MDR) modulator tenuates PI3K-AKT signaling pathway in T-cell lymphoma exposed
and thus a potential chemosensitizer [52]. It is involved in reversion to hydrogen peroxide [3]. Quercetin attenuates prostate cancer by
of MDR via transporter-mediated active efflux by modulating P-gp inhibiting anti-apoptotic and survival signaling [76]. Warburg’s
protein (glycoprotein encoded by the human MDR1 gene) and effect demonstrates the necessity of aerobic glycolysis or glycolytic
MRP1 (Multidrug Resistance Protein). Quercetin has been shown to metabolism to provide enough energy for maintenance of cancer
improve its therapeutic efficacy in combination with drugs against growth. Quercetin decreases glycolytic metabolism by lowering
different types of cancer. Combinatorial effect of quercetin and LDH-A activity in ascites cells of Dalton’s lymphoma bearing
resveratrol has been found to be antiproliferative against HT-29 mice, causing their increased susceptibility to death [41]. Glycolytic
colon cancer via induction of zinc finger protein ZBTB10 and sup- metabolism maintains acidic pH in the tumor microenvironment,
pression of oncogenic microRNA [53]. A recent study demon- which facilitates cancer growth. Acidic pH stimulates disruption of
strated that quercetin improves the synergistic anticancer efficacy in adherence junctions, metastatic potential, proteinase activity, and
combination with PI-103, rottlerin, and G0 6983 in MCF-7 and drug resistance.
RAW 264.7 cells [17]. The lower and higher dose of quercetin
Quercetin attenuates the major survival signals, like PI3K,
(0.1% or 1% quercetin, respectively) enhanced the anti-tumor ef-
AKT, and ERK in carcinoma HepG2 cells [77]. PI3K is implicated
fects of Trichostatin A (TSA) in mice [54]. The combination of
in cell survival, proliferation, and angiogenesis via activation of
quercetin with gemcitabine has been shown a synergistic effect in
AKT. Quercetin treatment leads to inactivation of PI3K suggesting
the inhibition of pancreatic cancer cells [55].
its role in the inhibition of ascite cell survival of lymphoma bearing
Targeting metastatic expansion by cell survival and prolifera- mice [41, 78]. Declined phosphorylation of BAD by quercetin in
tion in murine T-cell lymphoma (Dalton’s lymphoma), a fast- lymphoma suggests modulation of PI3K-PDK1-AKT-BAD signal-
growing and highly metastasized cancer, is a crucial target of can- ing pathway by quercetin in regression of cell survival [44, 79].
cer therapy. We highlighted a multi-factorial approach of quercetin Apart from this pathway, BAD is also modulated via ERK activated
towards lymphoma prevention in mice via modulation of cell cycle p90 ribosomal S6 kinase. GSK-3β is another downstream effector
proteins, oncogenes, and tumor suppressor genes. Quercetin is re- 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- 5. BIOAVAILABILITY OF QUERCETIN
phorylation by AKT promotes cell proliferation, whereas dephos-
phorylation of GSK-3β promotes apoptosis and suppresses tumor Nanoparticles have been serving as a tool to enhance the effec-
growth [80-82]. We reported a declined phosphorylation of GSK- tiveness of phytochemicals by increasing bioavailability [102].
3β by quercetin suggesting inhibition of cell proliferation as well as Digestion of quercetin in the human body occurs in the mouth,
NF-κB mediated cell survival in Dalton’s lymphoma ascite cells small intestine, liver, and kidneys where it undergoes glucuronida-
[44]. AKT also activates mTOR and promotes cell survival by in- tion, sulfation or methylation [8, 103-108]. Distribution of quercetin
creasing mTOR-dependant nutrient uptake [82, 83]. Hyperactivated after the intravenous and oral administration has been shown in rat.
PI3K-AKT-mTOR signaling has been well documented in a wide It entails that the intake of quercetin from daily diet can lead to the
range of cancer [3, 84-86]. AKT has been identified as master regu- accumulation of quercetin throughout the body [109]. Bioavailabil-
lators of IKK activity which activates NF-κB [87]. Oxidative stress ity of quercetin is related to its bio-accessibility. However, the pres-
is one of the major causes of NF-κB activation. Antioxidant quer- ence of sugar moieties increases bioavailability and differences in
cetin inhibits cell survival by downregulating the phosphorylation quercetin conjugated glycosides influence its bioavailability [110,
of IκBα and by declining survival factor NF-κB in DL ascite cells. 111]. Quercetin bioavailability increases when it is consumed as an
Quercetin has been shown to inhibit aflatoxin B1-induced hepatic integral food component. The size and polarities of these com-
damage in mice [88]. It protects against liver injury induced by pounds can cause difficulty crossing membranes in the gut. How-
alcohol in rats. Dietary supplement of quercetin inhibits the devel- ever, the efficacy of quercetin is limited due to its hydrophobicity,
opment of the carcinogen-induced rat mammary cancer, colonic poor gastrointestinal absorption, and instability in physiological
neoplasia, and oral carcinogenesis. PI3K-AKT-BAD pathway is medium, extensive xenobiotic metabolism in liver and intestine via
deactivated by PTEN, a phosphatase which regulates the level of glucuronidation or sulfation; which collectively contribute to low
PIP3 by its de-phosphorylation. Enhanced level of tumor suppressor oral bioavailability of quercetin [40, 112-114]. Bioavailability of
PTEN by quercetin supports the decreased activity of PI3K and quercetin obtained from onion is more due to its greater absorption
subsequent signaling pathway. PTEN is frequently lost or mutated [110]. Quercetin ingestion with brief chain fructooligosaccharide
in cancer. Induced level of PTEN by quercetin is correlated with progresses quercetin bioavailability [115]. Solid dispersions have a
upregulation of tumor suppressor p53, a principal mediator of greater surface area that promotes dissolution in the intestinal lu-
growth arrest, senescence, and apoptosis in response to a broad men, thereby promoting bioavailability of quercetin with cereal
array of cellular damage. AKT mediated oncogenic activation acts ingredients [116]. Variations in gut microbiota metabolism, dietary
as an anti-apoptotic signal via rapid destabilization of p53 [89-91]. history, genetic polymorphisms, and interindividual variation play a
We reported the anticancer effect of quercetin via induced mRNA significant role in the bioavailability of quercetin [117]. Available
expression and protein level of p53 [41]. Cancer preventive effect literature highlights that despite the tremendous anticarcinogenic
of quercetin is also validated against fatty acid synthase in liver potential, quercetin could not show clinical utility. Efficient deliv-
cancer [92]. Quercetin downregulates the Notch/ AKT/mTOR sig- ery of quercetin to target cells and its retention at the site is neces-
naling pathway in U937 leukemia cell [73]. Quercetin attenuates sary for its utilization as a therapeutical agent.
PI3K-AKT signaling pathway exposed to hydrogen peroxide in T-
cell lymphoma [3]. Quercetin attenuates AKT-mTOR pathway 6. APPROACHES FOR QUERCETIN DELIVERY
mediated autophagy induction by inhibiting cell mobility and gly- Nanomedicines have broad research and application prospects.
colysis in breast cancer [93]. There are more than 51 FDA approved nanomedicines and 77
Angiogenesis is an essential characteristic adaptation of tumor products are under consideration of clinical trials [118]. A better
growth for a continuous supply of glucose and oxygen as well as to understanding of the molecular mechanism of controlled and regu-
get rid of its waste products [94]. The angiogenic factor VEGF-A is lated drug delivery is essential for the development of novel and
required for neovascularization and for angiogenic remodeling [95]. effective therapies. Nanotechnology provides a method to create
PI3K-AKT signaling is reported to increase VEGF-A secretion by novel formulations for numerous hydrophobic drugs. Quercetin
HIF-1 dependent, or by HIF-1 independent mechanism [95]. Down- delivery through nanoparticles has pulled in much consideration for
regulated level of VEGF-A by quercetin suggests its ability to re- its improved anticancer potential and promising clinical application
gress angiogenesis in Dalton’s lymphoma bearing mice via PDK1 [40]. Utilization of delivery systems including nanoparticles, lipid-
[44]. PDK1 is vital to endothelial cell migration in response to based carriers, micelles, inclusion complexes, and conjugates-based
VEGF stimulation in a PI3K dependent manner. Further, overex- encapsulation has the potential to improve both stability and
pression of PTEN inhibits angiogenesis and tumor growth [44, 96, bioavailability and thus imposes health benefits of quercetin. These
97]. Loss of AKT1 is reported to reduce NO production and impair delivery carriers are widely used for enhancing drug solubility in
tumor angiogenesis. The inflammatory enzymes iNOS and COX-2 water, absorption, circulation, retention time, and ultimately target
are implicated in inflammatory diseases and tumor progression specificity.
[98]. We have further reported that quercetin reduces inflammatory Quercetin-containing self-nanoemulsifying drug delivery sys-
response by decreasing the activity of iNOS and COX-2 in ascite tem has improved oral bioavailability [112]. Curcio et al. showed
cells of Dalton’s lymphoma bearing mice as well as in HepG2 cells the suitability of molecularly imprinted nanospheres for con-
[15, 44]. However, both promoting and deterring actions of iNOS trolled/sustained release of quercetin using methacrylic acid and
during tumor development have been reported, probably depending ethylene glycoldymethacrylate as a functional monomer and cross-
upon the local concentration of NO within the tumor microenvi- linking agent, respectively. The antiproliferative activity of quer-
ronment [16, 99]. COX-2 and tumor suppressor p53 have been cetin has maintained without interfering with the cell viability
reciprocally regulated by quercetin in the prevention of cancer [15]. [119]. Semi-engineered water-soluble isoquercetin and maltooligo-
Such reciprocal relation is reported earlier [100]. Hepatocellular syl isoquercetin are produced by manufactured glycosylation to
carcinoma is one of the major health threats and the third leading defeat solvency challenges and restorative methodologies. Liposo-
cause among cancer-related deaths globally. Quercetin attenuates mal quercetin has significantly improved the solubility and
cell survival and cell proliferation of HepG2 cells and elicits apop- bioavailability of quercetin in MCF-7 breast cancer cell [120].
tosis by enhancing the expression of BAX and p53 through down- Regulated metabolism for nanosponge conjugated quercetin has
regulation of ROS, PI3K, PKC, and COX-2 [15]. The independent been demonstrated in breast cancer cells [121]. These delivery sys-
regulation of p53 by quercetin has been reported against hepatocel- tems are generally designed to efficiently encapsulate an apprecia-
lular carcinoma [101]. 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 biochemical target of anticancer effects of nanoconjugated quercetin.

Sr. Delivery Advantages Chemical/Polymer Effects/Targets Refs.


No. System

1 Liposomes Transporters for both Cross-linked chitosan (TPP- Quercetin shows resistance to acidic conditions and promoted [125]
lipophilic and hydro- chitosomes) the release in alkaline pH mimicking the intestinal environment [126]
philic Chitosan-quinoline pH-sensitive release behavior and promising anticancer activity [127]
particles, targetability Fucoidan/chitosan against HeLa cells
Controlled release, preventing quercetin degradation, strong
antioxidant activity
Piperine or polyethylene glycol 400 Inhibition of anti-inflammatory activities in terms of COX-2 [128, 129]
(PEG) or alginate and NF-κB in MCF-10A cells
Liposomal quercetin Improved bioavailability and cellular uptake of quercetin in [120]
MCF-7
1,2-distearoyl-sn-glycero-3- Enhanced cellular uptake and effective nanocarrier for drug [63]
phosphoethanolamine-N- delivery to a brain tumor
[methoxy(polyethylene glycol)-2000]
Phospholipids Improved intestinal absorption of quercetin [130]
Polyethyleneglycol Declination of cyclin D1, NF-κB, histone deacetylase 1 and [131]
Magnetoliposomes induction of caspase 3 in esophageal squamous cell carcinomas [30]
Efficient antitumoral agent against glioma cells
2 Silver High reactivity, Assembly of siRNA/silver Inhibition of bacterial propagation [24]
nanoparticles powerful antimicro-
Polyoxyethylene glycerol trioleate and Reduction in cytotoxicity, oxidative stress as well as improved [132]
bial activity, and low
polyoxyethylene metabolic activity of Caco-2 cells
propensity to induce
bacterial resistance Selenium Reduced cell viability of DL cells [133]
3 Poly lactic-co- Good biocompatibil- Dichloromethane Sustained release of quercetin as well as decreased cell viability [134]
glycolic acid ity, biodegradable of breast cancer cells MDA-MB231
(PLGA) polymer, better load-
Poly (ecaprolactone)- Improved cytotoxicity, cellular uptake and delivery of quercetin [135, 136]
ing, and encapsulat-
co-d-a-tocopheryl polyethylene glycol to SKBR3 breast cancer cells
ing efficacy
1000 succinate
Mannose / folate Improved cytotoxicity of quercetin on HaCaT cells and A431 [137]
Poly lactide-co-glycolide cells
Biotin Inhibits the activity and expression of P-glycoprotein in MCF- [138]
7/ADR cells
D-α-tocopheryl Induced apoptosis in HepG2 cells [139]
Arginine- glycineaspartic acid Growth inhibition of HepG2 cells [140]
Gold nanoparticles Improved cell cytotoxicity, apoptosis, and reduced histone [141]
deacetylases, reduced cell proliferation of HepG2 cells and
modulation of p21, cdk1, and AKT
Etoposide Significant increase in cytotoxicity on human lung adenocarci- [142]
noma epithelial cells A549
4 Polymeric Small size (com- Monomethoxy poly(ethylene glycol)– Improved apoptosis and cell growth inhibition in CT26 cells [139]
micelles monly <10nm), poly(ε-caprolactone)
thermodynamic
DSPE-PEG2000 Induced apoptosis as well as enhanced permeability and reten- [143]
dependability, colloi-
tion in PC-3 cells
dal dependability
Lecithin, Pluronic P123, and 1,2- Increased cellular uptake of quercetin and longer half-life in [52]
distearoyl-sn-glycero-3- MCF-7
phosphoethanolamine-N-
methoxy[poly(ethylene glycol)-2000]
Hyaluronic acid Increased cytotoxicity of MCF-7 and growth inhibition on H22 [144]
tumor-bearing mice
Monomethoxy poly(ethylene glycol)- Induced apoptosis, mitochondrial transmembrane potential [39]
poly(ε-caprolactone) change, decreased phosphorylation of p44/42 and AKT in
A2780S cells
Phosphatidylethanolamine Improved drug accumulation in A549 cancer cell line and [145]
murine xenograft model
Pluronic P123 and D-a-tocopheryl Increased IC50 values in MCF-7 cells [146]
polyethylene glycol succinate
Naphthyl or benzyl Enhanced delivery and inhibition of growth of human erythro- [147]
myelogenous leukemia cells (K562) and small lung carcinoma
cells (GLC4))
Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 5

chemical or thermal degradation. Quercetin can be released at a phatic arrangement of tumors, the nanoparticles are accumulated
controlled rate and at the site of action or within a region of the and held longer in the tumors. This phenomenon is the so-called
gastrointestinal tract [122, 123]. Therefore, the development of Enhanced Permeability and Retention (EPR) effect. In this manner,
nano-conjugated quercetin via nano-based carriers has gained much the polymeric nanoparticles are utilized to specifically deliver anti-
attention for the improvement of delivery and bioavailability of cancer drugs to the tumors. Nanoconjugated quercetin prompts EPR
quercetin. through the dual mood of delivery i.e., active or passive transport.
Subsequently, it enhances drug release, solubility & stability, pro-
7. NANO-CONJUGATED QUERCETIN tection against endosomal degradation, inhibition of MDR as well
as absorption of free quercetin in the blood vessel which leads to
Nanoparticle drug delivery system has increased impressive DNA fragmentation and programmed cell death or apoptosis at
consideration throughout the decades. Therapeutics can be captured metastatic sites as depicted in Fig. (1). Encapsulation of anti-cancer
in the nanoparticles to enhance bioavailability, to improve watery agent in the nanoparticle is recognized as a novel strategy to over-
dissolvability, to build dependability, and to control delivery [1, 16, come resistance through several mechanisms including increasing
124]. These delivery vehicles are broadly pondered in translational drug penetration into cancer cells, modulation of drug release, and
application for the revolution of anticancer action of quercetin high endocytosis phenomenon. Several approaches have been gen-
(Table 1). erated to increase the efficacy of quercetin for cancer treatment
Nanomedicine delivery system is preferred in cerebrum tumor with a unique advantage and drawback of each formulation. To
treatment utilizing glioma-particular nanoparticles [148] and fer- overcome the burden or limitations of accessibility, quercetin can
rocenyl complex (ansa-FcdiOH) through stealth Lipid Nanocap- be delivered through polymeric micelles [114, 155], liposomes [63,
sules (LNCs) [149] because of their tunable physicochemical prop- 129, 156], chitosan nanoparticles [125], silver nanoparticles [24],
erties. Quercetin stacked self-nanoemulsifying drug conveyance PLGA (Poly lactic-co-glycolic acid) [137], PLA (poly(D,L-lactic
framework (QT-SNEDDS) advances cell reinforcement movement acid)) nanoparticles, and silica nanoparticles. In addition, delivery
of quercetin against breast cancer [150]. Oral organization of quer- systems may likewise shield the bioactive compounds from being
cetin stacked polymeric nanocapsules (N1QC) has been accounted enzymatically metabolized or thermal/ light degradation and expand
for expanded cerebrum take up and mitochondrial restriction [151]. their stability [157]. Quercetin stacked solid lipid nanoparticles
Additionally, the enhanced calming impact of Zein nanoparticles enhance osteoprotective activity in postmenopausal osteoporosis
implanted quercetin was shown in the mouse model of incited en- [158]. Quercetin based solid lipid nanoparticle treatment recovers
dotoxemia [152]. Improved bioavailability of quercetin conjugated bone loss over quercetin treatment group in ovariectomized rats.
from almond gum nanoparticles without any toxicity has been stud- Co-delivery with nanoconjugated quercetin enhances doxorubicin
ied in Caco-2 cells [153]. Specifically, nanoparticles have been mediated cytotoxicity against MCF-7 breast cancer cells. Phyto-
broadly explored to deliver anticancer drugs by methods for passive some technology can improve the efficacy of chemotherapeutics
targeting. The higher amount of nano-sized particles passively per- potential of anticancerous drugs by increasing the permeability of
meates through the leaky tumor blood vessel than the tight capillary tumor cells. Poly (β-amino esters) polymers have emerged as highly
in healthy tissues [154]. Together with the insufficiency of the lym- 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. cetin liposomes to instigate the delivery of quercetin and increase
These polymeric systems can serve as pro-drug carriers for the its retention in the small intestinal surface. Alginate, an anionic
delivery of bioactive compounds which are biologically unstable biopolymer can expand encapsulation proficiency of liposome.
and suffer from poor aqueous solubility, low bioavailability such as
Formulations of nano-sized quercetin liposomes inhibit the
the antioxidant quercetin [155].
expression of COX-2 and NF-kB in MCF-10A Cells [128]. The
nano-sized quercetin encapsulated by liposomes enhanced the cellu-
7.1. Liposomes
lar uptake in MCF-7 breast cancer cells [120]. These polymers
Among different approaches to achieve intestinal drug delivery, penetrate the polar medium inside the cells and to protect them
the use of natural polymers holds clinical promises. Oral admini- against the highly toxic effect induced by cumene hydroperoxide.
stration of polymer-coated liposomes has been proposed for the Liposomes build up especially at tumor sites due to their ability to
targeted delivery of drugs to the inflamed intestinal mucosa [159]. extravasate through pores in the capillary endothelium. These pores
Liposomes have attractive biological properties including general seem to be important for rapid angiogenesis in tumors. Quercetin
biocompatibility, biodegradability, and ability to encapsulate both liposomes and spherical nanoparticles consisting of a phospholipid
hydrophilic and hydrophobic therapeutic agents. Encapsulation of bilayer with an aqueous compartment, own the unique capability to
hydrophobic drug into liposome can create aqueous intravenously effectually deliver both water-soluble chemical antioxidants in the
injectable formulations. In the meantime, liposomes can regulate aqueous phase and lipid-soluble chemical antioxidants in the lipid
drug release and protect the encapsulated drugs from undesired bilayer. Their surface can be conjugated with specific ligands that
effects of external conditions. Doxil is the first liposomal drug for- target liposomes to their location of action. The size, composition,
mulation approved by the Food and Drug Authority for the treat- surface charge, and other formulation possessions of liposomes is
ment of AIDS associated with Kaposi’s sarcoma. Encapsulation of well documented to encounter the necessities of specific conditions.
quercetin by liposome enhances quercetin solubility and improves Additionally, it is believable that gene therapy medications might
anticancer activity through longer exposure to cancer cells with a be delivered by liposomes. Nanoparticles with entrapped siRNA
high drug concentration. Cancer therapy with a combination of inhibit bacterial propagation [24, 162]. Quercetin loaded magnetol-
drugs is an important strategy in clinical use. Liposomal quercetin iposomes are designed to study as a stable and efficient antitumoral
enhances the sensitivity of cancer to thermotherapy and thermo- agent against glioma cells [30].
chemotherapy [64]. Liposome conjugated quercetin inhibits the
upregulation of hsp70 and enhances apoptosis induced by hyper- 7.2. Silver Nanoparticles
thermia and thermochemotherapy. However, systemic administra-
Silver nanoparticles (AgNPs) have numerous applications in
tion of liposomal quercetin could sensitize CT26 cells to ther-
healthcare, food packaging and conservation, and household mate-
motherapy and chemothermotherapy. Liposomal quercetin demon-
strates the most effective tumor growth inhibition in the JIMT-1 rials. With their increasing use, human exposures to silver nanopar-
human breast tumor xenograft as compared to monotherapy and ticles become the current concern with potential unexpected health
free vincristine/quercetin combinations [160]. The most widely complications mainly due to their high reactivity. The toxic effects
used strategy is to develop controlled drug released liposomes by of silver nanoparticles on human cells and the environment have
coating the surface with inert and biocompatible polymers such as been extensively studied [163]. Combined treatment with gemcit-
Polyethylene Glycol (PEG). PEG is an exceptionally hydrated abine and AgNPs caused increased cytotoxicity and apoptosis in
adaptable polymer chain that can enhance retention time by stabiliz- A2780 cells [163]. However, normally silver toxicity has been well
ing and shielding liposome from opsonisation. Moreover, PEG is detoxified by small periplasmic silver-binding proteins, which bind
non-toxic and non-immunogenic, making it suitable for clinical silver at the cell surface and by efflux pumps propels the incoming
applications. Liposomal quercetin is highly accumulated and has metals and protects the cytoplasm from toxicity. The organic matrix
long retention in liver, tumor, and spleen rather than kidney and contains silver binding proteins that provide amino acid moieties,
lung. In addition, liposomal quercetin demonstrated significant which serve as nucleation sites for the formation of silver nanopar-
tumor growth inhibition activity in vivo [64]. Many cancer active ticles. Silver nanoparticles are the most widely explored nano-
targeting liposomes are developed to deliver quercetin and to fur- agents because of their broad antimicrobial spectrum with powerful
ther improve the accumulation of quercetin in cancer cells. Liver- antimicrobial activity and low propensity to induce bacterial resis-
specific liposomal quercetin is prepared by galactosylation. Target- tance [25]. AgNPs provide extremely attractive scaffolds for the
ing the galactosyl receptors on the surface of hepatic cells allowed creation of transfection agents as they exhibit several advantages
liver-specific delivery of quercetin, whereas intravenous admini- including bioinertia, ready synthesis, and easy functionalization
stration of galactosylated liposomal quercetin demonstrated higher [138, 164-166]. Quercetin conjugated with silver nanoparticles
inhibitory activity on the development of hepatocarcinoma and on shows a synergistic effect on antibacterial activity [24]. However,
oxidative damage in rat liver as compared to free quercetin [156]. silver nanoparticles conjugated quercetin applies negligible impact
on numerous sorts of drug-resistant bacteria including Pseudo-
Customary liposomes can be barely utilized, due to their resis-
monas aeruginosa and Bacillus subtilis. Quercetin being a charac-
tance to gastric pH, enzymatic debasement, biocompatibility, bio-
teristic natural antioxidant compound can possibly conjugate with
degradability, biorenewability, and bioadhesion but they can be
silver nanoparticles pertinent in the treatment of cancer. Numerous
effortlessly ensured by a polymeric coating [161]. Chitosan is util-
ized to coat polyethylene glycol containing vesicles for controlled parameters like reducing agents, temperature, concentration of salt
delivery of quercetin in colon cancer [159, 161]. Cross-linked chi- and microenvironment affect the conjugation of quercetin and silver
tosan-liposome combination system speaks to a promising combi- nanoparticles. Quercetin loaded AgNPs reduces cytotoxicity, oxida-
nation of nanovesicles and for delivery of quercetin into the diges- tive stress as well as increases the metabolic activity of quercetin in
tive system [125]. Quercetin loaded chitosan-quinoline nanoparti- Caco-2 cells [132]. Silver nanoparticles in conjugation with sele-
cles display pH-sensitive release behavior and promising anticancer nium improve drug delivery of quercetin and reduce cell viability of
activity against HeLa cells [126]. The quercetin loaded fu- Dalton’s lymphoma cells [133].
coidan/chitosan nanoparticles has shown to exhibit controlled re- AgNPs not only induce apoptosis but also sensitize cancer cells.
lease, preventing quercetin degradation, strong antioxidant activity, The anticancer property of starch-coated silver nanoparticles was
and increasing its oral bioavailability [127]. Quercetin conjugated studied in normal human lung fibroblast cells (IMR-90) and human
liposomes have been utilized to provide quercetin in rodent brain glioblastoma cells (U251). AgNPs induced alterations in metabolic
[63, 129]. Piperine is among different ingredients joined with quer- 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- be considered such as the compatibility and interaction of polymer
age. Among these two cell types, U251 cells show more sensitivity and drug [135]. A few studies have established the investigation of
than IMR-90 [167]. The cellular uptake of AgNPs occurred mainly P (CL)-TPGS copolymers with and without other co-monomers for
through endocytosis. AgNPs treated cells exhibited various abnor- the delivery of genistein, paclitaxel, and TRAIL/endostatin [172-
malities, including upregulation of metallothionein, downregulation 176]. Quercetin-PLGA nanoparticles can be used as effective drug
of major actin-binding protein, filamin, and mitotic arrest [167]. delivery systems for skin cancer treatment encompassing natural
The analysis of cancer cell morphology suggests that biologically drugs [137].
synthesized AgNPs could significantly induce cell death. Silver
Metastatic breast cancer is the fundamental driver of death from
nanoparticles were used to target breast-cancer cells (SKBR3) and
breast cancer. We have reported the efficacy of trans-copper (II) β-
leukemia cells (SP2/O). Chitosan-based nanocarrier (NC) delivery
dithioester complexes and homoleptic zinc (II) complexes against
of AgNPs induces apoptosis at very low concentrations [168].
breast cancer [177, 178]. However, medicines are not focused on or
Lower concentrations of nanocarriers with AgNPs showed better
successful at this stage probably because of the presence of Breast
inhibitory results than AgNPs alone. Chitosan-coated silver
Cancer Stem Cells (BCSCs). The propinquity of BCSC is the criti-
nanotriangles (Chit-AgNTs) show an increased cell mortality rate.
cal explanation behind resistance and failure of therapy. As BCSC
The anticancer property of bacterial -AgNPs and fungal extract-
originates from normal breast stem cells and having self-renewal,
conjugated AgNPs (F-AgNPs) has been demonstrated in human
high proliferation rate, ability to generate heterogeneity, etc. fur-
breast cancer MDA-MB-231 cells. Both biologically produced
ther, Mesenchymal to Epithelial Transition (MET) is important for
AgNPs exhibited significant cytotoxicity. Recent studies indicate
invasion, intravasation, circulation and extravasation; and ulti-
cell-specific toxicity by AgNPs. Plant extract-mediated synthesis of
mately leads to colonization of metastatic cells. Afterward, Lungs,
AgNPs showed more pronounced toxic effect in human lung carci-
bone, liver, and brain are the main site of metastasis for breast can-
noma cells (A549) than non-cancer cells like human lung cells,
cer. Nanoconjugated quercetin in this regard has attracted much of
indicating that AgNPs could target cell-specific toxicity, probably
interest due to effective therapeutic approaches against various
based on the lower pH in the cancer cells [169]. AgNPs showed
molecular targets during breast cancer metastasis in vitro as well as
significant toxicity in MCF7 and T47D cancer cells by higher en-
in vivo system [120, 134, 157, 179-185] as depicted in Fig. (2). The
docytic activity than MCF10-A normal breast cell line [170]. The
combination of a chemotherapeutic drug with a chemosensitizer has
nanoconjugated quercetin through silver nanoparticles can be em-
emerged as a promising strategy for cancers showing multidrug
ployed in the treatment of tumor models via influencing its cytotox-
resistance. The improved synergistic efficacy of doxorubicin with
icity, delivery, and targeting site. However, further preclinical and
Boron Nitride Quantum Dots (BNQDs) has been reported in terms
clinical study is needed to establish a chemotherapeutic agent
of anticancer activity via DNA cleavage, ROS accumulation, and
against a certain form of cancer.
interaction of doxorubicin-BNQDs with DNA in MCF-7 cells [186].
Food-related applications of silver nanoparticles need to gain a Biotin conjugated poly (ethylene glycol)-b-poly(ε-caprolactone)
better understanding of the cellular and molecular mechanisms nanoparticles encapsulating the chemotherapeutic drug doxorubicin
involved in their interaction with the cells of the gastrointestinal and the chemosensitizer quercetin (BNDQ) has a potential role in
tract as well as with gastrointestinal fluids, food matrix, microflora, the treatment of drug-resistant breast cancer [138]. BNDQ is more
etc. Therefore, the safety of nanoparticles in the food matrix and effectively taken up with less efflux by doxorubicin-resistant MCF-
cancer cells remains to be explored and further understood. 7 breast cancer cells (MCF-7/ADR cells) than by the cells treated
with the free drugs or non-biotin conjugated nanoparticles. BNDQ
7.3. PLGA (Poly Lactic-Co-Glycolic Acid) exhibited clear inhibition of the activity and expression of p-
glycoprotein, an MDR marker in MCF-7/ADR cells [138]. Quer-
Combination of arginine-glycineaspartic acid - sorafenib- quer- cetin-chitosan conjugate has been studied for oral delivery of
cetin (RGD-SRF-QT) nanoparticles could provide a promising doxorubicin to improve its oral bioavailability by increasing its
platform for co-delivery of multiple anticancer drugs for the water solubility, opening tight junction and bypassing the P-
achievement of combinational therapy and could offer the potential glycoprotein in Caco-2 cells [118]. Quercetin loaded within
for enhancing the therapeutic efficacy on hepatocellular carcinoma poly(lactic‐co‐glycolic acid) biodegradable nanoparticles, plays
[140]. Anticancer efficacy of poly (lactic acid)-quercetin nanoparti- a major role in drug targeting to tumors due to its ability to interact
cles has been reported for sustained release kinetics revealing novel with CD44 receptor [55]. Chitosan is used to coat polyethylene
vehicle for the treatment of breast cancer [134, 171]. Quercetin- glycol containing vesicles for controlled delivery of quercetin in
loaded poly (lactic-co-glycolic acid)-d-α-tocopheryl polyethylene colon cancer [159]. Cross-linked chitosan-liposome fusion system
glycol succinate nanoparticles could be used as a potential intrave- represents a promising combination of nanovesicles and for delivery
nous dosage for treatment of liver cancer owing to the enhanced of quercetin into intestine [125]. Quercetin conjugated liposomes
pharmacological effects of quercetin with increased liver targeting have been used to deliver quercetin in rat brain [129].
[139]. Tamoxifen (Tmx) embedded PLGA nanoparticles (PLGA-
Tmx) is prepared to evaluate its better DNA cleavage potential, 7.4. Polymeric Micelles
cytotoxicity using Dalton’s lymphoma ascite cells and MDA-
MB231 breast cancer cells. PLGA-Tmx shows excellent DNA Polymeric micelles have attracted enormous attention with suc-
cleavage potential as compared to pure Tamoxifen raising better cess in clinical studies of cancer targeting. Polymeric micelles are
bioavailability. Sustained-release kinetics of PLGA-Tmx nanoparti- constructed through self-assembly of block copolymers as nano-
cles shows much better anticancer efficacy through enhanced DNA scaled drug carriers. These micelles have a core-shell structure
cleavage potential and nuclear fragmentation and, thereby, reveals a where the drug-loaded core is encompassed by a biocompatible
novel vehicle for the treatment of cancer [134]. Poly (e- PEG shell of 10-100nm [187]. Engineering the micelle-forming
caprolactone) (P(CL)) is one of the biodegradable and biocompati- block copolymers endowed polymeric micelles with smart on-
ble polyester polymers. The cellular uptake of P (CL)-TPGS demand functionalities such as environment-sensitivity and target-
nanoparticles by SKBR3 cells is reported through cholesterol- ability. Polymeric micelles have been considered as the most prom-
dependent endocytosis. The P (CL)-TPGS nanoparticles show im- ising nano-carriers because their critical size, drug incorporation
proved toxicity and uptake efficiency and could be potentially used efficiency, stability, and release rate can be squeezed by setting up
for the delivery of quercetin to breast cancer cells [136]. The retar- the basic block copolymers. Utilization of polymeric micelles has
dation of drug release from the nanoparticles depends on tempera- been reported in some tumor models and in some clinical studies
ture and crystallinity of the polymer. However, other factors must [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

Fig. (2). Targeting metastasis breast cancer through nanoconjugated quercetin. Metastasis breast cancer is the fundamental driver of death from breast cancer.
Medications are not focused 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 important for invasion, intravasation, circulation, 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 system.

the third leading cause of cancer-related death globally. The en- cells, avoiding the recognition of the micelle by the reticulo-
hanced delivery by quercetin conjugated monomethoxy poly (ethyl- endothelial system in the bloodstream [195]. Therefore, polymeric
ene glycol)–poly(ε-caprolactone) MPEG-PCL nanomicellar system micelles can display prolonged circulation with a half-life longer
in the CT26 tumor model, indicates a promising potential applica- than 10h. The polymeric micelles separate into the constituent
tion against colorectal tumor chemotherapy [114]. Further, quer- square copolymers, the extent of which is beneath the edge of
cetin loaded nanomicelles significantly increase drug accumulation glomerular discharge, in this way keeping away from long haul
at the tumor site and exhibit superior anticancer activity in prostate amassing in the body. Long-circulating polymeric micelles success-
cancer [143]. fully aggregate in large tumor because of the expanded brokenness
of tumor neo vasculature and impeded lymphatic seepage. Subse-
Biodegradable polyesters such as poly (D, L-lactide-co-
quently, polymeric micelles release incorporated drugs in a sus-
glycolide), poly (D, L-lactide), poly (ecaprolactone), and long-chain
tained or microenvironment responsive manner [187, 191]. In this
alkyl derivatives appear to be widely used as a core-forming poly-
manner, polymeric micelles can accomplish tumor-specific drug
mer [189, 190]. The utilities of polymeric micelles have been ac-
activity while limiting side effects in normal tissues [187]. Contin-
counted for in experimental tumor models in mice as well as in
gent upon the definitions, polymeric micelles are masked by malig-
clinical examinations. In this manner, polymeric micelles can un-
nancy cells, and after that apply the impact of drug in an organelle-
derstand protected and successful treatment, and offer personalized
specific way [191]. In such a way, polymeric micelles potentially
meds for individual patients. Various molecular interactions such as
circumvent drug efflux or intracellular detoxification mechanisms,
hydrophobic interactions, hydrogen bonding, electrostatic interac-
overcoming drug resistance in cancer cells [196]. Encapsulated
tion and metal complex formation in the core-forming segments can
polymeric micelle in conjugation with curcumin attenuates the pro-
be a driving force of the formation of polymeric micelles [191-193].
gression of colon cancer. Polymeric micelles are formed from vari-
A wide range of therapeutic molecules including hydrophobic sub-
ous block copolymers. Poly (ethylene glycol) is generally utilized
stances, charged compounds, and metal complexes can be stably
as a shell-shaping polymer because of its hydration property and
and efficiently incorporated into the micellar core, and their release
large excluded volume impact averting cooperation with serum
can be controlled in a sustained or environment-sensitive manner.
proteins. The micellar core is composed of a variety of synthetic
Several micelles formulations of anticancer drugs are currently
polymers, which critically affect the critical properties of polymeric
under evaluation in preclinical and clinical studies [187]. Novel
micelles as drug vehicles, including size, association number, criti-
nanoparticles containing polymeric microspheres loaded with pacli-
cal micelle concentration, drug loading and release, and stability in
taxel and quercetin utilize as a promising pneumonic delivery sys-
the bloodstream [187]. Development of α-helix of poly (L-
tem for combined chemotherapy [194].
glutamate) may incredibly add to delayed blood distribution and
Because of characteristic core-shell structures and limited size accumulation of cisplatin-stacked micelles in tumors under stage III
dispersions in the scope of 10-100nm, the drug-loaded center of clinical assessment [197]. The technique for drug incorporation can
polymeric micelles can hinder interaction with plasma proteins and be characterized into 'non-covalent' and 'covalent' convention. In
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- vitro and in vivo antitumor activities against oxaliplatin-safe tumor
pounds are physically ensnared into the micelle center by dialysis, cells [196].
ultrasound-helped scattering or as oil in water emulsion. A rela-
tively high drug loading capacity of approximately 20% can be 7.4.3. Nucleic Acid Conjugated Micelles
achieved without a chemical modification of drug molecules. For Nucleic acids-based drugs such as plasmid, antisense DNA, and
successful drug incorporation, the similarity between a drug mole- siRNA can be incorporated into polymeric micelles through polyion
cule and the core shaping fragments should be compared. Addition- complex formation between negatively charged nucleic acids and
ally, the properties of the center shaping sections, for example, positively charged PEG-β-PAA copolymers. Polymeric micelles
hydrophobicity, the glass transition temperature, level of crystallin- immensely enhance the stability of nucleic acids-based drugs under
ity, and optional structure (α-helix development) are vital as they in vivo conditions, prompting delayed blood circulation. The clini-
fundamentally influence the productivity and limit of drug stacking cally approved polyethylene glycol-coated (PEGylated) liposomes
and its discharge conduct. such as Doxil and albumin nanoparticles named Abraxane have the
Drug molecules are chemically conjugated to the core-forming measure of roughly 100nm. These polymeric micelles are accumu-
segments in the covalent drug loading system. For drug conjuga- lated in the tumor due to the EPR impact. Transport of nanoparti-
tion, the environment-responsive cleavable linkage is mistreated to cles depends on the origin of tumor cells and the microenvironment.
guarantee the drug release at the target site [191]. Because the tu- Solid tumors have a pore cut-off size bigger than 200nm apart from
mor microenvironment is known to develop an acidic condition due some malignancies, for example, glioblastoma (GBM). In such
to the production of lactate by predominant anaerobic glycolysis in manner, pancreatic tumors and diffuse-type gastric cancers (scir-
cancer cells (Warburg effect), the corrosive cleavable linkage, for rhous gastric diseases) have trademark histological highlights by
example, hydrazone bond is valuable for tumor-particular drug less porous vasculature with pericyte scope and thick fibrosis,
discharge [191]. The PEG-β-PAA copolymers are helpful for on- which may be a hindrance to extravasation and intrusion of
request drug incorporation because of the opportunity of the choice nanoparticles [200]. Co-administration of 30 and 70nm micelles in
of amino acids and flexible side chain adjustment. mice bearing tumors uncovered that 30-nm micelles demonstrate a
uniform intratumoral microdistribution while 70nm micelles indi-
7.4.1. DOX Conjugated Micelles cate heterogeneous limitation at perivascular districts [200]. The
The ‘non-covalent’ drug incorporation through chemical conju- gathering and entrance of polymeric micelles in pancreatic malig-
gation of doxorubicin (DOX) to the side chain of PAA by a stable nancy models rely upon their size, as the 30nm micelles can avoid
amide linkage resulted in loss of cytotoxic movement of conjugated the difficulties in transvascular transport and infiltrate tumor
DOX yet stabilized the physical capture of free DOX through the p- stroma. The 30nm DACHPt-stacked micelles demonstrated strik-
p bond between the anthracycline structures of conjugated and un- ingly drawn out survival in malignancy bearing mice and the com-
conjugated drugs [188, 198]. In this way, improvement of micellar parable impact was seen in gastric tumor. The extent of polymeric
core-forming portions relying upon drug molecule is possible. DOX micelles is additionally vital for focusing on tumor metastasis.
was conjugated through the hydrazone bond between the carbonyl Lymph nodes are regular courses for metastasis in a few tumors. It
gathering at C13 of DOX and the hydrazide bunch acquainted with is realized that nearby organization of nanoparticles to primary
poly (D, L-aspartate). The DOX conjugated square copolymers tumors prompts collection in the neighboring lymph node; however,
shaped polymeric micelles, which indicated acidic pH-responsive the lymphatic vessels are clinically associated in the sentinel lymph
DOX discharge. Currently, the micellar formulation of a less car- node biopsy [201]. The sentinel lymph node is defined as the first
diotoxic epimer, epirubicin (code name NC-6300/ K-912) is under lymph node or group of nodes draining metastasizing cancer cells
phase I clinical study [199]. Co-delivery with nanoconjugated quer- from the tumor. Lymph node metastasis is one of the most impor-
cetin upgrades DOX mediated cytotoxicity against MCF-7 cells. tant prognostic signs as it determines if cancer has spread beyond a
Monoclonal antibody (MAb604.107) builds the affectability of primary tumor into the very first draining lymph node or not. Senti-
doxorubicin and shows higher affection for Notch1 in T acute lym- nel lymph node biopsy has been considered as a standard of care in
phoblastic leukemia (T-ALL) [157]. early breast cancers [201]. Polymeric micelles can accumulate se-
lectively in lymph node metastases through the blood vascular
7.4.2. Metal Conjugated Micelles route, which is believed to be specific to the active recruitment of
Notwithstanding hydrophilic particles, metal complexes incor- lymphocytes to lymph nodes. Organization of COX-2 inhibitor
porated PEG-β-PAA micelles are complexed with PEG-β-poly(L- (celecoxib) brought about a huge reduction in collection of polym-
glutamate) through Pt(II)-carboxylate complex formation, leading eric micelles in pre angiogenic metastases; therefore, the inflamma-
to the formation of narrowly distributed micelles with the size of 30 tory microenvironment seems to be a mechanism for the retention
nm [187, 200]. In these frameworks, the reversible ligand exchange of micelles in the metastatic niche.
reaction of Pt (II) allows the ideal influx of dynamic platinum com- 7.4.4. Antibody Conjugated Micelles
plexes from the micelles, warranting their intense cytotoxic activity.
After systemic administration, cisplatin-loaded micelles were re- Antibody and its fragments are likewise valuable as targetable
vealed to show prolonged circulation and effective tumor accumu- ligands to design effectively targetable micelles. Antibody conju-
lation, achieving remarkable in vivo antitumor efficacies with re- gated micelles (immunomicelles) have been accounted against os-
duced side-effects. Currently, CDDP and DACHPt-loaded micelles teopontin, an epidermal development factor receptor for delivery of
(code names NC-6004 and NC-4016) are under phase III and phase antitumor drugs. Immunomicelles are conjugated with antagonistic
I clinical studies, respectively [197]. Intracellular drug release using to Tissue Factor (TF) antibody parts, a known essential initiator of
the vehicles may enhance the drug potency. For example, N-(2- blood coagulation, and assumes an imperative part in tumor multi-
hydroxypropyl) methacrylamide copolymer-DOX conjugate and plication, intrusion, and metastasis. Articulation level of TF on
pH-responsive DOX stacked polymeric micelles were accounted to cancer is related to the patient's prognosis. Anti-TF antibody frag-
conquest the DOX-hindrance in cancer cells because of intracellular ment conjugated micelles incorporating epirubicin and DACHPt
drug release. Micelles accelerate drug release in low pH conditions were efficiently internalized by TF-over expressing cancer cells and
emulating the late endosomal condition. Confocal microscopic showed superior in vitro and in vivo antitumor activity to non-
observation uncovered that DACHPt loaded micelles accomplished targeted micelles [202]. Immunomicelles can deliver several drug
intracellular drug release under both in vitro and in vivo conditions particles per antibody which increase the choice of anticancer drugs
[196]. Thusly, DACHPt-stacked micelles demonstrated excellent in 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- imaging due to small size with special electrical and electronic
ing Antibody Drug Conjugates (ADCs) have indicated much poten- properties. Quantum dots have been connected in the biomedical
tial in growth treatment by tumor-focused on delivery of cytotoxic field because of subordinate optical properties, high fluorescence
drugs. Targeted delivery of DOX through immune polymeric mi- quantum yields, amazing reliability against photobleaching, and
celles where antibody C225 against EGFR is coupled with poly (L- lower toxicity. Certain types of quantum dots such as boron nitride
glutamic acid)-co-PEG, shows improved activity in inhibiting the quantum dots [165, 166, 186] and graphene oxide quantum dots
growth of A431 cells than free DOX [203]. Coordination of anti- [213, 216, 217] are being studied for their fluorescence properties,
bodies to drug stacked nanocarriers expands the appropriateness of lower cytotoxicity, and increased accumulation in cancer cells;
antibodies to an extensive variety of therapeutics. Antibody frag- which can provide sufficient information for drug delivery. Gra-
ment-installed polymeric micelle in conjugation with maleimide- phene oxide has been implicated for controlled loading and targeted
thiol has been studied for the treatment of pancreatic cancer [202]. delivery of doxorubicin in MCF-7 breast cancer cells [218]. Re-
However, there is a limitation of loads that can be delivered through cently, silicon quantum dots possess strong luminescence character-
antibody drug conjugates. The measure of drugs that can be deliv- istics as well as low inherent cytotoxicity compared to conventional
ered by a solitary antibody is the real test being developed of anti- heavy metal quantum dots, thus making them a potential for bio-
body drug conjugates, since over-burdening may lessen the binding logical applications. Quercetin encapsulated silicon quantum dots
affinity of the antibody or affect pharmacokinetics, solubility, and results in red fluorescent, inhibits hydrogen peroxide-induced DNA
stability of the drug. Generally, 2 to 4 drugs for each antibody can damage, allows monitoring of delivery, improvement of aqueous
be presented in a neutralizer drug conjugate for achieving a suc- solubility and enhanced biocompatibility in HepG2 hepatocarci-
cessful therapeutic response. Certain cytotoxic drugs such as aur- noma cells [213]. Quercetin encapsulated silicon quantum dots emit
istatins, maytansines, and calicheamicins have been conjugated to red fluorescence allowing monitoring of delivery, inhibits hydrogen
the antibody, which are thousand-fold higher cytotoxic than typical peroxide-induced DNA damage, improves aqueous solubility,
anticancer drugs; however, there are some concerns of side effects and enhanced biocompatibility in HepG2 hepatocarcinoma cells
[196, 200, 204]. This antibody conjugated nanoparticles offer better [213, 216].
delivery system achieving superior therapeutic efficacy while re-
Utilization of silica nanoparticles as controlled drug delivery
ducing side effects.
system has been studied because of their high stability, high surface
During the last two decades, polymeric micelles as drug vehi- area and better ability to functionalize by various ligands, high drug
cles have made rapid progress and several formulations are already loading efficiency and low in vivo toxicity. The increased oral
being evaluated in late-stage clinical trials. The approval of these bioavailability of drug after loading with mesoporous silica
micellar anticancer drugs and generation of innovative micellar nanoparticles has been studied [219]. The US Food and Drug Ad-
nanomedicines with smart functionalities for a clinical trial is ex- ministration (FDA) permitted silica nanoparticles for the first hu-
pected soon. Polymeric micelle such as Paclitaxel (PTX)-loaded man clinical trial against cancer. Quercetin incorporated with
polymeric micelles, SP1049C, Genexol-PM, NK012, NK105, aminopropyl functionalized mesoporous silica nanoparticles pro-
NC6300, K-912, NK 6004 are now under clinical trial against cer- vides better nanocarrier for quercetin delivery. It is reported as an
tain forms of cancer like breast cancer, non-small-cell lung cancer improved chemo-preventive agent for in vitro system [220]. Al-
and ovarian cancer, adenocarcinoma of oesophagus, gastroesophag- though silica nanoparticles conjugated quercetin delivery system
eal junction and stomach, NSCLC, renal, stomach and pancreatic requires the evaluation of toxicity so that it may be utilized against
cancer, advance stomach cancer, liver cancer and solid tumors, cancer in the near future. Further, there is a need for monitored
respectively [197-199, 202, 205-212] (Table 2). bioimaging to understand the function of the delivery system more
precisely. Phytosome innovation can enhance the adequacy of che-
7.5. Other Delivery System for Nanoconjugated Quercetin motherapeutics capability of anticancerous tranquilizes by expand-
ing the permeability of tumor cells. Poly (β-amino esters) polymers
Apart from several nano-sized platforms including polymeric have emerged as highly promising candidates for drug delivery and
micelle, liposomes, PLGA (Poly lactic-co-glycolic acid), silver biomedical applications due to their pH sensitivity, tuneable and
nanoparticles; other carriers are also evolved for delivery of quer- degradable properties. These polymeric frameworks can fill in
cetin. In this series, nanosponges, poly (β-amino esters) nanogels, as professional drug transporters for the delivery of bioactive
solid lipid, carbon nanotubes, and silicon quantum dots are under compounds such as quercetin which experience the poor watery
evaluation nowadays for safe and efficient delivery of quercetin to solvency and low bioavailability. Treatment of quercetin based
target various cancerous cells [121, 137, 158, 213-215] (Table 3). solid lipid nanoparticles recovers bone loss in ovariectomized rats
In recent research, a certain new combination of quantum dots [158].
is being explored for clinical imaging and drug targeting. Use of Additionally, the discovery, characterization, and implication of
quantum dots plays a central theme in nanotechnology for medical carbon nanotubes having unexpected electrical, mechanical, and

Table 2. Polymeric micelles under clinical evaluation.

Sr. No. Polymeric Micelle Cancer Clinical Evaluation Refs.

1 Paclitaxel (PTX)-loaded polymeric micelles Breast cancer, non-small-cell lung cancer, and ovarian cancer Approved [206, 207]
2 SP1049C Adenocarcinoma of oesophagus, gastroesophageal junction and stomach Clinical phase III [208]
3 Genexol-PM NSCLC Clinical phase II [202]
4 NK012 Renal, stomach and Pancreatic cancer Clinical phase I/II [209]
5 NK105 Advance stomach cancer Clinical phase II [210]
6 NC6300 Liver cancer Clinical phase I [199, 211]
7 K-912 Solid tumors Clinical phase I [198]
8 NK 6004 Solid tumors Clinical phase 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

Table 3. Other delivery systems involved in anticancer effects of nanoconjugated quercetin.

Sr. No. Chemical/Polymer Advantages Effects/Targets Refs.

1 Silicon quantum dots Measure subordinate optical properties, high fluo- Inhibition of hydrogen peroxide induced DNA damage, monitoring [213]
rescence quantum yields, astounding soundness of delivery, improvement of aqueous solubility and enhanced
against photobleaching and lower toxicity biocompatibility in HepG2 cells
2 Carbon nanotubes Surprising electrical, mechanical and thermal prop- Increased hydrophilicity and solubility of quercetin at acidic cancer [214]
erties, targetability microenvironment [215]
Improved efficiency and reduced toxicity of quercetin-carbon
nanotube composite implicated in HeLa cells
3 Solid lipid Great tolerability and biodegradability, high encap- Improved osteoprotective activity in post-menopausal osteoporosis [158]
sulation effectiveness, targetability
4 Poly (β-amino esters) pH sensitivity, tuneable and degradable properties Protection against induced cellular oxidative stress [137]
nanogels
5 Nanosponges Regulated metabolism and protection against in- Regulated metabolism of quercetin in cancer therapy [121]
duced cellular oxidative stress

thermal properties further explore the possibility of drug delivery AUTHOR CONTRIBUTIONS
and targeting of anticancerous drugs. The release of quercetin from
carbon nanotubes attributes the increased hydrophilicity and solu- All authors have participated in planning or drafting of the
bility of quercetin at acid microenvironment of cancer [214]. Im- manuscript and approved the final version.
proved efficiency and reduced toxicity of quercetin carbon nano-
tube composite implicated in HeLa cells, suggests the considerable CONSENT FOR PUBLICATION
potential advantages of quercetin-carbon nanotube composite in Not applicable.
cancer therapy [215]. Further, the suitability of the nanosponges has
been implicated not only as a dual release drug delivery system but FUNDING
also with regulated metabolism through nano network [137]. The
variety in crosslinking allows a dual release with regulated release This work was supported by Department of Science and Tech-
kinetics and recommends enhanced bioavailability supported by a nology, India (Grant No. SR/S0/AS-97/2007); and University
reduced metabolism. Dual drug delivery of tamoxifen and quercetin Grants Commission, India (Grant Nos. F.31-217/2005; F.40-
with nanosponges in regulated metabolism has been suggested for 209/2011 SR).
anticancer treatment [121]. An embodiment of polyester nano-
sponge co-stacked with quercetin is examined for successful load- CONFLICT OF INTEREST
ing, release, and in vitro digestion. Lockhart et al. reported that The authors declare no conflict of interest, financial or other-
improved bioavailability of the nanoparticles formulations of nano- wise.
sponge is supported by drug release, cytotoxicity, and enhanced
anti-cancer effects in the recovery condition [121]. ACKNOWLEDGEMENTS
CONCLUSION AND FUTURE PERSPECTIVE Declared none.
Nanoparticles have been extensively investigated to deliver REFERENCES
anticancer drugs due to their enhanced anticancer potential and
promising clinical application. Recent research in nanoconjugated [1] Senapati, S.; Mahanta, A.K.; Kumar, S.; Maiti, P. Controlled drug
quercetin is proposed to overcome its limitations of low bioavail- delivery vehicles for cancer treatment and their performance. Sig-
ability, chemical instability, and short biological half-life. The nal Transduct. Target. Ther., 2018, 3, 7.
pharmacological effect of quercetin conjugated nanoparticles [http://dx.doi.org/10.1038/s41392-017-0004-3] [PMID: 29560283]
[2] Maurya, A.K.; Vinayak, M. Breast cancer stem cell mediated aber-
mostly depends on drug carriers used. Quercetin conjugated polym- rant signaling and epithelial-mesenchymal transition targets: hope
eric micelles, liposomes, silver nanoparticles, PLGA (Poly lactic- for breast cancer therapy. Int. J. Cancer Oncol., 2016, 3(3), 1-7.
co-glycolic acid) have shown better inhibition of tumor growth in [http://dx.doi.org/10.15436/2377-0902.16.1035]
vivo. Controlled drug release, long retention in tumor, improved [3] Maurya, A.K.; Vinayak, M. PI-103 and Quercetin Attenuate PI3K-
efficiency in invagination and extravagation through pores in the AKT Signaling Pathway in T- Cell Lymphoma Exposed to Hydro-
capillary endothelium of tumor by nanoconjugated drugs have built gen Peroxide. PLoS One, 2016, 11(8), e0160686.
the future hope of their clinical application in metastatic cancer. [http://dx.doi.org/10.1371/journal.pone.0160686] [PMID: 27494022]
Delivery of quercetin nanoparticles and their anticancer efficacy are [4] Das, L.; Vinayak, M. Curcumin Modulates Glycolytic Metabolism
mostly confined to laboratory animals and in vitro system. How- and Inflammatory Cytokines via Nrf 2 in Dalton’s Lymphoma As-
cites Cells In Vivo. Anticancer. Agents Med. Chem., 2018, 18(12),
ever, certain other nanoconjugated anticancer drugs are under clini- 1779-1791.
cal trials. Despite numerous in vitro and in vivo anti-cancer studies [http://dx.doi.org/10.2174/1871520618666180604093802] [PMID:
of quercetin formulations, there exist certain limitations for clinical 29866021]
translation such as cost, safety, and side effects. Locating key target [5] Vinayak, M. Molecular Action of Herbal Antioxidants in Regula-
molecules, safe and stable delivery systems with superior strategy tion of Cancer Growth: Scope for Novel Anticancer Drugs. Nutr.
to overcome resistance and reduce the side effects of chemothera- Cancer, 2018, 70(8), 1199-1209.
peutic agents are the main goals of ideal cancer treatment. Addition [http://dx.doi.org/10.1080/01635581.2018.1539187] [PMID:
of cancer cell-specific targeting moieties on the nanoparticles has 30463447]
been evolved to not only enhance target-specific delivery of nano- [6] Neuhouser, M.L. Dietary flavonoids and cancer risk: evidence from
human population studies. Nutr. Cancer, 2004, 50(1), 1-7.
formulations but also to reduce their interaction with the normal [http://dx.doi.org/10.1207/s15327914nc5001_1] [PMID: 15572291]
cells, preventing side effects of drugs.
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- [http://dx.doi.org/10.1016/j.antiviral.2012.03.005] [PMID:
take and risk of lung cancer: a meta-analysis. Jpn. J. Clin. Oncol., 22465313]
2009, 39(6), 352-359. [24] Sun, D.; Zhang, W.; Li, N.; Zhao, Z.; Mou, Z.; Yang, E.; Wang, W.
[http://dx.doi.org/10.1093/jjco/hyp028] [PMID: 19351659] Silver nanoparticles-quercetin conjugation to siRNA against drug-
[8] Cui, Y.; Morgenstern, H.; Greenland, S.; Tashkin, D.P.; Mao, J.T.; resistant Bacillus subtilis for effective gene silencing: in vitro and
Cai, L.; Cozen, W.; Mack, T.M.; Lu, Q.Y.; Zhang, Z.F. Dietary in vivo. Mater. Sci. Eng. C, 2016, 63, 522-534.
flavonoid intake and lung cancer--a population-based case-control [http://dx.doi.org/10.1016/j.msec.2016.03.024] [PMID: 27040247]
study. Cancer, 2008, 112(10), 2241-2248. [25] Rai, M.; Yadav, A.; Gade, A. Silver nanoparticles as a new genera-
[http://dx.doi.org/10.1002/cncr.23398] [PMID: 18327817] tion of antimicrobials. Biotechnol. Adv., 2009, 27(1), 76-83.
[9] Forcados, G.E.; James, D.B.; Sallau, A.B.; Muhammad, A.; Ma- [http://dx.doi.org/10.1016/j.biotechadv.2008.09.002] [PMID:
beta, P. Oxidative Stress and Carcinogenesis: Potential of Phyto- 18854209]
chemicals in Breast Cancer Therapy. Nutr. Cancer, 2017, 69(3), [26] Han, Q.; Wang, X.; Cai, S.; Liu, X.; Zhang, Y.; Yang, L.; Wang,
365-374. C.; Yang, R. Quercetin nanoparticles with enhanced bioavailability
[http://dx.doi.org/10.1080/01635581.2017.1267777] [PMID: as multifunctional agents toward amyloid induced neurotoxicity. J.
28103111] Mater. Chem. B Mater. Biol. Med., 2018, 6(9), 1387-1393.
[10] Abraham, A.N.; Sharma, T.K.; Bansal, V.; Shukla, R. Phytochemi- [http://dx.doi.org/10.1039/C7TB03053C]
cals as Dynamic Surface Ligands To Control Nanoparticle-Protein [27] George, D.; Maheswari, P.U.; Begum, K.M.M.S. Synergic formu-
Interactions. ACS Omega, 2018, 3(2), 2220-2229. lation of onion peel quercetin loaded chitosan-cellulose hydrogel
[http://dx.doi.org/10.1021/acsomega.7b01878] [PMID: 30023827] with green zinc oxide nanoparticles towards controlled release,
[11] Maurya, A.K.; Vinayak, M. Modulation of PKC signaling and biocompatibility, antimicrobial and anticancer activity. Int. J. Biol.
induction of apoptosis through suppression of reactive oxygen spe- Macromol., 2019, 132, 784-794.
cies and tumor necrosis factor receptor 1 (TNFR1): key role of [http://dx.doi.org/10.1016/j.ijbiomac.2019.04.008] [PMID:
quercetin in cancer prevention. Tumour Biol., 2015, 36(11), 8913- 30951778]
8924. [28] Alidadi, H.; Khorsandi, L.; Shirani, M. Effects of Quercetin on
[http://dx.doi.org/10.1007/s13277-015-3634-5] [PMID: 26076811] Tubular Cell Apoptosis and Kidney Damage in Rats Induced by Ti-
[12] D’Andrea, G. Quercetin: A flavonol with multifaceted therapeutic tanium Dioxide Nanoparticles. Malays. J. Med. Sci., 2018, 25(2),
applications? Fitoterapia, 2015, 106, 256-271. 72-81.
[http://dx.doi.org/10.1016/j.fitote.2015.09.018] [PMID: 26393898] [http://dx.doi.org/10.21315/mjms2018.25.2.8] [PMID: 30918457]
[13] Amanzadeh, E.; Esmaeili, A.; Rahgozar, S.; Nourbakhshnia, M. In. [29] Abdelhalim, M.A.K.; Qaid, H.A.; Al-Mohy, Y.; Al-Ayed, M.S.
Rev. Neurosci., 2019, 0 Effects of quercetin and arginine on the nephrotoxicity and lipid
[14] Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.; peroxidation induced by gold nanoparticles in vivo. Int. J.
Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A Nanomedicine, 2018, 13, 7765-7770.
comprehensive review. Phytother. Res., 2018, 32(11), 2109-2130. [http://dx.doi.org/10.2147/IJN.S183281] [PMID: 30538457]
[http://dx.doi.org/10.1002/ptr.6155] [PMID: 30039547] [30] Cruz Dos Santos, S.; Osti Silva, N.; Dos Santos Espinelli, J.B.J.;
[15] Maurya, A.K.; Vinayak, M. Anticarcinogenic action of quercetin Germani Marinho, M.A.; Vieira Borges, Z.; Bruzamarello Caon
by downregulation of phosphatidylinositol 3-kinase (PI3K) and Branco, N.; Faita, F.L.; Meira Soares, B.; Horn, A.P.; Parize, A.L.;
protein kinase C (PKC) via induction of p53 in hepatocellular car- Rodrigues de Lima, V. Molecular interactions and physico-
cinoma (HepG2) cell line. Mol. Biol. Rep., 2015, 42(9), 1419-1429. chemical characterization of quercetin-loaded magnetoliposomes.
[http://dx.doi.org/10.1007/s11033-015-3921-7] [PMID: 26311153] Chem. Phys. Lipids, 2019, 218, 22-33.
[16] Kumari, A.; Yadav, S.K.; Pakade, Y.B.; Kumar, V.; Singh, B.; [http://dx.doi.org/10.1016/j.chemphyslip.2018.11.010] [PMID:
Chaudhary, A.; Yadav, S.C. Nanoencapsulation and characteriza- 30508514]
tion of Albizia chinensis isolated antioxidant quercitrin on PLA [31] Wen, P.; Hu, T.G.; Li, L.; Zong, M.H.; Wu, H. A colon-specific
nanoparticles. Colloids Surf. B Biointerfaces, 2011, 82(1), 224-232. delivery system for quercetin with enhanced cancer prevention
[http://dx.doi.org/10.1016/j.colsurfb.2010.08.046] [PMID: 20870396] based on co-axial electrospinning. Food Funct., 2018, 9(11), 5999-
[17] Maurya, A.K.; Vinayak, M. Improved synergistic anticancer effi- 6009.
cacy of quercetin in combination with PI-103, rottlerin, and G0 [http://dx.doi.org/10.1039/C8FO01216D] [PMID: 30382268]
6983 against MCF-7 and RAW 264.7 cells. In Vitro Cell. Dev. [32] Ma, J.J.; Yu, Y.G.; Yin, S.W.; Tang, C.H.; Yang, X.Q. Cellular
Biol. Anim., 2019, 55(1), 36-44. Uptake and Intracellular Antioxidant Activity of Zein/Chitosan
[http://dx.doi.org/10.1007/s11626-018-0309-8] [PMID: 30413935] Nanoparticles Incorporated with Quercetin. J. Agric. Food Chem.,
[18] Kleemann, R.; Verschuren, L.; Morrison, M.; Zadelaar, S.; van Erk, 2018, 66(48), 12783-12793.
M.J.; Wielinga, P.Y.; Kooistra, T. Anti-inflammatory, anti- [http://dx.doi.org/10.1021/acs.jafc.8b04571] [PMID: 30406660]
proliferative and anti-atherosclerotic effects of quercetin in human [33] Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.F.; Flamm,
in vitro and in vivo models. Atherosclerosis, 2011, 218(1), 44-52. G.W.; Williams, G.M.; Lines, T.C. A critical review of the data re-
[http://dx.doi.org/10.1016/j.atherosclerosis.2011.04.023] [PMID: lated to the safety of quercetin and lack of evidence of in vivo tox-
21601209] icity, including lack of genotoxic/carcinogenic properties. Food
[19] Nabavi, S.F.; Russo, G.L.; Daglia, M.; Nabavi, S.M. Role of quer- Chem. Toxicol., 2007, 45(11), 2179-2205.
cetin as an alternative for obesity treatment: you are what you eat! [http://dx.doi.org/10.1016/j.fct.2007.05.015] [PMID: 17698276]
Food Chem., 2015, 179, 305-310. [34] Reinboth, M.; Wolffram, S.; Abraham, G.; Ungemach, F.R.; Cer-
[http://dx.doi.org/10.1016/j.foodchem.2015.02.006] [PMID: mak, R. Oral bioavailability of quercetin from different quercetin
25722169] glycosides in dogs. Br. J. Nutr., 2010, 104(2), 198-203.
[20] Xu, D.; Hu, M-J.; Wang, Y-Q.; Cui, Y-L. Antioxidant Activities of [http://dx.doi.org/10.1017/S000711451000053X] [PMID: 20230651]
Quercetin and Its Complexes for Medicinal Application. Mole- [35] Hirpara, K.V.; Aggarwal, P.; Mukherjee, A.J.; Joshi, N.; Burman,
cules, 2019, 24(6), 1123. A.C. Quercetin and its derivatives: synthesis, pharmacological uses
[http://dx.doi.org/10.3390/molecules24061123] [PMID: 30901869] with special emphasis on anti-tumor properties and prodrug with
[21] Dueñas, M.; González-Manzano, S.; González-Paramás, A.; San- enhanced bio-availability. Anticancer. Agents Med. Chem., 2009,
tos-Buelga, C. Antioxidant evaluation of O-methylated metabolites 9(2), 138-161.
of catechin, epicatechin and quercetin. J. Pharm. Biomed. Anal., [http://dx.doi.org/10.2174/187152009787313855] [PMID:
2010, 51(2), 443-449. 19199862]
[http://dx.doi.org/10.1016/j.jpba.2009.04.007] [PMID: 19442472] [36] Mulholland, P.J.; Ferry, D.R.; Anderson, D.; Hussain, S.A.; Young,
[22] Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: A.M.; Cook, J.E.; Hodgkin, E.; Seymour, L.W.; Kerr, D.J. Pre-
from antioxidant to nutraceutical. Eur. J. Pharmacol., 2008, 585(2-3), clinical and clinical study of QC12, a water-soluble, pro-drug of
325-337. quercetin. Ann. Oncol., 2001, 12(2), 245-248.
[http://dx.doi.org/10.1016/j.ejphar.2008.03.008] [PMID: 18417116] [http://dx.doi.org/10.1023/A:1008372017097] [PMID: 11300332]
[23] Ganesan, S.; Faris, A.N.; Comstock, A.T.; Wang, Q.; Nanua, S.; [37] Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of
Hershenson, M.B.; Sajjan, U.S. Quercetin inhibits rhinovirus repli- Biological Importance of Quercetin: A Bioactive Flavonoid. Phar-
cation in vitro and in vivo. Antiviral Res., 2012, 94(3), 258-271. 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] based mixed polymeric micelles containing quercetin in cancer
[38] Ezzati Nazhad Dolatabadi, J.; Mokhtarzadeh, A.; Ghareghoran, treatment and an in vivo pharmacokinetic study. Int. J. Nanomedi-
S.M.; Dehghan, G. Synthesis, Characterization and Antioxidant cine, 2016, 11, 1557-1566.
Property of Quercetin-Tb(III) Complex. Adv. Pharm. Bull., 2014, [PMID: 27143878]
4(2), 101-104. [53] Del Follo-Martinez, A.; Banerjee, N.; Li, X.; Safe, S.; Mertens-
[PMID: 24511472] Talcott, S. Resveratrol and quercetin in combination have antican-
[39] Gao, X.; Wang, B.; Wei, X.; Men, K.; Zheng, F.; Zhou, Y.; Zheng, cer activity in colon cancer cells and repress oncogenic microRNA-
Y.; Gou, M.; Huang, M.; Guo, G.; Huang, N.; Qian, Z.; Wei, Y. 27a. Nutr. Cancer, 2013, 65(3), 494-504.
Anticancer effect and mechanism of polymer micelle-encapsulated [http://dx.doi.org/10.1080/01635581.2012.725194] [PMID:
quercetin on ovarian cancer. Nanoscale, 2012, 4(22), 7021-7030. 23530649]
[http://dx.doi.org/10.1039/c2nr32181e] [PMID: 23044718] [54] Chan, S-T.; Chuang, C-H.; Lin, Y-C.; Liao, J-W.; Lii, C-K.; Yeh,
[40] Nam, J.S.; Sharma, A.R.; Nguyen, L.T.; Chakraborty, C.; Sharma, S-L. Quercetin enhances the antitumor effect of trichostatin A and
G.; Lee, S.S. Application of Bioactive Quercetin in Oncotherapy: suppresses muscle wasting in tumor-bearing mice. Food Funct.,
From Nutrition to Nanomedicine. Molecules, 2016, 21(1), E108. 2018, 9(2), 871-879.
[http://dx.doi.org/10.3390/molecules21010108] [PMID: 26797598] [http://dx.doi.org/10.1039/C7FO01444A] [PMID: 29292417]
[41] Maurya, A.K.; Vinayak, M. Quercetin regresses Dalton’s lym- [55] Serri, C.; Quagliariello, V.; Iaffaioli, R.V.; Fusco, S.; Botti, G.;
phoma growth via suppression of PI3K/AKT signaling leading to Mayol, L.; Biondi, M. Combination therapy for the treatment of
upregulation of p53 and decrease in energy metabolism. Nutr. Can- pancreatic cancer through hyaluronic acid-decorated nanoparticles
cer, 2015, 67(2), 354-363. loaded with quercetin and gemcitabine: A preliminary in vitro
[http://dx.doi.org/10.1080/01635581.2015.990574] [PMID: study. J. Cell. Physiol., 2019, 234(4), 4959-4969.
25658812] [http://dx.doi.org/10.1002/jcp.27297] [PMID: 30334571]
[42] Zhu, B.; Yu, L.; Yue, Q. Co-delivery of vincristine and quercetin [56] Flusberg, D.A.; Sorger, P.K. Surviving apoptosis: life-death signal-
by nanocarriers for lymphoma combination chemotherapy. Biomed. ing in single cells. Trends Cell Biol., 2015, 25(8), 446-458.
Pharmacother., 2017, 91, 287-294. [http://dx.doi.org/10.1016/j.tcb.2015.03.003] [PMID: 25920803]
[http://dx.doi.org/10.1016/j.biopha.2017.02.112] [PMID: [57] Pobezinskaya, Y.L.; Liu, Z. The role of TRADD in death receptor
28463792] signaling. Cell Cycle, 2012, 11(5), 871-876.
[43] Maurya, A.K.; Vinayak, M. PI-103 attenuates PI3K-AKT signaling [http://dx.doi.org/10.4161/cc.11.5.19300] [PMID: 22333735]
and induces apoptosis in murineT-cell lymphoma. Leuk. Lym- [58] Russo, M.; Spagnuolo, C.; Bilotto, S.; Tedesco, I.; Maiani, G.;
phoma, 2017, 58(5), 1153-1161. Russo, G.L.; Russo, M.; Spagnuolo, C.; Bilotto, S.; Tedesco, I.;
[http://dx.doi.org/10.1080/10428194.2016.1225207] [PMID: Maiani, G.; Russo, G.L. Inhibition of protein kinase CK2 by quer-
27658642] cetin enhances CD95-mediated apoptotis in a human thymus-
[44] Maurya, A.K.; Vinayak, M. Quercetin Attenuates Cell Survival, derived T cell line. Food Res. Int., 2014.
Inflammation, and Angiogenesis via Modulation of AKT Signaling [http://dx.doi.org/10.1016/j.foodres.2014.05.022]
in Murine T-Cell Lymphoma. Nutr. Cancer, 2017, 69(3), 470-480. [59] Chou, C.C.; Yang, J.S.; Lu, H.F.; Ip, S.W.; Lo, C.; Wu, C.C.; Lin,
[http://dx.doi.org/10.1080/01635581.2017.1267775] [PMID: J.P.; Tang, N.Y.; Chung, J.G.; Chou, M.J.; Teng, Y.H.; Chen, D.R.
28107044] Quercetin-mediated cell cycle arrest and apoptosis involving acti-
[45] Rivera Rivera, A.; Castillo-Pichardo, L.; Gerena, Y.; Dharmaward- vation of a caspase cascade through the mitochondrial pathway in
hane, S. Anti-Breast Cancer Potential of Quercetin via the human breast cancer MCF-7 cells. Arch. Pharm. Res., 2010, 33(8),
Akt/AMPK/Mammalian Target of Rapamycin (mTOR) Signaling 1181-1191.
Cascade. PLoS One, 2016, 11(6), e0157251. [http://dx.doi.org/10.1007/s12272-010-0808-y] [PMID: 20803121]
[http://dx.doi.org/10.1371/journal.pone.0157251] [PMID: 27285995] [60] Haghiac, M.; Walle, T. Quercetin induces necrosis and apoptosis in
[46] Seo, H.S.; Ku, J.M.; Choi, H.S.; Choi, Y.K.; Woo, J.K.; Kim, M.; SCC-9 oral cancer cells. Nutr. Cancer, 2005, 53(2), 220-231.
Kim, I.; Na, C.H.; Hur, H.; Jang, B.H.; Shin, Y.C.; Ko, S.G. Quer- [http://dx.doi.org/10.1207/s15327914nc5302_11] [PMID:
cetin induces caspase-dependent extrinsic apoptosis through inhibi- 16573383]
tion of signal transducer and activator of transcription 3 signaling [61] Mozhgan Farzami Sepehr, S.B.J.B.H. The Cuscuta kotschyana
in HER2-overexpressing BT-474 breast cancer cells. Oncol. Rep., effects on breast cancer cells line MCF7. J. Med. Plants Res., 2011,
2016, 36(1), 31-42. 5(27), 6344-6351.
[http://dx.doi.org/10.3892/or.2016.4786] [PMID: 27175602] [http://dx.doi.org/10.5897/JMPR11.1048]
[47] Subramaniam, D.; Kaushik, G.; Dandawate, P.; Anant, S. Targeting [62] Niu, G.; Yin, S.; Xie, S.; Li, Y.; Nie, D.; Ma, L.; Wang, X.; Wu, Y.
cancer stem cells for chemoprevention of pancreatic cancer. Curr. Quercetin induces apoptosis by activating caspase-3 and regulating
Med. Chem., 2017. Bcl-2 and cyclooxygenase-2 pathways in human HL-60 cells. Acta
[PMID: 28137215] Biochim. Biophys. Sin. (Shanghai), 2011, 43(1), 30-37.
[48] Gavrilas, L.I.; Ionescu, C.; Tudoran, O.; Lisencu, C.; Balacescu, [http://dx.doi.org/10.1093/abbs/gmq107] [PMID: 21173056]
O.; Miere, D. The Role of Bioactive Dietary Components in Modu- [63] Hu, J.; Wang, J.; Wang, G.; Yao, Z.; Dang, X. Pharmacokinetics
lating miRNA Expression in Colorectal Cancer. Nutrients, 2016, and antitumor efficacy of DSPE-PEG2000 polymeric liposomes
8(10), E590. [http://dx.doi.org/10.3390/nu8100590] [PMID: loaded with quercetin and temozolomide: Analysis of their effec-
27681738] tiveness in enhancing the chemosensitization of drug-resistant
[49] Saleem, T.H.; Attya, A.M.; Ahmed, E.A.; Ragab, S.M.; Ali Abdal- glioma cells. Int. J. Mol. Med., 2016, 37(3), 690-702.
lah, M.A.; Omar, H.M. Possible Protective Effects of Quercetin [http://dx.doi.org/10.3892/ijmm.2016.2458] [PMID: 26782731]
and Sodium Gluconate Against Colon Cancer Induction by Di- [64] Hu, J.; Yu, Q.; Zhao, F.; Ji, J.; Jiang, Z.; Chen, X.; Gao, P.; Ren,
methylhydrazine in Mice. Asian Pac. J. Cancer Prev., 2015, Y.; Shao, S.; Zhang, L.; Yan, M. Protection of Quercetin against
16(14), 5823-5828. Triptolide-induced apoptosis by suppressing oxidative stress in rat
[http://dx.doi.org/10.7314/APJCP.2015.16.14.5823] [PMID: Leydig cells. Chem. Biol. Interact., 2015, 240, 38-46.
26320457] [http://dx.doi.org/10.1016/j.cbi.2015.08.004] [PMID: 26277538]
[50] Engen, A.; Maeda, J.; Wozniak, D.E.; Brents, C.A.; Bell, J.J.; Ue- [65] Shen, S.C.; Lee, W.R.; Yang, L.Y.; Tsai, H.H.; Yang, L.L.; Chen,
saka, M.; Aizawa, Y.; Kato, T.A. Induction of cytotoxic and geno- Y.C. Quercetin enhancement of arsenic-induced apoptosis via
toxic responses by natural and novel quercetin glycosides. Mutat. stimulating ROS-dependent p53 protein ubiquitination in human
Res. Genet. Toxicol. Environ. Mutagen., 2015, 784-785, 15-22. HaCaT keratinocytes. Exp. Dermatol., 2012, 21(5), 370-375.
[http://dx.doi.org/10.1016/j.mrgentox.2015.04.007] [PMID: [http://dx.doi.org/10.1111/j.1600-0625.2012.01479.x] [PMID:
26046972] 22509835]
[51] Maurya, A.K.; Vinayak, M. Abstract A07: Decline in the growth of [66] Zhang, J.Y.; Yi, T.; Liu, J.; Zhao, Z.Z.; Chen, H.B. Quercetin in-
murine T-cell lymphoma via modulation of PI3K signaling path- duces apoptosis via the mitochondrial pathway in KB and KBv200
way: Key role of quercetin and PI-103. Mol. Cancer Ther., 2015, cells. J. Agric. Food Chem., 2013, 61(9), 2188-2195.
14(7)(Suppl.), A07-A07. [http://dx.doi.org/10.1021/jf305263r] [PMID: 23410218]
[52] Chen, L.C.; Chen, Y.C.; Su, C.Y.; Hong, C.S.; Ho, H.O.; Sheu, [67] Zhang, Q.; Zhao, X.H.; Wang, Z.J. Cytotoxicity of flavones and
M.T. Development and characterization of self-assembling lecithin- 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 [82] Chiang, G.G.; Abraham, R.T. Phosphorylation of mammalian
Vitro, 2009, 23(5), 797-807. target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6
[http://dx.doi.org/10.1016/j.tiv.2009.04.007] [PMID: 19397994] kinase. J. Biol. Chem., 2005, 280(27), 25485-25490.
[68] Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo, [http://dx.doi.org/10.1074/jbc.M501707200] [PMID: 15899889]
Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest [83] Hung, C.M.; Garcia-Haro, L.; Sparks, C.A.; Guertin, D.A. mTOR-
and apoptosis in human breast cancer cells by quercetin. Int. J. On- dependent cell survival mechanisms. Cold Spring Harb. Perspect.
col., 2001, 19(4), 837-844. Biol., 2012, 4(12), a008771.
[http://dx.doi.org/10.3892/ijo.19.4.837] [PMID: 11562764] [http://dx.doi.org/10.1101/cshperspect.a008771] [PMID:
[69] Lee, Y.K.; Hwang, J.T.; Kwon, D.Y.; Surh, Y.J.; Park, O.J. Induc- 23124837]
tion of apoptosis by quercetin is mediated through AMPKal- [84] Simpson, D.R.; Mell, L.K.; Cohen, E.E. Targeting the
pha1/ASK1/p38 pathway. Cancer Lett., 2010, 292(2), 228-236. PI3K/AKT/mTOR pathway in squamous cell carcinoma of the
[http://dx.doi.org/10.1016/j.canlet.2009.12.005] [PMID: 20083342] head and neck. Oral Oncol., 2015, 51(4), 291-298.
[70] Kim, M.C.; Lee, H.J.; Lim, B.; Ha, K.T.; Kim, S.Y.; So, I.; Kim, [http://dx.doi.org/10.1016/j.oraloncology.2014.11.012] [PMID:
B.J. Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 25532816]
channels in AGS cells. Int. J. Mol. Med., 2014, 33(6), 1657-1663. [85] Mabuchi, S.; Kuroda, H.; Takahashi, R.; Sasano, T. The
[http://dx.doi.org/10.3892/ijmm.2014.1704] [PMID: 24647664] PI3K/AKT/mTOR pathway as a therapeutic target in ovarian can-
[71] Lee, K.H.; Yoo, C.G. Simultaneous inactivation of GSK-3β sup- cer. Gynecol. Oncol., 2015, 137(1), 173-179.
presses quercetin-induced apoptosis by inhibiting the JNK path- [http://dx.doi.org/10.1016/j.ygyno.2015.02.003] [PMID:
way. Am. J. Physiol. Lung Cell. Mol. Physiol., 2013, 304(11), 25677064]
L782-L789. [86] Martelli, A.M.; Evangelisti, C.; Chiarini, F.; Grimaldi, C.; Cappel-
[http://dx.doi.org/10.1152/ajplung.00348.2012] [PMID: 23564507] lini, A.; Ognibene, A.; McCubrey, J.A. The emerging role of the
[72] Vidya Priyadarsini, R.; Senthil Murugan, R.; Maitreyi, S.; Rama- phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin
lingam, K.; Karunagaran, D.; Nagini, S. The flavonoid quercetin signaling network in normal myelopoiesis and leukemogenesis.
induces cell cycle arrest and mitochondria-mediated apoptosis in Biochim. Biophys. Acta, 2010, 1803(9), 991-1002.
human cervical cancer (HeLa) cells through p53 induction and NF- [http://dx.doi.org/10.1016/j.bbamcr.2010.04.005] [PMID:
κB inhibition. Eur. J. Pharmacol., 2010, 649(1-3), 84-91. 20399811]
[http://dx.doi.org/10.1016/j.ejphar.2010.09.020] [PMID: [87] Bai, D.; Ueno, L.; Vogt, P.K. Akt-mediated regulation of NFkap-
20858478] paB and the essentialness of NFkappaB for the oncogenicity of
[73] Chen, X.; Dong, X.S.; Gao, H.Y.; Jiang, Y.F.; Jin, Y.L.; Chang, PI3K and Akt. Int. J. Cancer, 2009, 125(12), 2863-2870.
Y.Y.; Chen, L.Y.; Wang, J.H. Suppression of HSP27 increases the [http://dx.doi.org/10.1002/ijc.24748] [PMID: 19609947]
anti-tumor effects of quercetin in human leukemia U937 cells. Mol. [88] Choi, K.C.; Chung, W.T.; Kwon, J.K.; Yu, J.Y.; Jang, Y.S.; Park,
Med. Rep., 2016, 13(1), 689-696. S.M.; Lee, S.Y.; Lee, J.C. Inhibitory effects of quercetin on afla-
[http://dx.doi.org/10.3892/mmr.2015.4600] [PMID: 26648539] toxin B1-induced hepatic damage in mice. Food Chem. Toxicol.,
[74] Mukherjee, A.; Khuda-Bukhsh, A.R. Quercetin Down-regulates IL- 2010, 48(10), 2747-2753.
6/STAT-3 Signals to Induce Mitochondrial-mediated Apoptosis in [http://dx.doi.org/10.1016/j.fct.2010.07.001] [PMID: 20621152]
a Nonsmall- cell Lung-cancer Cell Line, A549. J. Pharmacopunc- [89] Ventura, A.; Kirsch, D.G.; McLaughlin, M.E.; Tuveson, D.A.;
ture, 2015, 18(1), 19-26. Grimm, J.; Lintault, L.; Newman, J.; Reczek, E.E.; Weissleder, R.;
[http://dx.doi.org/10.3831/KPI.2015.18.002] [PMID: 25830055] Jacks, T. Restoration of p53 function leads to tumour regression in
[75] Ranganathan, S.; Halagowder, D.; Sivasithambaram, N.D. Quer- vivo. Nature, 2007, 445(7128), 661-665.
cetin Suppresses Twist to Induce Apoptosis in MCF-7 Breast Can- [http://dx.doi.org/10.1038/nature05541] [PMID: 17251932]
cer Cells. PLoS One, 2015, 10(10), e0141370. [90] Moll, U.M.; Petrenko, O. The MDM2-p53 interaction. Mol. Cancer
[http://dx.doi.org/10.1371/journal.pone.0141370] [PMID: Res., 2003, 1(14), 1001-1008.
26491966] [PMID: 14707283]
[76] Ward, A.B.; Mir, H.; Kapur, N.; Gales, D.N.; Carriere, P.P.; Singh, [91] Mayo, L.D.; Donner, D.B. The PTEN, Mdm2, p53 tumor suppres-
S. Quercetin inhibits prostate cancer by attenuating cell survival sor-oncoprotein network. Trends Biochem. Sci., 2002, 27(9), 462-
and inhibiting anti-apoptotic pathways. World J. Surg. Oncol., 467.
2018, 16(1), 108. [http://dx.doi.org/10.1016/S0968-0004(02)02166-7] [PMID:
[http://dx.doi.org/10.1186/s12957-018-1400-z] [PMID: 29898731] 12217521]
[77] Granado-Serrano, A.B.; Martín, M.A.; Bravo, L.; Goya, L.; Ramos, [92] Zhao, P.; Mao, J.M.; Zhang, S.Y.; Zhou, Z.Q.; Tan, Y.; Zhang, Y.
S. Quercetin induces apoptosis via caspase activation, regulation of Quercetin induces HepG2 cell apoptosis by inhibiting fatty acid
Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a biosynthesis. Oncol. Lett., 2014, 8(2), 765-769.
human hepatoma cell line (HepG2). J. Nutr., 2006, 136(11), 2715- [http://dx.doi.org/10.3892/ol.2014.2159] [PMID: 25009654]
2721. [93] Jia, L.; Huang, S.; Yin, X.; Zan, Y.; Guo, Y.; Han, L. Quercetin
[http://dx.doi.org/10.1093/jn/136.11.2715] [PMID: 17056790] suppresses the mobility of breast cancer by suppressing glycolysis
[78] Walker, E.H.; Pacold, M.E.; Perisic, O.; Stephens, L.; Hawkins, through Akt-mTOR pathway mediated autophagy induction. Life
P.T.; Wymann, M.P.; Williams, R.L. Structural determinants of Sci., 2018, 208, 123-130.
phosphoinositide 3-kinase inhibition by wortmannin, LY294002, [http://dx.doi.org/10.1016/j.lfs.2018.07.027] [PMID: 30025823]
quercetin, myricetin, and staurosporine. Mol. Cell, 2000, 6(4), 909- [94] Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and
919. lymphangiogenesis. Nat. Rev. Mol. Cell Biol., 2007, 8(6), 464-478.
[http://dx.doi.org/10.1016/S1097-2765(05)00089-4] [PMID: [http://dx.doi.org/10.1038/nrm2183] [PMID: 17522591]
11090628] [95] Graupera, M.; Potente, M. Regulation of angiogenesis by PI3K
[79] Datta, S.R.; Dudek, H.; Tao, X.; Masters, S.; Fu, H.; Gotoh, Y.; signaling networks. Exp. Cell Res., 2013, 319(9), 1348-1355.
Greenberg, M.E. Akt phosphorylation of BAD couples survival [http://dx.doi.org/10.1016/j.yexcr.2013.02.021] [PMID: 23500680]
signals to the cell-intrinsic death machinery. Cell, 1997, 91(2), 231- [96] Ma, F.; Zhang, L.; Westlund, K.N. Reactive oxygen species medi-
241. ate TNFR1 increase after TRPV1 activation in mouse DRG neu-
[http://dx.doi.org/10.1016/S0092-8674(00)80405-5] [PMID: rons. Mol. Pain, 2009, 5, 31.
9346240] [http://dx.doi.org/10.1186/1744-8069-5-31] [PMID: 19531269]
[80] Pap, M.; Cooper, G.M. Role of glycogen synthase kinase-3 in the [97] Fang, J.; Ding, M.; Yang, L.; Liu, L.Z.; Jiang, B.H.
phosphatidylinositol 3-Kinase/Akt cell survival pathway. J. Biol. PI3K/PTEN/AKT signaling regulates prostate tumor angiogenesis.
Chem., 1998, 273(32), 19929-19932. Cell. Signal., 2007, 19(12), 2487-2497.
[http://dx.doi.org/10.1074/jbc.273.32.19929] [PMID: 9685326] [http://dx.doi.org/10.1016/j.cellsig.2007.07.025] [PMID:
[81] Dickey, A.; Schleicher, S.; Leahy, K.; Hu, R.; Hallahan, D.; Tho- 17826033]
tala, D.K. GSK-3β inhibition promotes cell death, apoptosis, and in [98] Sobolewski, C.; Cerella, C.; Dicato, M.; Ghibelli, L.; Diederich, M.
vivo tumor growth delay in neuroblastoma Neuro-2A cell line. J. The role of cyclooxygenase-2 in cell proliferation and cell death in
Neurooncol., 2011, 104(1), 145-153. human malignancies. Int. J. Cell Biol., 2010, 2010, 215158.
[http://dx.doi.org/10.1007/s11060-010-0491-3] [PMID: 21161565] [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 [115] Matsumura, Y.; Kataoka, K. Preclinical and clinical studies of
of tumour suppressor p53 and phase-II antioxidant enzymes via ac- anticancer agent-incorporating polymer micelles. Cancer Sci.,
tivation of Nrf2 signalling and modulation of inflammation in pre- 2009, 100(4), 572-579.
vention of cancer. PLoS One, 2015, 10(4), e0124000. [http://dx.doi.org/10.1111/j.1349-7006.2009.01103.x] [PMID:
[http://dx.doi.org/10.1371/journal.pone.0124000] [PMID: 19462526]
25860911] [116] Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of
[100] Subbaramaiah, K.; Altorki, N.; Chung, W.J.; Mestre, J.R.; Sampat, quercetin bioavailability. J. Nutr. Biochem., 2015, 26(3), 201-210.
A.; Dannenberg, A.J. Inhibition of cyclooxygenase-2 gene expres- [http://dx.doi.org/10.1016/j.jnutbio.2014.10.008] [PMID:
sion by p53. J. Biol. Chem., 1999, 274(16), 10911-10915. 25468612]
[http://dx.doi.org/10.1074/jbc.274.16.10911] [PMID: 10196169] [117] Santos, A.F.A.G.I.A.B.M.P.A.T.L.T.K.V.G.W.C.N. Bioavailability
[101] Jeon, J-S.; Kwon, S.; Ban, K.; Kwon Hong, Y.; Ahn, C.; Sung, J- of Quercetin in Humans with a Focus on Interindividual Variation.
S.; Choi, I. Regulation of the Intracellular ROS Level Is Critical for Compr. Rev. Food Sci. Food Saf., 2018, 17, 714-731.
the Antiproliferative Effect of Quercetin in the Hepatocellular Car- [http://dx.doi.org/10.1111/1541-4337.12342]
cinoma Cell Line HepG2. Nutr. Cancer, 2019, 71(5), 861-869. [118] Mu, Y.; Fu, Y.; Li, J.; Yu, X.; Li, Y.; Wang, Y.; Wu, X.; Zhang,
[http://dx.doi.org/10.1080/01635581.2018.1559929] [PMID: K.; Kong, M.; Feng, C.; Chen, X. Multifunctional quercetin conju-
30661409] gated chitosan nano-micelles with P-gp inhibition and permeation
[102] Jadhav, N.R.; Nadaf, S.J.; Lohar, D.A.; Ghagare, P.S.; Powar, T.A. enhancement of anticancer drug. Carbohydr. Polym., 2019, 203,
Phytochemicals Formulated As Nanoparticles: Inventions, Recent 10-18.
Patents and Future Prospects. Recent Pat. Drug Deliv. Formul., [http://dx.doi.org/10.1016/j.carbpol.2018.09.020] [PMID:
2017, 11(3), 173-186. 30318192]
[http://dx.doi.org/10.2174/1872211311666171120102531] [PMID: [119] Ahmad, N.; Ahmad, R.; Naqvi, A.A.; Alam, M.A.; Abdur Rub, R.;
29165100] Ahmad, F.J. Enhancement of Quercetin Oral Bioavailability by
[103] Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Self-Nanoemulsifying Drug Delivery System and their Quantifica-
Polyphenols: food sources and bioavailability. Am. J. Clin. Nutr., tion Through Ultra High Performance Liquid Chromatography and
2004, 79(5), 727-747. Mass Spectrometry in Cerebral Ischemia. Drug Res. (Stuttg.), 2017,
[http://dx.doi.org/10.1093/ajcn/79.5.727] [PMID: 15113710] 67(10), 564-575.
[104] Weldin, J.; Jack, R.; Dugaw, K.; Kapur, R.P. Quercetin, an over- [http://dx.doi.org/10.1055/s-0043-109564] [PMID: 28561230]
the-counter supplement, causes neuroblastoma-like elevation of [120] Rezaei-Sadabady, R.; Eidi, A.; Zarghami, N.; Barzegar, A. Intracel-
plasma homovanillic acid. Pediatr. Dev. Pathol., 2003, 6(6), 547- lular ROS protection efficiency and free radical-scavenging activity
551. of quercetin and quercetin-encapsulated liposomes. Artif. Cells
[http://dx.doi.org/10.1007/s10024-003-5061-7] [PMID: 15018454] Nanomed. Biotechnol., 2016, 44(1), 128-134.
[105] Konishi, Y.; Zhao, Z.; Shimizu, M. Phenolic acids are absorbed [http://dx.doi.org/10.3109/21691401.2014.926456] [PMID:
from the rat stomach with different absorption rates. J. Agric. Food 24959911]
Chem., 2006, 54(20), 7539-7543. [121] Lockhart, J.N.; Stevens, D.M.; Beezer, D.B.; Kravitz, A.; Harth, E.
[http://dx.doi.org/10.1021/jf061554+] [PMID: 17002419] Dual drug delivery of tamoxifen and quercetin: Regulated metabo-
[106] Németh, K.; Plumb, G.W.; Berrin, J.G.; Juge, N.; Jacob, R.; Naim, lism for anticancer treatment with nanosponges. J Control Release.,
H.Y.; Williamson, G.; Swallow, D.M.; Kroon, P.A. Deglycosyla- 2015, 220(Pt B), 751-757.
tion by small intestinal epithelial cell beta-glucosidases is a critical [122] des Rieux, A.; Fievez, V.; Garinot, M.; Schneider, Y.J.; Préat, V.
step in the absorption and metabolism of dietary flavonoid gly- Nanoparticles as potential oral delivery systems of proteins and
cosides in humans. Eur. J. Nutr., 2003, 42(1), 29-42. vaccines: a mechanistic approach. J. Control. Release, 2006,
[http://dx.doi.org/10.1007/s00394-003-0397-3] [PMID: 12594539] 116(1), 1-27.
[107] Murota, K.; Terao, J. Quercetin appears in the lymph of unanesthe- [http://dx.doi.org/10.1016/j.jconrel.2006.08.013] [PMID:
tized rats as its phase II metabolites after administered into the 17050027]
stomach. FEBS Lett., 2005, 579(24), 5343-5346. [123] McClements, D.J.; Decker, E.A.; Park, Y.; Weiss, J. Structural
[http://dx.doi.org/10.1016/j.febslet.2005.08.060] [PMID: design principles for delivery of bioactive components in nutraceu-
16194534] ticals and functional foods. Crit. Rev. Food Sci. Nutr., 2009, 49(6),
[108] Spencer, J.P.; Kuhnle, G.G.; Williams, R.J.; Rice-Evans, C. Intra- 577-606.
cellular metabolism and bioactivity of quercetin and its in vivo me- [http://dx.doi.org/10.1080/10408390902841529] [PMID:
tabolites. Biochem. J., 2003, 372(Pt 1), 173-181. 19484636]
[http://dx.doi.org/10.1042/bj20021972] [PMID: 12578560] [124] Rodriguez-Nogales, C.; Noguera, R.; Patrick, C.; Blanco-Prieto,
[109] de Boer, V.C.; Dihal, A.A.; van der Woude, H.; Arts, I.C.; Wolf- M.J.J. Therapeutic opportunities in neuroblastoma using nanotech-
fram, S.; Alink, G.M.; Rietjens, I.M.; Keijer, J.; Hollman, P.C. Tis- nology. Journal of Pharmacology and Experimental Therapeutics,
sue distribution of quercetin in rats and pigs. J. Nutr., 2005, 135(7), 2019, jpet.118.255067.
1718-1725. [http://dx.doi.org/10.1124/jpet.118.255067]
[http://dx.doi.org/10.1093/jn/135.7.1718] [PMID: 15987855] [125] Caddeo, C.; Díez-Sales, O.; Pons, R.; Carbone, C.; Ennas, G.;
[110] Lee, J.; Mitchell, A.E. Pharmacokinetics of quercetin absorption Puglisi, G.; Fadda, A.M.; Manconi, M. Cross-linked chito-
from apples and onions in healthy humans. J. Agric. Food Chem., san/liposome hybrid system for the intestinal delivery of quercetin.
2012, 60(15), 3874-3881. J. Colloid Interface Sci., 2016, 461, 69-78.
[http://dx.doi.org/10.1021/jf3001857] [PMID: 22439822] [http://dx.doi.org/10.1016/j.jcis.2015.09.013] [PMID: 26397912]
[111] Kaşıkcı, M.B. B.N., Bioavailability of Quercetin. Curr. Res. Nutr. [126] Rahimi, S.; Khoee, S.; Ghandi, M. Preparation and characterization
Food Sci., 2016, 4, 146-151. of rod-like chitosan-quinoline nanoparticles as pH-responsive
[http://dx.doi.org/10.12944/CRNFSJ.4.Special-Issue-October.20] nanocarriers for quercetin delivery. Int. J. Biol. Macromol., 2019,
[112] Tran, T.H.; Guo, Y.; Song, D.; Bruno, R.S.; Lu, X. Quercetin- 128, 279-289.
containing self-nanoemulsifying drug delivery system for improv- [http://dx.doi.org/10.1016/j.ijbiomac.2019.01.137] [PMID:
ing oral bioavailability. J. Pharm. Sci., 2014, 103(3), 840-852. 30695722]
[http://dx.doi.org/10.1002/jps.23858] [PMID: 24464737] [127] Barbosa, A.I.; Costa Lima, S.A.; Reis, S. Application of pH-
[113] Dian, L.; Yu, E.; Chen, X.; Wen, X.; Zhang, Z.; Qin, L.; Wang, Q.; Responsive Fucoidan/Chitosan Nanoparticles to Improve Oral
Li, G.; Wu, C. Enhancing oral bioavailability of quercetin using Quercetin Delivery. Molecules, 2019, 24(2), 346.
novel soluplus polymeric micelles. Nanoscale Res. Lett., 2014, [http://dx.doi.org/10.3390/molecules24020346] [PMID: 30669398]
9(1), 2406. [128] Aroonsri Priprem, S.S. Hye-Kyung Na, Young-Joon Surh, Malyn
[http://dx.doi.org/10.1186/1556-276X-9-684] [PMID: 26088982] Chulasiri, Effect of Formulations of Nanosized Quercetin
[114] Xu, G.; Shi, H.; Ren, L.; Gou, H.; Gong, D.; Gao, X.; Huang, N. Liposomes on COX-2 and NF-kB in MCF-10A Cells. Pharm.
Enhancing the anti-colon cancer activity of quercetin by self- Nanotechnol., 2013, 1(1), 26-34.
assembled micelles. Int. J. Nanomedicine, 2015, 10, 2051-2063. [http://dx.doi.org/10.2174/2211738511301010026]
[PMID: 25844036]
16 Anti-Cancer Agents in Medicinal Chemistry, 2019, Vol. 19, No. 0 Vinayak and Maurya

[129] Priprem, A.; Watanatorn, J.; Sutthiparinyanont, S.; Phachonpai, [142] Pimple, S.; Manjappa, A.S.; Ukawala, M.; Murthy, R.S. PLGA
W.; Muchimapura, S. Anxiety and cognitive effects of quercetin nanoparticles loaded with etoposide and quercetin dihydrate indi-
liposomes in rats. Nanomedicine (Lond.), 2008, 4(1), 70-78. vidually: in vitro cell line study to ensure advantage of combination
[http://dx.doi.org/10.1016/j.nano.2007.12.001] [PMID: 18249157] therapy. Cancer Nanotechnol., 2012, 3(1-6), 25-36.
[130] Ren, J.; Fang, Z.; Jiang, L.; Du, Q. Quercetin-containing self- [http://dx.doi.org/10.1007/s12645-012-0027-y] [PMID: 26069494]
assemble proliposome preparation and evaluation. J. Liposome [143] Zhao, J.; Liu, J.; Wei, T.; Ma, X.; Cheng, Q.; Huo, S.; Zhang, C.;
Res., 2017, 27(4), 335-342. Zhang, Y.; Duan, X.; Liang, X.J. Quercetin-loaded nanomicelles to
[http://dx.doi.org/10.1080/08982104.2016.1239635] [PMID: circumvent human castration-resistant prostate cancer in vitro and
27667265] in vivo. Nanoscale, 2016, 8(9), 5126-5138.
[131] Zheng, N.G.; Mo, S.J.; Li, J.P.; Wu, J.L. Anti-CSC effects in hu- [http://dx.doi.org/10.1039/C5NR08966B] [PMID: 26875690]
man esophageal squamous cell carcinomas and Eca109/9706 cells [144] Pang, X.; Lu, Z.; Du, H.; Yang, X.; Zhai, G. Hyaluronic acid-
induced by nanoliposomal quercetin alone or combined with CD quercetin conjugate micelles: synthesis, characterization, in vitro
133 antiserum. Asian Pac. J. Cancer Prev., 2014, 15(20), 8679- and in vivo evaluation. Colloids Surf. B Biointerfaces, 2014, 123,
8684. 778-786.
[http://dx.doi.org/10.7314/APJCP.2014.15.20.8679] [PMID: [http://dx.doi.org/10.1016/j.colsurfb.2014.10.025] [PMID: 25454664]
25374189] [145] Tan, B.J.; Liu, Y.; Chang, K.L.; Lim, B.K.; Chiu, G.N. Perorally
[132] Martirosyan, A.; Grintzalis, K.; Polet, M.; Laloux, L.; Schneider, active nanomicellar formulation of quercetin in the treatment of
Y.J. Tuning the inflammatory response to silver nanoparticles via lung cancer. Int. J. Nanomedicine, 2012, 7, 651-661.
quercetin in Caco-2 (co-)cultures as model of the human intestinal [PMID: 22334787]
mucosa. Toxicol. Lett., 2016, 253, 36-45. [146] Zhao, L.; Shi, Y.; Zou, S.; Sun, M.; Lil, L.; Zhail, G. Formulation
[http://dx.doi.org/10.1016/j.toxlet.2016.04.018] [PMID: 27113704] and in vitro evaluation of quercetin loaded polymeric micelles
[133] Mittal, A.K.; Kumar, S.; Banerjee, U.C. Quercetin and gallic acid composed of pluronic P123 and D-a-tocopheryl polyethylene gly-
mediated synthesis of bimetallic (silver and selenium) nanoparti- col succinate. J. Biomed. Nanotechnol., 2011, 7(3), 358-365.
cles and their antitumor and antimicrobial potential. J. Colloid In- [http://dx.doi.org/10.1166/jbn.2011.1298] [PMID: 21830476]
terface Sci., 2014, 431, 194-199. [147] Khonkarn, R.; Mankhetkorn, S.; Hennink, W.E.; Okonogi, S. PEG-
[http://dx.doi.org/10.1016/j.jcis.2014.06.030] [PMID: 25000181] OCL micelles for quercetin solubilization and inhibition of cancer
[134] Pandey, S.K.; Patel, D.K.; Maurya, A.K.; Thakur, R.; Mishra, D.P.; cell growth. Eur. J. Pharm. Biopharm., 2011, 79(2), 268-275.
Vinayak, M.; Haldar, C.; Maiti, P. Controlled release of drug and [http://dx.doi.org/10.1016/j.ejpb.2011.04.011] [PMID: 21596135]
better bioavailability using poly(lactic acid-co-glycolic acid) [148] Sun, C.; Ding, Y.; Zhou, L.; Shi, D.; Sun, L.; Webster, T.J.; Shen,
nanoparticles. Int. J. Biol. Macromol., 2016, 89, 99-110. Y. Noninvasive nanoparticle strategies for brain tumor targeting.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.04.065] [PMID: Nanomedicine., 2017, 13(8), 2605-2621.
27112980] [http://dx.doi.org/10.1016/j.nano.2017.07.009] [PMID: 28756093]
[135] Suksiriworapong, J.; Sripha, K.; Kreuter, J.; Junyaprasert, V.B. [149] Lainé, A.L.; Clavreul, A.; Rousseau, A.; Tétaud, C.; Vessieres, A.;
Functionalized (poly(ɛ-caprolactone))₂-poly(ethylene glycol) Garcion, E.; Jaouen, G.; Aubert, L.; Guilbert, M.; Benoit, J.P.;
nanoparticles with grafting nicotinic acid as drug carriers. Int. J. Toillon, R.A.; Passirani, C. Inhibition of ectopic glioma tumor
Pharm., 2012, 423(2), 562-570. growth by a potent ferrocenyl drug loaded into stealth lipid nano-
[http://dx.doi.org/10.1016/j.ijpharm.2011.11.033] [PMID: capsules. Nanomedicine., 2014, 10(8), 1667-1677.
22155410] [http://dx.doi.org/10.1016/j.nano.2014.05.002] [PMID: 24842766]
[136] Suksiriworapong, J.; Phoca, K.; Ngamsom, S.; Sripha, K.; Moong- [150] Jain, A.K.; Thanki, K.; Jain, S. Novel self-nanoemulsifying formu-
karndi, P.; Junyaprasert, V.B. Comparison of poly(ε-caprolactone) lation of quercetin: Implications of pro-oxidant activity on the anti-
chain lengths of poly(ε-caprolactone)-co-d-α-tocopheryl- cancer efficacy. Nanomedicine., 2014, 10(5), 959-969.
poly(ethylene glycol) 1000 succinate nanoparticles for enhance- [http://dx.doi.org/10.1016/j.nano.2013.12.010] [PMID: 24407148]
ment of quercetin delivery to SKBR3 breast cancer cells. Eur. J. [151] Ghosh, S.; Sarkar, S.; Choudhury, S.T.; Ghosh, T.; Das, N.
Pharm. Biopharm., 2016, 101, 15-24. Triphenyl phosphonium coated nano-quercetin for oral delivery:
[http://dx.doi.org/10.1016/j.ejpb.2016.01.008] [PMID: 26802701] Neuroprotective effects in attenuating age related global moderate
[137] Gupta, A.; Kaur, C.D.; Saraf, S.; Saraf, S. Formulation, characteri- cerebral ischemia reperfusion injury in rats. Nanomedicine., 2017,
zation, and evaluation of ligand-conjugated biodegradable quer- 13(8), 2439-2450.
cetin nanoparticles for active targeting. Artif. Cells Nanomed. Bio- [http://dx.doi.org/10.1016/j.nano.2017.08.002] [PMID: 28822845]
technol., 2016, 44(3), 960-970. [152] Penalva, R.; González-Navarro, C.J.; Gamazo, C.; Esparza, I.;
[PMID: 25813566] Irache, J.M. Zein nanoparticles for oral delivery of quercetin:
[138] Lv, L.; Liu, C.; Chen, C.; Yu, X.; Chen, G.; Shi, Y.; Qin, F.; Ou, J.; Pharmacokinetic studies and preventive anti-inflammatory effects
Qiu, K.; Li, G. Quercetin and doxorubicin co-encapsulated biotin in a mouse model of endotoxemia. Nanomedicine (Lond.), 2017,
receptor-targeting nanoparticles for minimizing drug resistance in 13(1), 103-110.
breast cancer. Oncotarget, 2016, 7(22), 32184-32199. [http://dx.doi.org/10.1016/j.nano.2016.08.033] [PMID: 27615118]
[http://dx.doi.org/10.18632/oncotarget.8607] [PMID: 27058756] [153] Sedaghat Doost, A.; Kassozi, V.; Grootaert, C.; Claeys, M.; Dewet-
[139] Guan, X.; Gao, M.; Xu, H.; Zhang, C.; Liu, H.; Lv, L.; Deng, S.; tinck, K.; Van Camp, J.; Van der Meeren, P. Self-assembly, func-
Gao, D.; Tian, Y. Quercetin-loaded poly (lactic-co-glycolic acid)- tionality, and in-vitro properties of quercetin loaded nanoparticles
d-α-tocopheryl polyethylene glycol 1000 succinate nanoparticles based on shellac-almond gum biological macromolecules. Int. J.
for the targeted treatment of liver cancer. Drug Deliv., 2016, 23(9), Biol. Macromol., 2019, 129, 1024-1033.
3307-3318. [http://dx.doi.org/10.1016/j.ijbiomac.2019.02.071] [PMID:
[http://dx.doi.org/10.1080/10717544.2016.1176087] [PMID: 30794898]
27067032] [154] Fang, J.; Nakamura, H.; Maeda, H. The EPR effect: Unique fea-
[140] Wang, C.; Su, L.; Wu, C.; Wu, J.; Zhu, C.; Yuan, G. RGD peptide tures of tumor blood vessels for drug delivery, factors involved,
targeted lipid-coated nanoparticles for combinatorial delivery of and limitations and augmentation of the effect. Adv. Drug Deliv.
sorafenib and quercetin against hepatocellular carcinoma. Drug Rev., 2011, 63(3), 136-151.
Dev. Ind. Pharm., 2016, 42(12), 1938-1944. [http://dx.doi.org/10.1016/j.addr.2010.04.009] [PMID: 20441782]
[http://dx.doi.org/10.1080/03639045.2016.1185435] [PMID: [155] Patra, A.; Satpathy, S.; Shenoy, A.K.; Bush, J.A.; Kazi, M.; Hus-
27142812] sain, M.D. Formulation and evaluation of mixed polymeric mi-
[141] Bishayee, K.; Khuda-Bukhsh, A.R.; Huh, S.O. PLGA-Loaded celles of quercetin for treatment of breast, ovarian, and multidrug
Gold-Nanoparticles Precipitated with Quercetin Downregulate resistant cancers. Int. J. Nanomedicine, 2018, 13, 2869-2881.
HDAC-Akt Activities Controlling Proliferation and Activate p53- [http://dx.doi.org/10.2147/IJN.S153094] [PMID: 29844670]
ROS Crosstalk to Induce Apoptosis in Hepatocarcinoma Cells. [156] Men, K.; Duan, X.; Wei, X.W.; Gou, M.L.; Huang, M.J.; Chen,
Mol. Cells, 2015, 38(6), 518-527. L.J.; Qian, Z.Y.; Wei, Y.Q. Nanoparticle-delivered quercetin for
[http://dx.doi.org/10.14348/molcells.2015.2339] [PMID: cancer therapy. Anticancer. Agents Med. Chem., 2014, 14(6), 826-
25947292] 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: lease of drug and better bioavailability using poly(lactic acid-co-
24851877] glycolic acid) nanoparticles" [Int. J. Biol. Macromol. 89 (2016) 99-
[157] Sharma, G.; Park, J.; Sharma, A.R.; Jung, J.S.; Kim, H.; Chak- 110]. Int J Biol Macromol., 2018, 114, 1361.
raborty, C.; Song, D.K.; Lee, S.S.; Nam, J.S. Methoxy [http://dx.doi.org/10.1016/j.ijbiomac.2018.03.038] [PMID:
poly(ethylene glycol)-poly(lactide) nanoparticles encapsulating 29674009]
quercetin act as an effective anticancer agent by inducing apoptosis [172] Zhang, H.; Liu, G.; Zeng, X.; Wu, Y.; Yang, C.; Mei, L.; Wang, Z.;
in breast cancer. Pharm. Res., 2015, 32(2), 723-735. Huang, L. Fabrication of genistein-loaded biodegradable TPGS-b-
[http://dx.doi.org/10.1007/s11095-014-1504-2] [PMID: 25186442] PCL nanoparticles for improved therapeutic effects in cervical can-
[158] Ahmad, N.; Banala, V.T.; Kushwaha, P.; Karvande, A.; Sharma, S.; cer cells. Int. J. Nanomedicine, 2015, 10, 2461-2473.
Tripathi, A.K.; Verma, A.; Trivedi, R.; Mishra, P.R. Quercetin- [PMID: 25848264]
loaded solid lipid nanoparticles improve osteoprotective activity in [173] Bernabeu, E.; Gonzalez, L.; Legaspi, M.J.; Moretton, M.A.; Chiap-
an ovariectomized rat model: a preventive strategy for post- petta, D.A. Paclitaxel-Loaded TPGS-b-PCL Nanoparticles: In Vitro
menopausal osteoporosis. RSC Advances, 2016, 6(100), 97613- Cytotoxicity and Cellular Uptake in MCF-7 and MDA-MB-231
97628. Cells versus mPEG-b-PCL Nanoparticles and Abraxane®. J. Nano-
[http://dx.doi.org/10.1039/C6RA17141A] sci. Nanotechnol., 2016, 16(1), 160-170.
[159] Castangia, I.; Nácher, A.; Caddeo, C.; Merino, V.; Díez-Sales, O.; [http://dx.doi.org/10.1166/jnn.2016.10739] [PMID: 27398441]
Catalán-Latorre, A.; Fernàndez-Busquets, X.; Fadda, A.M.; Man- [174] Bernabeu, E.; Cagel, M.; Lagomarsino, E.; Moretton, M.; Chiap-
coni, M. Therapeutic efficacy of quercetin enzyme-responsive petta, D.A. Paclitaxel: What has been done and the challenges re-
nanovesicles for the treatment of experimental colitis in rats. Acta main ahead. Int. J. Pharm., 2017, 526(1-2), 474-495.
Biomater., 2015, 13, 216-227. [http://dx.doi.org/10.1016/j.ijpharm.2017.05.016] [PMID:
[http://dx.doi.org/10.1016/j.actbio.2014.11.017] [PMID: 25463498] 28501439]
[160] Singh, S.K.; Singh, S.; Lillard, J.W., Jr; Singh, R. Drug delivery [175] Zheng, Y.; Chen, H.; Zeng, X.; Liu, Z.; Xiao, X.; Zhu, Y.; Gu, D.;
approaches for breast cancer. Int. J. Nanomedicine, 2017, 12, 6205- Mei, L. Surface modification of TPGS-b-(PCL-ran-PGA) nanopar-
6218. ticles with polyethyleneimine as a co-delivery system of TRAIL
[http://dx.doi.org/10.2147/IJN.S140325] [PMID: 28883730] and endostatin for cervical cancer gene therapy. Nanoscale Res.
[161] Gulbake, A.; Jain, S.K. Chitosan: a potential polymer for colon- Lett., 2013, 8(1), 161.
specific drug delivery system. Expert Opin. Drug Deliv., 2012, [http://dx.doi.org/10.1186/1556-276X-8-161] [PMID: 23570619]
9(6), 713-729. [176] Jiang, L.; Li, X.; Liu, L.; Zhang, Q. Thiolated chitosan-modified
[http://dx.doi.org/10.1517/17425247.2012.682148] [PMID: PLA-PCL-TPGS nanoparticles for oral chemotherapy of lung can-
22530707] cer. Nanoscale Res. Lett., 2013, 8(1), 66.
[162] Han, L.; Zhao, J.; Zhang, X.; Cao, W.; Hu, X.; Zou, G.; Duan, X.; [http://dx.doi.org/10.1186/1556-276X-8-66] [PMID: 23394588]
Liang, X.J. Enhanced siRNA delivery and silencing gold-chitosan [177] Yadav, M.K.; Maurya, A.K.; Rajput, G.; Manar, K.K.; Vinayak,
nanosystem with surface charge-reversal polymer assembly and M.; Drew, M.G.B.; Singh, N. Synthesis, characterization, DNA
good biocompatibility. ACS Nano, 2012, 6(8), 7340-7351. binding and cleavage activity of homoleptic zinc(II) β-
[http://dx.doi.org/10.1021/nn3024688] [PMID: 22838646] oxodithioester chelate complexes. J. Coord. Chem., 2017, 70(18),
[163] Yuan, Y.G.; Peng, Q.L.; Gurunathan, S. Silver nanoparticles en- 3171-3185.
hance the apoptotic potential of gemcitabine in human ovarian can- [http://dx.doi.org/10.1080/00958972.2017.1377835]
cer cells: combination therapy for effective cancer treatment. Int. J. [178] Yadav, M.K.; Maurya, A.K.; Rajput, G.; Manar, K.K.; Vinayak,
Nanomedicine, 2017, 12, 6487-6502. M.; Drew, M.G.B.; Singh, N. New planar trans-copper(II) β-
[http://dx.doi.org/10.2147/IJN.S135482] [PMID: 28919750] dithioester chelate complexes: synthesis, characterization, antican-
[164] Yoo, G.; Jeong, S.H.; Ryu, W.I.; Lee, H.; Kim, J.H.; Bae, H.C.; cer activity and DNA-binding/cleavage studies. J. Coord. Chem.,
Son, S.W. Gene expression analysis reveals a functional role for 2017, 70(4), 565-583.
the Ag-NPs-induced Egr-1 transcriptional factor in human kerati- [http://dx.doi.org/10.1080/00958972.2016.1275589]
nocytes. Mol. Cell. Toxicol., 2014, 10(2), 149-156. [179] Pool, H.; Quintanar, D.; Figueroa, J.D.; Marinho Mano, C.;
[http://dx.doi.org/10.1007/s13273-014-0016-9] Bechara, J.E.H. God; #xed; nez, L.A.; Mendoza, S., Antioxidant
[165] Lin, J.; Huang, Z.; Wu, H.; Zhou, W.; Jin, P.; Wei, P.; Zhang, Y.; Effects of Quercetin and Catechin Encapsulated into PLGA
Zheng, F.; Zhang, J.; Xu, J.; Hu, Y.; Wang, Y.; Li, Y.; Gu, N.; Nanoparticles. J. Nanomater., 2012, 2012, 12.
Wen, L. Inhibition of autophagy enhances the anticancer activity of [http://dx.doi.org/10.1155/2012/145380]
silver nanoparticles. Autophagy, 2014, 10(11), 2006-2020. [180] Jain, A.K.; Thanki, K.; Jain, S. Co-encapsulation of tamoxifen and
[http://dx.doi.org/10.4161/auto.36293] [PMID: 25484080] quercetin in polymeric nanoparticles: implications on oral bioavail-
[166] Lin, L.; Xu, Y.; Zhang, S.; Ross, I.M.; Ong, A.C.; Allwood, D.A. ability, antitumor efficacy, and drug-induced toxicity. Mol. Pharm.,
Fabrication and luminescence of monolayered boron nitride quan- 2013, 10(9), 3459-3474.
tum dots. Small, 2014, 10(1), 60-65. [http://dx.doi.org/10.1021/mp400311j] [PMID: 23927416]
[http://dx.doi.org/10.1002/smll.201301001] [PMID: 23839969] [181] Balakrishnan, S.; Mukherjee, S.; Das, S.; Bhat, F.A.; Raja Singh,
[167] AshaRani, P.V.; Low Kah Mun, G.; Hande, M.P.; Valiyaveettil, S. P.; Patra, C.R.; Arunakaran, J. Gold nanoparticles-conjugated quer-
Cytotoxicity and genotoxicity of silver nanoparticles in human cetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated
cells. ACS Nano, 2009, 3(2), 279-290. pathway in breast cancer cell lines (MCF-7 and MDA-MB-231).
[http://dx.doi.org/10.1021/nn800596w] [PMID: 19236062] Cell Biochem. Funct., 2017, 35(4), 217-231.
[168] Sanpui, P.; Chattopadhyay, A.; Ghosh, S.S. Induction of apoptosis [http://dx.doi.org/10.1002/cbf.3266] [PMID: 28498520]
in cancer cells at low silver nanoparticle concentrations using chi- [182] Sun, M.; Nie, S.; Pan, X.; Zhang, R.; Fan, Z.; Wang, S. Quercetin-
tosan nanocarrier. ACS Appl. Mater. Interfaces, 2011, 3(2), 218- nanostructured lipid carriers: characteristics and anti-breast cancer
228. activities in vitro. Colloids Surf. B Biointerfaces, 2014, 113, 15-24.
[http://dx.doi.org/10.1021/am100840c] [PMID: 21280584] [http://dx.doi.org/10.1016/j.colsurfb.2013.08.032] [PMID:
[169] Gurunathan, S.; Jeong, J.K.; Han, J.W.; Zhang, X.F.; Park, J.H.; 24060926]
Kim, J.H. Multidimensional effects of biologically synthesized sil- [183] Sarkar, A.; Ghosh, S.; Chowdhury, S.; Pandey, B.; Sil, P.C. Tar-
ver nanoparticles in Helicobacter pylori, Helicobacter felis, and geted delivery of quercetin loaded mesoporous silica nanoparticles
human lung (L132) and lung carcinoma A549 cells. Nanoscale Res. to the breast cancer cells. Biochim. Biophys. Acta, 2016, 1860(10),
Lett., 2015, 10, 35. 2065-2075.
[http://dx.doi.org/10.1186/s11671-015-0747-0] [PMID: 25852332] [http://dx.doi.org/10.1016/j.bbagen.2016.07.001] [PMID:
[170] Ortega, F.G.; Fernández-Baldo, M.A.; Fernández, J.G.; Serrano, 27392941]
M.J.; Sanz, M.I.; Diaz-Mochón, J.J.; Lorente, J.A.; Raba, J. Study [184] Aghapour, F.; Moghadamnia, A.A.; Nicolini, A.; Kani, S.N.M.;
of antitumor activity in breast cell lines using silver nanoparticles Barari, L.; Morakabati, P.; Rezazadeh, L.; Kazemi, S. Quercetin
produced by yeast. Int. J. Nanomedicine, 2015, 10, 2021-2031. conjugated with silica nanoparticles inhibits tumor growth in MCF-
[PMID: 25844035] 7 breast cancer cell lines. Biochem. Biophys. Res. Commun., 2018,
[171] Pandey, S.K.; Patel, D.K.; Maurya, A.K.; Thakur, R.; Mishra, D.P.; 500(4), 860-865.
Vinayak, M.; Haldar, C.; Maiti, P. Corrigendum to "Controlled re- [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, [199] Mukai, H.; Kogawa, T.; Matsubara, N.; Naito, Y.; Sasaki, M.;
Q. Synergistic inhibition of migration and invasion of breast cancer Hosono, A. A first-in-human Phase 1 study of epirubicin-
cells by dual docetaxel/quercetin-loaded nanoparticles via conjugated polymer micelles (K-912/NC-6300) in patients with ad-
Akt/MMP-9 pathway. Int. J. Pharm., 2017, 523(1), 300-309. vanced or recurrent solid tumors. Invest. New Drugs, 2017, 35(3),
[http://dx.doi.org/10.1016/j.ijpharm.2017.03.040] [PMID: 307-314.
28336457] [http://dx.doi.org/10.1007/s10637-016-0422-z] [PMID: 28054329]
[186] Umrao, S.; Maurya, A.K.; Shukla, V.; Grigoriev, A.; Ahuja, R.; [200] Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.;
Vinayak, M.; Srivastava, R.R.; Saxena, P.S.; Oh, I.K.; Srivastava, Kimura, M.; Terada, Y.; Kano, M.R.; Miyazono, K.; Uesaka, M.;
A. Anti-carcinogenic Activity of Blue Fluorescent h-BN Quantum Nishiyama, N.; Kataoka, K. Accumulation of sub-100 nm polym-
Dots: As an Effective Enhancer for DNA Cleavage Activity of eric micelles in poorly permeable tumours depends on size. Nat.
Anti-cancer Drug Doxorubicin In: Materials Today Bio; , 2019. Nanotechnol., 2011, 6(12), 815-823.
[187] Cabral, H.; Kataoka, K. Progress of drug-loaded polymeric mi- [http://dx.doi.org/10.1038/nnano.2011.166] [PMID: 22020122]
celles into clinical studies. J. Control. Release, 2014, 190, 465-476. [201] Chintamani, ; Tandon, M.; Mishra, A.; Agarwal, U.; Saxena, S.
[http://dx.doi.org/10.1016/j.jconrel.2014.06.042] [PMID: Sentinel lymph node biopsy using dye alone method is reliable and
24993430] accurate even after neo-adjuvant chemotherapy in locally advanced
[188] Nishiyama, N.; Matsumura, Y.; Kataoka, K. Development of po- breast cancer--a prospective study. World J. Surg. Oncol., 2011, 9,
lymeric micelles for targeting intractable cancers. Cancer Sci., 19.
2016, 107(7), 867-874. [http://dx.doi.org/10.1186/1477-7819-9-19] [PMID: 21396137]
[http://dx.doi.org/10.1111/cas.12960] [PMID: 27116635] [202] Ahn, H.K.; Jung, M.; Sym, S.J.; Shin, D.B.; Kang, S.M.; Kyung,
[189] Soleymani Abyaneh, H.; Vakili, M.R.; Zhang, F.; Choi, P.; La- S.Y.; Park, J.W.; Jeong, S.H.; Cho, E.K. A phase II trial of Cre-
vasanifar, A. Rational design of block copolymer micelles to con- morphor EL-free paclitaxel (Genexol-PM) and gemcitabine in pa-
trol burst drug release at a nanoscale dimension. Acta Biomater., tients with advanced non-small cell lung cancer. Cancer Che-
2015, 24, 127-139. mother. Pharmacol., 2014, 74(2), 277-282.
[http://dx.doi.org/10.1016/j.actbio.2015.06.017] [PMID: 26093068] [http://dx.doi.org/10.1007/s00280-014-2498-5] [PMID: 24906423]
[190] Fonge, H.; Huang, H.; Scollard, D.; Reilly, R.M.; Allen, C. Influ- [203] Vega, J.; Ke, S.; Fan, Z.; Wallace, S.; Charsangavej, C.; Li, C.
ence of formulation variables on the biodistribution of multifunc- Targeting doxorubicin to epidermal growth factor receptors by site-
tional block copolymer micelles. J. Control. Release, 2012, 157(3), specific conjugation of C225 to poly(L-glutamic acid) through a
366-374. polyethylene glycol spacer. Pharm. Res., 2003, 20(5), 826-832.
[http://dx.doi.org/10.1016/j.jconrel.2011.09.088] [PMID: [http://dx.doi.org/10.1023/A:1023454107190] [PMID: 12751641]
21982897] [204] Reddy, S.T.; van der Vlies, A.J.; Simeoni, E.; Angeli, V.;
[191] Bae, Y.; Kataoka, K. Intelligent polymeric micelles from functional Randolph, G.J.; O’Neil, C.P.; Lee, L.K.; Swartz, M.A.; Hubbell,
poly(ethylene glycol)-poly(amino acid) block copolymers. Adv. J.A. Exploiting lymphatic transport and complement activation in
Drug Deliv. Rev., 2009, 61(10), 768-784. nanoparticle vaccines. Nat. Biotechnol., 2007, 25(10), 1159-1164.
[http://dx.doi.org/10.1016/j.addr.2009.04.016] [PMID: 19422866] [http://dx.doi.org/10.1038/nbt1332] [PMID: 17873867]
[192] Matsukawa, N.; Matsumoto, M.; Shinoki, A.; Hagio, M.; Inoue, R.; [205] Houdaihed, L.; Evans, J.C.; Allen, C. Overcoming the Road
Hara, H. Nondigestible saccharides suppress the bacterial degrada- Blocks: Advancement of Block Copolymer Micelles for Cancer
tion of quercetin aglycone in the large intestine and enhance the Therapy in the Clinic. Mol. Pharm., 2017, 14(8), 2503-2517.
bioavailability of quercetin glucoside in rats. J. Agric. Food Chem., [http://dx.doi.org/10.1021/acs.molpharmaceut.7b00188] [PMID:
2009, 57(20), 9462-9468. 28481116]
[http://dx.doi.org/10.1021/jf9024079] [PMID: 19807098] [206] Lee, K.S.; Chung, H.C.; Im, S.A.; Park, Y.H.; Kim, C.S.; Kim,
[193] Miyata, K.; Nishiyama, N.; Kataoka, K. Rational design of smart S.B.; Rha, S.Y.; Lee, M.Y.; Ro, J. Multicenter phase II trial of
supramolecular assemblies for gene delivery: chemical challenges Genexol-PM, a Cremophor-free, polymeric micelle formulation of
in the creation of artificial viruses. Chem. Soc. Rev., 2012, 41(7), paclitaxel, in patients with metastatic breast cancer. Breast Cancer
2562-2574. Res. Treat., 2008, 108(2), 241-250.
[http://dx.doi.org/10.1039/C1CS15258K] [PMID: 22105545] [http://dx.doi.org/10.1007/s10549-007-9591-y] [PMID: 17476588]
[194] Liu, K.; Chen, W.; Yang, T.; Wen, B.; Ding, D.; Keidar, M.; Tang, [207] Kim, J.Y.; Do, Y.R.; Song, H.S.; Cho, Y.Y.; Ryoo, H.M.; Bae,
J.; Zhang, W. Paclitaxel and quercetin nanoparticles co-loaded in S.H.; Kim, J.G.; Chae, Y.S.; Kang, B.W.; Baek, J.H.; Kim, M.K.;
microspheres to prolong retention time for pulmonary drug deliv- Lee, K.H.; Park, K. Multicenter Phase II Clinical Trial of Genexol-
ery. Int. J. Nanomedicine, 2017, 12, 8239-8255. PM® with Gemcitabine in Advanced Biliary Tract Cancer. Anti-
[http://dx.doi.org/10.2147/IJN.S147028] [PMID: 29180863] cancer Res., 2017, 37(3), 1467-1473.
[195] Mosqueira, V.C.; Legrand, P.; Gulik, A.; Bourdon, O.; Gref, R.; [http://dx.doi.org/10.21873/anticanres.11471] [PMID: 28314319]
Labarre, D.; Barratt, G. Relationship between complement activa- [208] Sutton, D.; Nasongkla, N.; Blanco, E.; Gao, J. Functionalized mi-
tion, cellular uptake and surface physicochemical aspects of novel cellar systems for cancer targeted drug delivery. Pharm. Res., 2007,
PEG-modified nanocapsules. Biomaterials, 2001, 22(22), 2967- 24(6), 1029-1046.
2979. [http://dx.doi.org/10.1007/s11095-006-9223-y] [PMID: 17385025]
[http://dx.doi.org/10.1016/S0142-9612(01)00043-6] [PMID: [209] Matsumura, Y. Preclinical and clinical studies of NK012, an SN-
11575471] 38-incorporating polymeric micelles, which is designed based on
[196] Murakami, M.; Cabral, H.; Matsumoto, Y.; Wu, S.; Kano, M.R.; EPR effect. Adv. Drug Deliv. Rev., 2011, 63(3), 184-192.
Yamori, T.; Nishiyama, N.; Kataoka, K. Improving drug potency [http://dx.doi.org/10.1016/j.addr.2010.05.008] [PMID: 20561951]
and efficacy by nanocarrier-mediated subcellular targeting. Sci. [210] Hamaguchi, T.; Kato, K.; Yasui, H.; Morizane, C.; Ikeda, M.;
Transl. Med., 2011, 3(64), 64ra2. Ueno, H.; Muro, K.; Yamada, Y.; Okusaka, T.; Shirao, K.; Shi-
[http://dx.doi.org/10.1126/scitranslmed.3001385] [PMID: mada, Y.; Nakahama, H.; Matsumura, Y. A phase I and pharma-
21209412] cokinetic study of NK105, a paclitaxel-incorporating micellar
[197] Mochida, Y.; Cabral, H.; Kataoka, K. Polymeric micelles for tar- nanoparticle formulation. Br. J. Cancer, 2007, 97(2), 170-176.
geted tumor therapy of platinum anticancer drugs. Expert Opin. [http://dx.doi.org/10.1038/sj.bjc.6603855] [PMID: 17595665]
Drug Deliv., 2017, 14(12), 1423-1438. [211] Matsumura, Y. The drug discovery by nanomedicine and its clini-
[http://dx.doi.org/10.1080/17425247.2017.1307338] [PMID: cal experience. Jpn. J. Clin. Oncol., 2014, 44(6), 515-525.
28290714] [http://dx.doi.org/10.1093/jjco/hyu046] [PMID: 24755547]
[198] Takahashi, A.; Yamamoto, Y.; Yasunaga, M.; Koga, Y.; Kuroda, [212] Wilson, R.H.; Plummer, R.; Adam, J.; Eatock, M.M.; Boddy, A.V.;
J.; Takigahira, M.; Harada, M.; Saito, H.; Hayashi, T.; Kato, Y.; Griffin, M.; Miller, R.; Matsumura, Y.; Shimizu, T.; Calvert, H.
Kinoshita, T.; Ohkohchi, N.; Hyodo, I.; Matsumura, Y. NC-6300, Phase I and pharmacokinetic study of NC-6004, a new platinum
an epirubicin-incorporating micelle, extends the antitumor effect entity of cisplatin-conjugated polymer forming micelles. Journal of
and reduces the cardiotoxicity of epirubicin. Cancer Sci., 2013, Clinical Oncology., 2008, 26(15_suppl), 2573-2573.
104(7), 920-925. [213] Wang, C.; Wu, C.; Zhou, X.; Han, T.; Xin, X.; Wu, J.; Zhang, J.;
[http://dx.doi.org/10.1111/cas.12153] [PMID: 23495762] 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., new strategy to prepare graphene quantum dots for DNA cleavage.
2013, 3, 2852. ACS Nano, 2012, 6(8), 6592-6599.
[http://dx.doi.org/10.1038/srep02852] [PMID: 24092333] [http://dx.doi.org/10.1021/nn301629v] [PMID: 22813062]
[214] Dolatabadi, J.E.N.; Jamali, A. A.; Hasanzadeh, M.; Omidi, Y [218] Zhang, L.; Xia, J.; Zhao, Q.; Liu, L.; Zhang, Z. Functional gra-
Quercetin Delivery into Cancer Cells with Single Walled Carbon phene oxide as a nanocarrier for controlled loading and targeted de-
Nanotubes. International Journal of Bioscience, Biochemistry and livery of mixed anticancer drugs. Small, 2010, 6(4), 537-544.
Bioinformatics., 2011, 21(a), 25. [http://dx.doi.org/10.1002/smll.200901680] [PMID: 20033930]
[http://dx.doi.org/10.7763/IJBBB.2011.V1.4] [219] Zhang, Y.; Wang, J.; Bai, X.; Jiang, T.; Zhang, Q.; Wang, S.
[215] Cirillo, G.; Vittorio, O.; Hampel, S.; Iemma, F.; Parchi, P.; Cec- Mesoporous silica nanoparticles for increasing the oral bioavail-
chini, M.; Puoci, F.; Picci, N. Quercetin nanocomposite as novel ability and permeation of poorly water soluble drugs. Mol. Pharm.,
anticancer therapeutic: improved efficiency and reduced toxicity. 2012, 9(3), 505-513.
Eur. J. Pharm. Sci., 2013, 49(3), 359-365. [http://dx.doi.org/10.1021/mp200287c] [PMID: 22217205]
[http://dx.doi.org/10.1016/j.ejps.2013.04.008] [PMID: 23602995] [220] Sapino, S.; Ugazio, E.; Gastaldi, L.; Miletto, I.; Berlier, G.; Zonari,
[216] Wang, Q.; Bao, Y.; Ahire, J.; Chao, Y. Co-encapsulation of biode- D.; Oliaro-Bosso, S. Mesoporous silica as topical nanocarriers for
gradable nanoparticles with silicon quantum dots and quercetin for quercetin: characterization and in vitro studies. Eur. J. Pharm. Bio-
monitored delivery. Adv. Healthc. Mater., 2013, 2(3), 459-466. pharm., 2015, 89, 116-125.
[http://dx.doi.org/10.1002/adhm.201200178] [PMID: 23184534] [http://dx.doi.org/10.1016/j.ejpb.2014.11.022] [PMID: 25478737]
[217] Zhou, X.; Zhang, Y.; Wang, C.; Wu, X.; Yang, Y.; Zheng, B.; Wu,
H.; Guo, S.; Zhang, J. Photo-Fenton reaction of graphene oxide: a

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.

View publication stats

You might also like